Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Vis ; 21: 1024-35, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26392741

RESUMEN

PURPOSE: The purpose of this study was to identify potential therapeutic strategies to slow down or prevent the expression of early-onset epithelial to mesenchymal transition (EMT) marker proteins (fibronectin and alpha smooth muscle actin, α-SMA) without sacrificing the synthesis and accumulation of the prosurvival protein vascular endothelial growth factor (VEGF) in cultured virally transformed human lens epithelial (HLE) cells. METHODS: HLE-B3 cells, maintained in a continuous hypoxic environment (1% oxygen), were treated with SB216763, a specific inhibitor of glycogen synthase kinase-3ß (GSK-3ß) catalytic activity. Western blot analysis was employed to detect the cytoplasmic and nuclear levels of ß-catenin, as well as the total lysate content of fibronectin and α-SMA. Enzyme-linked immunosorbent assay (ELISA) was used to measure the levels of VEGF in cell culture medium. A hypoxia-inducible factor-1α (HIF-1α) translation inhibitor and an HIF-2α translation inhibitor were independently employed to evaluate the effect of hypoxia inducible factor inhibition on EMT marker protein and VEGF expression. XAV932 was used to assess the suppression of nuclear ß-catenin and its downstream effect on EMT marker proteins and VEGF expression. RESULTS: SB216763-treated HLE-B3 cells caused marked inhibition of GSK-3ß activity prompting a significant increase in the translocation of cytoplasmic ß-catenin to the nucleus. The enhancement of nuclear ß-catenin looked as if it positively correlated with a significant increase in the basal expression of VEGF as well as increased expression of fibronectin and α-SMA. In conjunction with SB216763, coadministration of an HIF-1α translation inhibitor, but not an HIF-2α translation inhibitor, markedly suppressed the expression of fibronectin and α-SMA without affecting VEGF levels. Treatment with XAV932 significantly reduced the level of nuclear ß-catenin, but the levels of neither the EMT marker proteins nor VEGF were changed. CONCLUSIONS: Recently, we reported that nuclear ß-catenin, but not HIF-2α, regulates the expression of fibronectin and α-SMA in atmospheric oxygen. In marked contrast, data from the hypoxic condition clearly establish that nuclear ß-catenin plays little apparent role in the expression of EMT marker proteins. Instead, the loss of HIF-1α (but not HIF-2α) decreases the expression of the EMT marker proteins without sacrificing the levels of the prosurvival protein VEGF. These findings support the development of a potentially relevant therapeutic strategy to undermine the progression of normal cells to the mesenchymal phenotype in the naturally hypoxic lens without subverting cell viability.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Oxígeno/farmacología , Actinas/genética , Actinas/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia de la Célula , Línea Celular Transformada , Supervivencia Celular , Disulfuros/farmacología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Indoles/farmacología , Cristalino/efectos de los fármacos , Cristalino/metabolismo , Cristalino/patología , Maleimidas/farmacología , Biosíntesis de Proteínas , Transporte de Proteínas , Transducción de Señal , Sulfonamidas/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/antagonistas & inhibidores , beta Catenina/genética , beta Catenina/metabolismo
2.
Mol Vis ; 20: 1758-75, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25593505

RESUMEN

PURPOSE: The inhibition of GSK-3ß blocks mitochondrial membrane permeability transition (mMPT) for HLE-B3 cells in atmospheric oxygen. GSK-3ß, as part of a multifactorial complex, also regulates nuclear levels of ß-catenin, a known coordinator of cell survival and adhesion. The purpose of these studies was to demonstrate a novel, but likely disadvantageous, link between ß-catenin's influence on the expression of the pro-survival protein, vascular endothelial growth factor (VEGF), resulting in enhanced lens epithelial cell mitochondrial protection against depolarization and nuclear ß-catenin as an inducer of epithelial to mesenchymal transition (EMT). METHODS: Virally transformed human lens epithelial cells (HLE-B3) were treated with SB216763, a specific inhibitor of GSK-3ß catalytic activity and XAV939, a specific ß-catenin inhibitor that bars the translocation of ß-catenin from cytoplasm to the nucleus. Western blot analysis was employed to detect the levels of cytoplasmic and nuclear ß-catenin and phospho-ß-catenin, pBcl-2 and the EMT proteins, α-smooth muscle actin (α-SMA), and fibronectin. ELISA was used to measure the levels of VEGF in cell culture supernatants. JC-1 analysis was performed to analyze the influence of either SB216763 or XAV939 on mitochondrial depolarization. RESULTS: Cultured lens epithelial cells maintained in hypoxia (1% oxygen) and subsequently reintroduced into atmospheric oxygen and treated with the GSK-3ß inhibitor SB216763 illustrated a marked inhibition of phosphorylation of glycogen synthase (downstream substrate of GSK-3ß) and significant increase in nuclear translocation of ß-catenin. The augmented nuclear ß-catenin levels positively correlated with increased expression of α-SMA and fibronectin, both marker proteins indicative of EMT. The enhanced nuclear ß-catenin activity also elicited increased VEGF and pBcl-2 expression, resulting in increased resistance to mitochondrial depolarization. Treatment of the cells with the ß-catenin inhibitor XAV939 resulted in decreased expression of nuclear ß-catenin, VEGF levels, pBcl-2, and EMT proteins, as well as increased mitochondrial depolarization. CONCLUSIONS: The data support a model whereby the onset of epithelial to mesenchymal transition may circuitously benefit from the enhanced synthesis of VEGF by setting up a potentially harmful situation whereby the resulting mesenchymal cell population may be more resistant to mitochondrial depolarization than the lens epithelial cell population from which it originated. These findings support the potential therapeutic relevance of developing strategies to undermine the progression of normal cells to mesenchymal transition without subverting cell viability.


Asunto(s)
Transición Epitelial-Mesenquimal , Glucógeno Sintasa Quinasa 3/metabolismo , Cristalino/metabolismo , Cristalino/patología , Potencial de la Membrana Mitocondrial , Animales , Bovinos , Hipoxia de la Célula , Línea Celular , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Indoles/farmacología , Cristalino/efectos de los fármacos , Maleimidas/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Modelos Biológicos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/metabolismo
3.
Mol Vis ; 19: 2451-67, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24319338

RESUMEN

PURPOSE: Loss of integrity of either the inner or outer mitochondrial membrane results in the dissipation of the mitochondrial electrochemical gradient that leads to mitochondrial membrane permeability transition (mMPT). This study emphasizes the role of glycogen synthase kinase 3beta (GSK-3ß) in maintaining mitochondrial membrane potential, thus preventing mitochondrial depolarization (hereafter termed mitoprotection). Using 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione (SB216763), an inhibitor of GSK-3ß, and drawing a distinction between it and 1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio] butadiene (UO126), an inhibitor of extracellular-signal-regulated kinase (ERK) phosphorylation, the means by which GSK-3ß influences mitoprotection in cultured human lens epithelial (HLE-B3) cells and normal, secondary cultures of bovine lens epithelial cells, maintained in atmospheric oxygen, was investigated. METHODS: Virally transfected human lens epithelial cells (HLE-B3) and normal cultures of bovine lens epithelial cells were exposed to acute hypoxic conditions (about 1% O2) followed by exposure to atmospheric oxygen (about 21% O2). Specific antisera and western blot analysis was used to examine the state of phosphorylation of ERK and GSK-3ß, as well as the phosphorylation of a downstream substrate of GSK-3ß, glycogen synthase (GS, useful in monitoring GSK-3ß activity). The potentiometric dye, 1H-benzimidazolium-5,6-dichloro-2-[3-(5,6-dichloro-1,3-diethyl-1,3-dihydro-2H-benzimidazol-2-ylidene)-1-propenyl]-1,3-diethyl-iodide (JC-1), was used to monitor mitochondrial depolarization upon exposure of inhibitor treatment relative to the control cells (mock inhibition) in atmospheric oxygen. Caspase-3 activation was scrutinized to determine whether mitochondrial depolarization inevitably leads to apoptosis. RESULTS: Treatment of HLE-B3 cells with SB216763 (12 µM) inactivated GSK-3ß activity as verified by the enzyme's inability to phosphorylate its substrate, GS. SB216763-treated cells were not depolarized relative to the control cells as demonstrated with JC-1 fluorescent dye analysis. The HLE-B3 cells treated with UO126, which similarly blocked phosphorylation of GS, were nevertheless prone to mMPT relative to the control cells. Western blot analysis determined that Bcl-2-associated X (BAX) levels were unchanged for SB216763-treated or UO126-treated HLE-B3 cells when compared to their respective control cells. However, unlike the SB216763-treated cells, the UO126-treated cells showed a marked absence of Bcl-2, as well as phosphorylated Bcl-2 relative to the controls. UO126 treatment of bovine lens epithelial cells showed similar results with pBcl-2 levels, while the Bcl-2 content appeared unchanged relative to the control cells. HLE-B3 and normal bovine lens cell cultures showed susceptibility to mMPT associated with the loss of pBcl-2 by UO126 treatment. CONCLUSIONS: MITOCHONDRIAL DEPOLARIZATION MAY OCCUR BY ONE OF TWO KEY OCCURRENCES: interruption of the electrochemical gradient across the inner mitochondrial membrane resulting in mMPT or by disruption of the integrity of the inner or outer mitochondrial membrane. The latter scenario is generally tightly regulated by members of the Bcl-2 family of proteins. Inhibition of GSK-3ß activity by SB216763 blocks mMPT by preventing the opening of the mitochondrial permeability transition pore. UO126, likewise, inhibits GSK-3ß activity, but unlike SB216763, inhibition of ERK phosphorylation induces the loss of intracellular pBcl-2 levels under conditions where intracellular BAX levels remain constant. These results suggest that the lenticular mitoprotection normally afforded by the inactivation of GSK-3ß activity may, however, be bypassed by a loss of pBcl-2, an anti-apoptotic member of the Bcl-2 family. Bcl-2 prevents the translocation of BAX to the mitochondrial outer membrane inhibiting depolarization by disrupting the normal electrochemical gradient leading to mMPT.


Asunto(s)
Células Epiteliales/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Cristalino/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Oxígeno/metabolismo , Animales , Bencimidazoles , Butadienos/farmacología , Carbocianinas , Bovinos , Hipoxia de la Célula/genética , Línea Celular , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Colorantes Fluorescentes , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Indoles/farmacología , Cristalino/citología , Cristalino/efectos de los fármacos , Maleimidas/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nitrilos/farmacología , Oxígeno/farmacología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína Letal Asociada a bcl/genética , Proteína Letal Asociada a bcl/metabolismo
4.
Mol Vis ; 19: 1-15, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23335846

RESUMEN

PURPOSE: The prosurvival signaling cascades that mediate the unique ability of human lens epithelial cells to survive in their naturally hypoxic environment are not well defined. Hypoxia induces the synthesis of the hypoxia inducible factor HIF-1α that in turn, plays a crucial role in modulating a downstream survival scheme, where vascular endothelial growth factor (VEGF) also plays a major role. To date, no published reports in the lens literature attest to the expression and functionality of HIF-2α and the role it might play in regulating VEGF expression. The aim of this study was to identify the functional expression of the hypoxia inducible factors HIF-1α and HIF-2α and establish their role in regulating VEGF expression. Furthermore, we demonstrate a link between sustained VEGF expression and the ability of the hypoxic human lens epithelial cell to thrive in low oxygen conditions and resist mitochondrial membrane permeability transition (also referred to as lenticular cytoprotection). METHODS: Hypoxia inducible factor translation inhibitors were used to demonstrate the role of HIF-1α and HIF-2α and the simultaneous expression of both hypoxic inducible factors to determine their role in regulating VEGF expression. Axitinib, which inhibits lenticular cell autophosphorylation of its VEGF receptor, was employed to demonstrate a role for the VEGF-VEGFR2 receptor complex in regulating Bcl-2 expression. Specific antisera and western blot analysis were used to detect the protein levels of HIF-1α and HIF-2α, as well as the proapoptotic protein, BAX and the prosurvival protein, Bcl-2. VEGF levels were analyzed with enzyme-linked immunosorbent assay (ELISA). The potentiometric dye, 5,5',6,6'-tetrachloro1,1',3,3'-tetraethyl-benzimidazolylcarbocyanine iodide, was used to determine the effect of the inhibitors on mitochondrial membrane permeability transition. RESULTS: Cultured human lens epithelial cells (HLE-B3) maintained under hypoxic condition (1% oxygen) displayed consistent accumulation of VEGF throughout the 72 h incubation period. Using hypoxia inducible factor translation inhibitors targeting HIF-1α or HIF-2α, the specific inhibition of each protein did not diminish VEGF synthesis. The combined inhibition of HIF-1α and HIF-2α expression, using a double hypoxia inducible factor translation inhibitor, markedly decreased the level of VEGF. The inhibition of VEGF synthesis was associated with a profound deficiency in the level of the prosurvival protein, Bcl-2. Axitinib also prevented the VEGF-mediated expression of Bcl-2. The loss of VEGF coupled with the decrease in intracellular Bcl-2 correlated with marked mitochondrial depolarization, an early predictor of cellular apoptosis. CONCLUSIONS: Our data support a model in which the sustained synthesis of VEGF in human lens epithelial cells, maintained under hypoxic condition, is regulated by a compensatory inter-relationship between HIF-1α and HIF-2α. VEGF acts as a prosurvival factor in hypoxic lens epithelial cells by maintaining consistent expression of the prosurvival protein Bcl-2, which likely prevents the translocation of cytosolic BAX to the outer mitochondrial membrane, thus preventing the initiation of mitochondrial depolarization.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Cristalino/citología , Cristalino/metabolismo , Modelos Moleculares , Axitinib , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Hipoxia de la Célula/fisiología , Línea Celular , Supervivencia Celular , Células Epiteliales/citología , Células Epiteliales/enzimología , Células Epiteliales/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Imidazoles/farmacología , Indazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal , Topotecan/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Mol Vis ; 19: 1406-12, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825920

RESUMEN

PURPOSE: Dissipation of the electrochemical gradient across the inner mitochondrial membrane results in mitochondrial membrane permeability transition (mMPT), a potential early marker for the onset of apoptosis. In this study, we demonstrate a role for glycogen synthase kinase-3ß (GSK-3ß) in regulating mMPT. Using direct inhibition of GSK-3ß with the GSK-3ß inhibitor SB216763, mitochondria may be prevented from depolarizing (hereafter referred to as mitoprotection). Cells treated with SB216763 showed an artifact of fluorescence similar to the green emission spectrum of the JC-1 dye. We demonstrate the novel use of spectral deconvolution to negate the interfering contributing fluorescence by SB216763, thus allowing an unfettered analysis of the JC-1 dye to determine the mitochondrial membrane potential. METHODS: Secondary cultures of virally transfected human lens epithelial cells (HLE-B3) were exposed to acute hypoxic conditions (approximately 1% O2) followed by exposure to atmospheric oxygen (approximately 21% O2). The fluorescent dye JC-1 was used to monitor the extent of mitochondrial depolarization upon exposure of inhibitor treatment relative to the control cells (mock inhibition) in atmospheric oxygen. Annexin V-fluorescein isothiocyanate/propidium iodide staining was implemented to determine cell viability. RESULTS: Treatment of HLE-B3 cells with SB216763 (12 µM), when challenged by oxidative stress, suppressed mitochondrial depolarization relative to control cells as demonstrated with JC-1 fluorescent dye analysis. Neither the control nor the SB216763-treated HLE-B3 cells tested positive with annexin V-fluorescein isothiocyanate/propidium iodide staining under the conditions of the experiment. CONCLUSIONS: Inhibition of GSK-3ß activity by SB216763 blocked mMPT relative to the slow but consistent depolarization observed with the control cells. We conclude that inhibition of GSK-3ß activity by the GSK-3ß inhibitor SB216763 provides positive protection against mitochondrial depolarization.


Asunto(s)
Bencimidazoles/farmacología , Carbocianinas/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Indoles/farmacología , Cristalino/metabolismo , Maleimidas/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Artefactos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Fluoresceína-5-Isotiocianato/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Cristalino/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Propidio/metabolismo , Espectrometría de Fluorescencia
6.
Biochim Biophys Acta ; 1793(10): 1540-70, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19559056

RESUMEN

There has been increasing evidence pointing to the mitochondrial respiratory chain (MRC) as a novel and important target for the actions of 17beta-estradiol (E(2)) and estrogen receptors (ER) in a number of cell types and tissues that have high demands for mitochondrial energy metabolism. This novel E(2)-mediated mitochondrial pathway involves the cooperation of both nuclear and mitochondrial ERalpha and ERbeta and their co-activators on the coordinate regulation of both nuclear DNA- and mitochondrial DNA-encoded genes for MRC proteins. In this paper, we have: 1) comprehensively reviewed studies that reveal a novel role of estrogens and ERs in the regulation of MRC biogenesis; 2) discussed their physiological, pathological and pharmacological implications in the control of cell proliferation and apoptosis in relation to estrogen-mediated carcinogenesis, anti-cancer drug resistance in human breast cancer cells, neuroprotection for Alzheimer's disease and Parkinson's disease in brain, cardiovascular protection in human heart and their beneficial effects in lens physiology related to cataract in the eye; and 3) pointed out new research directions to address the key questions in this important and newly emerging area. We also suggest a novel conceptual approach that will contribute to innovative regimens for the prevention or treatment of a wide variety of medical complications based on E(2)/ER-mediated MRC biogenesis pathway.


Asunto(s)
Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Estrógenos/fisiología , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Receptores de Estrógenos/fisiología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Animales , Apoptosis/genética , Apoptosis/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/etiología , Enfermedades Cardiovasculares/prevención & control , Proliferación Celular , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Resistencia a Antineoplásicos , Transporte de Electrón/genética , Estradiol/farmacología , Estradiol/fisiología , Femenino , Genoma Mitocondrial , Humanos , Cristalino/efectos de los fármacos , Cristalino/fisiología , Masculino , Mitocondrias/genética , Proteínas Mitocondriales/fisiología , ATPasas de Translocación de Protón Mitocondriales/fisiología , Modelos Biológicos , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/etiología , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/etiología , Biosíntesis de Proteínas/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
7.
Mol Vis ; 16: 630-8, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20700369

RESUMEN

PURPOSE: The family of natriuretic peptides (NPs); atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP) as well as three associated receptors (NPRs); natriuretic peptide receptor A (NPR-A), natriuretic peptide receptor B (NPR-B), and natriuretic peptide receptor C (NPR-C) has never been documented in human lens epithelial cells. The study described herein was designed to demonstrate both expression and functionality of components of the natriuretic peptides and natriuretic peptide receptors in the human lens epithelial cell line, HLE-B3 and in normal human lens epithelial cell cultures (nHLE). METHODS: Reverse transcriptase-polymerase chain reaction (RT-PCR) along with confirmation by DNA sequencing and real-time quantitative RT-PCR was used to identify and demonstrate expression of mRNA for the natriuretic peptide family. Authentication of protein expression of the natriuretic peptide receptors was determined by using formaldehyde-fixed, Saponin-permeabilized cells (HLE-B3) or methanol:acetone-fixed and permeabilized cells (nHLE) using conventional immunofluorescence techniques. Enzyme-linked immunosorbent assay was used to determine cyclic GMP (cGMP) activity as stimulated by exogenous addition of natriuretic peptides. RESULTS: Using RT-PCR with confirmation by DNA sequencing and real-time quantitative RT-PCR, HLE-B3 cells were shown to express mRNA for ANP, BNP, and CNP along with their associated receptors. Conventional immunofluorescence on the permeabilized cells confirmed positive diffuse staining indicating the presence of the three natriuretic peptide receptors in both HLE-B3 and nHLE cells. All three natriuretic peptides educe a cGMP response in the rank order CNP>>ANP approximately BNP indicating that the natriuretic peptide family is functional in HLE-B3 cells. CONCLUSIONS: The data indicates that ANP, BNP, and CNP and natriuretic peptide receptor transcripts are expressed and are functional in human lens epithelial cells. The cellular expression of NPs and NPRs, as well as the demonstration that all three NPs activate guanylyl cyclase suggests a potential role in maintaining lens epithelial cell homeostasis.


Asunto(s)
Células Epiteliales/metabolismo , Cristalino/citología , Péptidos Natriuréticos/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Bioensayo , Línea Celular , GMP Cíclico/biosíntesis , Células Epiteliales/enzimología , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , Péptido Natriurético Tipo-C/genética , Péptido Natriurético Tipo-C/metabolismo , Péptidos Natriuréticos/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Factor Natriurético Atrial/genética , Receptores del Factor Natriurético Atrial/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/genética
8.
Mol Vis ; 14: 898-905, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18490963

RESUMEN

PURPOSE: 17 beta-estradiol (17beta-E(2)) protects human lens epithelial cells against oxidative stress by preserving mitochondrial function in part via the non-genomic rapid activation of prosurvival signal transduction pathways. The study described herein examined whether 17beta-E(2) also elicits genomic protection by influencing the expression (and activity) of mitochondrial-associated manganese superoxide dismutase (MnSOD) as a possible parallel mechanism by which 17beta-E(2) protects against oxidative stress. METHODS: Virally-transformed human lens epithelial cells (HLE-B3) were pre-incubated with 17beta-E(2), and mRNA or protein lysates were collected over a time course ranging from 90 min to 24 h. Positive expression of lens epithelial cell MnSOD mRNA was determined by semi-quantitative reverse transcriptase polymerase chain reaction (RT-PCR), and its levels were monitored by real-time PCR up to 24 h after 17beta-E(2) administration. Western blot analysis was used to examine the pattern of protein expression as influenced by 17beta-E(2) treatment. MnSOD activity as influenced by 17beta-E(2) was determined by measuring enzymatic activity. RESULTS: A significant rapid increase in the activity of MnSOD was observed with HLE-B3 cells by 90 min post-bolus addition of 17beta-E(2), which returned to control level by 240 min. Neither an increase in MnSOD mRNA nor in protein expression was detected up through 24 h. CONCLUSIONS: These data demonstrate that 17beta-E(2) rapidly and transiently increases the activity of MnSOD but influences neither its mRNA expression nor its protein expression. The results suggest that (estrogen-activated) MnSOD plays an important role against mitochondrial oxidative stress by diminishing reactive oxygen species, thus promoting cell survival.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Estradiol/farmacología , Cristalino/citología , Cristalino/enzimología , Mitocondrias/enzimología , Superóxido Dismutasa/metabolismo , Western Blotting , Línea Celular , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Cristalino/efectos de los fármacos , Mitocondrias/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/genética , Regulación hacia Arriba/efectos de los fármacos
9.
Mol Vis ; 12: 271-82, 2006 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-16617294

RESUMEN

PURPOSE: This study examined the state of mitochondrial physiology subsequent to exposing lens epithelium to high ambient galactose (Gal), which upon conversion to galactitol (GalOH) and resultant intracellular accumulation thereof, leads to profound destabilization of mitochondrial membrane potential (Deltapsim). Further, we determined whether the aldose reductase (AR) inhibitor, Sorbinil, or estrogen (17beta-E2, and its isomer, 17alpha-E2, which exhibits marginal binding affinity for estrogen receptor), administered prior to and concomitant with Gal exposure might prevent or delay mitochondrial membrane depolarization. METHODS: Secondary cultures of bovine lens epithelial cells (BLECs), as well as a virally-transformed human lens epithelial cell line (HLE-B3), were maintained in 40 mM galactose (Gal) for up to seven days in the presence and absence of Sorbinil, 17beta-E2 or 17alpha-E2. Endogenous accumulation of reactive oxygen species (ROS) was assessed by loading cells with H2DCF-DA, which upon oxidation in the presence of ROS transitions to the fluorescent compound, DCF. To assess Deltapsim, confocal microscopy was employed in conjunction with the potentiometric dye, JC-1. Intracellular polyol content was determined by gas chromatography. Cells were monitored for apoptosis and necrosis as determined by annexin V-propidium iodide staining and visualized by confocal fluorescence microscopy. RESULTS: BLECs, more so than HLE-B3 cells, accumulate high intracellular levels of GalOH upon exposure to high ambient Gal. BLECs were significantly depolarized while HLE-B3 cells showed little depolarization over the same course of Gal exposure. The addition of either 17alpha-E2 or 17beta-E2 to BLECs, over a dose range of 0.01 microM to 1.0 microM, prevented mitochondrial membrane depolarization as did the addition of 0.1 mM Sorbinil. The polyol content in BLECs after 3 days of exposure to Gal was 282 nmol/mg protein. Co-addition of Sorbinil during the 3-day exposure period prevented any significant accumulation of GalOH. Co-administration of either isoform of estrogen did not block GalOH synthesis and the level of attained intracellular accumulation was similar to that of Gal alone. The observed accumulation of ROS from HLE-B3 cells subsequent to 3 days of Gal exposure was negligible and consistent with that of control cells maintained in physiological medium. Intracellular accumulation of ROS with 3-day, Gal-maintained BLECs, exhibited a marginal but statistically significant increase over control cells maintained in physiological medium (5.5 mM glucose) and similar levels of ROS were generated irrespective of the presence of estrogen with Gal. Bolus addition of 100 microM hydrogen peroxide to 3-day, Gal plus Sorbinil-maintained BLECs failed to induce a change in mitochondrial membrane potential. Evidence of apoptosis or necrosis was negligible through 7 days of sustained exposure to high ambient Gal. CONCLUSIONS: Polyol accumulation promotes mitochondrial membrane depolarization and the decrease in Deltapsim is prevented by prior addition and co-administration of Sorbinil or estrogen with Gal. Unlike Sorbinil, estrogens' mode of action is not via the inhibition of aldose reductase activity. The data supports the theory that with Gal plus estradiol-treated cells, at a given intracellular polyol load, a larger portion of the mitochondrial population retains Deltapsim, and hence continues to function relative to Gal-treated cells. Results with 17alpha-E2 indicate that maintaining Deltapsim, in the face of chronic polyol accumulation, is likely to be mediated via a nuclear estrogen receptor-independent mechanism. The failure of supraphysiological levels of hydrogen peroxide added to Gal plus Sorbinil-maintained BLECs to depolarize mitochondria indicates that polyol accumulation, not ROS generation, is the causative factor responsible for the loss of mitochondrial membrane potential.


Asunto(s)
Estradiol/farmacología , Cristalino/efectos de los fármacos , Cristalino/fisiología , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Polímeros/metabolismo , Aldehído Reductasa/antagonistas & inhibidores , Animales , Apoptosis , Bencimidazoles , Carbocianinas , Bovinos , Células Cultivadas , Sinergismo Farmacológico , Electrofisiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Colorantes Fluorescentes , Galactosa/farmacología , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/fisiología , Cristalino/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Microscopía Confocal , Especies Reactivas de Oxígeno/metabolismo
10.
Mitochondrion ; 5(4): 235-47, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16050986

RESUMEN

17beta-estradiol (17beta-E2) protects against H2O2-mediated depletion of intracellular ATP and lessens the degree of depolarization of mitochondrial membrane potential (DeltaPsi(m)) in cultured lens epithelial cells consequential to oxidative insult. We now report that 17beta-E2 acts as a positive regulator of the survival signal transduction pathway, MAPK which, in turn, acts to stabilize DeltaPsi(m) in effect, attenuating the extent of depolarization of mitochondrial membrane potential in the face of acute oxidative stress. The SV-40 viral transformed human cell line, HLE-B3 was treated with 17beta-E2 over a time course of 60 min and phosphorylation of ERK1/2 was analyzed by Western blot. ERK1/2 was phosphorylated within 5-15 min in the presence of 17beta-E2. Cell cultures were exposed to the MEK1/2 inhibitor, UO126, subsequent to H2O2+/-17beta-E2 treatment and the DeltaPsi(m) examined using JC-1, a potentiometric dye which serves as an indicator for the state of mitochondrial membrane potential. UO126 treatment attenuated ERK1/2 phosphorylation irrespective of whether estradiol was administered. Mitochondrial membrane depolarization resulting from H2O2 stress was substantially greater in the presence of UO126. The greater the extent of depolarization, the less effective 17beta-E2 treatment was in checking mitochondrial membrane depolarization, indicating that the relative degree of ERK phosphorylation influences mitochondrial stability with oxidative insult. The data support a positive correlation between 17beta-E2 stimulation of ERK1/2 phosphorylation and mitochondrial stabilization that would otherwise cause a complete collapse of DeltaPsi(m).


Asunto(s)
Estradiol/farmacología , Cristalino/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/fisiología , Estrés Oxidativo , Animales , Butadienos/farmacología , Proteínas Portadoras/metabolismo , Bovinos , Línea Celular Transformada , Citoprotección/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Epiteliales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Cristalino/efectos de los fármacos , Microscopía Confocal , Mitocondrias/efectos de los fármacos , Nitrilos/farmacología , Fosforilación , Proteína Letal Asociada a bcl
11.
Invest Ophthalmol Vis Sci ; 44(5): 2067-75, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12714645

RESUMEN

PURPOSE: Epidemiologic studies demonstrate a higher incidence of cataracts in estrogen-deprived postmenopausal women, but the mechanism for the increased risk of cataracts is unclear. An elevated level of H(2)O(2) in aqueous humor and whole lenses has been associated with cataractogenesis. In the present study, for the first time, the protective effect of estrogens against oxidative stress were tested in cultured human lens epithelial cells (HLECs). METHODS: To investigate the involvement of 17beta-estradiol (17beta-E(2)) in protection against oxidative stress, HLECs were exposed to insult with H(2)O(2) at a physiological level (100 microM) over a time course of several hours, with and without pretreatment with 17beta-E(2). Cell viability was measured by calcein AM assay, and 2',7'-dichlorofluorescein diacetate (DCFH-DA) was used to determine intracellular reactive oxygen species (ROS). Intracellular adenosine triphosphate (ATP) level was quantified with a luciferin- and luciferase-based assay and mitochondrial potential (deltapsi(m)) was monitored by a fluorescence resonance energy-transfer technique. RESULTS: H(2)O(2) caused a dose-dependent decrease in mitochondrial membrane potential, intracellular ATP levels, and cell viability. Dose-dependent increases in cell viability and intracellular ATP level were observed with pretreatment of 17beta-E(2) for 2 hours before oxidative insult. At 1 nM, 17beta-E(2) increased cell viability from 39% +/- 4% to 75% +/- 3%, and at 100 nM or higher, it increased survival to greater than 95%. The level of intracellular ATP approached normal with 17beta-E(2) at 100 nM or higher. Pretreatment with 17beta-E(2) did not diminish intracellular ROS accumulation after exposure to H(2)O(2). Moreover, two nonfeminizing estrogens, 17alpha-E(2) and ent-E(2), both of which do not bind to either estrogen receptor alpha or beta, were as effective as 17beta-E(2) in the recovery of cell viability. The estrogen receptor antagonist, ICI 182,780, did not block protection by 17beta-E(2). Both 17beta- and 17alpha-E(2) moderated the collapse of deltapsi(m) in response to either H(2)O(2) or excessive Ca(2+) loading. CONCLUSIONS: The present study indicates that both 17alpha- and 17beta-E(2) can preserve mitochondrial function, cell viability, and ATP levels in human lens cells during oxidative stress. Although the precise mechanism responsible for protection by the estradiols against oxidative stress remains to be determined, the ability of nonfeminizing estrogens, which do not bind to estrogen receptors, to protect against H(2)O(2) toxicity indicates that this conservation is not likely to be mediated through classic estrogen receptors.


Asunto(s)
Adenosina Trifosfato/metabolismo , Citoprotección/efectos de los fármacos , Células Epiteliales/citología , Estradiol/análogos & derivados , Estradiol/farmacología , Cristalino/citología , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/fisiología , Estrés Oxidativo , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Antagonistas de Estrógenos/farmacología , Fluoresceínas , Fulvestrant , Humanos , Peróxido de Hidrógeno/toxicidad , Cristalino/efectos de los fármacos , Cristalino/metabolismo , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
12.
Invest Ophthalmol Vis Sci ; 43(2): 425-33, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11818387

RESUMEN

PURPOSE: Comparative assessment of cultured human lens epithelial cells (HLECs) and bovine lens epithelial cells (BLECs) established the nature of the relationship between taurine-concentrating capability and intracellular polyol accumulation or extracellular hypertonicity. METHODS: The kinetic characteristics of active taurine accumulation based on the measurement of in vitro [3H]-taurine uptake were resolved by side-to-side review of cultured HLECs and BLECs pre-exposed to either galactose-supplemented medium or extracellular hypertonicity. Competitive RT-PCR was used to appraise variation in taurine transporter (TauT) mRNA abundance from cells maintained in hyperosmotic medium over a 72-hour exposure period. RESULTS: The capacity to accumulate [3H]-taurine was significantly lowered after prolonged (20-hour) incubation of cultured BLECs in 40 mM galactose in contrast to HLECs, the latter cells' velocity curve being indistinguishable from control cells in physiological medium. Inhibition of the intracellular taurine transport site appeared to be noncompetitive, in that there was a marked reduction in the V(max) without significant alteration in the K(m) to a high-affinity transport site. Galactitol content in BLECs exceeded five times that found in HLECs. The coadministration of the aldose reductase inhibitor, sorbinil, with 40 mM galactose completely prevented the inhibitory effect of galactose on [3H]-taurine uptake. Acute exposure (3 hours) of HLECs and BLECs to a range of 10 to 40 mM galactitol or 10 to 40 mM galactose plus sorbinil-supplemented medium suggested by Dixon plot that neither galactitol nor galactose interacted with the extracellular taurine transport site. In contrast, [3H]-taurine accumulation was markedly elevated in both HLECs and BLECs after prolonged exposure to galactose-free medium made hyperosmotic by supplementation with sodium chloride. The enhanced taurine uptake capacity involved increase in peak velocity (V(max)) without significant change in Michaelis-Menten constant (K(m)). Cultured HLECs and BLECs responded to hypertonicity with an inducible but transitory upregulation of TauT mRNA. CONCLUSIONS: These results demonstrate that lens epithelial cells express a high-affinity TauT protein capable of active uptake, but predisposed to inhibition by intracellular galactitol when the sugar alcohol is present in sufficiently high concentration to interfere with cell metabolism. Furthermore, lens epithelial cells respond to hypertonic stress by raising taurine transport activity. The increase in taurine uptake is due to an increase in the number of high-affinity TauTs expressed as a result of an increase in the manifestation of taurine mRNA stemming from exposure to hypertonic medium.


Asunto(s)
Células Epiteliales/metabolismo , Imidazolidinas , Cristalino/metabolismo , Proteínas de Transporte de Membrana , Taurina/metabolismo , Equilibrio Hidroelectrolítico , Aldehído Reductasa/antagonistas & inhibidores , Animales , Transporte Biológico/efectos de los fármacos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Bovinos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Galactitol/farmacología , Galactosa/farmacología , Humanos , Imidazoles/farmacología , Cristalino/efectos de los fármacos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
13.
Am J Physiol Endocrinol Metab ; 294(3): E589-99, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18171912

RESUMEN

17beta-Estradiol (E(2)) reduces oxidative stress-induced depolarization of mitochondrial membrane potential (MMP) in cultured human lens epithelial cells (HLE-B3). The mechanism by which the nongenomic effects of E(2) contributed to the protection against mitochondrial membrane depolarization was investigated. Mitochondrial membrane integrity is regulated by phosphorylation of BAD, and it is known that phosphorylation of Ser(112) inactivates BAD and prevents its participation in the mitochondrial death pathway. We found that E(2) rapidly increased both the phosphorylation of ERK2 and Ser(112) in BAD. Ser(112) is phosphorylated by p90 ribosomal S6 kinase (RSK), a Ser/Thr kinase, which is a downstream effector of ERK1/2. Inhibition of RSK by the RSK-specific inhibitor SL0101 did not reduce the level of E(2)-induced phosphorylation of Ser(112). Silencing BAD using small interfering RNA did not alter mitochondrial membrane depolarization elicited by peroxide insult. However, under the same conditions, silencing ERK2 dramatically increased membrane depolarization compared with the control small interfering RNA. Therefore, ERK2, functioning through a BAD-independent mechanism regulates MMP in humans lens epithelial cells. We propose that estrogen-induced activation of ERK2 acts to protect cells from acute oxidative stress. Moreover, despite the fact that ERK2 plays a regulatory role in mitochondrial membrane potential, estrogen was found to block mitochondrial membrane depolarization via an ERK-independent mechanism.


Asunto(s)
Estradiol/farmacología , Cristalino/ultraestructura , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Células Epiteliales/ultraestructura , Humanos , Fosforilación/efectos de los fármacos , Serina/metabolismo , Proteína Letal Asociada a bcl/metabolismo
14.
Exp Eye Res ; 81(2): 165-75, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16080910

RESUMEN

A number of variants of the wild-type (wt) estrogen receptor beta (ERbeta-1) coexist in a wide range of tissues. In the human these include, together with others, the expression of several isoforms (ERbeta-2-ERbeta-5) due to alternative splicing of exons encoding the carboxy terminus. In this study, we determined whether virally transformed cell cultures of human lens epithelial cells (HLE-B3) express both full length (or wt) and variant isoforms of ERbeta in comparison to normal secondary cultures of human lens epithelial cells (nHLE) and furthermore, identify the subcellular localization of the wtERbeta-1 and ERbeta isoform variants in HLE-B3 and nHLE cells, as well as from human breast adenocarcinoma cells (MCF-7) which provided a positive control. ERbeta isoform mRNA expression was evaluated by coupled RT-PCR. Subcellular localization of ERbeta isoforms was determined on formaldehyde-fixed, Saponin-permeabilized cells using conventional immunofluorescence techniques and affinity purified polyclonal antibodies specific for ERbeta-1 as well as to two of the truncated carboxy terminus isoforms (beta-2 and beta-5). Total RNA was extracted from HLE-B3 and nHLE cells and lens tissue, as well as from human breast adenocarcinoma cells (MCF-7) and subjected to RT-PCR using specific estrogen receptor primers intended to distinguish ERbeta-1-ERbeta-5 mRNA. The PCR products corresponded to wtERbeta-1 as well as to the isoform variants beta-2 and beta-5. The proportional distribution of wtERbeta-1, beta-2 and beta-5 PCR products differed between the normal lens epithelial cells and the SV-40 transformed lens epithelial cell line; the nHLE being similar to lens tissue with respect to relative expression of ERbeta isoform cDNAs. Confocal microscopy and immunofluorescence revealed ERbeta-2 was distributed throughout the cytosol and was associated with the nucleus of all cells examined, although sporadic immunostaining was observed with the nuclei of MCF-7. Prominent immunostaining of ERbeta-1 appeared in the mitochondria (along with weaker staining in the nucleus) of all cell types as authenticated by co-localization with Mitotrack-633. ERbeta-5 immunostaining was diffuse in the cytosol and also associated with the nuclei of all cell types. The differential subcellular partitioning of ERbeta-1 to the mitochondria and ERbeta-2 to the nucleus suggests a new aspect of regulation and function of the estrogen signalling system.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Cristalino/metabolismo , Núcleo Celular/metabolismo , Transformación Celular Viral , Células Cultivadas , Células Epiteliales/metabolismo , Receptor beta de Estrógeno/genética , Humanos , Microscopía Confocal , Mitocondrias/metabolismo , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
15.
Exp Eye Res ; 78(5): 933-44, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15051475

RESUMEN

It has been demonstrated that estrogens are potent antioxidants and protect against H2O2-mediated depletion of intracellular ATP in human lens epithelial cells (HLE-B3) [Invest. Ophthalmol. Vis. Sci. 44 (2003) 2067]. To investigate the mechanism by which 17beta-estradiol (17beta-E2) protects against oxidative stress, HLE-B3 cells were exposed to insult with H2O2 at physiological (50 microm) and moderately supra- physiological (100 microm) levels over a time course of several hours, with and without pretreatment with 17beta-E2. The ability of 17beta-E2 to prevent H2O2-induced injury to several oxidant susceptible components of the cellular ATP generating machinery, including abundances of mitochondrial gene transcripts encoding respiratory chain subunits and cytochrome c, the glycolytic pathway enzyme, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and the energy-shuttling creatine kinase (CK) system, and mitochondrial membrane potential (deltapsi(m)) a measure of mitochondrial membrane integrity, were determined 3 hr after oxidative insult. Northern blot analysis revealed H2O2-induced reductions in mitochondrial transcripts for nicotinamide adenine dinucleotide dehydrogenase (NADH) subunits 4 and 5 and cytochrome c. H2O2 also inactivated GAPDH but did not alter CK activity. Pretreatment and simultaneous addition of 17beta-E2 with H2O2 did not prevent the reductions in mitochondrial transcript levels and GAPDH activity. 17beta-Estradiol did moderate the collapse of mitochondrial membrane potential (deltapsi(m)) in response to H2O2 as demonstrated by JC-1 staining and fluorescence microscopy. Although the precise mode of action responsible for protection by estradiols against oxidative stress remains to be determined, these results indicate that the hormone stabilizes the mitochondrial membrane, thereby preserving the driving force for oxidative ATP synthesis.


Asunto(s)
Citoprotección , Estradiol/farmacología , Cristalino/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Adenosina Trifosfato/biosíntesis , Células Cultivadas , Creatina Quinasa/metabolismo , Medios de Cultivo , Medio de Cultivo Libre de Suero , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Cristalino/metabolismo , Cristalino/fisiología , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/fisiología , Estrés Oxidativo/efectos de los fármacos
16.
Exp Eye Res ; 78(4): 861-71, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15037120

RESUMEN

The expression and intracellular localization of the estrogen receptor (ER) subtypes, ERalpha and ERbeta were examined in cultured human lens epithelial cells (HLE-B3). ERalpha and ERbeta mRNA expression was evaluated by coupled reverse transcription-polymerase chain reaction (RT-PCR) and Southern blot analysis. Subcellular localization of ERalpha and ERbeta was determined on formaldehyde-fixed, Saponin-permeabilized cells using conventional immunofluorescence techniques, as well as immunodetection of differential cellular components after subjecting the cultured cells to fractionation by sucrose gradient centrifugation. Using RT-PCR, ER species specific primers distinguished mRNA from total RNA extracted from HLE-B3 cells, as well as from human breast adenocarcinoma cells (MCF-7), which provided a positive control. The 286-bp (ERalpha) and 167-bp (ERbeta) PCR products were verified by sequence analysis. Southern blot analysis using internal oligonucleotides directed to specific primer pairs for ERalpha and ERbeta, respectively, further confirmed the authenticity of the PCR products. HLE-B3 cells expressed ERalpha and ERbeta in association with the nucleus and ERbeta in the mitochondria. That the mitochondrial-enriched subfraction correlated with the presence of the ERbeta subtype was confirmed by Western blot analysis. The differential subcellular partitioning of ERalpha and ERbeta subtypes lends a new aspect to the estrogen signalling system wherein mitochondrial stabilization, possibly through ER binding, may play a causal role in the maintenance of cellular integrity. The occurrence of ER subtypes in human lens epithelial cells suggests that estrogen plays a role in the physiology of the lens. The comparative nuclear and mitochondrial distribution of ERalpha and ERbeta raises the interesting probability that the two receptors, dependent upon subcellular localization, may have differential cytoprotective potential, and by inference, disparity in their mechanisms of action.


Asunto(s)
Células Epiteliales/química , Cristalino/química , Receptores de Estrógenos/análisis , Secuencia de Bases , Línea Celular Transformada , Núcleo Celular/química , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Humanos , Inmunohistoquímica/métodos , Mitocondrias/química , Datos de Secuencia Molecular , ARN Mensajero/análisis , Receptores de Estrógenos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA