Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 8(1): e2525, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28055018

RESUMEN

Aberrant Sumoylation of protein(s) in response to oxidative stress or during aging is known to be involved in etiopathogenesis of many diseases. Upon oxidative stress, Peroxiredoxin (Prdx) 6 is aberrantly Sumoylated by Sumo1, resulting in loss of functions and cell death. We identified lysines (K) 122 and 142 as the major Sumo1 conjugation sites in Prdx6. Intriguingly, the mutant Prdx6 K122/142 R (arginine) gained protective efficacy, increasing in abundance and promoting glutathione (GSH) peroxidase and acidic calcium-independent phospholipase A2 (aiPLA2) activities. Using lens epithelial cells derived from targeted inactivation of Prdx6-/- gene and relative enzymatic and stability assays, we discovered dramatic increases in GSH-peroxidase (30%) and aiPLA2 (37%) activities and stability in the K122/142 R mutant, suggesting Sumo1 destabilized Prdx6 integrity. Prdx6-/-LECs with EGFP-Sumo1 transduced or co-expressed with mutant TAT-HA-Prdx6K122/142 R or pGFP-Prdx6K122/142 R were highly resistant to oxidative stress, demonstrating mutant protein escaped and interrupted the Prdx6 aberrant Sumoylation-mediated cell death pathway. Mutational analysis of functional sites showed that both peroxidase and PLA2 active sites were necessary for mutant Prdx6 function, and that Prdx6 phosphorylation (at T177 residue) was essential for optimum PLA2 activity. Our work reveals the involvement of oxidative stress-induced aberrant Sumoylation in dysregulation of Prdx6 function. Mutant Prdx6 at its Sumo1 sites escapes and abates this adverse process by maintaining its integrity and gaining function. We propose that the K122/142R mutant of Prdx6 in the form of a TAT-fusion protein may be an easily applicable intervention for pathobiology of cells related to aberrant Sumoylation signaling in aging or oxidative stress.


Asunto(s)
Envejecimiento/genética , Glutatión Peroxidasa/genética , Fosfolipasas A2 Grupo VI/genética , Proteínas Mutantes/genética , Peroxiredoxina VI/genética , Proteína SUMO-1/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Apoptosis/genética , Estabilidad de Enzimas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Glutatión Peroxidasa/metabolismo , Fosfolipasas A2 Grupo VI/metabolismo , Humanos , Lisina/química , Lisina/genética , Proteínas Mutantes/metabolismo , Estrés Oxidativo/genética , Peroxiredoxina VI/metabolismo , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Proteína SUMO-1/metabolismo , Transducción de Señal/genética , Sumoilación/genética
2.
Am J Physiol Cell Physiol ; 310(1): C1-16, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26447207

RESUMEN

Oxidative stress or reduced expression of naturally occurring antioxidants during aging has been identified as a major culprit in neuronal cell/tissue degeneration. Peroxiredoxin (Prdx) 6, a protective protein with GSH peroxidase and acidic calcium-independent phospholipase A2 activities, acts as a rheostat in regulating cellular physiology by clearing reactive oxygen species (ROS) and thereby optimizing gene regulation. We found that under stress, the neuronal cells displayed reduced expression of Prdx6 protein and mRNA with increased levels of ROS, and the cells subsequently underwent apoptosis. Using Prdx6 fused to TAT transduction domain, we showed evidence that Prdx6 was internalized in human brain cortical neuronal cells, HCN-2, and mouse hippocampal cells, HT22. The cells transduced with Prdx6 conferred resistance against the oxidative stress inducers paraquat, H2O2, and glutamate. Furthermore, Prdx6 delivery ameliorated damage to neuronal cells by optimizing ROS levels and overstimulation of NF-κB. Intriguingly, transduction of Prdx6 increased the expression of endogenous Prdx6, suggesting that protection against oxidative stress was mediated by both extrinsic and intrinsic Prdx6. The results demonstrate that Prdx6 expression is critical to protecting oxidative stress-evoked neuronal cell death. We propose that local or systemic application of Prdx6 can be an effective means of delaying/postponing neuronal degeneration.


Asunto(s)
Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo , Peroxiredoxina VI/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Niño , Citoprotección , Relación Dosis-Respuesta a Droga , Endocitosis , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/metabolismo , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Peroxiredoxina VI/genética , Peroxiredoxina VI/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal , Transfección
3.
FEBS J ; 281(15): 3357-81, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24910119

RESUMEN

Loss of the cytoprotective protein peroxiredoxin 6 (Prdx6) in cells that are aging or under oxidative stress is known to be linked to the pathobiology of many age-related diseases. However, the mechanism by which Prdx6 activity goes awry is largely unknown. Using Prdx6-deficient (Prdx6(-/-) ) cells as a model for aging or redox active cells, human/mouse lens epithelial cells (LECs) facing oxidative stress and aging lenses, we found a significant increase in the levels of small ubiquitin-like modifier (Sumo)1 conjugates. These cells displayed increased levels of Sumo1 and reduced the expression of Prdx6. Specifically, we observed that Prdx6 is a target for aberrant sumoylation signaling, and that Sumo1 modification reduces its cellular abundance. LECs overexpressing Sumo1 showed reduced expression and activity of Prdx6 and its transactivator specificity protein 1 (Sp1), mRNA and protein with increased levels of reactive oxygen species; those cells were vulnerable to oxidative stress-induced cell death. A significant reduction in Prdx6, Sp1 protein and mRNA expression was observed in redox active Prdx6(-/-) cells and in aging lenses/LECs. The reduction was correlated with increased expression of Sumo1 and enrichment of the inactive form (dimeric) of Sumo-specific protease (Senp)1. Experiments with Sumo1-fused Prdx6 and Prdx6 promoter-linked to chloramphenicol acetyltransferase reporter gene constructs indicated that Sumo1 dysregulated Prdx6 activity by reducing its abundance and attenuating its transcription; in contrast, the delivery of Senp1 or Prdx6 reversed the process. The data show that reactive oxygen species-evoked aberrant sumoylation signaling affects Prdx6 activity by reducing Prdx6 abundance, as well as transcription. The findings of the present study may provide a foundation for a strategy to repair deleterious oxidative signaling generated by a reduced activity of Prdx6.


Asunto(s)
Peroxiredoxina VI/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteína SUMO-1/metabolismo , Sumoilación , Transcripción Genética , Envejecimiento , Animales , Supervivencia Celular , Células Cultivadas , Cisteína Endopeptidasas , Endopeptidasas/genética , Endopeptidasas/metabolismo , Represión Enzimática , Estabilidad de Enzimas , Humanos , Ratones , Estrés Oxidativo , Transducción de Señal , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo
4.
Am J Physiol Cell Physiol ; 304(7): C636-55, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23364261

RESUMEN

Oxidative stress and endoplasmic reticulum (ER) stress are emerging as crucial events in the etiopathology of many neurodegenerative diseases. While the neuroprotective contributions of the dietary compound curcumin has been recognized, the molecular mechanisms underlying curcumin's neuroprotection under oxidative and ER stresses remains elusive. Herein, we show that curcumin protects HT22 from oxidative and ER stresses evoked by the hypoxia (1% O(2) or CoCl(2) treatment) by enhancing peroxiredoxin 6 (Prdx6) expression. Cells exposed to CoCl(2) displayed reduced expression of Prdx6 with higher reactive oxygen species (ROS) expression and activation of NF-κB with IκB phosphorylation. When NF-κB activity was blocked by using SN50, an inhibitor of NF-κB, or cells treated with curcumin, the repression of Prdx6 expression was restored, suggesting the involvement of NF-κB in modulating Prdx6 expression. These cells were enriched with an accumulation of ER stress proteins, C/EBP homologous protein (CHOP), GRP/78, and calreticulin, and had activated states of caspases 12, 9, and 3. Reinforced expression of Prdx6 in HT22 cells by curcumin reestablished survival signaling by reducing propagation of ROS and blunting ER stress signaling. Intriguingly, knockdown of Prdx6 by antisense revealed that loss of Prdx6 contributed to cell death by sustaining enhanced levels of ER stress-responsive proapoptotic proteins, which was due to elevated ROS production, suggesting that Prdx6 deficiency is a cause of initiation of ROS-mediated ER stress-induced apoptosis. We propose that using curcumin to reinforce the naturally occurring Prdx6 expression and attenuate ROS-based ER stress and NF-κB-mediated aberrant signaling improves cell survival and may provide an avenue to treat and/or postpone diseases associated with ROS or ER stress.


Asunto(s)
Curcumina/metabolismo , Hipocampo/citología , Hipoxia/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Peroxiredoxina VI/metabolismo , Animales , Muerte Celular , Línea Celular , Cobalto , Retículo Endoplásmico , Regulación de la Expresión Génica/fisiología , Ratones , FN-kappa B/genética , Péptidos/genética , Péptidos/metabolismo , Peroxiredoxina VI/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
5.
Epigenetics ; 8(3): 268-80, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23386123

RESUMEN

Expression level of lens epithelial derived growth factor (LEDGF) is vital for LEDGF-mediated cell survival and cytoprotection against proapoptotic stimuli. We previously demonstrated that LEDGF is transcriptionally regulated by Sp1-responsive elements within a CpG island in the LEDGF promoter. Herein, we report on the existence of epigenetic signaling involved in the repression of LEDGF transcription in lens epithelial cells (LECs) facing UVB. UVB exposure led to histone H3 dimethylation and deacetylation at its CpG island, where a histone deacetylase/histone methylase (HDAC1/SUV39H1) complex was recruited. Exposure of LECs to UVB stress altered LEDGF protein and mRNA expression as well as promoter activity, while failing to methylate the CpG island. These events were correlated with increased reactive oxygen species (ROS) and increased cell death. LEDGF promoter activity and expression remained unaltered after 5-Aza treatment, but were relieved with tricostatin A, an inhibitor of HDACs. Expression analysis disclosed that UVB radiation altered the global expression levels of acetylated histone proteins, diminished total histone acetyltransferase (HAT) activity and increased HDAC activity and HDAC1 expression. In silico analysis of LEDGF proximal promoter and ChIP analyses disclosed HDAC1/SUV39H1 complex anchored to the -170/-10 nt promoter regions at Sp1-responsive elements and also attenuated Sp1 binding, resulting in HDAC1- and SUV39H1-dependent deacetylation and dimethylation of H3 at K9. Acetylation of H3K9 was essential for LEDGF active transcription, while enrichment of H3K9me2 at Sp1-responsive elements within CpGs (-170/-10) by UVB radiation repressed LEDGF transcription. Our study may contribute to understanding diseases associated with LEDGF aberrant expression due to specific epigenetic modifications, including blinding disorders.


Asunto(s)
Islas de CpG , Epigénesis Genética , Histona Desacetilasa 1/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Elementos de Respuesta , Rayos Ultravioleta , Acetilación , Apoptosis , Epitelio Corneal/metabolismo , Epitelio Corneal/efectos de la radiación , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metilación , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción Sp1/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación
6.
J Cell Mol Med ; 17(1): 212-21, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23205574

RESUMEN

Injury to lens epithelial cells (LECs) leads to epithelial-mesenchymal transition (EMT) with resultant fibrosis. The tropomyosin (Tpm) family of cytoskeleton proteins is involved in regulating and stabilizing actin microfilaments. Aberrant expression of Tpms leads to abnormal morphological changes with disintegration of epithelial integrity. The EMT of LECs has been proposed as a major cause of posterior capsule opacification (PCO) after cataract surgery. Using in vivo rodent PCO and human cataractous LECs, we demonstrated that the aberrant expression of rat Tpm and human Tpm1α/2ß suggested their association in remodelling of the actin cytoskeleton during EMT of LECs. Expression analysis from abnormally growing LECs after lens extraction revealed elevated expression of α-smooth muscle actin (α-SMA), a marker for EMT. Importantly, these cells displayed increased expression of Tpm1α/2ß following EMT/PCO formation. Expression of Tpm1α/2ß was up-regulated in LECs isolated from cataractous lenses of Shumiya Cataract Rats (SCRs), compared with non-cataractous lenses. Also, LECs from human patients with nuclear cataract and anterior subcapsular fibrosis (ASF) displayed significantly increased expression of Tpm2ß mRNA, suggesting that similar signalling invokes the expression of these molecules in LECs of cataractous SCR and human lenses. EMT was observed in LECs overexpressed with Tpm1α/2ß, as evidenced by increased expression of α-SMA. These conditions were correlated with remodelling of actin filaments, possibly leading to EMT/PCO and ASF. The present findings may help clarify the condition of the actin cytoskeleton during morphogenetic EMT, and may contribute to development of Tpm-based inhibitors for postponing PCO and cataractogenesis.


Asunto(s)
Opacificación Capsular/genética , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Cápsula del Cristalino/metabolismo , Tropomiosina/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Opacificación Capsular/metabolismo , Opacificación Capsular/patología , Células Cultivadas , Células Epiteliales/patología , Femenino , Expresión Génica , Humanos , Cápsula del Cristalino/patología , Masculino , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Transfección , Tropomiosina/metabolismo
7.
FEBS J ; 279(17): 3048-70, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22748127

RESUMEN

Lens epithelium-derived growth factor (LEDGF), a ubiquitously expressed nuclear protein, acts by interacting with DNA and protein and is involved in widely varying cellular functions. Despite its importance, the mechanism(s) that regulate naturally occurring LEDGF activity are unidentified. In the present study, we report that LEDGF is constitutively Sumoylated, and that the dynamical regulatory mechanism(s) (i.e. Sumoylation and deSumoylation) act as a molecular switch in modulating the DNA-binding and transcriptional activity of LEDGF with the functional consequences. Using bioinformatics analysis coupled with in vitro and in vivo Sumoylation assays, we found that lysine (K) 364 of LEDGF was Sumoylated, repressing its transcriptional activity. Conversely, mutation of K364 to arginine (R) or deSumoylation by small ubiquitin-like modifier (Sumo)-specific protease-1, a nuclear deSumoylase, enhanced the transactivation capacity of LEDGF and its cellular abundance. The enhancements were directly correlated with an increase in the DNA-binding activity and small heat shock protein transcription of LEDGF, whereas the process was reversed in cells overexpressing Sumo1. Interestingly, cells expressing Sumoylation-deficient pEGFP-K364R protein showed increased cellular survival compared to wild-type LEDGF protein. The findings provide insights into the regulation and regulatory functions of LEDGF in Sumoylation-dependent transcriptional control that may be essential for modifying the physiology of cells to maintain cellular homeostasis. These studies also provide new evidence of the important role of post-translational modification in controlling LEDGF function.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas de Choque Térmico/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Activación Transcripcional , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Cisteína Endopeptidasas , Cartilla de ADN , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Sumoilación
8.
PLoS One ; 7(5): e37012, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615874

RESUMEN

LEDGF/p75 interacts with DNA/protein to regulate gene expression and function. Despite the recognized diversity of function of LEDGF/p75, knowledge of its transregulation is in its infancy. Here we report that LEDGF/p75 gene is TATA-less, contains GC-rich cis elements and is transcriptionally regulated by Sp1 involving small ubiquitin-like modifier (Sumo1). Using different cell lines, we showed that Sp1 overexpression increased the level of LEDGF/p75 protein and mRNA expression in a concentration-dependent fashion. In contrast, RNA interference depletion of intrinsic Sp1 or treatment with artemisinin, a Sp1 inhibitor, reduced expression of LEDGF/p75, suggesting Sp1-mediated regulation of LEDGF/p75. In silico analysis disclosed three evolutionarily conserved, putative Sp1 sites within LEDGF/p75 proximal promoter (-170/+1 nt). DNA-binding and transactivation assays using deletion and point mutation constructs of LEDGF/p75 promoter-CAT revealed that all Sp1 sites (-50/-43, -109/-102 and -146/-139) differentially regulate LEDGF/p75. Cotransfection studies with Sp1 in Drosophila cells that were Sp1-deficient, showed increased LEDGF/p75 transcription, while in lens epithelial cells (LECs) promoter activity was inhibited by artemisinin. These events were correlated with levels of endogenous Sp1-dependent LEDGF/p75 expression, and higher resistance to UVB-induced cell death. ChIP and transactivation assays showed that Sumoylation of Sp1 repressed its transcriptional activity as evidenced through its reduced binding to GC-box and reduced ability to activate LEDGF/p75 transcription. As whole, results revealed the importance of Sp1 in regulating expression of LEDGF/p75 gene and add to our knowledge of the factors that control LEDGF/p75 within cellular microenvironments, potentially providing a foundation for LEDGF/p75 expression-based transcription therapy.


Asunto(s)
Secuencia Rica en GC , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Adolescente , Adulto , Anciano , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Drosophila , Humanos , Persona de Mediana Edad , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Activación Transcripcional , Adulto Joven
9.
Am J Physiol Cell Physiol ; 301(4): C954-67, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21677259

RESUMEN

The multifunctional cytoprotective protein peroxiredoxin 6 (Prdx6) maintains cellular homeostasis and membrane integrity by regulating expression of intracellular reactive oxygen species (ROS) and phospholipid turnover. Using cells derived from targeted inactivation of Prdx6 gene or its depletion by RNA interference or aging, we showed that Prdx6 deficiency in cells evoked unfolded protein response (UPR), evidenced by increased expression or activation of proapoptotic factors, CHOP, ATF4, PERK, IRE-α and eIF2-α and by increased caspases 3 and 12 processing. Those cells displayed enhanced and sustained expression of endoplasmic reticulum (ER) stress-related chaperon proteins, Bip/glucose-regulated protein 78, calnexin, and calreticulin. Under cellular stress induced by hypoxia (1% O(2) or CoCl(2) treatment) or tunicamycin, Prdx6-deficient cells exhibited aberrant activation of ER stress-responsive genes/protein with higher expression of ROS, and died with apoptosis. Wild-type cells exposed to tunicamycin or hypoxia remained relatively insensitive with lower expression of ROS and ER-responsive genes than did Prdx6-deficient cells, but upregulation of ER stress responsive proteins or chaperones mimicked the UPR response of Prdx6-deficient or aging cells. Expression of Prdx6 blocked ER stress-induced deleterious signaling by optimizing physiologically aberrant expression of ER stress responsive genes/proteins in Prdx6-deficient cells or cells facing stressors, and rescued the cells from apoptosis. These findings demonstrate that impaired homeostasis and progression of pathogenesis in Prdx6-deficient lens epithelial cells or in aging cells should be blocked by a supply of Prdx6. The results provide a new molecular basis for understanding the etiology of several age-associated degenerative disorders, and potentially for developing antioxidant Prdx6-based therapeutics.


Asunto(s)
Retículo Endoplásmico/metabolismo , Células Epiteliales/metabolismo , Cristalino/citología , Peroxiredoxina VI/genética , Peroxiredoxina VI/metabolismo , Animales , Apoptosis , Células Cultivadas , Regulación de la Expresión Génica , Homeostasis , Ratones , Ratones Noqueados , Estrés Fisiológico
10.
BMC Neurosci ; 11: 125, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20923568

RESUMEN

BACKGROUND: The ability to respond to changes in the extra-intracellular environment is prerequisite for cell survival. Cellular responses to the environment include elevating defense systems, such as the antioxidant defense system. Hypoxia-evoked reactive oxygen species (ROS)-driven oxidative stress is an underlying mechanism of retinal ganglion cell (RGC) death that leads to blinding disorders. The protein peroxiredoxin 6 (PRDX6) plays a pleiotropic role in negatively regulating death signaling in response to stressors, and thereby stabilizes cellular homeostasis. RESULTS: We have shown that RGCs exposed to hypoxia (1%) or hypoxia mimetic cobalt chloride display reduced expression of PRDX6 with higher ROS expression and activation of NF-κB. These cells undergo apoptosis, while cells with over-expression of PRDX6 demonstrate resistance against hypoxia-driven RGC death. The RGCs exposed to hypoxia either with 1% oxygen or cobalt chloride (0-400 µM), revealed ~30%-70% apoptotic cell death after 48 and 72 h of exposure. Western analysis and real-time PCR showed elevated expression of PRDX6 during hypoxia at 24 h, while PRDX6 protein and mRNA expression declined from 48 h onwards following hypoxia exposure. Concomitant with this, RGCs showed increased ROS expression and activation of NF-κB with IkB phosphorylation/degradation, as examined with H2DCF-DA and transactivation assays. These hypoxia-induced adverse reactions could be reversed by over-expression of PRDX6. CONCLUSION: Because an abundance of PRDX6 in cells was able to attenuate hypoxia-induced RGC death, the protein could possibly be developed as a novel therapeutic agent acting to postpone RGC injury and delay the progression of glaucoma and other disorders caused by the increased-ROS-generated death signaling related to hypoxia.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Fármacos Neuroprotectores , Peroxiredoxina VI/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Cobalto/farmacología , Genes Reporteros , Proteínas I-kappa B/metabolismo , Etiquetado Corte-Fin in Situ , FN-kappa B/metabolismo , Peroxiredoxina VI/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
PLoS One ; 5(1): e8602, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20066046

RESUMEN

L-type calcium currents (I(Ca)) are influenced by changes in extracellular chloride, but sites of anion effects have not been identified. Our experiments showed that CaV1.2 currents expressed in HEK293 cells are strongly inhibited by replacing extracellular chloride with gluconate or perchlorate. Variance-mean analysis of I(Ca) and cell-attached patch single channel recordings indicate that gluconate-induced inhibition is due to intracellular anion effects on Ca(2+) channel open probability, not conductance. Inhibition of CaV1.2 currents produced by replacing chloride with gluconate was reduced from approximately 75%-80% to approximately 50% by omitting beta subunits but unaffected by omitting alpha(2)delta subunits. Similarly, gluconate inhibition was reduced to approximately 50% by deleting an alpha1 subunit N-terminal region of 15 residues critical for beta subunit interactions regulating open probability. Omitting beta subunits with this mutant alpha1 subunit did not further diminish inhibition. Gluconate inhibition was unchanged with expression of different beta subunits. Truncating the C terminus at AA1665 reduced gluconate inhibition from approximately 75%-80% to approximately 50% whereas truncating it at AA1700 had no effect. Neutralizing arginines at AA1696 and 1697 by replacement with glutamines reduced gluconate inhibition to approximately 60% indicating these residues are particularly important for anion effects. Expressing CaV1.2 channels that lacked both N and C termini reduced gluconate inhibition to approximately 25% consistent with additive interactions between the two tail regions. Our results suggest that modest changes in intracellular anion concentration can produce significant effects on CaV1.2 currents mediated by changes in channel open probability involving beta subunit interactions with the N terminus and a short C terminal region.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Secuencia de Aminoácidos , Aniones , Canales de Calcio Tipo L/química , Línea Celular , Humanos , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Probabilidad
12.
Am J Physiol Cell Physiol ; 298(2): C342-54, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19889963

RESUMEN

Oxidative stress is one of the causative factors in progression and etiology of age-related cataract. Peroxiredoxin 6 (Prdx6), a savior for cells from internal or external environmental stresses, plays a role in cellular signaling by detoxifying reactive oxygen species (ROS) and thereby controlling gene regulation. Using targeted inactivation of the Prdx6 gene, we show that Prdx6-deficient lens epithelial cells (LECs) are more vulnerable to UV-triggered cell death, a major cause of skin disorders including cataractogenesis, and these cells display abnormal protein profiles. PRDX6-depleted LECs showed phenotypic changes and formed lentoid body, a characteristic of terminal cell differentiation and epithelial-mesenchymal transition. Prdx6(-/-) LECs exposed to UV-B showed higher ROS expression and were prone to apoptosis compared with wild-type LECs, underscoring a protective role for Prdx6. Comparative proteomic analysis using fluorescence-based difference gel electrophoresis along with mass spectrometry and database searching revealed a total of 13 proteins that were differentially expressed in Prdx6(-/-) cells. Six proteins were upregulated, whereas expression of seven proteins was decreased compared with Prdx6(+/+) LECs. Among the cytoskeleton-associated proteins that were highly expressed in Prdx6-deficient LECs was tropomyosin (Tm)2beta. Protein blot and real-time PCR validated dramatic increase of Tm2beta and Tm1alpha expression in these cells. Importantly, Prdx6(+/+) LECs showed a similar pattern of Tm2beta protein expression after transforming growth factor (TGF)-beta or H(2)O(2) treatment. An extrinsic supply of PRDX6 could restore Tm2beta expression, demonstrating that PRDX6 may attenuate adverse signaling in cells and thereby maintain cellular homeostasis. Exploring redox-proteomics (Prdx6(-/-)) and characterization and identification of abnormally expressed proteins and their attenuation by PRDX6 delivery should provide a basis for development of novel therapeutic interventions to postpone ROS-mediated abnormal signaling deleterious to cells or tissues.


Asunto(s)
Citoprotección , Células Epiteliales/efectos de la radiación , Proteínas del Ojo/metabolismo , Cristalino/efectos de la radiación , Peroxiredoxina VI/deficiencia , Proteómica/métodos , Rayos Ultravioleta , Animales , Apoptosis/efectos de la radiación , Western Blotting , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Electroforesis en Gel Bidimensional , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas del Ojo/genética , Regulación de la Expresión Génica/efectos de la radiación , Peróxido de Hidrógeno/toxicidad , Cristalino/efectos de los fármacos , Cristalino/metabolismo , Cristalino/patología , Ratones , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo/efectos de la radiación , Mapeo Peptídico , Peroxiredoxina VI/genética , Peroxiredoxina VI/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tropomiosina/metabolismo
13.
Free Radic Res ; 43(9): 783-95, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19572226

RESUMEN

Oxidative stress and TGFbeta-induced disturbance of cells and tissues are implicated in initiation and progression of pathophysiology of cells/tissues. Using primary human trabecular meshwork (TM) cells from normal and glaucomatous subjects, this study demonstrated that peroxiredoxin (PRDX) 6, an antioxidant, offsets the deleterious effects of oxidative stress on TM cells by optimizing ROS and TGFbeta levels. An analysis of glaucomatous TM cells revealed a reduced expression of PRDX6 mRNA and protein. Biochemical assays disclosed enhanced levels of ROS, as well as high levels of TGFbetas and these cells expressed elevated extracellular matrix (ECM) and Tsp1 proteins with reduced MMP2; conditions implicated in the pathophysiology of glaucoma. Non-glaucomatous TM cells exposed to TGFbetas/ROS showed similar features as in glaucomatous cells. The abnormalities induced were reversed by delivery of PRDX6. The data provide evidence that oxidative stress-induced abnormality in TM may be related to reduced PRDX6 expression and provide a foundation for antioxidant-based therapeutics for treating glaucoma.


Asunto(s)
Glaucoma/metabolismo , Estrés Oxidativo , Peroxiredoxina VI/metabolismo , Malla Trabecular/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Actinas/metabolismo , Células Cultivadas , Senescencia Celular , Daño del ADN , Relación Dosis-Respuesta a Droga , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Glaucoma/patología , Humanos , Peróxido de Hidrógeno/toxicidad , Metaloproteinasa 2 de la Matriz/metabolismo , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Peroxiredoxina VI/genética , Fenotipo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Trombospondina 1/metabolismo , Malla Trabecular/efectos de los fármacos , Malla Trabecular/patología , Transducción Genética , Factor de Crecimiento Transformador beta/metabolismo , Transglutaminasas/metabolismo , Tropomiosina/metabolismo
15.
Life Sci ; 84(23-24): 857-64, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19351539

RESUMEN

AIMS: Hyperglycemia-induced oxidative stress is implicated in pericyte apoptosis seen in diabetic retinopathy. The six mammalian Peroxiredoxins (PRDXs) comprise a novel family of antioxidative proteins that negatively regulate oxidative stress-induced apoptosis by controlling reactive oxygen species (ROS) levels. MAIN METHODS: Sprague-Dawley rats were used to detect the retinal expressions of PRDXs1-6. Pig pericytes cultured in high-glucose medium were used to monitor the protective effect of PRDX5 and 6 against high-glucose-associated change. Recombinant PRDX5 and 6 proteins were linked to the Trans-Activating Transduction (TAT) domain from HIV-1 TAT protein for their efficient delivery into cells/tissues. KEY FINDINGS: We found higher expression of PRDX5 and 6 mRNAs and PRDX5 and 6 proteins in retina than the other Prdxs (Prdx1-4). Western blotting affirmed the intracellular presence of TAT-linked proteins and revealed the efficient transduction of TAT-HA-PRDX5 and 6 in these cells. Extrinsic supply of TAT-HA-PRDX5 and 6 proteins inhibited the oxidative stress-induced DNA damage after high-glucose exposure in pig pericytes. The cell survival and apoptosis assay revealed that extrinsic supply of TAT-HA-PRDX5 and 6 proteins was responsible for inhibiting hyperglycemia-induced pericyte apoptosis. SIGNIFICANCE: Results suggest that delivery of PRDX5 and 6 might protect hyperglycemia-induced pericyte loss to inhibit oxidative stress.


Asunto(s)
Glucosa/toxicidad , VIH-1/fisiología , Pericitos/metabolismo , Peroxiredoxina VI/genética , Peroxirredoxinas/genética , Retina/metabolismo , Transducción Genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología , Animales , Apoptosis/genética , Células Cultivadas , Femenino , VIH-1/genética , Humanos , Pericitos/patología , Peroxiredoxina VI/administración & dosificación , Peroxiredoxina VI/biosíntesis , Peroxirredoxinas/administración & dosificación , Peroxirredoxinas/biosíntesis , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Retina/patología , Porcinos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
16.
Brain Res ; 1233: 63-78, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18694738

RESUMEN

Higher expression of reactive oxygen species (ROS) is implicated in neurological disorders. A major event in glaucoma, the death of retinal ganglion cells (RGCs), has been associated with elevated levels of glutamate and TNF-alpha in the RGCs' local microenvironment. Herein we show that the transduction of Peroxiredoxin 6 (PRDX6) attenuates TNF-alpha- and glutamate-induced RGC death, by limiting ROS and maintaining Ca2+ homeostasis. Immunohistochemical staining of rat retina disclosed the presence of PRDX6 in RGCs, and Western and real-time PCR analysis revealed an abundance of PRDX6 protein and mRNA. RGCs treated with glutamate and/or TNF-alpha displayed elevated levels of ROS and reduced expression of PRDX6, and underwent apoptosis. A supply of PRDX6 protected RGCs from glutamate and TNF-alpha induced cytotoxicity by reducing ROS level and NF-kappaB activation, and limiting increased intracellular Ca2+ influx. Results provide a rationale for use of PRDX6 for blocking ROS-mediated pathophysiology in glaucoma and other neuronal disorders.


Asunto(s)
Apoptosis/fisiología , Calcio/metabolismo , Peroxiredoxina VI/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Células Cultivadas , Ácido Glutámico/metabolismo , Estrés Oxidativo/fisiología , Peroxiredoxina VI/genética , ARN Mensajero/análisis , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
17.
Am J Physiol Cell Physiol ; 294(3): C842-55, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18184874

RESUMEN

A diminished level of endogenous antioxidant in cells/tissues is associated with reduced resistance to oxidative stress. Peroxiredoxin 6 (PRDX6), a protective molecule, regulates gene expression/function by controlling reactive oxygen species (ROS) levels. Using PRDX6 protein linked to TAT, the transduction domain from human immunodeficiency virus type 1 TAT protein, we demonstrated that PRDX6 was transduced into lens epithelial cells derived from rat or mouse lenses. The protein was biologically active, negatively regulating apoptosis and delaying progression of cataractogenesis by attenuating deleterious signaling. Lens epithelial cells from cataractous lenses bore elevated levels of ROS and were susceptible to oxidative stress. These cells harbored increased levels of active transforming growth factor (TGF)-beta 1 and of alpha-smooth muscle actin and beta ig-h3, markers for cataractogenesis. Importantly, cataractous lenses showed a 10-fold reduction in PRDX6 expression, whereas TGF-beta1 mRNA and protein levels were elevated. The changes were reversed, and cataractogenesis was delayed when PRDX6 was supplied. Results suggest that delivery of PRDX6 can postpone cataractogenesis, and this should be an effective approach to delaying cataracts and other degenerative diseases that are associated with increased ROS.


Asunto(s)
Antioxidantes/metabolismo , Apoptosis , Catarata/terapia , Células Epiteliales/metabolismo , Terapia Genética/métodos , Cristalino/metabolismo , Peroxiredoxina VI/metabolismo , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Catarata/genética , Catarata/metabolismo , Catarata/patología , Catarata/prevención & control , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Proteínas de la Matriz Extracelular/metabolismo , Peróxido de Hidrógeno/toxicidad , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cristalino/efectos de los fármacos , Cristalino/patología , Peroxidación de Lípido , Ratones , Ratones Noqueados , Oxidantes/toxicidad , Estrés Oxidativo , Peroxiredoxina VI/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción Genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
18.
Am J Physiol Cell Physiol ; 290(2): C554-66, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16403949

RESUMEN

TNF-alpha induces oxidative stress by generating reactive oxygen species (ROS). This molecule elevates the expression of gamma-glutamylcysteine synthetase heavy subunit (gamma-GCS-HS). Lens epithelium-derived growth factor (LEDGF)/p75, a transcriptional protein, is inducible by oxidative stress and protects cells from various stresses by upregulating stress-responsive genes. This paper presents evidence that TNF-alpha elevates the expression of LEDGF and that LEDGF is one of the transactivators of gamma-GCS-HS gene. An analysis of the gamma-GCS-HS promoter sequence (-819 to +518 nt) revealed the presence of putative sites for LEDGF binding. Gel mobility assay confirmed the binding of LEDGF to the heat shock element (nGAAn) and the stress response element (A/TGGGGA/T) present in gamma-GCS-HS promoter. Transactivation experiments showed activation of gamma-GCS-HS promoter in cells overexpressing LEDGF or treated with a sublethal dose of TNF-alpha (20 ng/ml). Downregulation of gamma-GCS-HS promoter activity in cells transfected with LEDGF small interfering RNA validated the finding. Notably, cells treated with TNF-alpha (20 ng/ml) for 24 h had an increased abundance of LEDGF and gamma-GCS-HS mRNA and protein. In contrast, cells treated with TNF-alpha for longer periods or with higher concentrations of TNF-alpha showed reduced expression of LEDGF and gamma-GCS-HS and increased cellular death with higher ROS levels. Cells overexpressing LEDGF revealed elevated GSH levels (10-15%), a condition that may potentially eliminate the insult to cells induced by TNF-alpha. Thus TNF-alpha regulation of LEDGF may be physiologically important, as elevated expression of LEDGF increases the expression of endogenous gamma-GCS-HS gene, the catalytic subunit of the regulating enzyme in GSH biosynthesis that may constitute a protective mechanism in limiting oxidative stress induced by inflammatory cytokines.


Asunto(s)
Células Epiteliales/metabolismo , Glutamato-Cisteína Ligasa/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cristalino/citología , Subunidades de Proteína/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/fisiología , Secuencia de Bases , Forma de la Célula , Supervivencia Celular , Células Cultivadas , Células Epiteliales/citología , Glutamato-Cisteína Ligasa/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Datos de Secuencia Molecular , Mutación , Regiones Promotoras Genéticas , Subunidades de Proteína/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Activación Transcripcional
19.
Biochem Pharmacol ; 71(6): 818-26, 2006 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-16434028

RESUMEN

Increasing data from epidemiological and in vitro studies show that the isoflavonoids, genistein and daidzein, and the flavonols, quercetin and kaempferol, are protective against postmenopausal bone loss. However, the physiological mechanisms for these effects are not well understood. We now report that kaempferol exerts profound antiosteoclastogenic effects by acting on both osteoblasts and osteoclasts. Kaempferol but not quercetin dose-dependently inhibited tumor necrosis factor alpha (TNFalpha)-induced production of the osteoclastogenic cytokines interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1/CCL2) in osteoblasts. The effect on IL-6 was posttranscriptional, whereas kaempferol reduced MCP-1 mRNA levels. In addition, in mouse primary calvarial osteoblasts, kaempferol but not quercetin blocked TNFalpha-induced translocation of the nuclear factor kappaB (NF-kappaB) subunit p65 from the cytoplasm to the nucleus. However, TNFalpha-stimulated intracellular ROS production was unaltered by kaempferol. In RAW264.7 cells, a monocyte/macrophage precursor for osteoclasts, both kaempferol and quercetin dose-dependently inhibited the receptor activator of NF-kappaB ligand (RANKL)-induced immediate-early oncogene c-fos expression at 6 h. After 3-5 days, both flavonols robustly inhibited RANKL-induced expression of the osteoclastic differentiation markers, RANK and calcitonin receptor. Consistent with down regulation of these osteoclastic differentiation markers, both flavonols strongly attenuated the RANKL-induced formation of multinucleated osteoclasts. However, kaempferol was more potent than quercetin in inhibiting RANKL-stimulated effects on RAW264.7 cells. Thus, our data indicate that kaempferol exerts profound antiosteoclastogenic effects by specifically antagonizing TNF receptor family action on bone cells at two distinct levels, by disrupting production of osteoclastogenic cytokines from osteoblasts and attenuating osteoclast precursor cell differentiation.


Asunto(s)
Quempferoles/farmacología , Osteoblastos/efectos de los fármacos , Quercetina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Animales Recién Nacidos , Línea Celular , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Osteoblastos/metabolismo , Osteoblastos/patología , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
20.
Mech Ageing Dev ; 127(3): 249-56, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16321424

RESUMEN

Peroxiredoxin (PRDX) 6 is a unique member of the PRDX family. Its antioxidant and signaling properties are related to its expression level in cells. We studied development- and age-associated changes in PRDX6 expression in the murine lens. We also investigated the effects of dexamethasone (Dex), transforming growth factor-beta1 (TGF-beta1) and tumor necrosis factor-alfa (TNF-alpha) on PRDX6 expression. Expression levels of PRDX6 mRNA in whole lenses isolated from postnatal day (PD) 1- to 18-month-old mice, and the effects of Dex, TGF-beta1 and TNF-alpha on the expression of PRDX6 in lens epithelial cells (LECs), were monitored using real-time reverse transcriptase-polymerase chain reaction (PCR) or Western blot. Localization of PRDX6 was studied using in situ hybridization and immunohistochemistry. PRDX6 expression gradually increased in the lenses of 4-week- to 6-month-old mice and declined thereafter. In situ hybridization and immunohistochemistry revealed that PRDX6 was localized in the cytoplasm of LECs and in lens fibers. Intense PRDX6 staining was present in the whole lens on gestational days 14 and 18. The lenses of PD1 mice showed diminished nuclear fiber staining, while those of 4-week-old mice revealed lack of nuclear fiber staining but intense staining of the germinative zone. LECs treated with TNF-alpha or Dex showed higher PRDX6 expression, while TGF-beta1 down regulated expression. Thus, our results provide a topographic basis for understanding the role of PRDX6 in the lens.


Asunto(s)
Envejecimiento/fisiología , Regulación de la Expresión Génica/fisiología , Cristalino/metabolismo , Peroxidasas/biosíntesis , Embarazo/fisiología , Animales , Animales Recién Nacidos , Femenino , Perfilación de la Expresión Génica/métodos , Cristalino/citología , Ratones , Ratones Endogámicos BALB C , Peroxiredoxina VI , Peroxirredoxinas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...