Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 356: 36-43, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30030096

RESUMEN

Cadmium (Cd) has been linked to a variety of cancers, including breast cancer; however, the molecular mechanism of its carcinogenic activity is not fully understood. To this end, the present study investigated the roles of ferroportin (FPN), a prognostic marker of breast cancer, in Cd-induced stimulation of cell proliferation and cell migration. Triple-negative MDA-MB-231 cells were treated with 1-3 µM Cd. The cells exhibited significant reduction in FPN expression and concomitant increase in iron concentration. Cells treated with Cd for 8 weeks displayed elevated proliferative and migratory activities which were inversely related with FPN expression. Reduced FPN expression also resulted in EMT as indicated by an increase in the expression of E-cadherin, and a decrease in the expression of N-cadherin, Twist and Slug. Further investigation revealed that Cd suppressed FPN expression at least partially by activating TGF-ß, a known regulator of FPN expression. Taken together, these results indicate that Cd-induced stimulation of MDA-MB-231 cell proliferation, EMT, and migration is brought about by suppression of FPN expression and associated disruption of iron homeostasis.


Asunto(s)
Cadmio/toxicidad , Proteínas de Transporte de Catión/antagonistas & inhibidores , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/patología , Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Proteínas de Transporte de Catión/biosíntesis , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Homeostasis/efectos de los fármacos , Humanos , Hierro/metabolismo , ARN Interferente Pequeño/farmacología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
2.
Toxicol Appl Pharmacol ; 344: 46-55, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29501589

RESUMEN

Epidemiological and experimental studies have implicated cadmium (Cd) with breast cancer. In breast epithelial MCF10A and MDA-MB-231 cells, Cd has been shown to promote cell growth. The present study examined whether Cd also promotes epithelial-mesenchymal transition (EMT), a hallmark of cancer progression. Human breast epithelial cells consisting of non-cancerous MCF10A, non-metastatic HCC 1937 and HCC 38, and metastatic MDA-MB-231 were treated with 1 or 3 µM Cd for 4 weeks. The MCF10A epithelial cells switched to a more mesenchymal-like morphology, which was accompanied by a decrease in the epithelial marker E-cadherin and an increase in the mesenchymal markers N-cadherin and vimentin. In both non-metastatic HCC 1937 and HCC 38 cells, treatment with Cd decreased the epithelial marker claudin-1. In addition, E-cadherin also decreased in the HCC 1937 cells. Even the mesenchymal-like MDA-MB-231 cells exhibited an increase in the mesenchymal marker vimentin. These changes indicated that prolonged treatment with Cd resulted in EMT in both normal and cancer-derived breast epithelial cells. Furthermore, both the MCF10A and MDA-MB-231 cells labeled with Zcad, a dual sensor for tracking EMT, demonstrated a decrease in the epithelial marker E-cadherin and an increase in the mesenchymal marker ZEB-1. Treatment of cells with Cd significantly increased the level of Snail, a transcription factor involved in the regulation of EMT. However, the Cd-induced Snail expression was completely abolished by actinomycin D. Luciferase reporter assay indicated that the expression of Snail was regulated by Cd at the promotor level. Snail was essential for Cd-induced promotion of EMT in the MDA-MB-231 cells, as knockdown of Snail expression blocked Cd-induced cell migration. Together, these results indicate that Cd promotes EMT in breast epithelial cells and does so by modulating the transcription of Snail.


Asunto(s)
Mama/fisiología , Cadmio/toxicidad , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal/fisiología , Factores de Transcripción de la Familia Snail/fisiología , Antígenos CD , Mama/citología , Mama/efectos de los fármacos , Cadherinas/fisiología , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos
3.
Toxicol Appl Pharmacol ; 328: 70-80, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28527916

RESUMEN

Cadmium (Cd) is a carcinogenic heavy metal which is implicated in breast cancer development. While the mechanisms of Cd-induced breast cancer initiation and promotion have been studied, the molecular processes involved in breast cancer progression remain to be investigated. The purpose of the present study was to evaluate the influence of Cd on metastasis-associated phenotypes, such as cell adhesion, migration, and invasion in triple-negative breast cancer cells. Treatment of MDA-MB-231 cells with 1µM Cd increased cell spreading and cell migration. This was associated with the activation of integrin ß1, FAK, Src, and Rac1. Treatment with Cd also inhibited GSK3ß activity and induced T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription, indicating the involvement of ß-catenin signaling. Furthermore, treatment with 3µM Cd for 4weeks increased the expression of ß-catenin and enhanced TCF/LEF-mediated transcription. Furthermore, enhanced expressions of integrins α5 and ß1, paxillin, and vimentin indicated that prolonged Cd treatment reorganized the cytoskeleton, which aided malignancy, as evidenced by enhanced matrix metalloprotease 2/9 (MMP2/9) secretion and cell invasion. Prolonged Cd treatment also caused an increase in cell growth. Together, these results indicate that Cd alters key signaling processes involved in the regulation of cytoskeleton to enhance cancer cell migration, invasion, adhesion, and proliferation.


Asunto(s)
Cadmio/toxicidad , Integrinas/efectos de los fármacos , Metástasis de la Neoplasia/patología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , beta Catenina/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Fenotipo , Cicatrización de Heridas/efectos de los fármacos
4.
Food Funct ; 8(2): 757-766, 2017 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-28112327

RESUMEN

Oxidative stress and free radical generation accelerate the formation of advanced glycation endproducts (AGEs) which are linked to several chronic diseases. Published data suggest that phenolic-rich plant foods, show promise as natural anti-AGEs agents due to their anti-oxidation capacities. A phenolic-enriched maple syrup extract (MSX) has previously been reported to show anti-inflammatory and neuroprotective effects but its anti-AGE effects remain unknown. Therefore, herein, we investigated the anti-glycation and anti-oxidation effects of MSX using biochemical and biophysical methods. MSX (500 µg mL-1) reduced the formation of AGEs by 40% in the bovine serum albumin (BSA)-fructose assay and by 30% in the BSA-methylglyoxal (MGO) assay. MSX also inhibited the formation of crosslinks typically seen in the late stage of glycation. Circular dichroism and differential scanning calorimeter analyses demonstrated that MSX maintained the structure of BSA during glycation. In the anti-oxidant assays, MSX (61.7 µg mL-1) scavenged 50% of free radicals (DPPH assay) and reduced free radical generation by 20% during the glycation process (electron paramagnetic resonance time scan). In addition, the intracellular levels of hydrogen peroxide induced reactive oxygen species were reduced by 27-58% with MSX (50-200 µg mL-1) in normal/non-tumorigenic human colon CCD-18Co cells. Moreover, in AGEs and MGO challenged CCD-18Co cells, higher cellular viabilities and rapid extracellular signal-regulated kinase (ERK) phosphorylation were observed in MSX treated cells, indicating its protective effects against AGEs-induced cytotoxicity. Overall, this study supports the biological effects of MSX, and warrants further investigation of its potential as a dietary agent against diseases mediated by oxidative stress and inflammation.


Asunto(s)
Acer/química , Antioxidantes/farmacología , Colon/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fenoles/farmacología , Extractos Vegetales/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Colon/citología , Colon/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Glicosilación/efectos de los fármacos , Humanos , Fenoles/análisis , Especies Reactivas de Oxígeno/metabolismo
5.
Neurochem Res ; 41(11): 2836-2847, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27418278

RESUMEN

Published data supports the neuroprotective effects of several phenolic-containing natural products, including certain fruit, berries, spices, nuts, green tea, and olive oil. However, limited data are available for phenolic-containing plant-derived natural sweeteners including maple syrup. Herein, we investigated the neuroprotective effects of a chemically standardized phenolic-enriched maple syrup extract (MSX) using a combination of biophysical, in vitro, and in vivo studies. Based on biophysical data (Thioflavin T assay, transmission electron microscopy, circular dichroism, dynamic light scattering, and zeta potential), MSX reduced amyloid ß1-42 peptide (Aß1-42) fibrillation in a concentration-dependent manner (50-500 µg/mL) with similar effects as the neuroprotective polyphenol, resveratrol, at its highest test concentration (63.5 % at 500 µg/mL vs. 77.3 % at 50 µg/mL, respectively). MSX (100 µg/mL) decreased H2O2-induced oxidative stress (16.1 % decrease in ROS levels compared to control), and down-regulated the production of lipopolysaccharide (LPS)-stimulated inflammatory markers (22.1, 19.9, 74.8, and 87.6 % decrease in NOS, IL-6, PGE2, and TNFα levels, respectively, compared to control) in murine BV-2 microglial cells. Moreover, in a non-contact co-culture cell model, differentiated human SH-SY5Y neuronal cells were exposed to conditioned media from BV-2 cells treated with MSX (100 µg/mL) and LPS or LPS alone. MSX-BV-2 media increased SH-SY5Y cell viability by 13.8 % compared to media collected from LPS-BV-2 treated cells. Also, MSX (10 µg/mL) showed protective effects against Aß1-42 induced neurotoxicity and paralysis in Caenorhabditis elegans in vivo. These data support the potential neuroprotective effects of MSX warranting further studies on this natural product.


Asunto(s)
Acer/química , Péptidos beta-Amiloides/farmacología , Supervivencia Celular/efectos de los fármacos , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/metabolismo , Animales , Caenorhabditis elegans , Técnicas de Cocultivo , Ratones , Fármacos Neuroprotectores/metabolismo , Fenoles/farmacología , Polifenoles
6.
Toxicol Appl Pharmacol ; 289(1): 98-108, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26385184

RESUMEN

Cadmium (Cd) is a carcinogenic metal which is implicated in breast cancer by epidemiological studies. It is reported to promote breast cancer cell growth in vitro through membrane receptors. The study described here examined Cd-mediated growth of non-metastatic human breast cancer derived cells that lack receptors for estrogen, progesterone, and HER2. Treatment of triple-negative HCC 1937 cells with 0.1-0.5 µM Cd increased cell growth by activation of AKT and ERK. Accelerated cell cycle progression was achieved by increasing the levels of cyclins A, B, and E, as well as those of CDKs 1 and 2. Although triple negative cells lack estrogen receptor, they express high levels of EGFR. Therefore, further studies on HCC 1937 and another triple-negative cell line, HCC 38, were conducted using specific siRNA and an inhibitor of EGFR to determine whether EGFR was responsible for mediating the effect of Cd. The results revealed that in both cell types EGFR was not only activated upon Cd treatment, but was also essential for the downstream activation of AKT and ERK. Based on these observations, it is concluded that, in breast cancer cells lacking estrogen receptor, sub-micromolar concentration of Cd can promote cell proliferation. Furthermore, that EGFR plays a critical role in this process.


Asunto(s)
Cadmio/toxicidad , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Receptores ErbB/genética , Femenino , Humanos , Sistema de Señalización de MAP Quinasas , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/metabolismo
7.
Toxicol Appl Pharmacol ; 287(1): 26-34, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26006730

RESUMEN

Cadmium (Cd) is a common environmental toxicant and an established carcinogen. Epidemiological studies implicate Cd with human breast cancer. Low micromolar concentrations of Cd promote proliferation of human breast cancer cells in vitro. The growth promotion of breast cancer cells is associated with the activation of MAPK/ERK pathway. This study explores the mechanism of Cd-induced activation of MAPK/ERK pathway. Specifically, the role of cell surface receptors ERα, EGFR, and Src kinase was evaluated in human breast cancer MCF-7 cells treated with 1-3µM Cd. The activation of ERK was studied using a serum response element (SRE) luciferase reporter assay. Receptor phosphorylation was detected by Western blot analyses. Cd treatment increased both the SRE reporter activity and ERK1/2 phosphorylation in a concentration-dependent manner. Cd treatment had no effect on reactive oxygen species (ROS) generation. Also, blocking the entry of Cd into the cells with manganese did not diminish Cd-induced activation of MAPK/ERK. These results suggest that the effect of Cd was likely not caused by intracellular ROS generation, but through interaction with the membrane receptors. While Cd did not appear to activate either EGFR or Src kinase, their inhibition completely blocked the Cd-induced activation of ERK as well as cell proliferation. Similarly, silencing ERα with siRNA or use of ERα antagonist blocked the effects of Cd. Based on these results, it is concluded that not only ERα, but also basal activities of EGFR and Src kinase are essential for Cd-induced signal transduction and activation of MAPK/ERK pathway for breast cancer cell proliferation.


Asunto(s)
Neoplasias de la Mama/enzimología , Cloruro de Cadmio/toxicidad , Carcinógenos Ambientales/toxicidad , Receptores ErbB/metabolismo , Receptor alfa de Estrógeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Familia-src Quinasas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática , Receptores ErbB/antagonistas & inhibidores , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Células MCF-7 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Factores de Tiempo , Transfección , Familia-src Quinasas/antagonistas & inhibidores
8.
Toxicol Appl Pharmacol ; 245(1): 83-90, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20153348

RESUMEN

Cadmium (Cd) is a nonessential metal that is dispersed throughout the environment. It is an endocrine-disrupting element which mimics estrogen, binds to estrogen receptor alpha (ERalpha), and promotes cell proliferation in breast cancer cells. We have previously published that Cd promotes activation of the extracellular regulated kinases, erk-1 and -2 in both ER-positive and ER-negative human breast cancer cells, suggesting that this estrogen-like effect of Cd is not associated with the ER. Here, we have investigated whether the newly appreciated transmembrane estrogen receptor, G-protein coupled receptor 30 (GPR30), may be involved in Cd-induced cell proliferation. Towards this end, we compared the effects of Cd in ER-negative human SKBR3 breast cancer cells in which endogenous GPR30 signaling was selectively inhibited using a GPR30 interfering mutant. We found that Cd concentrations from 50 to 500 nM induced a proliferative response in control vector-transfected SKBR3 cells but not in SKBR3 cells stably expressing interfering mutant. Similarly, intracellular cAMP levels increased about 2.4-fold in the vector transfectants but not in cells in which GPR30 was inactivated within 2.5 min after treatment with 500 nM Cd. Furthermore, Cd treatment rapidly activated (within 2.5 min) raf-1, mitogen-activated protein kinase kinase, mek-1, extracellular signal regulated kinases, erk-1/2, ribosomal S6 kinase, rsk, and E-26 like protein kinase, elk, about 4-fold in vector transfectants. In contrast, the activation of these signaling molecules in SKBR3 cells expressing the GPR30 mutant was only about 1.4-fold. These results demonstrate that Cd-induced breast cancer cell proliferation occurs through GPR30-mediated activation in a manner that is similar to that achieved by estrogen in these cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Cadmio/toxicidad , Disruptores Endocrinos/toxicidad , Receptores Acoplados a Proteínas G/metabolismo , Carcinógenos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Receptores de Estrógenos
9.
Toxicol Appl Pharmacol ; 241(1): 81-9, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19682480

RESUMEN

Kidney is the primary target organ in chronic cadmium (Cd) toxicity, and oxidative stress plays an important role in this process. The nuclear transcription factor Nrf2 binds to antioxidant response elements (AREs) and regulates genes involved in protecting cells from oxidative damage. Whether kidney cells respond to Cd by activating Nrf2 is unknown. This study was designed to examine the Cd-induced activation of Nrf2 transcriptional activity in a stable rat kidney cell line, NRK-52E, and to investigate the protection this might offer against apoptosis. The cells were treated with 5-20 microM CdCl(2) for 5 h, followed by a recovery period of up to 24 h. A concentration-dependent increase (up to 2.9-fold) in the level of reactive oxygen species was noted upon termination of 5-h Cd treatment. The Nrf2-ARE binding activity also increased and peaked (6.1-fold) at 10 microM Cd concentration. Time-course study revealed that the binding activity increased at 1 h of Cd treatment and peaked 2 h post Cd treatment. Apoptosis was detected 6 h post treatment with Cd and a concentration- and time-dependent increase in the apoptotic cell population occurred during the next 18 h. Over-expression of Nrf2 by transient transfection conferred resistance against Cd-induced apoptosis. Conversely, suppression of Nrf2 expression by specific siRNA resulted in greater sensitivity of the cells to Cd by decreasing the levels of two antioxidant enzymes, hemeoxygenase-1 and glutamate-cysteine ligase. Taken together, these results suggest that in kidney cells the activation of Nrf2 is an adaptive intracellular response to Cd-induced oxidative stress, and that Nrf2 is protective against Cd-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Cloruro de Cadmio/toxicidad , Túbulos Renales Proximales/efectos de los fármacos , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Cloruro de Cadmio/administración & dosificación , Línea Celular , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Glutamato-Cisteína Ligasa/metabolismo , Hemo-Oxigenasa 1/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , ARN Interferente Pequeño/administración & dosificación , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Transfección
10.
Toxicol Appl Pharmacol ; 228(3): 286-94, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18275979

RESUMEN

Cadmium (Cd), an endocrine disruptor, can induce a variety of signaling events including the activation of ERK1/2 and AKT. In this study, the involvement of estrogen receptors (ER) in these events was evaluated in three human breast cancer cell lines, MCF-7, MDA-MB-231, and SK-BR-3. The Cd-induced signal activation patterns in the three cell lines mimicked those exhibited in response to 17 beta-estradiol. Specifically, treatment of MCF-7 cells, that express ER alpha, ER beta and GPR30, to 0.5-10 microM Cd for only 2.5 min resulted in transient phosphorylation of ERK1/2. Cd also triggered a gradual increase and sustained activation of AKT during the 60 min treatment period. In SK-BR-3 cells, that express only GPR30, Cd also caused a transient activation of ERK1/2, but not of AKT. In contrast, in MDA-MB-231 cells, that express only ER beta, Cd was unable to cause rapid activation of either ERK1/2 or AKT. A transient phosphorylation of ER alpha was also observed within 2.5 min of Cd exposure in the MCF-7 cells. While the estrogen receptor antagonist, ICI 182,780, did not prevent the effect of Cd on these signals, specific siRNA against hER alpha significantly reduced Cd-induced activation of ERK1/2 and completely blocked the activation of AKT. It is concluded that Cd, like estradiol, can cause rapid activation of ERK1/2 and AKT and that these signaling events are mediated by possible interaction with membrane ER alpha and GPR30, but not ER beta.


Asunto(s)
Neoplasias de la Mama/metabolismo , Cadmio/toxicidad , Disruptores Endocrinos/toxicidad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Fosforilación , ARN Interferente Pequeño/farmacología
11.
Toxicology ; 223(3): 202-8, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16682108

RESUMEN

Glycine treatment is reported to protect against the nephrotoxicity of cadmium (Cd) in rats. The purpose of the present study was to explore the mechanism of this protection using a renal epithelial cell line, LLC-PK(1). The cells were incubated with 10-30 microM Cd in serum-free DMEM and cytotoxicity was evaluated by LDH leakage into the incubation medium. Under these conditions, 20 and 30 microM Cd concentrations were cytotoxic. As compared to the non-Cd-exposed cells, the LDH release was elevated more than six-fold in cells exposed to 30 microM Cd for 24h. Co-treatment with 5-50mM glycine was cytoprotective in a concentration-dependent manner. Prior treatment with 50 mM glycine for 16 h, or co-treatment for 24h, reduced LDH leakage due to 30 microM Cd exposure by 60 and 66%, respectively. Co-incubation with 50 mM alanine was also protective but only about half as effective as with glycine. During the first 4h, prior to the onset of any significant cell membrane damage, the Cd-exposed cells accumulated 0.55 microg Cd/mg protein. Glycine pre-treatment or co-treatment reduced Cd accumulation by about one-quarter or one-half, respectively. To delineate the mechanism of glycine's effect on Cd accumulation, the efflux of Cd was studied after a 30 min pulse exposure. The results suggested that pre-treatment reduced Cd accumulation by increasing its efflux from the cells. In contrast, co-treatment reduced Cd efflux, suggesting that the co-treatment lowered Cd accumulation by suppressing its uptake. When co-incubated, Cd and glycine formed a complex that was apparently responsible for the marked reduction in Cd uptake. It is concluded that, regardless of the mode of treatment, glycine is cytoprotective against Cd and that it may do so by lowering the intracellular Cd burden.


Asunto(s)
Cloruro de Cadmio/toxicidad , Células Epiteliales/efectos de los fármacos , Glicinérgicos/farmacología , Glicina/farmacología , Túbulos Renales/efectos de los fármacos , Alanina/farmacología , Animales , Cloruro de Cadmio/metabolismo , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Túbulos Renales/citología , Túbulos Renales/metabolismo , Células LLC-PK1 , Porcinos
12.
Toxicology ; 224(1-2): 56-65, 2006 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-16730872

RESUMEN

Cadmium (Cd) has been reported to cause cell cycle arrest in various cell types by p53-dependent and -independent mechanisms. This study was designed to investigate cell cycle progression in kidney cells that are the target of chronic Cd toxicity. Rat renal proximal tubular epithelial cells, NRK-52E, were treated with up to 20 microM CdCl2 in DMEM containing 10% calf serum for up to 24 h. Flow cytometric analysis revealed time- and concentration-dependent increases in cells in G2/M phase of the cell cycle. As compared to the control cells, the cells exposed to 20 microM Cd showed a doubling of the number of cells in this phase after 24 h. The cell cycle arrest was associated with a decrease in protein levels of both cyclins A and B. Further investigation into the mechanism revealed that Cd treatment led to down-modulation of cyclin-dependent kinases, Cdk1 and Cdk2, apparently by elevating the expression of cyclin kinase inhibitors, KIP1/p27 and WAF1/p21. Furthermore, the wild-type p53 DNA-binding activity was up-regulated. Based on these observations, it appears that Cd causes G2/M phase arrest in NRK-52E cells via elevation of p53 activity, increasing the expression of cyclin kinase inhibitors p27 and p21, and decreasing the expression of cyclin-dependent kinases Cdk1 and 2, and of cyclins A and B.


Asunto(s)
Cadmio/farmacología , División Celular/efectos de los fármacos , Fase G2/efectos de los fármacos , Riñón/citología , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/farmacología , Citoplasma/metabolismo , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Riñón/efectos de los fármacos , Ratas , Proteína p53 Supresora de Tumor/metabolismo
13.
Toxicol Sci ; 91(1): 299-308, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16478757

RESUMEN

Renal epithelial cells undergo apoptosis upon exposure to cadmium (Cd). Transcription factors, such as nuclear factor-kappa B (NF-kappaB), mediate the expression of a number of genes involved in apoptosis. The present study was designed to examine the involvement of this transcription factor in Cd-induced apoptosis. Rat kidney proximal tubular epithelial cells, NRK-52E, were incubated with up to 20 microM CdCl2 in serum-free medium for 5 h followed by incubation in serum-containing medium (without Cd) for an additional 12 h. The cells accumulated 582 +/- 19 ng Cd/mg protein after 5-h exposure to 20 microM Cd. As a result of Cd exposure, the DNA-binding activity of the p65 subunit of NF-kappaB was decreased in a concentration- and time-dependent manner. The activity of tumor necrosis factor-alpha-induced inhibitor of kappa B (IkappaB) kinase alpha was also inhibited by Cd. In addition, the phosphorylation of IkappaB-alpha and NF-kappaB p65, as well as the levels of NF-kappaB target gene products, cIAP-1 and cIAP-2, were reduced. Pretreatment of the cells with the antioxidant U83836E or butylated hydroxytoluene preserved the DNA-binding activity and blocked the Cd-induced decease in IkappaB-alpha phosphorylation. Cd exposure caused the activation of caspase-3, -7, and -9 and DNA fragmentation. By flow cytometry, 14.6 and 30.5% apoptosis was detected at 6 and 12 h after stopping the Cd exposure. Overexpression of NF-kappaB p65 by transient transfection protected the cells from the Cd-induced apoptosis. Conversely, attenuation of NF-kappaB activity by pretreatment with SN50, an NF-kappaB nuclear translocation inhibitor, potentiated apoptosis. These results suggest that Cd-induced apoptosis involves suppression of NF-kappaB activity which may be mediated by oxidative stress.


Asunto(s)
Apoptosis/efectos de los fármacos , Cadmio/toxicidad , Riñón/efectos de los fármacos , FN-kappa B/metabolismo , Animales , Secuencia de Bases , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Citometría de Flujo , Riñón/citología , Riñón/metabolismo , Oligonucleótidos , Fosforilación , Ratas
14.
Biol Trace Elem Res ; 108(1-3): 17-31, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16327056

RESUMEN

The objectives of this study were to examine the association between urinary excretion of cadmium (U-Cd), copper (U-Cu), and zinc (U-Zn) and the severity of two different indicators of renal toxicity (urinary excretion of beta2-microglobulin [U-beta2-MG] and metallothionein [U-MT]) in Cd-exposed subjects compared to controls, and to assess the physiologic mechanisms by which the exposure to environmental Cd affects U-Cd, U-Cu, and U-Zn. The target population included 3508 Cd-exposed and 294 nonexposed participants who received a health survey conducted among the population of the Kakehashi River basin. Increases of U-Cd, U-beta2-MG, and U-MT in the Cd-exposed population were observed relative to excretion of these substances in controls. Regression analysis using a general linear model revealed that the correlations between U-Cd or U-Cu, and U-beta2-MG and between U-Cd, U-Cu or U-Zn, and U-MT were statistically significant in both sexes, but the correlation between U-Zn and U-beta2-MG excretion was significant only in men. These results suggest U-Cd and U-Cu is affected by dysfunction in renal tubular absorption (indicated by U-beta2-MG), whereas not only U-Cd and U-Cu but also U-Zn appear to be a function of renal cellular desquamation (indicated by U-MT).


Asunto(s)
Cadmio/farmacología , Cadmio/orina , Cobre/orina , Metalotioneína/orina , Zinc/orina , Microglobulina beta-2/orina , Anciano , Anciano de 80 o más Años , Exposición a Riesgos Ambientales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Contaminantes Químicos del Agua/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...