Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chem Biol Interact ; 264: 43-51, 2017 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-28108222

RESUMO

Previously we have shown that the redox cycler menadione (MQ) induced cellular pyridine nucleotide redox imbalance that was linked to a decrease in aerobic glycolysis and perturbation of the mitochondrial respiratory activity due to the redox cycling of the compound; these processes were potentiated by low glucose. In this study, we investigated how colonic epithelial cells maintained pyridine nucleotide (NAD+/NADH and NADP+/NADPH) redox homeostasis upon acute metabolic variation and exposure to the redox cycling diquat (DQ). Our results show that DQ challenge disrupted cellular NADH/NAD+ redox status and enhanced cellular NADPH generation. Notably, DQ-induced NADH decrease was associated with enhanced lactate production, a process that was potentiated by glucose availability, but not by the mitochondrial substrates, succinate or malate/glutamate. In addition, DQ increased glucose 6-phoshate dehydrogenase (G6PDH) activity consistent with glucose diversion towards pentose phosphate pathway. As a consequence, steady-state NADPH levels were maintained during MQ challenge at normal glucose. In contrast and despite increased G6PDH and malic enzyme (ME) activities, DQ induced cellular NADPH-to-NADP+ shift at low glucose, a situation that was reversed by mitochondrial substrates. Collectively, these results are consistent with increased aerobic glycolysis by DQ and specific metabolic changes leading to enhanced NADPH generation upon oxidative challenge.


Assuntos
Neoplasias do Colo/metabolismo , Diquat/metabolismo , Glucose/metabolismo , NADP/metabolismo , NAD/metabolismo , Linhagem Celular Tumoral , Colo/citologia , Colo/metabolismo , Células Epiteliais/metabolismo , Glicólise , Células HT29 , Humanos , Mitocôndrias/metabolismo , Oxirredução
2.
Chem Biol Interact ; 264: 16-24, 2017 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-28087461

RESUMO

In this study, we investigated how colonic epithelial cells maintained pyridine nucleotide (NADH/NAD+) redox homeostasis upon acute metabolic variation imposed by glucose deprivation or supplementation with mitochondrial substrates, succinate and malate/glutamate (M/G). Our results showed that low glucose caused cellular NADH/NAD+ redox imbalance that diminished lactate dehydrogenase (LDH) activity and resulted in lower lactate contents. The concurrent activation of malic enzyme (ME) suggested a role for malate in preserving cellular pyruvate that remained unchanged at low glucose. Mitochondrial substrates restored cellular NADH/NAD+ redox homeostasis at low glucose in association with specific compartmental catabolism of mitochondrial substrates. As compared with normal glucose, M/G and low glucose promoted glycolytic ATP production but inhibited mitochondrial-derived ATP generation in association with decreased glucose availability for mitochondrial respiration. At normal glucose, succinate and M/G enhanced mitochondrial respiratory activity, but had minimal impact on mitochondrial-derived ATP production. Collectively, these results are consistent with low glucose-induced NADH/NAD+ redox imbalance in association with decreased aerobic glycolysis that is reversed by supplementation with M/G but not succinate.


Assuntos
Trifosfato de Adenosina/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , Células Epiteliais/metabolismo , Glucose/metabolismo , Mitocôndrias/metabolismo , NAD/metabolismo , Colo/patologia , Neoplasias do Colo/patologia , Células Epiteliais/patologia , Ácido Glutâmico/metabolismo , Glicólise , Células HT29 , Humanos , Malatos/metabolismo , Mitocôndrias/patologia , Oxirredução , Ácido Succínico/metabolismo
4.
PLoS One ; 11(1): e0146931, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26784896

RESUMO

Lysosome trafficking plays a significant role in tumor invasion, a key event for the development of metastasis. Previous studies from our laboratory have demonstrated that the anterograde (outward) movement of lysosomes to the cell surface in response to certain tumor microenvironment stimulus, such as hepatocyte growth factor (HGF) or acidic extracellular pH (pHe), increases cathepsin B secretion and tumor cell invasion. Anterograde lysosome trafficking depends on sodium-proton exchanger activity and can be reversed by blocking these ion pumps with Troglitazone or EIPA. Since these drugs cannot be advanced into the clinic due to toxicity, we have designed a high-content assay to discover drugs that block peripheral lysosome trafficking with the goal of identifying novel drugs that inhibit tumor cell invasion. An automated high-content imaging system (Cellomics) was used to measure the position of lysosomes relative to the nucleus. Among a total of 2210 repurposed and natural product drugs screened, 18 "hits" were identified. One of the compounds identified as an anterograde lysosome trafficking inhibitor was niclosamide, a marketed human anti-helminthic drug. Further studies revealed that niclosamide blocked acidic pHe, HGF, and epidermal growth factor (EGF)-induced anterograde lysosome redistribution, protease secretion, motility, and invasion of DU145 castrate resistant prostate cancer cells at clinically relevant concentrations. In an effort to identify the mechanism by which niclosamide prevented anterograde lysosome movement, we found that this drug exhibited no significant effect on the level of ATP, microtubules or actin filaments, and had minimal effect on the PI3K and MAPK pathways. Niclosamide collapsed intralysosomal pH without disruption of the lysosome membrane, while bafilomycin, an agent that impairs lysosome acidification, was also found to induce JLA in our model. Taken together, these data suggest that niclosamide promotes juxtanuclear lysosome aggregation (JLA) via modulation of pathways involved in lysosome acidification. In conclusion, we have designed a validated reproducible high-content assay to screen for drugs that inhibit lysosome trafficking and reduce tumor invasion and we summarize the action of one of these drugs.


Assuntos
Antinematódeos/farmacologia , Lisossomos/efeitos dos fármacos , Niclosamida/farmacologia , Neoplasias da Próstata/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Masculino , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos
5.
Free Radic Biol Med ; 54: 51-61, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23108103

RESUMO

We recently demonstrated that methylglyoxal (MG) induced apoptosis of brain microvascular endothelial cells (IHECs) that was preceded by glutathione (GSH) depletion. Here, we test the hypothesis that MG induces occludin glycation and disrupts IHEC barrier function, which is prevented by GSH-dependent MG metabolism. Exposure of IHECs to MG decreased transendothelial electrical resistance (TEER) in association with MG-adduct formation. A 65-kDa MG-glycated protein corresponded to occludin, which was confirmed by immunoprecipitation. Moreover, immunofluorescence staining showed that MG disrupted the architectural organization of ZO-1. Occludin glycation and ZO-1 disruption were prevented by N-acetylcysteine (NAC). Accordingly, TEER loss was abrogated by NAC (via GSH synthesis) and exacerbated by buthionine sulfoximine (BSO; GSH synthesis inhibitor). BSO treatment attenuated D-lactate production, consistent with a role for GSH in glyoxalase I-catalyzed MG elimination. Although MG increased reactive oxygen species (ROS) generation, the ROS scavengers tempol and tiron did not block barrier disruption. This suggests that endogenously generated ROS were unlikely to be a major cause of or did not reach a threshold to elicit barrier failure as elicited by exogenous hydrogen peroxide (300-400 µM). Immunohistochemistry revealed a lower percentage of microvessels stained with anti-occludin, but a higher percentage stained with anti-MG in diabetic rat brain compared to controls. Western analyses confirmed the decrease in diabetic brain occludin expression, but an increase in glycated occludin levels. These results provide novel evidence that reactive carbonyl species can mediate occludin glycation in cerebral microvessels and in microvascular endothelial cells that contribute to barrier dysfunction, a process that was prevented by GSH through enhanced MG catabolism.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/efeitos dos fármacos , Diabetes Mellitus Experimental/metabolismo , Endotélio/efeitos dos fármacos , Ocludina/metabolismo , Aldeído Pirúvico/metabolismo , Animais , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Células Cultivadas , Endotélio/patologia , Glutationa/metabolismo , Produtos Finais de Glicação Avançada/química , Humanos , Masculino , Microvasos/efeitos dos fármacos , Ocludina/química , Estresse Oxidativo , Polimerização/efeitos dos fármacos , Carbonilação Proteica , Aldeído Pirúvico/farmacologia , Ratos , Ratos Wistar , Proteína da Zônula de Oclusão-1/metabolismo
6.
Int J Cell Biol ; 2012: 434971, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22745639

RESUMO

The integrity of the vascular endothelium of the blood-brain barrier (BBB) is central to cerebrovascular homeostasis. Given the function of the BBB as a physical and metabolic barrier that buffers the systemic environment, oxidative damage to the endothelial monolayer will have significant deleterious impact on the metabolic, immunological, and neurological functions of the brain. Glutathione (GSH) is a ubiquitous major thiol within mammalian cells that plays important roles in antioxidant defense, oxidation-reduction reactions in metabolic pathways, and redox signaling. The existence of distinct GSH pools within the subcellular organelles supports an elegant mode for independent redox regulation of metabolic processes, including those that control cell fate. GSH-dependent homeostatic control of neurovascular function is relatively unexplored. Significantly, GSH regulation of two aspects of endothelial function is paramount to barrier preservation, namely, GSH protection against oxidative endothelial cell injury and GSH control of postdamage cell proliferation in endothelial repair and/or wound healing. This paper highlights our current insights and hypotheses into the role of GSH in cerebral microvascular biology and pathobiology with special focus on endothelial GSH and vascular integrity, oxidative disruption of endothelial barrier function, GSH regulation of endothelial cell proliferation, and the pathological implications of GSH disruption in oxidative stress-associated neurovascular disorders, such as diabetes and stroke.

7.
Biochim Biophys Acta ; 1823(10): 1767-77, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22732297

RESUMO

Apoptosis is a highly organized form of cell death that is important for tissue homeostasis, organ development and senescence. To date, the extrinsic (death receptor mediated) and intrinsic (mitochondria derived) apoptotic pathways have been characterized in mammalian cells. Reduced glutathione, is the most prevalent cellular thiol that plays an essential role in preserving a reduced intracellular environment. glutathione protection of cellular macromolecules like deoxyribose nucleic acid proteins and lipids against oxidizing, environmental and cytotoxic agents, underscores its central anti-apoptotic function. Reactive oxygen and nitrogen species can oxidize cellular glutathione or induce its extracellular export leading to the loss of intracellular redox homeostasis and activation of the apoptotic signaling cascade. Recent evidence uncovered a novel role for glutathione involvement in apoptotic signaling pathways wherein post-translational S-glutathiolation of protein redox active cysteines is implicated in the potentiation of apoptosis. In the present review we focus on the key aspects of glutathione redox mechanisms associated with apoptotic signaling that includes: (a) changes in cellular glutathione redox homeostasis through glutathione oxidation or GSH transport in relation to the initiation or propagation of the apoptotic cascade, and (b) evidence for S-glutathiolation in protein modulation and apoptotic initiation.


Assuntos
Apoptose , Células/citologia , Glutationa/metabolismo , Animais , Células/metabolismo , Humanos , Modelos Biológicos , Oxirredução , Transdução de Sinais
8.
Cancer Sci ; 103(8): 1429-33, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22533343

RESUMO

Isocitrate dehydrogenase 1 (IDH1), a cytosolic enzyme that converts isocitrate to alpha-ketoglutarate, has been shown to be dysregulated during tumorigenesis. However, at what stage of cancer development IDH1 is dysregulated and how IDH1 may affect cell transformation and tumor promotion during early stages of cancer development are unclear. We used a skin cell transformation model and mouse skin epidermal tissues to study the role of IDH1 in early skin tumorigenesis. Our studies demonstrate that both the tumor promoter TPA and UVC irradiation decreased expression and activity levels of IDH1, not IDH2, in the tumor promotable JB6 P+ cell model. Skin epidermal tissues treated with dimethylbenz[α]anthracene/TPA also showed decreases in IDH1 expression and activity. In non-promotable JB6 P-cells, IDH1 was upregulated upon TPA treatment, whereas IDH2 was maintained at similar levels with TPA treatment. Interestingly, IDH1 knockdown enhanced, whereas IDH1 overexpression suppressed, TPA-induced cell transformation. Finally, manganese superoxide dismutase overexpression suppressed tumor promoter induced decreases in IDH1 expression and mitochondrial respiration, while intracellular alpha-ketoglutarate levels were unchanged. These results suggest that decreased IDH1 expression in early stage skin tumorigenesis is highly correlated with tumor promotion. In addition, oxidative stress might contribute to IDH1 inactivation, because manganese superoxide dismutase, a mitochondrial antioxidant enzyme, blocked decreases in IDH1 expression and activity.


Assuntos
Antioxidantes/metabolismo , Transformação Celular Neoplásica/metabolismo , Isocitrato Desidrogenase/metabolismo , Queratinócitos/metabolismo , Papiloma/metabolismo , Neoplasias Cutâneas/metabolismo , Pele/metabolismo , Superóxido Dismutase/metabolismo , Animais , Western Blotting , Regulação para Baixo , Imunofluorescência , Camundongos , Camundongos Endogâmicos DBA , Estresse Oxidativo , Consumo de Oxigênio , RNA Interferente Pequeno , Pele/patologia , Transfecção
9.
Semin Cell Dev Biol ; 23(7): 729-37, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22484611

RESUMO

The intestinal epithelium sits at the interface between an organism and its luminal environment, and as such is prone to oxidative damage induced by luminal oxidants. Mucosal integrity is maintained by the luminal redox status of the glutathione/glutathione disulfide (GSH/GSSG) and cysteine/cystine (Cys/CySS) couples which also support luminal nutrient absorption, mucus fluidity, and a diverse microbiota. The epithelial layer is uniquely organized for rapid self-renewal that is achieved by the well-regulated processes of crypt stem cell proliferation and crypt-to-villus cell differentiation. The GSH/GSSG and Cys/CySS redox couples, known to modulate intestinal cell transition through proliferation, differentiation or apoptosis, could govern the regenerative potential of the mucosa. These two couples, together with that of the thioredoxin/thioredoxin disulfide (Trx/TrxSS) couple are the major intracellular redox systems, and it is proposed that they each function as distinctive redox control nodes or circuitry in the control of metabolic processes and networks of enzymatic reactions. Specificity of redox signaling is accomplished in part by subcellular compartmentation of the individual redox systems within the mitochondria, nucleus, endoplasmic reticulum, and cytosol wherein each defined redox environment is suited to the specific metabolic function within that compartment. Mucosal oxidative stress would result from the disruption of these unique redox control nodes, and the subsequent alteration in redox signaling can contribute to the development of degenerative pathologies of the intestine, such as inflammation and cancer.


Assuntos
Mucosa Intestinal/metabolismo , Estresse Oxidativo , Animais , Espaço Extracelular/metabolismo , Glutationa/metabolismo , Homeostase , Humanos , Intestinos/microbiologia , Oxirredução
10.
Anal Bioanal Chem ; 401(7): 2165-75, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21842197

RESUMO

S-glutathionylation (Pr-SSG) is a specific post-translational modification of cysteine residues by the addition of glutathione. S-Glutathionylated proteins induced by oxidative or nitrosative stress play an essential role in understanding the pathogenesis of the aging and age-related disorder, such as Alzheimer's disease (AD). The purpose of this research is to develop a novel and ultrasensitive method to accurately and rapidly quantify the Pr-SSG by using capillary gel electrophoresis with laser-induced fluorescence detection (CGE-LIF). The derivatization method is based on the specific reduction of protein-bound S-glutathionylation with glutaredoxin (Grx) and labeling with thiol-reactive fluorescent dye (Dylight 488 maleimide). The experiments were performed by coupling the derivatization method with CGE-LIF to study electrophoretic profiling in in vitro oxidative stress model-S-glutathionylated bovine serum albumin (BSA-SSG), oxidant-induced human colon adenocarcinoma (HT-29) cells, brain tissues, and whole blood samples from an AD transgenic (Tg) mouse model. The results showed almost an eightfold increase in S-glutathionyl abundance when subjecting HT-29 cells in an oxidant environment, resulting in Pr-SSG at 232 ± 10.64 (average ±SD; n=3) nmol/mg. In the AD-Tg mouse model, an initial quantitative measurement demonstrated the extent of protein S-glutathionylation in three brain regions (hippocampus, cerebellum, and cerebrum), ranging from 1 to 10 nmol/mg. Additionally, we described our developed method to potentially serve as a highly desirable diagnostic tool for monitoring S-glutathionylated protein profile in minuscule amount of whole blood. The whole blood samples for S-glutathionyl expression of 5-month-old AD-Tg mice are quantified as 16.3 µmol/L (=7.2 nmol/mg protein). Altogether, this is a fast, easy, and accurate method, reaching the lowest limit of Pr-SSG detection at 1.8 attomole (amol) level, reported to date.


Assuntos
Cisteína/química , Eletroforese Capilar/métodos , Glutarredoxinas/metabolismo , Glutationa/metabolismo , Lasers , Processamento de Proteína Pós-Traducional , Doença de Alzheimer/diagnóstico , Animais , Bovinos , Modelos Animais de Doenças , Eletroforese Capilar/instrumentação , Fluorescência , Células HT29 , Humanos , Camundongos , Oxirredução , Soroalbumina Bovina/metabolismo
11.
Free Radic Res ; 45(11-12): 1245-66, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21831010

RESUMO

The intestinal tract, known for its capability for self-renew, represents the first barrier of defence between the organism and its luminal environment. The thiol/disulfide redox systems comprising the glutathione/glutathione disulfide (GSH/GSSG), cysteine/cystine (Cys/CySS) and reduced and oxidized thioredoxin (Trx/TrxSS) redox couples play important roles in preserving tissue redox homeostasis, metabolic functions, and cellular integrity. Control of the thiol-disulfide status at the luminal surface is essential for maintaining mucus fluidity and absorption of nutrients, and protection against chemical-induced oxidant injury. Within intestinal cells, these redox couples preserve an environment that supports physiological processes and orchestrates networks of enzymatic reactions against oxidative stress. In this review, we focus on the intestinal redox and antioxidant systems, their subcellular compartmentation, redox signalling and epithelial turnover, and contribution of luminal microbiota, key aspects that are relevant to understanding redox-dependent processes in gut biology with implications for degenerative digestive disorders, such as inflammation and cancer.


Assuntos
Cisteína/metabolismo , Dissulfeto de Glutationa/metabolismo , Glutationa/metabolismo , Mucosa Intestinal/metabolismo , Estresse Oxidativo , Tiorredoxinas/metabolismo , Cistina/metabolismo , Humanos , Intestinos/enzimologia , Oxirredução , Transdução de Sinais
12.
J Chromatogr A ; 1218(38): 6756-62, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21820121

RESUMO

S-nitrosylated proteins are biomarkers of oxidative damage in aging and Alzheimer's disease (AD). Here, we report a new method for detecting and quantifying nitrosylated proteins by capillary gel electrophoresis with laser induced fluorescence detection (CGE-LIF). Dylight 488 maleimide was used to specifically label thiol group (SH) after switching the S-nitrosothiol (S-NO) to SH in cysteine using the "fluorescence switch" assay. In vitro nitrosylation model-BSA subjected to S-nitrosoglutathione (GSNO) optimized the labeling reactions and characterized the response of the LIF detector. The method proves to be highly sensitive, detecting 1.3 picomolar (pM) concentration of nitrosothiols in nanograms of proteins, which is the lowest limit of detection of nitrosothiols reported to date. We further demonstrated the direct application of this method in monitoring protein nitrosylation damage in MQ mediated human colon adenocarcinoma cells. The nitrosothiol amounts in MQ treated and untreated cells are 14.8±0.2 and 10.4±0.5 pmol/mg of proteins, respectively. We also depicted nitrosylated protein electrophoretic profiles of brain cerebrum of 5-month-old AD transgenic (Tg) mice model. In Tg mice brain, 15.5±0.4 pmol of nitrosothiols/mg of proteins was quantified while wild type contained 11.7±0.3 pmol/mg proteins. The methodology is validated to quantify low levels of S-nitrosylated protein in complex protein mixtures from both physiological and pathological conditions.


Assuntos
Doença de Alzheimer/diagnóstico , Eletroforese Capilar/métodos , Proteínas/análise , S-Nitrosotióis/análise , Doença de Alzheimer/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Eletroforese Capilar/instrumentação , Fluorescência , Humanos , Lasers , Camundongos , Proteínas/metabolismo , S-Nitrosotióis/metabolismo
13.
J Biol Chem ; 286(23): 20267-74, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21515692

RESUMO

Stationary-phase Saccharomyces cerevisiae cells transferred from spent rich media into water live for weeks, whereas the same cells die within hours if transferred into water with 2% glucose in a process called sugar-induced cell death (SICD). Our hypothesis is that SICD is due to a dysregulated Crabtree effect, which is the phenomenon whereby glucose transiently inhibits respiration and ATP synthesis. We found that stationary-phase cells in glucose/water consume 21 times more O(2) per cell than exponential-phase cells in rich media, and such excessive O(2) consumption causes reactive oxygen species to accumulate. We also found that inorganic phosphate and succinate protect against SICD but by different mechanisms. Phosphate protects by triggering the synthesis of Fru-1,6-P(2), which inhibits respiration in isolated mitochondria. Succinate protects in wild-type cells but fails to protect in dic1Δ cells. DIC1 codes for a mitochondrial inner membrane protein that exchanges cytosolic succinate for matrix phosphate. We propose that succinate depletes matrix phosphate, which in turn inhibits respiration and ATP synthesis. In sum, restoring the Crabtree effect, whether with phosphate or succinate, protects cells from SICD.


Assuntos
Trifosfato de Adenosina/biossíntese , Glucose/metabolismo , Consumo de Oxigênio/fisiologia , Fosfatos/metabolismo , Saccharomyces cerevisiae/metabolismo , Ácido Succínico/metabolismo , Trifosfato de Adenosina/genética , Frutosedifosfatos/genética , Frutosedifosfatos/metabolismo , Saccharomyces cerevisiae/genética
14.
Antioxid Redox Signal ; 14(11): 2151-62, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21083422

RESUMO

We recently demonstrated that menadione (MQ), a redox cycling quinone, mediated the loss of mitochondrial glutathione/glutathione disulfide redox balance. In this study, we showed that MQ significantly disrupted cellular pyridine nucleotide (NAD(+)/NADH, NADP(+)/NADPH) redox balance that compromised cellular ATP, mitochondrial respiratory activity, and NADPH-dependent reducing capacity in colonic epithelial cells, a scenario that was exaggerated by low glucose. In the cytosol, MQ induced NAD(+) loss concurrent with increased NADP(+) and NAD kinase activity, but decreased NADPH. In the mitochondria, NADH loss occurred in conjunction with increased nicotinamide nucleotide transhydrogenase activity and NADP(+), and decreased NADPH. These results are consistent with cytosolic NAD(+)-to-NADP(+) and mitochondrial NADH-to-NADPH shifts, but compromised NADPH availability. Thus, despite the sacrifice of NAD(+)/NADH in favor of NADPH generation, steady-state NADPH levels were not maintained during MQ challenge. Impairments of cellular bioenergetics were evidenced by ATP losses and increased mitochondrial O(2) dependence of pyridine nucleotide oxidation-reduction; half-maximal oxidation (P(50)) was 10-fold higher in low glucose, which was lowered by glutamate or succinate supplementation. This exaggerated O(2) dependence is consistent with increased O(2) diversion to nonmitochondrial O(2) consumption by MQ-semiquinone redox cycling secondary to decreased NADPH-dependent MQ detoxication at low glucose, a situation that was corrected by glucose-sparing mitochondrial substrates.


Assuntos
Trifosfato de Adenosina/metabolismo , Colo/metabolismo , Células Epiteliais/metabolismo , Glucose/metabolismo , Mitocôndrias/metabolismo , NADP/metabolismo , NAD/metabolismo , Estresse Oxidativo , Respiração Celular , Colo/citologia , Citoplasma/enzimologia , Ensaios Enzimáticos , Glucose/deficiência , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Células HT29 , Homeostase , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Ácido Láctico/metabolismo , NADH NADPH Oxirredutases/metabolismo , Oxirredução , Consumo de Oxigênio , Vitamina K 3/farmacologia
15.
J Immunol ; 185(6): 3217-26, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20733204

RESUMO

The major cellular antioxidant glutathione is depleted during HIV infection and in obesity. Although the consequence of glutathione depletion on immune function is starting to emerge, it is currently not known whether glutathione dysregulation influences the differentiation and maturation of dendritic cells (DCs). Moreover, the effect of glutathione depletion on DC effector functions, such as Ag presentation, is poorly understood. Glutathione synthesis depends on the cystine/glutamate antiporter, which transports the rate-limiting precursor cystine into the cell in exchange for glutamate. In this paper, we present a detailed study of antiporter function in DCs and demonstrate a role for the antiporter in DC differentiation and cross-presentation. We show that the antiporter is the major mechanism for transport of cystine and glutamate and modulates the intracellular glutathione content and glutathione efflux from DCs. Blocking antiporter-dependent cystine transport decreases intracellular glutathione levels, and these effects correlate with reduced transcription of the functional subunit of the antiporter. We further demonstrate that blocking antiporter activity interferes with DC differentiation from monocyte precursors, but antiporter activity is not required for LPS-induced phenotypic maturation. Finally, we show that inhibiting antiporter uptake of cystine interferes with presentation of exogenous Ag to class II MHC-restricted T cells and blocks cross-presentation on MHC class I. We conclude that aberrant antiporter function disrupts glutathione homeostasis in DCs and may contribute to impaired immunity in the diseased host.


Assuntos
Sistema y+ de Transporte de Aminoácidos/fisiologia , Apresentação de Antígeno/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Sistema y+ de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Apresentação de Antígeno/genética , Transporte Biológico/imunologia , Diferenciação Celular/genética , Células Cultivadas , Apresentação Cruzada/genética , Apresentação Cruzada/imunologia , Cistina/metabolismo , Células Dendríticas/metabolismo , Ácido Glutâmico/metabolismo , Glutationa/antagonistas & inibidores , Glutationa/metabolismo , Homeostase/imunologia , Humanos , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Lipopolissacarídeos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia , Ovalbumina/metabolismo
16.
Free Radic Biol Med ; 48(6): 749-62, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20045723

RESUMO

Reactive oxygen species (ROS) are products of normal metabolism and xenobiotic exposure, and depending on their concentration, ROS can be beneficial or harmful to cells and tissues. At physiological low levels, ROS function as "redox messengers" in intracellular signaling and regulation, whereas excess ROS induce oxidative modification of cellular macromolecules, inhibit protein function, and promote cell death. Additionally, various redox systems, such as the glutathione, thioredoxin, and pyridine nucleotide redox couples, participate in cell signaling and modulation of cell function, including apoptotic cell death. Cell apoptosis is initiated by extracellular and intracellular signals via two main pathways, the death receptor- and the mitochondria-mediated pathways. Various pathologies can result from oxidative stress-induced apoptotic signaling that is consequent to ROS increases and/or antioxidant decreases, disruption of intracellular redox homeostasis, and irreversible oxidative modifications of lipid, protein, or DNA. In this review, we focus on several key aspects of ROS and redox mechanisms in apoptotic signaling and highlight the gaps in knowledge and potential avenues for further investigation. A full understanding of the redox control of apoptotic initiation and execution could underpin the development of therapeutic interventions targeted at oxidative stress-associated disorders.


Assuntos
Apoptose , Espécies Reativas de Oxigênio/metabolismo , Animais , Humanos , Oxirredução , Transdução de Sinais
17.
Free Radic Biol Med ; 47(8): 1190-8, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19647792

RESUMO

Although oxidative stress induces mitochondrial DNA (mtDNA) damage, a role for redox in modulating mtDNA oxidation and repair is relatively unexplored. This study examines the contribution of cellular glutathione (GSH) redox status to menadione (MQ)-induced mtDNA damage and postoxidant mtDNA recovery in a nontransformed NCM460 colonic cell line. We show that MQ caused dose-dependent increases in mtDNA damage that were blunted by N-acetylcysteine, a thiol antioxidant. Damage to mtDNA paralleled mitochondrial protein disulfide formation and glutathione disulfide increases in the cytosol and mitochondria and was exacerbated by inhibition of GSH synthesis in accordance with decreased cytosolic and mitochondrial GSH. Blockade of mitochondrial GSH (mtGSH) transport potentiated mtDNA damage, which was prevented by overexpression of the oxoglutarate mtGSH carrier, underscoring a link between mtGSH and mtDNA responsiveness to oxidative stress. The removal of MQ posttreatment elicited mtDNA recovery to basal levels by 4 h, indicating complete repair. Notably, mtDNA recovery was preceded by restored cytosolic and mtGSH levels at 2 h, suggesting a connection between the maintenance of cell GSH and effective mtDNA repair. The MQ-induced dose-dependent increase in mtDNA damage was attenuated by overexpressing mitochondrial 8-oxoguanine DNA glycosylase (Ogg1), consistent with 7,8-dihydro-8-oxoguanine being a major oxidative mtDNA lesion. Collectively, the results show that oxidative mtDNA damage in colonic cells is highly responsive to the mtGSH status and that postoxidant mtDNA recovery may also be GSH sensitive.


Assuntos
Colo/efeitos dos fármacos , Colo/metabolismo , Dano ao DNA , DNA Mitocondrial/genética , Glutationa/metabolismo , Oxidantes/farmacologia , Células Cultivadas , DNA Glicosilases/metabolismo , Células Epiteliais/metabolismo , Dissulfeto de Glutationa/metabolismo , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Oxirredução , Vitamina K 3/farmacologia
18.
Biochem Pharmacol ; 77(1): 76-85, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18840413

RESUMO

Staurosporine (STP) was shown to induce cell apoptosis through formation of reactive oxygen species, but a role for cellular redox has not been defined. In this study, we report that STP (2 microM) caused apoptosis (24+/-3% at 24 h) of human colon adenocarcinoma epithelial cell line HT29 that was preceded by significant glutathione (GSH) and glutathione disulfide (GSSG) efflux (6 h), but independent of changes in cellular glutathione/glutathione disulfide (GSH/GSSG) redox status. The blockade of GSH efflux by gamma-glutamyl glutamate (gamma-GG) or ophthalmic acid was associated with apoptosis attenuation; however, gamma-GG administration after peak GSH efflux (8 h) did not confer cytoprotection. Moreover, lowering cellular GSH through inhibition of its synthesis prevented extracellular GSH accumulation and cell apoptosis, thus validating a link between cellular GSH export and the trigger of cell apoptosis. Inhibition of gamma-glutamyl transferase (GGT1, EC 2.3.2.2)-catalyzed extracellular GSH degradation with acivicin significantly blocked GSH efflux, suggesting that GSH breakdown is a driving force for GSH export. Interestingly, acivicin treatment enhanced extracellular GSSG accumulation, consistent with GSH oxidation. STP-induced HT29 cell apoptosis was associated with caspase-3 activation independent of caspase-8 or caspase-9 activity; accordingly, inhibitors of the latter caspases were without effect on STP-induced apoptosis. STP similarly induced GSH efflux and apoptosis in a non-malignant human NCM460 colonic cell line in association with caspase-3 activation. Collectively, our results demonstrate that STP induction of apoptosis in malignant and non-malignant colonic cells is temporally linked to the export of cellular GSH and the activation of caspase-3 without caspase-8 or -9 involvement.


Assuntos
Apoptose/fisiologia , Colo/metabolismo , Glutationa/metabolismo , Mucosa Intestinal/metabolismo , Estaurosporina/farmacologia , Apoptose/efeitos dos fármacos , Colo/citologia , Colo/efeitos dos fármacos , Feminino , Glutationa/fisiologia , Células HT29 , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Oxirredução
19.
Free Radic Biol Med ; 45(11): 1591-9, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18926903

RESUMO

Previously, we demonstrated an important role for insulin in the protection of endothelial cells against hyperglycemic stress through maintaining cellular glutathione (GSH) redox balance. The current study focuses on the contribution of insulin to transcriptional control of endothelial cell GSH recovery during acute oxidative challenge and the influence of low glucose. The results show that insulin induced an approximate 2-fold increase in expression of gamma-glutamylcysteine ligase catalytic subunit (GCLc) mRNA and protein; interestingly, cellular GSH levels were not elevated accordingly. However, on tert-butylhydroperoxide challenge, insulin-treated cells demonstrated a robust GSH recovery that was attributed to a greater capacity for de novo synthesis via elevated GCLc levels. Notably, the effects of insulin were observed under low, but not normal, glucose conditions. Our results implicate a role for Nrf2 involvement in both constitutive and inducible endothelial GCLc expression and GSH synthesis, while PI3K/Akt/mTOR signaling appears to participate only in insulin-inducible GSH synthesis. Collectively, these results support the functional importance of insulin in Nrf2-dependent transcriptional upregulation of GCLc in GSH recovery during oxidative challenge and suggest a possible role for hypoglycemia in promoting insulin-mediated GCLc upregulation.


Assuntos
Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Insulina/fisiologia , Estresse Oxidativo/fisiologia , Análise de Variância , Domínio Catalítico , Linhagem Celular , Sobrevivência Celular , Cromatografia Líquida de Alta Pressão , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Expressão Gênica , Glucose/metabolismo , Glutamato-Cisteína Ligase/química , Glutamato-Cisteína Ligase/genética , Humanos , Insulina/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Transdução de Sinais , terc-Butil Hidroperóxido/farmacologia
20.
Free Radic Res ; 42(8): 689-706, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18671159

RESUMO

Apoptosis or programmed cell death represents a physiologically conserved mechanism of cell death that is pivotal in normal development and tissue homeostasis in all organisms. As a key modulator of cell functions, the most abundant non-protein thiol, glutathione (GSH), has important roles in cellular defense against oxidant aggression, redox regulation of proteins thiols and maintaining redox homeostasis that is critical for proper function of cellular processes, including apoptosis. Thus, a shift in the cellular GSH-to-GSSG redox balance in favour of the oxidized species, GSSG, constitutes an important signal that could decide the fate of a cell. The current review will focus on three main areas: (1) general description of cellular apoptotic pathways, (2) cellular compartmentation of GSH and the contribution of mitochondrial GSH and redox proteins to apoptotic signalling and (3) role of redox mechanisms in the initiation and execution phases of apoptosis.


Assuntos
Apoptose/fisiologia , Glutationa/metabolismo , Animais , Caspases/metabolismo , Dissulfeto de Glutationa/metabolismo , Humanos , Mitocôndrias/metabolismo , Estresse Oxidativo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...