Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Egypt J Immunol ; 31(2): 102-111, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38615267

RESUMO

Hepatocellular carcinoma (HCC) is one of the most prevalent cancers in the world. Two risk factors that cause 80-90% of HCC cases globally are chronic infection with hepatitis B virus (HBV) and hepatitis C virus (HCV). The diagnostic value of circulating microRNAs (miRNAs) in numerous tumors has been described. Our research assessed microRNA-16 (miR-16) as a novel biomarker in patients with HCV-induced HCC. The study included three groups. Group 1 included 55 individuals with cirrhosis caused by liver HCV infection in addition to HCC. Group 2 included 55 individuals with cirrhosis brought on by HCV infection. Group 3 included 55 normal control individuals. Expression of miR-16 in blood was assessed by real-time polymerase chain reaction (RT-PCR). The mean level of miR-16 was significantly different in the three groups, with group 1 having the greatest value (1.098 ± 0.647), followed by group 2 (1.1035 ± 0.8567) and group 3 (control subjects) having the lowest value (0.3842 ± 0.21485). The receiver operating characteristic (ROC) curve analysis showed that miR-16 had a higher diagnostic value at area under the curve (AUC) of 0.935 than alpha-feto protein (AUC of 0.859) to differentiate between HCC and control subjects. MiR-16 has a sensitivity of 81.82 % and a specificity of 69.09%, to distinguish between patients with liver cirrhosis and HCC patients. Our findings illustrated that circulating miR-16 can be proposed as a marker for detection of patients with HCV-induced HCC.


Assuntos
Carcinoma Hepatocelular , MicroRNA Circulante , Hepatite C , Neoplasias Hepáticas , MicroRNAs , Humanos , Hepacivirus/genética , Carcinoma Hepatocelular/diagnóstico , Egito , Neoplasias Hepáticas/diagnóstico , Hepatite C/complicações , Cirrose Hepática/diagnóstico , Biomarcadores
2.
Biomed Res Int ; 2022: 8962149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35528172

RESUMO

This study examined the protective effect of quercetin against high-altitude-induced brain damage in rats. A molecular docking study was performed to investigate the potential effect of quercetin in reducing brain damages through its ability to target the oxidative stress enzymes. Biomarker assessment screening assays were also performed then followed by in vivo studies. Three groups of rats were divided into the control group, an untreated animal model group with induced brain damage, and finally, the quercetin treated group that received quercetin dose equal to 20 mg/kg of their body weights. Molecular docking studies and biomarker assessment screening assays proved the potential effect of quercetin to affect the level of representative biomarkers glutathione (GSH), glutathione reductase (GR), glutathione-S-transferase (GST), glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT), and malondialdehyde (MDA). Additionally, the protective effect of quercetin against high altitude, low pressure, and low oxygen was also investigated by exploring the brain histopathology of experimental rats. Brain damage was observed in the untreated animal model group. After treatment with quercetin, the cerebral edema in the brain tissues was improved significantly, confirming the protective effects of quercetin. Therefore, quercetin can be used as a natural food additive to protect from the highaltitude-induced brain damage.


Assuntos
Lesões Encefálicas , Quercetina , Animais , Antioxidantes/farmacologia , Encéfalo/metabolismo , Lesões Encefálicas/tratamento farmacológico , Catalase/metabolismo , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Transferase/metabolismo , Simulação de Acoplamento Molecular , Estresse Oxidativo , Quercetina/farmacologia , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
3.
Biomarkers ; 27(2): 138-150, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34927500

RESUMO

INTRODUCTION: Bladder cancer is still of unknown initiation and progression, it is difficult to treat the patient once bladder cancer have a distant metastasis. MATERIALS AND METHODS: In the present study, propolis extract was evaluated against bladder cancer cells (T24). Two independent pathways were investigated, apoptosis and angiogenesis, Bax, Bcl-2, P53, and caspase-3 for apoptosis, vascular endothelial growth factor receptor and protein kinase A as angiogenesis potential targets. OBJECTIVES: Molecular docking studies will be conducted for the major known constituents of Egyptian propolis into apoptotic and angiogenic protein targets, to give better insights to the possible binding mode and interactions and investigate the ability of propolis constituents to target both apoptotic and angiogenic pathways. RESULTS: Propolis showed anti-proliferative activity against T24 cancer cell line, the IC50 value was 6.36 µg/ml. Also significant effects of propolis on Bax, Bcl-2, P53, and caspase-3 were observed. DISCUSSION: These obtained results proved the ability of propolis to induce cell death. Also it has revealed noticeable effects on protein kinase A and vascular endothelial growth factor receptor. CONCLUSION: The obtained results can encourage us to say that propolis extract can induce a programmed cell death in human bladder cancer cells, and also affect angiogenesis.


Assuntos
Antineoplásicos , Própole , Neoplasias da Bexiga Urinária , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Detecção Precoce de Câncer , Humanos , Simulação de Acoplamento Molecular , Própole/farmacologia , Própole/uso terapêutico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/patologia , Fator A de Crescimento do Endotélio Vascular
4.
Br J Radiol ; 94(1120): 20201353, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33571018

RESUMO

OBJECTIVES: To set age-specific normal reference values for brainstem, cerebellar vermis, and peduncles measurements and characterize values' variations according to gender, age, and age by gender interaction. METHODS: 565 normal brain magnetic resonance examinations with normal anatomy and signal intensity of the supra- and infratentorial structures were categorized into six age groups (infant, child, adolescent, young adult, middle-age adult, and old aged adults). Patients with congenital malformations, gross pathology of the supra- or infratentorial brain, brain volume loss, developmental delay, metabolic disorders, and neuropsychological disorders (n = 2.839) were excluded. On midsagittal T1 weighted and axial T2 weighted images specific linear diameters and ratios of the brainstem, cerebellar vermis, and peduncles were attained. Two observers assessed a random sample of 100 subjects to evaluate the inter- and intraobserver reproducibility. Intraclass correlation coefficients, means ± standard deviation, one and two-way analysis of variance tests were used in the statistical analysis. RESULTS: Good to excellent inter- and intraobserver measurements' reproducibility were observed, except for the transverse diameter of the midbrain, the anteroposterior diameter of the medulla oblongata at the pontomedullary and cervicomedullary junctions, cerebellar vermis anteroposterior diameter, and thickness of the superior cerebellar peduncle. Age-specific mean values of the investigated measurements were established. A significant gender-related variation was recorded in the anteroposterior diameter of the basis pontis (p = 0.044), the anteroposterior diameter of the medulla oblongata at the cervicomedullary junction (p = 0.044), and cerebellar vermis height (p = 0.018). A significant age-related change was detected in all measurements except the tectal ratio. Age by gender interaction had a statistically significant effect on the tectal ratio, inferior, and middle cerebellar peduncles' thickness (p = 0.001, 0.022, and 0.028, respectively). CONCLUSION: This study provides age-specific normal mean values for various linear dimensions and ratios of the posterior fossa structures with documentation of measurements' variability according to gender, age, and their interaction. ADVANCES IN KNOWLEDGE: It provides a valuable reference in the clinical practice for easier differentiation between physiological and pathological conditions of the posterior fossa structures especially various neurodegenerative diseases and congenital anomalies.


Assuntos
Mapeamento Encefálico/métodos , Tronco Encefálico/anatomia & histologia , Vermis Cerebelar/anatomia & histologia , Imageamento por Ressonância Magnética/métodos , Córtex Olfatório/anatomia & histologia , Adolescente , Adulto , Fatores Etários , Idoso , Pedúnculo Cerebral/anatomia & histologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Pedúnculo Cerebelar Médio/anatomia & histologia , Valores de Referência , Reprodutibilidade dos Testes , Adulto Jovem
5.
Sci Rep ; 8(1): 2780, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29426916

RESUMO

Numerous experimental studies have supported the evidence that 2-methoxyestradiol (2 ME) is a biologically active metabolite that mediates multiple effects on the cardiovascular system, largely independent of the estrogen receptor. 2 ME is a major cytochrome P450 1B1 (CYP1B1) metabolite and has been reported to have vasoprotective and anti-inflammatory actions. However, whether 2 ME would prevent cardiac hypertrophy induced by abdominal aortic constriction (AAC) has not been investigated yet. Therefore, the overall objectives of the present study were to elucidate the potential antihypertrophic effect of 2 ME and explore the mechanism(s) involved. Our results showed that 2 ME significantly inhibited AAC-induced left ventricular hypertrophy using echocardiography. The antihypertrophic effect of 2 ME was associated with a significant inhibition of CYP1B1 and mid-chain hydroxyeicosatetraenoic acids. Based on proteomics data, the protective effect of 2 ME is linked to the induction of antioxidant and anti-inflammatory proteins in addition to the modulation of proteins involved in myocardial energy metabolism. In vitro, 2 ME has shown a direct antihypertrophic effect through mitogen-activated protein kinases- and nuclear factor-κB-dependent mechanisms. The present work shows a strong evidence that 2 ME protects against left ventricular hypertrophy. Our data suggest the potential of repurposing 2 ME as a selective CYP1B1 inhibitor for the treatment of heart failure.


Assuntos
2-Metoxiestradiol/farmacologia , Citocromo P-450 CYP1B1 , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Substâncias Protetoras/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Citocromo P-450 CYP1B1/antagonistas & inibidores , Citocromo P-450 CYP1B1/metabolismo , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/citologia , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley
6.
Cardiovasc Toxicol ; 18(3): 268-283, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29196978

RESUMO

Several studies have demonstrated the role of cytochrome P450 (CYP) and its associated arachidonic acid (AA) metabolites in the anthracyclines-induced cardiac toxicity. However, the ability of daunorubicin (DNR) to induce cardiotoxicity through the modulation of CYP and its associated AA metabolites has not been investigated yet. Therefore, we hypothesized that DNR-induced cardiotoxicity is mediated through the induction of cardiotoxic hydroxyeicosatetraenoic acids and/or the inhibition of cardioprotctive epoxyeicosatrienoic acids (EETs). To test our hypothesis, Sprague-Dawley rats were treated with DNR (5 mg/kg i.p.) for 24 h, whereas human ventricular cardiomyocytes RL-14 cells were exposed to DNR in the presence and absence of 4-[[trans-4-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]cyclohexyl]oxy]-benzoic acid (tAUCB), a soluble epoxide hydrolase (sEH) inhibitor. Thereafter, real-time PCR, Western blot analysis and liquid chromatography-electron spray ionization mass spectroscopy were used to determine the level of gene expression, protein expression and AA metabolites, respectively. Our results showed that DNR-induced cardiotoxicity in vivo and in vitro as evidenced by the induction of hypertrophic and fibrotic markers. Moreover, the DNR-induced cardiotoxicity was associated with a dramatic increase in the formation of cardiac DHET/EET metabolites both in vivo and in RL-14 cells suggesting a sEH enzyme dependent mechanism. Interestingly, inhibition of sEH using tAUCB, a selective sEH inhibitor, significantly protects against DNR-induced cardiotoxicity. Mechanistically, the protective effect tAUCB was mediated through the induction of P50 nuclear factor-κB and the inhibition of phosphorylated p38. In conclusion, our study provides the first evidence that DNR induces cardiotoxicity through a sEH-mediated EETs degradation-dependent mechanism.


Assuntos
Antibióticos Antineoplásicos , Cardiomegalia/induzido quimicamente , Cardiomegalia/enzimologia , Daunorrubicina , Epóxido Hidrolases/metabolismo , Miócitos Cardíacos/enzimologia , Animais , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Cardiotoxicidade , Células Cultivadas , Sistema Enzimático do Citocromo P-450/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Epóxido Hidrolases/genética , Fibrose , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Subunidade p50 de NF-kappa B/metabolismo , Fosforilação , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Xenobiotica ; 48(2): 124-134, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28134025

RESUMO

1. Dimethylarsinic acid (DMA(V)) is the major metabolite of inorganic arsenic in human body. Thus we investigated the effect of DMA(V) on the alteration of phase I (typified by Cyp1a) and phase II (typified by Nqo1) AhR-regulated genes in vivo. C57BL/6 mice received DMA(V) (13.3 mg/kg, i.p.) with or without TCDD (15 µg/kg, i.p.), thereafter the liver, lung, and kidney were harvested at 6 and 24 h post-treatment. 2. Results demonstrated that DMA(V) has no significant effect on Cyp1a mRNA and protein expression or catalytic activity in the liver. On the other hand, DMA(V) significantly potentiated the TCDD-mediated induction of Cyp1a mRNA and protein expression, with a subsequent potentiation of catalytic activity in the lung. Moreover, DMA(V) significantly inhibited the TCDD-mediated induction of Cyp1a mRNA and protein expression with subsequent inhibition of catalytic activity in the kidney. 3. Regarding to phase II AhR-regulated genes, DMA(V) has no significant effect on Nqo1 mRNA and protein expression, or activity neither in the liver, lung, or kidney. 4. In conclusion, the present work demonstrates for the first time that DMA(V) modulates AhR-regulated genes in a tissue- and enzyme-specific manner. This modulation may play a crucial role in arsenic-induced toxicity and carcinogenicity.


Assuntos
Ácido Cacodílico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Herbicidas/farmacologia , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , RNA Mensageiro/metabolismo
8.
J Cardiovasc Pharmacol ; 70(1): 16-24, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28437282

RESUMO

Recent data demonstrated the role of CYP1B1 in cardiovascular disease. It was, therefore, necessary to examine whether the inhibition of CYP1B1 and hence inhibiting the formation of its metabolites, using 2,4,3',5'-tetramethoxystilbene (TMS), would have a cardioprotective effect against angiotensin II (Ang II)-induced cardiac hypertrophy. For this purpose, male Sprague Dawley rats were treated with Ang II with or without TMS (300 µg/kg every third day i.p.). Thereafter, cardiac hypertrophy and the formation of mid-chain HETEs and arachidonic acid were assessed. In vitro, RL-14 cells were treated with Ang II (10 µM) in the presence and absence of TMS (0.5 µM). Then, reactive oxygen species, mitogen-activated protein kinase phosphorylation levels, and nuclear factor-kappa B-binding activity were determined. Our results demonstrated that TMS protects against Ang II-induced cardiac hypertrophy as indicated by the improvement in cardiac functions shown by the echocardiography as well as by reversing the increase in heart weight to tibial length ratio caused by Ang II. In addition, the cardioprotective effect of TMS was associated with a significant decrease in cardiac mid-chain HETEs levels. Mechanistically, TMS inhibited reactive oxygen species formation, the phosphorylation of ERK1/2, p38 mitogen-activated protein kinase, and the binding of p65 NF-κB.


Assuntos
Angiotensina II/toxicidade , Cardiomegalia/metabolismo , Cardiomegalia/prevenção & controle , Citocromo P-450 CYP1B1/antagonistas & inibidores , Citocromo P-450 CYP1B1/metabolismo , Ácidos Hidroxieicosatetraenoicos/antagonistas & inibidores , Ácidos Hidroxieicosatetraenoicos/metabolismo , Animais , Cardiomegalia/induzido quimicamente , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Células Cultivadas , Humanos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Estilbenos/farmacologia , Estilbenos/uso terapêutico , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Mol Cell Biochem ; 429(1-2): 151-165, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28251434

RESUMO

Numerous experimental studies have demonstrated the role of cytochrome P450 1B1 (CYP1B1) and its associated mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) metabolite in the pathogenesis of cardiac hypertrophy. However, the ability of isoproterenol (ISO) to induce cardiac hypertrophy through mid-chain HETEs has not been investigated yet. Therefore, we hypothesized that ISO induces cardiac hypertrophy through the induction of CYP1B1 and its associated mid-chain HETE metabolites. To test our hypothesis, Sprague-Dawley rats were treated with ISO (5 mg/kg i.p.) for 12 and 72 h whereas, human ventricular cardiomyocytes RL-14 cells were exposed to 100 µM ISO in the presence and absence of 0.5 µM tetramethoxystilbene (TMS) a selective CYP1B1 inhibitor, or 25 nM CYP1B1-siRNA. Moreover, RL-14 cells were transiently transfected with the CRISPR-CYP1B1 plasmid. Thereafter, real-time PCR, western blot analysis, and liquid chromatography-electrospray ionization mass spectroscopy were used to determine the level of gene expression, protein expression, and mid-chain HETEs, respectively. Our results showed that ISO induced CYP1B1 protein expression and the level of cardiac mid-chain HETEs in vivo at pre-hypertrophic and hypertrophic stage. In vitro, inhibition of CYP1B1 using TMS or CYP1B1-siRNA significantly attenuates ISO-induced hypertrophy. Furthermore, overexpression of CYP1B1 significantly induced cellular hypertrophy and mid-chain HETEs metabolite. Mechanistically, the protective effect of TMS against cardiac hypertrophy was mediated through the modulation of superoxide anion, mitogen-activated protein kinases (MAPKs), and nuclear factor-κB (NF-κB). In conclusion, our study provides the first evidence that CYP1B1 and its associated mid-chain HETE metabolites are directly involved in the ISO-induced cardiac hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Citocromo P-450 CYP1B1/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Isoproterenol/efeitos adversos , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Linhagem Celular , Citocromo P-450 CYP1B1/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley
10.
Toxicol Lett ; 270: 34-50, 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28189647

RESUMO

Inorganic arsenic is a human toxicant and carcinogen that has been extensively studied over decades; however, no definitive understanding of the underlying mechanisms has been established yet. Arsenic is capable of modulating the expression of aryl hydrocarbon receptor (AhR)-regulated genes, nevertheless, whether its trivalent organic metabolites have similar effects or not need to be investigated. Therefore, in this study we examined the effects of monomethylarsonous acid (MMA(III)) as compared to its parent compound sodium arsenite (As(III)) on the expression of CYP1A1 in HepG2 cells. HepG2 cells were treated with MMA(III) (5µM) or its parents compound, As(III) (5µM), in the absence and presence of the prototypical AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 1nM). Experiments were conducted at 6h for gene expression; 24h for XRE-driven luciferase activity, protein expression, and EROD activity. Our results showed that both MMA(III) and As(III) decreased CYP1A1 mRNA, protein, and catalytic activity levels; and inhibit the TCDD-mediated induction of CYP1A1 mRNA, protein, and catalytic activity levels. MMA(III) and As(III) significantly inhibited XRE-driven luciferase activity and it inhibited the TCDD-mediated induction of XRE-driven luciferase reporter gene expression. Although MMA(III) and As(III) were not shown to be AhR ligands, both compounds showed inhibition of nuclear accumulation of AhR transcription factor as evidenced by immunocytochemical analysis. MMA(III) and As(III) had no effect on CYP1A1 mRNA stability; however MMA(III), but not As(III), decreased the protein stability of CYP1A1. As(III), but not MMA(III), induced HO-1 mRNA levels. Both MMA(III) and As(III) increased ROS production. Our results demonstrate for the first time that, MMA(III) down-regulates CYP1A1 mainly through transcriptional and post-translational mechanisms. This modulation of CYP1A1 proves that trivalent metabolites of arsenic are highly reactive and could participate in arsenic toxicity.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Compostos Organometálicos/toxicidade , Arsenitos/toxicidade , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citocromo P-450 CYP1A1/genética , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Células Hep G2 , Humanos , Estresse Oxidativo/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Processamento de Proteína Pós-Traducional , Estabilidade Proteica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Compostos de Sódio/toxicidade
11.
Pharmacol Res ; 105: 28-43, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26772815

RESUMO

Doxorubicin (DOX) has been reported to be a very potent and effective anticancer agent. However, clinical treatment with DOX has been greatly limited due to its cardiotoxicity. Furthermore, several studies have suggested a role for cytochrome P450 1B1 (CYP1B1) and mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) in DOX-induced cardiac toxicity. Therefore, we hypothesized that DOX induced cardiotoxicity is mediated through the induction of CYP1B1 and its associated mid-chain HETEs metabolite. To test our hypothesis, Sprague-Dawley rats and RL-14 cells were treated with DOX in the presence and absence of 2,3',4,5'-tetramethoxystilbene (TMS), a selective CYP1B1 inhibitor. Thereafter, cardiotoxicity parameters were determined using echocardiography, histopathology, and gene expression. Further, the level of mid-chain HETEs was quantified using liquid chromatography-electron spray ionization-mass spectrometry. Our results showed that DOX induced cardiotoxicity in vivo and in vitro as evidenced by deleterious changes in echocardiography, histopathology, and hypertrophic markers. Importantly, the TMS significantly reversed these changes. Moreover, the DOX-induced cardiotoxicity was associated with a proportional increase in the formation of cardiac mid-chain HETEs both in vivo and in our cell culture model. Interestingly, the inhibition of cardiotoxicity by TMS was associated with a dramatic decrease in the formation of cardiac mid-chain HETEs suggesting a mid-chain HETEs-dependent mechanism. Mechanistically, the protective effect of TMS against DOX-induced cardiotoxicity was mediated through the inhibition of mitogen activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB). In conclusion, our study provides the first evidence that the inhibition of CYP1B1 and mid-chain HETE formation attenuate DOX-induced cardiotoxicity.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Cardiotoxicidade/tratamento farmacológico , Citocromo P-450 CYP1B1/antagonistas & inibidores , Doxorrubicina/toxicidade , Inibidores Enzimáticos/uso terapêutico , Ácidos Hidroxieicosatetraenoicos/metabolismo , Estilbenos/uso terapêutico , Animais , Cardiotoxicidade/metabolismo , Cardiotoxicidade/patologia , Linhagem Celular , Citocromo P-450 CYP1B1/metabolismo , Humanos , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Ratos Sprague-Dawley
12.
Cell Biol Toxicol ; 31(4-5): 241-59, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26493311

RESUMO

Recent studies have established the role of mid-chain hydroxyeicosatetraenoic acids (mid-chain HETEs) in the development of cardiovascular disease. Among these mid-chains, 8-HETE has been reported to have a proliferator and proinflammatory action. However, whether 8-HETE can induce cardiac hypertrophy has never been investigated before. Therefore, the overall objectives of the present study are to elucidate the potential hypertrophic effect of 8-HETE in the human ventricular cardiomyocytes, RL-14 cells, and to explore the mechanism(s) involved. Our results showed that 8-HETE induced cellular hypertrophy in RL-14 cells as evidenced by the induction of cardiac hypertrophy markers ANP, BNP, α-MHC, and ß-MHC in a concentration- and time-dependent manner as well as the increase in cell surface area. Mechanistically, 8-HETE was able to induce the NF-κB activity as well as it significantly induced the phosphorylation of ERK1/2. The induction of cellular hypertrophy was associated with a proportional increase in the formation of dihydroxyeicosatrienoic acids (DHETs) parallel to the increase of soluble epoxide hydrolase (sEH) enzyme activity. Blocking the induction of NF-κB, ERK1/2, and sEH signaling pathways significantly inhibited 8-HETE-induced cellular hypertrophy. Our study provides the first evidence that 8-HETE induces cellular hypertrophy in RL-14 cells through MAPK- and NF-κB-dependent mechanism


Assuntos
Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Ácidos Hidroxieicosatetraenoicos/toxicidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , NF-kappa B/metabolismo , Adulto , Animais , Cardiomegalia/enzimologia , Cardiomegalia/patologia , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação , Ratos
13.
Toxicol Appl Pharmacol ; 289(3): 550-9, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26454030

RESUMO

We have recently demonstrated that 19-hydroxyeicosatetraenoic acid (19-HETE) is the major subterminal-HETE formed in the heart tissue, and its formation was decreased during cardiac hypertrophy. In the current study, we examined whether 19-HETE confers cardioprotection against angiotensin II (Ang II)-induced cardiac hypertrophy. The effect of Ang II, with and without 19-HETE (20 µM), on the development of cellular hypertrophy in cardiomyocyte RL-14 cells was assessed by real-time PCR. Also, cardiac hypertrophy was induced in Sprague-Dawley rats by Ang II, and the effect of increasing 19-HETE by isoniazid (INH; 200mg/kg/day) was assessed by heart weight and echocardiography. Also, alterations in cardiac cytochrome P450 (CYP) and their associated arachidonic acid (AA) metabolites were determined by real-time PCR, Western blotting and liquid-chromatography-mass-spectrometry. Our results demonstrated that 19-HETE conferred a cardioprotective effect against Ang II-induced cellular hypertrophy in vitro, as indicated by the significant reduction in ß/α-myosin heavy chain ratio. In vivo, INH improved heart dimensions, and reversed the increase in heart weight to tibia length ratio caused by Ang II. We found a significant increase in cardiac 19-HETE, as well as a significant reduction in AA and its metabolite, 20-HETE. In conclusion, 19-HETE, incubated with cardiomyocytes in vitro or induced in the heart by INH in vivo, provides cardioprotection against Ang II-induced hypertrophy. This further confirms the role of CYP, and their associated AA metabolites in the development of cardiac hypertrophy.


Assuntos
Angiotensina II/farmacologia , Cardiomegalia/induzido quimicamente , Cardiomegalia/tratamento farmacológico , Ácidos Hidroxieicosatetraenoicos/farmacologia , Isoniazida/farmacologia , Substâncias Protetoras/farmacologia , Animais , Ácido Araquidônico/metabolismo , Cardiomegalia/metabolismo , Células Cultivadas , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley
14.
Free Radic Biol Med ; 82: 1-12, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25614461

RESUMO

Evidence suggests that upregulation of soluble epoxide hydrolase (sEH) is associated with the development of myocardial infarction, dilated cardiomyopathy, cardiac hypertrophy, and heart failure. However, the upregulation mechanism is still unknown. In this study, we treated H9C2 cells with buthionine sulfoximine (BSO) to explore whether oxidative stress upregulates sEH gene expression and to identify the molecular and cellular mechanisms behind this upregulatory response. Real-time PCR and Western blot analyses were used to measure mRNA and protein expression, respectively. We demonstrated that BSO significantly upregulated sEH at mRNA levels in a concentration- and time-dependent manner, leading to a significant increase in the cellular hypertrophic markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). Furthermore, BSO significantly increased the cytosolic phosphorylated IκB-α and translocation of NF-κB p50 subunits, as measured by Western blot analysis. This level of translocation was paralleled by an increase in the DNA-binding activity of NF-κB P50 subunits. Moreover, our results demonstrated that pretreatment with the NF-κB inhibitor PDTC significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression in a dose-dependent manner. Additionally, mitogen-activated protein kinases (MAPKs) were transiently phosphorylated by BSO treatment. To understand further the role of MAPKs pathway in BSO-mediated induction of sEH mRNA, we examined the role of extracellular signal-regulated kinase (ERK), c-JunN-terminal kinase (JNK), and p38 MAPK. Indeed, treatment with the MEK/ERK signal transduction inhibitor, PD98059, partially blocked the activation of IκB-α and translocation of NF-κB p50 subunits induced by BSO. Moreover, pretreatment with MEK/ERK signal transduction inhibitors, PD98059 and U0126, significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression. These results clearly demonstrated that the NF-κB signaling pathway is involved in BSO-mediated induction of sEH gene expression, and appears to be associated with the activation of the MAPK pathway. Furthermore, our findings provide a strong link between sEH-induced cardiac dysfunction and involvement of NF-κB in the development of cellular hypertrophy.


Assuntos
Butionina Sulfoximina/farmacologia , Cardiomegalia/patologia , Epóxido Hidrolases/biossíntese , Sistema de Sinalização das MAP Quinases/fisiologia , Subunidade p50 de NF-kappa B/metabolismo , Animais , Antioxidantes/farmacologia , Fator Natriurético Atrial/biossíntese , Butadienos/farmacologia , Linhagem Celular , Sobrevivência Celular , Ativação Enzimática , Epóxido Hidrolases/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Regulação da Expressão Gênica/fisiologia , Glutationa/biossíntese , Insuficiência Cardíaca/patologia , Proteínas I-kappa B/biossíntese , Proteínas I-kappa B/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mioblastos Cardíacos/metabolismo , Inibidor de NF-kappaB alfa , Subunidade p50 de NF-kappa B/antagonistas & inibidores , Subunidade p50 de NF-kappa B/biossíntese , Peptídeo Natriurético Encefálico/biossíntese , Nitrilas/farmacologia , Estresse Oxidativo , Fosforilação , Prolina/análogos & derivados , Prolina/farmacologia , RNA Mensageiro/biossíntese , Ratos , Tiocarbamatos/farmacologia , Fator de Transcrição RelA/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Artigo em Inglês | MEDLINE | ID: mdl-25454080

RESUMO

INTRODUCTION: RL-14 cells, human fetal ventricular cardiomyocytes, are a commercially available cell line that has been established from non-proliferating primary cultures derived from human fetal heart tissue. However, the expression of different drug metabolizing enzymes (DMEs) in RL-14 cells has not been elucidated yet. Therefore, the main objectives of the current work were to investigate the capacity of RL-14 cells to express different cytochrome P450 (CYP) isoenzymes and correlate this expression to primary cardiomyocytes. METHODS: The expression of CYP isoenzymes was determined at mRNA, protein and catalytic activity levels using real time-PCR, Western blot analysis and liquid chromatography-electron spray ionization-mass spectrometry (LC-ESI-MS), respectively. RESULTS: Our results showed that RL-14 cells constitutively express CYP ω-hydroxylases, CYP1A, 1B, 4A and 4F; CYP epoxygenases, CYP2B, 2C and 2J; in addition to soluble epoxide hydrolayse (EPHX2) at mRNA and protein levels. The basal expression of CYP ω-hydroxylases, epoxygenases and EPHX2 was supported by the ability of RL-14 cells to convert arachidonic acid to its biologically active metabolites, 20-hydroxyeicosatetraenoic acids (20-HETEs), 14,15-epoxyeicosatrienoic acids (14,15-EET), 11,12-EET, 8,9-EET, 5,6-EET, 14,15-dihydroxyeicosatrienoic acid (14,15-DHET), 11,12-DHET, 8,9-DHET and 5,6-DHET. Furthermore, RL-14 cells express CYP epoxygenases and ω-hydroxylase at comparable levels to those expressed in adult and fetal human primary cardiomyocytes cells implying the importance of RL-14 cells as a model for studying DMEs in vitro. Lastly, different CYP families were induced in RL-14 cells using 2,3,7,8-tetrachlorodibenzo-p-dioxin and fenofibrate at mRNA and protein levels. DISCUSSION: The current study provides the first evidence that RL-14 cells express CYP isoenzymes at comparable levels to those expressed in the primary cells and thus offers a unique in vitro model to study DMEs in the heart.


Assuntos
Ácido Araquidônico/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Fenofibrato/farmacologia , Modelos Biológicos , Dibenzodioxinas Policloradas/farmacologia , Células Cultivadas , Sistema Enzimático do Citocromo P-450/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Vascul Pharmacol ; 58(5-6): 363-73, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23466634

RESUMO

We have previously shown that isoproterenol-induced cardiac hypertrophy causes significant changes to cytochromes P450 (CYPs) and soluble epoxide hydrolase (sEH) gene expression. Therefore, in this study, we examined the effect of isoproterenol in H9c2 cells, and the protective effects of 14,15-EET against isoproterenol-induced cellular hypertrophy. Isoproterenol was incubated with H9c2 cells for 24 and 48 h. To determine the protective effects of 14,15-EET, H9c2 cells were incubated with isoproterenol in the absence and presence of 14,15-EET. Thereafter, the expression of hypertrophic markers and different CYP genes were determined by real time-PCR. Our results demonstrated that isoproterenol significantly increased the expression of hypertrophic marker, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), parallel to a significant increase in cell surface area. Also, isoproterenol increased the mRNA expression of CYP1A1, CYP1B1, CYP2J3, CYP4F4 and CYP4F5, as well as the gene encoding sEH, EPHX2. On other hand, 14,15-EET significantly attenuated the isoproterenol-mediated induction of ANP, BNP, CYP1A1, CYP2J3, CYP4F4, CYP4F5 and EPHX2. Moreover 14,15-EET prevented the isoproterenol-mediated increase in cell surface area. Interestingly, 20-hydroxyeicosatetraenoic acid (20-HETE) treatment caused similar effects to that of isoproterenol treatment and induced cellular hypertrophy in H9c2 cells. In conclusion, isoproterenol induces cellular hypertrophy and modulates the expression of CYPs and EPHX2 in H9c2 cells. Furthermore, 14,15-EET exerts a protective effect against isoproterenol-induced cellular hypertrophy whereas, 20-HETE induced cellular hypertrophy in H9c2 cells.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Cardiomegalia/prevenção & controle , Isoproterenol/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Cardiomegalia/induzido quimicamente , Linhagem Celular , Sistema Enzimático do Citocromo P-450/genética , Epóxido Hidrolases/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Ácidos Hidroxieicosatetraenoicos/toxicidade , Miócitos Cardíacos/patologia , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo
17.
Free Radic Biol Med ; 58: 52-63, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23369756

RESUMO

Heavy metals, typified by arsenite (As(III)), have been implicated in altering the carcinogenicity of aryl hydrocarbon receptor (AhR) ligands, typified by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), by modulating the induction of the Cyp1a1 enzyme, but the mechanism remains unresolved. In this study, the effects of As(III) on Cyp1a1 expression and activity were investigated in C57BL/6 mouse livers and isolated hepatocytes. For this purpose, C57BL/6 mice were injected intraperitoneally with As(III) (12.5 mg/kg) in the absence and presence of TCDD (15 µg/kg) for 6 and 24 h. Furthermore, isolated hepatocytes from C57BL/6 mice were treated with As(III) (1, 5, and 10 µM) in the absence and presence of TCDD (1 nM) for 3, 6, 12, and 24 h. At the in vivo level, As(III) decreased the TCDD-mediated induction of Cyp1a1 mRNA at 6h while potentiating its mRNA, protein, and catalytic activity levels at 24 h. At the in vitro level, As(III) decreased the TCDD-mediated induction of Cyp1a1 mRNA in a concentration- and time-dependent manner. Moreover, As(III) decreased the TCDD-mediated induction of Cyp1a1 protein and catalytic activity levels at 24 h. Interestingly, As(III) increased the serum hemoglobin (Hb) levels in animals treated for 24 h. Upon treatment of isolated hepatocytes with Hb alone, there was an increase in the nuclear accumulation of AhR and AhR-dependent luciferase activity. Furthermore, Hb potentiated the TCDD-induced AhR-dependent luciferase activity. Importantly, when isolated hepatocytes were treated for 5h with As(III) in the presence of TCDD and the medium was then replaced with new medium containing Hb, there was potentiation of the TCDD-mediated effect. Taken together, these results demonstrate for the first time that there is a differential modulation of the TCDD-mediated induction of Cyp1a1 by As(III) in C57BL/6 mouse livers and isolated hepatocytes. Thus, this study implicates Hb as an in vivo-specific modulator.


Assuntos
Arsenitos/farmacologia , Citocromo P-450 CYP1A1/metabolismo , Hemoglobinas/metabolismo , Dibenzodioxinas Policloradas/farmacologia , Animais , Células Cultivadas , Radicais Livres/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Proteínas de Membrana/metabolismo , Metais Pesados/farmacologia , Camundongos , RNA Mensageiro/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/metabolismo
18.
Arch Toxicol ; 87(8): 1531-43, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23423713

RESUMO

The objective of the current study was to investigate the effect of vanadium (V(5+)) on Cyp1 expression and activity in C57BL/6 mice liver and isolated hepatocytes. For this purpose, C57BL6 mice were injected intraperitoneally with V(5+) (5 mg/kg) in the absence and presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (15 µg/kg) for 6 and 24 h. Furthermore, isolated hepatocytes from C57BL6 mice were treated with V(5+) (5, 10, and 20 µM) in the absence and presence of TCDD (1 nM) for 3, 6, 12, and 24 h. In vivo, V(5+) alone did not significantly alter Cyp1a1, Cyp1a2, or Cyp1b1 mRNA, protein, or catalytic activity levels. Upon co-exposure to V(5+) and TCDD, V(5+) significantly potentiated the TCDD-mediated induction of the Cyp1a1, Cyp1a2, and Cyp1b1 mRNA, protein, and catalytic activity levels at 24 h. In vitro, V(5+) decreased the TCDD-mediated induction of Cyp1a1 mRNA, protein, and catalytic activity levels. Furthermore, V(5+) significantly inhibited the TCDD-induced AhR-dependent luciferase activity. V(5+) also increased serum hemoglobin (Hb) levels in animals treated for 24 h. Upon treatment of isolated hepatocytes with Hb alone or in the presence of TCDD, there was an increase in the AhR-dependent luciferase activity. When isolated hepatocytes were treated for 2 h with V(5+) in the presence of TCDD, followed by replacement of the medium with new medium containing Hb, there was further potentiation to the TCDD-mediated effect. The present study demonstrates that there is a differential modulation of Cyp1a1 by V(5+) in C57BL/6 mice livers and isolated hepatocytes and demonstrates Hb as an in vivo specific modulator.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Dibenzodioxinas Policloradas/farmacologia , Vanádio/farmacologia , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1B1 , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica , Heme Oxigenase-1/genética , Hemoglobinas/metabolismo , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Hidrocarboneto Arílico/metabolismo
19.
Vascul Pharmacol ; 58(4): 326-33, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23268359

RESUMO

HL-1 cells are currently the only cells that spontaneously contract while maintaining a differentiated cardiac phenotype. Thus, our objective was to examine murine HL-1 cells as a new in vitro model to study drug metabolizing enzymes. We examined the expression of cytochrome P450s (Cyps), phase II enzymes, and nuclear receptors and compared their levels to mice hearts. Our results demonstrated that except for Cyp4a12 and Cyp4a14 all Cyps, phase II enzymes: glutathione-S-transferases (Gsts), heme oxygenase-1 (HO-1), and NAD(P)H: quinone oxidoreductase (Nqo1), nuclear receptors: aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR), pregnane X receptor (PXR), and peroxisome proliferator activated receptor (PPAR-alpha) were all constitutively expressed in HL-1 cells. Cyp2b19, Cyp2c29, Cyp2c38, Cyp2c40, and Cyp4f16 mRNA levels were higher in HL-1 cells compared to mice hearts. Cyp2b9, Cyp2c44, Cyp2j9, Cyp2j11, Cyp2j13, Cyp4f13, Cyp4f15 mRNA levels were expressed to the same extent to that of mice hearts. Cyp1a1, Cyp1a2, Cyp1b1, Cyp2b10, Cyp2d10, Cyp2d22, Cyp2e1, Cyp2j5, Cyp2j6, Cyp3a11, Cyp4a10, and Cyp4f18 mRNA levels were lower in HL-1 cells compared to mice hearts. Moreover, 3-methylcholanthrene induced Cyp1a1 while fenofibrate induced Cyp2j9 and Cyp4f13 mRNA levels in HL-1 cells. Examining the metabolism of arachidonic acid (AA) by HL-1 cells, our results demonstrated that HL-1 cells metabolize AA to epoxyeicosatrienoic acids, dihydroxyeicosatrienoic acids, and 20-hydroxyeicosatetraenoic acids. In conclusion, HL-1 cells provide a valuable in vitro model to study the role of Cyps and their associated AA metabolites in addition to phase II enzymes in cardiovascular disease states.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Regulação Enzimológica da Expressão Gênica , Átrios do Coração/enzimologia , Miocárdio/enzimologia , Animais , Ácido Araquidônico/metabolismo , Linhagem Celular , Citocromo P-450 CYP2J2 , Indução Enzimática/efeitos dos fármacos , Enzimas/metabolismo , Átrios do Coração/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Reprodutibilidade dos Testes
20.
J Appl Toxicol ; 33(11): 1230-40, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22611038

RESUMO

We recently reported that vanadium (V(5+) ) was able to decrease the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated induction of Cyp1a1 and Nqo1 at mRNA, protein and catalytic activity levels in mouse hepatoma Hepa 1c1c7 and human hepatoma HepG2 cells. However, little is known regarding the in vivo effects. Thus, the objective of this study was to investigate whether similar effects would occur at the in vivo level. Therefore, we examined the effect of exposure to V(5+) (5 mg kg(-1) ) with or without TCDD (15 µg kg(-1) ) on the AhR-regulated genes in kidney, lung and heart of C57BL/6 J mice. Our results demonstrated that V(5+) alone significantly decreased Cyp1b1 protein and catalytic activity levels in kidney at 24 h. Moreover, it significantly potentiated Nqo1 and Gsta1 gene expression in the heart, and only Gsta1 gene expression in the lung. Upon co-exposure, we found that V(5+) significantly inhibited the TCDD-mediated induction of Cyp1a1, Cyp1a2 and Cyp1b1 mRNA, protein and catalytic activity levels in the kidney at 24 h. On the other hand, V(5+) significantly potentiated the TCDD-mediated induction of Nqo1 and Gsta1 protein and activity levels in the kidney. Cyp1a1, Cyp1b1, Nqo1 mRNA, protein and catalytic activity levels in the lung were significantly potentiated at 6 h. Interestingly, all tested genes in the heart were significantly decreased at 6 h with the exception of Gsta1 mRNA. The present study demonstrates that V(5+) modulates TCDD-induced AhR-regulated genes. Furthermore, the effect on one of these enzymes could not be generalized to other enzymes even if it was in the same organ.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Rim/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/genética , Vanadatos/farmacologia , Animais , Western Blotting , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1B1/genética , Interações Medicamentosas , Glutationa Transferase/genética , Isoenzimas/genética , Rim/metabolismo , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microssomos/efeitos dos fármacos , Microssomos/metabolismo , Miocárdio/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...