Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 151(4): 794-806, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23141538

RESUMEN

PDI catalyzes the oxidative folding of disulfide-containing proteins. However, the sequence of reactions leading to a natively folded and oxidized protein remains unknown. Here we demonstrate a technique that enables independent measurements of disulfide formation and protein folding. We find that non-native disulfides are formed early in the folding pathway and can trigger misfolding. In contrast, a PDI domain favors native disulfides by catalyzing oxidation at a late stage of folding. We propose a model for cotranslational oxidative folding wherein PDI acts as a placeholder that is relieved by the pairing of cysteines caused by substrate folding. This general mechanism can explain how PDI catalyzes oxidative folding in a variety of structurally unrelated substrates.


Asunto(s)
Procolágeno-Prolina Dioxigenasa/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Pliegue de Proteína , Disulfuros , Microscopía de Fuerza Atómica , Modelos Moleculares , Oxidación-Reducción , Proteínas/química , Proteínas/metabolismo
2.
Mol Ther ; 31(3): 825-846, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36638800

RESUMEN

Blindness caused by advanced stages of inherited retinal diseases and age-related macular degeneration are characterized by photoreceptor loss. Cell therapy involving replacement with functional photoreceptor-like cells generated from human pluripotent stem cells holds great promise. Here, we generated a human recombinant retina-specific laminin isoform, LN523, and demonstrated the role in promoting the differentiation of human embryonic stem cells into photoreceptor progenitors. This chemically defined and xenogen-free method enables reproducible production of photoreceptor progenitors within 32 days. We observed that the transplantation into rd10 mice were able to protect the host photoreceptor outer nuclear layer (ONL) up to 2 weeks post transplantation as measured by full-field electroretinogram. At 4 weeks post transplantation, the engrafted cells were found to survive, mature, and associate with the host's rod bipolar cells. Visual behavioral assessment using the water maze swimming test demonstrated visual improvement in the cell-transplanted rodents. At 20 weeks post transplantation, the maturing engrafted cells were able to replace the loss of host ONL by extensive association with host bipolar cells and synapses. Post-transplanted rabbit model also provided congruent evidence for synaptic connectivity with the degenerated host retina. The results may pave the way for the development of stem cell-based therapeutics for retina degeneration.


Asunto(s)
Células Madre Pluripotentes , Degeneración Retiniana , Humanos , Ratones , Animales , Conejos , Laminina/genética , Retina , Células Fotorreceptoras , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , Diferenciación Celular
3.
Physiol Rev ; 96(1): 307-64, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26681794

RESUMEN

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated from aerobic metabolism, as a result of accidental electron leakage as well as regulated enzymatic processes. Because ROS/RNS can induce oxidative injury and act in redox signaling, enzymes metabolizing them will inherently promote either health or disease, depending on the physiological context. It is thus misleading to consider conventionally called antioxidant enzymes to be largely, if not exclusively, health protective. Because such a notion is nonetheless common, we herein attempt to rationalize why this simplistic view should be avoided. First we give an updated summary of physiological phenotypes triggered in mouse models of overexpression or knockout of major antioxidant enzymes. Subsequently, we focus on a series of striking cases that demonstrate "paradoxical" outcomes, i.e., increased fitness upon deletion of antioxidant enzymes or disease triggered by their overexpression. We elaborate mechanisms by which these phenotypes are mediated via chemical, biological, and metabolic interactions of the antioxidant enzymes with their substrates, downstream events, and cellular context. Furthermore, we propose that novel treatments of antioxidant enzyme-related human diseases may be enabled by deliberate targeting of dual roles of the pertaining enzymes. We also discuss the potential of "antioxidant" nutrients and phytochemicals, via regulating the expression or function of antioxidant enzymes, in preventing, treating, or aggravating chronic diseases. We conclude that "paradoxical" roles of antioxidant enzymes in physiology, health, and disease derive from sophisticated molecular mechanisms of redox biology and metabolic homeostasis. Simply viewing antioxidant enzymes as always being beneficial is not only conceptually misleading but also clinically hazardous if such notions underpin medical treatment protocols based on modulation of redox pathways.


Asunto(s)
Antioxidantes/metabolismo , Enzimas/metabolismo , Estado de Salud , Estrés Oxidativo , Animales , Modelos Animales de Enfermedad , Inducción Enzimática , Represión Enzimática , Enzimas/biosíntesis , Enzimas/genética , Técnicas de Silenciamiento del Gen , Predisposición Genética a la Enfermedad , Humanos , Ratones Transgénicos , Estado Nutricional , Oxidación-Reducción , Fenotipo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo
4.
Nitric Oxide ; 118: 26-30, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34742907

RESUMEN

The intracellular concentration of reduced glutathione (GSH) lies in the range of 1-10 mM, thereby indisputably making it the most abundant intracellular thiol. Such a copious amount of GSH makes it the most potent and robust cellular antioxidant that plays a crucial role in cellular defence against redox stress. The role of GSH as a denitrosylating agent is well established; in this study, we demonstrate GSH mediated denitrosylation of HepG2 cell-derived protein nitrosothiols (PSNOs), by a unique spin-trapping mechanism, using 5,5-dimethyl-1-pyrroline N-oxide (DMPO) as the spin trapping agent, followed by a western blot analysis. We also report our findings of two, hitherto unidentified substrates of GSH mediated S-denitrosylation, namely S-nitrosoglutaredoxin 1 (Grx1-SNO) and S-nitrosylated R1 subunit of ribonucleotide reductase (R1-SNO).


Asunto(s)
Glutarredoxinas/metabolismo , Glutatión/metabolismo , Ribonucleósido Difosfato Reductasa/metabolismo , S-Nitrosotioles/metabolismo , Óxidos N-Cíclicos/química , Glutarredoxinas/química , Células Hep G2 , Humanos , Ribonucleósido Difosfato Reductasa/química , S-Nitrosotioles/química , Marcadores de Spin , Detección de Spin , Tiorredoxinas/química , Tiorredoxinas/metabolismo
5.
Acta Neuropathol ; 141(5): 725-754, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33694021

RESUMEN

The mechanisms by which parkin protects the adult human brain from Parkinson disease remain incompletely understood. We hypothesized that parkin cysteines participate in redox reactions and that these are reflected in its posttranslational modifications. We found that in post mortem human brain, including in the Substantia nigra, parkin is largely insoluble after age 40 years; this transition is linked to its oxidation, such as at residues Cys95 and Cys253. In mice, oxidative stress induces posttranslational modifications of parkin cysteines that lower its solubility in vivo. Similarly, oxidation of recombinant parkin by hydrogen peroxide (H2O2) promotes its insolubility and aggregate formation, and in exchange leads to the reduction of H2O2. This thiol-based redox activity is diminished by parkin point mutants, e.g., p.C431F and p.G328E. In prkn-null mice, H2O2 levels are increased under oxidative stress conditions, such as acutely by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxin exposure or chronically due to a second, genetic hit; H2O2 levels are also significantly increased in parkin-deficient human brain. In dopamine toxicity studies, wild-type parkin, but not disease-linked mutants, protects human dopaminergic cells, in part through lowering H2O2. Parkin also neutralizes reactive, electrophilic dopamine metabolites via adduct formation, which occurs foremost at the primate-specific residue Cys95. Further, wild-type but not p.C95A-mutant parkin augments melanin formation in vitro. By probing sections of adult, human midbrain from control individuals with epitope-mapped, monoclonal antibodies, we found specific and robust parkin reactivity that co-localizes with neuromelanin pigment, frequently within LAMP-3/CD63+ lysosomes. We conclude that oxidative modifications of parkin cysteines are associated with protective outcomes, which include the reduction of H2O2, conjugation of reactive dopamine metabolites, sequestration of radicals within insoluble aggregates, and increased melanin formation. The loss of these complementary redox effects may augment oxidative stress during ageing in dopamine-producing cells of mutant PRKN allele carriers, thereby enhancing the risk of Parkinson's-linked neurodegeneration.


Asunto(s)
Envejecimiento/metabolismo , Dopamina/metabolismo , Mesencéfalo/metabolismo , Degeneración Nerviosa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , Animales , Niño , Preescolar , Femenino , Humanos , Masculino , Mesencéfalo/patología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Degeneración Nerviosa/patología , Oxidación-Reducción , Adulto Joven
6.
Mol Biol Rep ; 48(11): 7621-7626, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34599703

RESUMEN

Ribonucleotide reductases (RNR) catalyze the rate-limiting step in DNA synthesis during the S-phase of the cell cycle. Its constant activity in order to maintain dNTP homeostasis is a fascinating area of research and an attractive candidate for cancer research and antiviral drugs. Redox modification such as S-glutathionylation of the R1 subunit of mammalian RNR protein has been presumed to regulate the activity of RNR during catalytic cycles. Herein, we report S-glutathionylation of the R2 subunit. We have also shown Grx1 system can efficiently deglutathionylate the S-glutathionylated R2 subunit. Additionally, our data also showed for the very first time S-glutathionylation of mammalian p53R2 subunit that regulates DNA synthesis outside S-phase during DNA damage and repair. Taken together, these data will open new avenues for future research relating to exact physiological significance, target thiols, and/or overall RNR activity due to S-glutathionylation of R2 and p53R2 subunits and provide valuable insights for effective treatment regimes.


Asunto(s)
Proteínas de Ciclo Celular , Replicación del ADN , Glutatión , Subunidades de Proteína , Ribonucleótido Reductasas , Fase S , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Glutatión/química , Glutatión/metabolismo , Ratones , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/metabolismo
7.
J Biol Chem ; 294(34): 12708-12716, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31266802

RESUMEN

Deoxyribonucleotides are DNA building blocks and are produced de novo by reduction of ribose to deoxyribose. This reduction is catalyzed by ribonucleotide reductase (RNR), a heterodimeric tetramer enzyme in mammalian cells, having one of two free radical-containing subunits called R2 and p53R2. R2 is S-phase specific and used for DNA replication, whereas p53R2 functions in DNA repair and mitochondrial DNA synthesis. The larger RNR subunit, R1, has catalytically active cysteine thiols in its buried active site and a C-terminal swinging arm, with a Cys-Leu-Met-Cys sequence suggested to act as a shuttle dithiol/disulfide for electron transport. After each catalytic cycle the active site contains a disulfide, which has to be reduced for turnover. Thioredoxin (Trx) and glutaredoxin (Grx) systems have been implicated as electron donors for the RNR disulfide reduction via the swinging arm. Using mouse R1-R2 and R1-p53R2 complexes, we found here that the catalytic efficiency of the GSH-Grx system is 4-6 times higher than that of the Trx1 system. For both complexes, the Vmax values for Grx are strongly depended on GSH concentrations. The GSH disulfide resulting from the Grx reaction was reduced by NADPH and GSH reductase and this enzyme was essential because reaction with GSH alone yielded only little activity. These results indicate that C-terminal shuttle dithiols of mammalian R1 have a crucial catalytic role and that the GSH-Grx system favors the R1-p53R2 enzyme for DNA replication in hypoxic conditions, mitochondrial DNA synthesis, and in DNA repair outside the S-phase.


Asunto(s)
Electrones , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Ribonucleótido Reductasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ratones , Modelos Moleculares , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética
8.
Chem Res Toxicol ; 33(9): 2441-2445, 2020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32786549

RESUMEN

Tumor cells are characterized by increased reactive oxygen species production in parallel with an enhanced antioxidant system to avoid oxidative damage. The inhibition of antioxidant systems is an effective way to kill cancer cells, and the thioredoxin system or, more specifically, the cytosolic selenocysteine-containing enzyme thioredoxin reductase (TrxR) has become an interesting target for cancer therapy. We show here that the known cytotoxic and apoptosis-inducing osmium carbonyl cluster Os3(CO)10(NCCH3)2 (1) is a nonsubstrate inhibitor of mammalian TrxR, with an IC50 of 5.3 ± 0.9 µM. It inhibits TrxR selectively over the closely related glutathione reductase (GR) and in the presence of excess reduced glutathione (GSH). This inhibition has also been demonstrated in cell lysates, suggesting that TrxR inhibition is a potential apoptotic pathway for 1.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Inhibidores Enzimáticos/farmacología , Osmio/farmacología , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/química , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Femenino , Humanos , Estructura Molecular , Osmio/química , Reductasa de Tiorredoxina-Disulfuro/metabolismo
9.
Anal Biochem ; 568: 24-30, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30597126

RESUMEN

Glutathione is an abundant low-molecular-weight thiol, up to 10 mM in mammalian cells, and exists in three major forms: reduced sulphydryl (GSH), glutathione disulfide (GSSG) or bound to Cys residues in proteins (PSSG). The ratio GSH/GSSG has been used as an indicator of the cells redox level but this parameter can also be estimated by the quantification of PSSG. In fact, PSSGs have the advantage of being more stable than GSSG. Here we present a highly sensitive fluorescent-based method for detection of low concentrations of glutathione in complex samples such as cell lysates, tissues and plasma. The method is based on our previously described protocol to study Glutaredoxin (Grx) activity. The whole procedure was optimized to measure the fluorescence increase of the di-eosin-glutathione disulfide (Di-E-GSSG) reduced by Grx in the presence of Glutathione Reductase and NADPH, keeping GSH as the limiting factor to drive the reaction. The methods to selectively measure PSSG are expensive and not widely accessible, therefore we optimized our glutaredoxin protocol to quantify this post-translational modification using common laboratory equipments. Overall, our method has simplicity and rapidity combined with high sensitivity as its main advantages; therefore, it may be particularly suitable for large-scale clinical studies.


Asunto(s)
Fluorescencia , Glutarredoxinas/metabolismo , Glutatión/análisis , Células Cultivadas , Glutarredoxinas/química , Glutatión/metabolismo , Humanos
10.
Cell Mol Life Sci ; 75(9): 1567-1586, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29327078

RESUMEN

Mammalian thioredoxin reductase (TrxR) is a selenoprotein with three existing isoenzymes (TrxR1, TrxR2, and TrxR3), which is found primarily intracellularly but also in extracellular fluids. The main substrate thioredoxin (Trx) is similarly found (as Trx1 and Trx2) in various intracellular compartments, in blood plasma, and is the cell's major disulfide reductase. Thioredoxin reductase is necessary as a NADPH-dependent reducing agent in biochemical reactions involving Trx. Genetic and environmental factors like selenium status influence the activity of TrxR. Research shows that the Trx/TrxR system plays a significant role in the physiology of the adipose tissue, in carbohydrate metabolism, insulin production and sensitivity, blood pressure regulation, inflammation, chemotactic activity of macrophages, and atherogenesis. Based on recent research, it has been reported that the modulation of the Trx/TrxR system may be considered as a new target in the management of the metabolic syndrome, insulin resistance, and type 2 diabetes, as well as in the treatment of hypertension and atherosclerosis. In this review evidence about a possible role of this system as a marker of the metabolic syndrome is reported.


Asunto(s)
Síndrome Metabólico/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Animales , Aterosclerosis/diagnóstico , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Aterosclerosis/terapia , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Diabetes Mellitus/terapia , Humanos , Hipertensión/diagnóstico , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipertensión/terapia , Síndrome Metabólico/diagnóstico , Síndrome Metabólico/fisiopatología , Síndrome Metabólico/terapia , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Enfermedad del Hígado Graso no Alcohólico/terapia , Obesidad/diagnóstico , Obesidad/metabolismo , Obesidad/fisiopatología , Obesidad/terapia , Pronóstico
11.
PLoS Pathog ; 12(2): e1005442, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26915097

RESUMEN

Nitric oxide (NO) generated by inducible NO synthase (iNOS) is critical for defense against intracellular pathogens but may mediate inflammatory tissue damage. To elucidate the role of iNOS in neuroinflammation, infections with encephalitogenic Trypanosoma brucei parasites were compared in inos(-/-) and wild-type mice. Inos(-/-) mice showed enhanced brain invasion by parasites and T cells, and elevated protein permeability of cerebral vessels, but similar parasitemia levels. Trypanosome infection stimulated T cell- and TNF-mediated iNOS expression in perivascular macrophages. NO nitrosylated and inactivated pro-inflammatory molecules such as NF-κΒp65, and reduced TNF expression and signalling. iNOS-derived NO hampered both TNF- and T cell-mediated parasite brain invasion. In inos(-/-) mice, TNF stimulated MMP, including MMP9 activity that increased cerebral vessel permeability. Thus, iNOS-generated NO by perivascular macrophages, strategically located at sites of leukocyte brain penetration, can serve as a negative feed-back regulator that prevents unlimited influx of inflammatory cells by restoring the integrity of the blood-brain barrier.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalitis/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Animales , Citocinas/metabolismo , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Trypanosoma brucei brucei/metabolismo
12.
J Biol Chem ; 291(33): 17197-208, 2016 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-27261461

RESUMEN

Protein misfolding is implicated in neurodegenerative diseases such as ALS, where mutations of superoxide dismutase 1 (SOD1) account for about 20% of the inherited mutations. Human SOD1 (hSOD1) contains four cysteines, including Cys(57) and Cys(146), which have been linked to protein stability and folding via forming a disulfide bond, and Cys(6) and Cys(111) as free thiols. But the roles of the cellular oxidation-reduction (redox) environment in SOD1 folding and aggregation are not well understood. Here we explore the effects of cellular redox systems on the aggregation of hSOD1 proteins. We found that the known hSOD1 mutations G93A and A4V increased the capability of the thioredoxin and glutaredoxin systems to reduce hSOD1 compared with wild-type hSOD1. Treatment with inhibitors of these redox systems resulted in an increase of hSOD1 aggregates in the cytoplasm of cells transfected with mutants but not in cells transfected with wild-type hSOD1 or those containing a secondary C111G mutation. This aggregation may be coupled to changes in the redox state of the G93A and A4V mutants upon mild oxidative stress. These results strongly suggest that the thioredoxin and glutaredoxin systems are the key regulators for hSOD1 aggregation and may play critical roles in the pathogenesis of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Estrés Oxidativo , Agregación Patológica de Proteínas , Pliegue de Proteína , Superóxido Dismutasa-1 , Sustitución de Aminoácidos , Esclerosis Amiotrófica Lateral/enzimología , Esclerosis Amiotrófica Lateral/genética , Animales , Línea Celular Tumoral , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Humanos , Mutación Missense , Oxidación-Reducción , Agregación Patológica de Proteínas/enzimología , Agregación Patológica de Proteínas/genética , Ratas , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
13.
J Biol Chem ; 291(46): 24036-24040, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27645994

RESUMEN

The human genome contains 25 genes coding for selenocysteine-containing proteins (selenoproteins). These proteins are involved in a variety of functions, most notably redox homeostasis. Selenoprotein enzymes with known functions are designated according to these functions: TXNRD1, TXNRD2, and TXNRD3 (thioredoxin reductases), GPX1, GPX2, GPX3, GPX4, and GPX6 (glutathione peroxidases), DIO1, DIO2, and DIO3 (iodothyronine deiodinases), MSRB1 (methionine sulfoxide reductase B1), and SEPHS2 (selenophosphate synthetase 2). Selenoproteins without known functions have traditionally been denoted by SEL or SEP symbols. However, these symbols are sometimes ambiguous and conflict with the approved nomenclature for several other genes. Therefore, there is a need to implement a rational and coherent nomenclature system for selenoprotein-encoding genes. Our solution is to use the root symbol SELENO followed by a letter. This nomenclature applies to SELENOF (selenoprotein F, the 15-kDa selenoprotein, SEP15), SELENOH (selenoprotein H, SELH, C11orf31), SELENOI (selenoprotein I, SELI, EPT1), SELENOK (selenoprotein K, SELK), SELENOM (selenoprotein M, SELM), SELENON (selenoprotein N, SEPN1, SELN), SELENOO (selenoprotein O, SELO), SELENOP (selenoprotein P, SeP, SEPP1, SELP), SELENOS (selenoprotein S, SELS, SEPS1, VIMP), SELENOT (selenoprotein T, SELT), SELENOV (selenoprotein V, SELV), and SELENOW (selenoprotein W, SELW, SEPW1). This system, approved by the HUGO Gene Nomenclature Committee, also resolves conflicting, missing, and ambiguous designations for selenoprotein genes and is applicable to selenoproteins across vertebrates.


Asunto(s)
Selenoproteínas/clasificación , Selenoproteínas/genética , Humanos , Terminología como Asunto
14.
Biochim Biophys Acta ; 1860(6): 1265-71, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26971857

RESUMEN

BACKGROUND: Bacillus anthracis is the causative agent of anthrax, a disease associated with a very high mortality rate in its invasive forms. METHODS: We studied a number of ebselen analogs as inhibitors of B. anthracis thioredoxin reductase and their antibacterial activity on Bacillus subtilis, Staphylococcus aureus, Bacillus cereus and Mycobacterium tuberculosis. RESULTS: The most potent compounds in the series gave IC(50) values down to 70 nM for the pure enzyme and minimal inhibitory concentrations (MICs) down to 0.4 µM (0.12 µg/ml) for B. subtilis, 1.5 µM (0.64 µg/ml) for S. aureus, 2 µM (0.86 µg/ml) for B. cereus and 10 µg/ml for M. tuberculosis. Minimal bactericidal concentrations (MBCs) were found at 1-1.5 times the MIC, indicating a general, class-dependent, bactericidal mode of action. The combined bacteriological and enzymological data were used to construct a preliminary structure-activity-relationship for the benzoisoselenazol class of compounds. When S. aureus and B. subtilis were exposed to ebselen, we were unable to isolate resistant mutants on both solid and in liquid medium suggesting a high resistance barrier. CONCLUSIONS: These results suggest that ebselen and analogs thereof could be developed into a novel antibiotic class, useful for the treatment of infections caused by B. anthracis, S. aureus, M. tuberculosis and other clinically important bacteria. Furthermore, the high barrier against resistance development is encouraging for further drug development. GENERAL SIGNIFICANCE: We have characterized the thioredoxin system from B. anthracis as a novel drug target and ebselen and analogs thereof as a potential new class of antibiotics targeting several important human pathogens.


Asunto(s)
Antibacterianos/farmacología , Azoles/farmacología , Bacillus anthracis/enzimología , Bacillus/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Staphylococcus aureus/efectos de los fármacos , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Secuencia de Aminoácidos , Isoindoles , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular
15.
Glia ; 65(9): 1521-1534, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28618115

RESUMEN

Demyelinated brain lesions, a hallmark of autoimmune neuroinflammatory diseases like multiple sclerosis, result from oligodendroglial cell damage. Activated microglia are considered a major source of nitric oxide and subsequent peroxynitrite-mediated damage of myelin. Here, we provide biochemical and biophysical evidence that the oxidoreductase glutaredoxin 2 inhibits peroxynitrite formation by transforming nitric oxide into dinitrosyl-diglutathionyl-iron-complexes. Glutaredoxin 2 levels influence both survival rates of primary oligodendrocyte progenitor cells and preservation of myelin structure in cerebellar organotypic slice cultures challenged with activated microglia or nitric oxide donors. Of note, glutaredoxin 2-mediated protection is not linked to its enzymatic activity as oxidoreductase, but to the disassembly of its uniquely coordinated iron-sulfur cluster using glutathione as non-protein ligand. The protective effect of glutaredoxin 2 is connected to decreased protein carbonylation and nitration. In line, brain lesions of mice suffering from experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, show decreased glutaredoxin 2 expression and increased nitrotyrosine formation indicating that this type of protection is missing in the inflamed central nervous system. Our findings link inorganic biochemistry to neuroinflammation and identify glutaredoxin 2 as a protective factor against neuroinflammation-mediated myelin damage. Thus, improved availability of glutathione-coordinated iron-sulfur clusters emerges as a potential therapeutic approach in inflammatory demyelination.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Glutarredoxinas/metabolismo , Microglía/metabolismo , Óxido Nítrico/metabolismo , Oligodendroglía/metabolismo , Animales , Cerebelo/metabolismo , Cerebelo/patología , Encefalomielitis Autoinmune Experimental/patología , Escherichia coli , Femenino , Glutarredoxinas/genética , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Inflamación/metabolismo , Inflamación/patología , Ratones Endogámicos C57BL , Microglía/patología , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuroprotección/fisiología , Oligodendroglía/patología , Ácido Peroxinitroso/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Schistosoma japonicum , Técnicas de Cultivo de Tejidos
16.
Proc Natl Acad Sci U S A ; 111(19): 6964-9, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24778250

RESUMEN

Thioredoxin-related protein of 14 kDa (TRP14, also called TXNDC17 for thioredoxin domain containing 17, or TXNL5 for thioredoxin-like 5) is an evolutionarily well-conserved member of the thioredoxin (Trx)-fold protein family that lacks activity with classical Trx1 substrates. However, we discovered here that human TRP14 has a high enzymatic activity in reduction of l-cystine, where the catalytic efficiency (2,217 min(-1)⋅µM(-1)) coupled to Trx reductase 1 (TrxR1) using NADPH was fivefold higher compared with Trx1 (418 min(-1)⋅µM(-1)). Moreover, the l-cystine reduction with TRP14 was in contrast to that of Trx1 fully maintained in the presence of a protein disulfide substrate of Trx1 such as insulin, suggesting that TRP14 is a more dedicated l-cystine reductase compared with Trx1. We also found that TRP14 is an efficient S-denitrosylase with similar efficiency as Trx1 in catalyzing TrxR1-dependent denitrosylation of S-nitrosylated glutathione or of HEK293 cell-derived S-nitrosoproteins. Consequently, nitrosylated and thereby inactivated caspase 3 or cathepsin B could be reactivated through either Trx1- or TRP14-catalyzed denitrosylation reactions. TRP14 was also, in contrast to Trx1, completely resistant to inactivation by high concentrations of hydrogen peroxide. The oxidoreductase activities of TRP14 thereby complement those of Trx1 and must therefore be considered for the full understanding of enzymatic control of cellular thiols and nitrosothiols.


Asunto(s)
Cistina/metabolismo , Estrés Oxidativo/fisiología , Tiorredoxina Reductasa 1/metabolismo , Tiorredoxinas/metabolismo , Carcinoma de Células Escamosas , Cisteína/metabolismo , Activación Enzimática/fisiología , Glutatión/metabolismo , Células HEK293 , Células HT29 , Humanos , Peróxido de Hidrógeno/farmacología , Neoplasias Pulmonares , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , NADP/metabolismo , Óxido Nítrico/metabolismo , Oxidantes/farmacología , Oxidación-Reducción , Especificidad por Sustrato , Azufre/metabolismo , Tiorredoxina Reductasa 1/genética , Tiorredoxinas/genética
17.
Anal Biochem ; 499: 24-33, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26836485

RESUMEN

Glutaredoxins catalyze glutathione-dependent disulfide oxidoreductions, particularly reduction of glutathione (GSH)-protein mixed disulfides. Mammalian glutaredoxins are present in the cytosol/nucleus as Grx1 or in mitochondria as Grx2a. Here we describe di-eosin-glutathione disulfide (Di-E-GSSG) as a new tool to study glutaredoxin (Grx) activity. Di-E-GSSG has almost no fluorescence in its disulfide form due to self-quenching, whereas the reduced form (E-GSH) has a large fluorescence emission at 545 nm after excitation at 520 nm. Di-E-GSSG was a very poor substrate for glutathione reductase, but we discovered that the molecule was an excellent substrate for glutaredoxin in a coupled assay system with GSH, nicotinamide adenine dinucleotide phosphate (NADPH), and glutathione reductase or with lipoamide, NADH, and lipoamide dehydrogenase. In addition, Di-E-GSSG was used to glutathionylate the free SH group of bovine serum albumin (BSA), yielding eosin-glutathionylated BSA (E-GS-BSA) readily observed in ultraviolet (UV) light. E-GS-BSA also displayed a quenched fluorescence, and its Grx-catalyzed reduction could be followed by the formation of E-GSH by fluorescence emission using microtiter plates. This way of measuring Grx activity provided an ultrasensitive method that detected Grx1 and Grx2 at picomolar levels. Human Grx1 was readily quantified in 40 µl of plasma and determined to be 680 ± 208 pM in healthy controls.


Asunto(s)
Disulfuros/metabolismo , Eosina Amarillenta-(YS)/química , Fluorescencia , Colorantes Fluorescentes/metabolismo , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Proteína S/metabolismo , Animales , Bovinos , Disulfuros/química , Colorantes Fluorescentes/química , Glutarredoxinas/sangre , Glutarredoxinas/química , Glutatión/química , Humanos , Estructura Molecular , Proteína S/química , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/metabolismo , Espectrometría de Fluorescencia
18.
Proc Natl Acad Sci U S A ; 110(50): 20057-62, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277839

RESUMEN

Embryonic development depends on complex and precisely orchestrated signaling pathways including specific reduction/oxidation cascades. Oxidoreductases of the thioredoxin family are key players conveying redox signals through reversible posttranslational modifications of protein thiols. The importance of this protein family during embryogenesis has recently been exemplified for glutaredoxin 2, a vertebrate-specific glutathione-disulfide oxidoreductase with a critical role for embryonic brain development. Here, we discovered an essential function of glutaredoxin 2 during vascular development. Confocal microscopy and time-lapse studies based on two-photon microscopy revealed that morpholino-based knockdown of glutaredoxin 2 in zebrafish, a model organism to study vertebrate embryogenesis, resulted in a delayed and disordered blood vessel network. We were able to show that formation of a functional vascular system requires glutaredoxin 2-dependent reversible S-glutathionylation of the NAD(+)-dependent protein deacetylase sirtuin 1. Using mass spectrometry, we identified a cysteine residue in the conserved catalytic region of sirtuin 1 as target for glutaredoxin 2-specific deglutathionylation. Thereby, glutaredoxin 2-mediated redox regulation controls enzymatic activity of sirtuin 1, a mechanism we found to be conserved between zebrafish and humans. These results link S-glutathionylation to vertebrate development and successful embryonic angiogenesis.


Asunto(s)
Sistema Cardiovascular/embriología , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Sirtuina 1/metabolismo , Animales , Western Blotting , Cartilla de ADN/genética , Técnicas de Silenciamiento del Gen , Glutarredoxinas/genética , Células HeLa , Humanos , Espectrometría de Masas , Microscopía Confocal , Oxidación-Reducción , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Imagen de Lapso de Tiempo , Pez Cebra
19.
Toxicol Appl Pharmacol ; 286(3): 216-23, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25981166

RESUMEN

Mercury (Hg) is a strong toxicant affecting mainly the central nervous, renal, cardiovascular and immune systems. Thiomersal (TM) is still in use in medical practice as a topical antiseptic and as a preservative in multiple dose vaccines, routinely given to young children in some developing countries, while other forms of mercury such as methylmercury represent an environmental and food hazard. The aim of the present study was to determine the effects of thiomersal (TM) and its breakdown product ethylmercury (EtHg) on the thioredoxin system and NADP(+)-dependent dehydrogenases of the pentose phosphate pathway. Results show that TM and EtHg inhibited the thioredoxin system enzymes in purified suspensions, being EtHg comparable to methylmercury (MeHg). Also, treatment of neuroblastoma and liver cells with TM or EtHg decreased cell viability (GI50: 1.5 to 20µM) and caused a significant (p<0.05) decrease in the overall activities of thioredoxin (Trx) and thioredoxin reductase (TrxR) in a concentration- and time-dependent manner in cell lysates. Compared to control, the activities of Trx and TrxR in neuroblastoma cells after EtHg incubation were reduced up to 60% and 80% respectively, whereas in hepatoma cells the reduction was almost 100%. In addition, the activities of glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase were also significantly inhibited by all mercurials, with inhibition intensity of Hg(2+)>MeHg≈EtHg>TM (p<0.05). Cell incubation with sodium selenite alleviated the inhibitory effects on TrxR and glucose-6-phosphate dehydrogenase. Thus, the molecular mechanism of toxicity of TM and especially of its metabolite EtHg encompasses the blockage of the electrons from NADPH via the thioredoxin system.


Asunto(s)
Compuestos de Etilmercurio/toxicidad , NADPH Deshidrogenasa/antagonistas & inhibidores , Vía de Pentosa Fosfato/efectos de los fármacos , Timerosal/toxicidad , Tiorredoxinas/antagonistas & inhibidores , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , NADPH Deshidrogenasa/metabolismo , Vía de Pentosa Fosfato/fisiología , Tiorredoxinas/metabolismo
20.
J Biol Chem ; 288(45): 32241-32247, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24062305

RESUMEN

The mammalian cytosolic thioredoxin system, comprising thioredoxin (Trx), Trx reductase, and NADPH, is the major protein-disulfide reductase of the cell and has numerous functions. Besides the active site thiols, human Trx1 contains three non-active site cysteine residues at positions 62, 69, and 73. A two-disulfide form of Trx1, containing an active site disulfide between Cys-32 and Cys-35 and a non-active site disulfide between Cys-62 and Cys-69, is inactive either as a disulfide reductase or as a substrate for Trx reductase. This could possibly provide a structural switch affecting Trx1 function during oxidative stress and redox signaling. We found that two-disulfide Trx1 was generated in A549 cells under oxidative stress. In vitro data showed that two-disulfide Trx1 was generated from oxidation of Trx1 catalyzed by peroxiredoxin 1 in the presence of H2O2. The redox Western blot data indicated that the glutaredoxin system protected Trx1 in HeLa cells from oxidation caused by ebselen, a superfast oxidant for Trx1. Our results also showed that physiological concentrations of glutathione, NADPH, and glutathione reductase reduced the non-active site disulfide in vitro. This reaction was stimulated by glutaredoxin 1 via the so-called monothiol mechanism. In conclusion, reversible oxidation of the non-active site disulfide of Trx1 is suggested to play an important role in redox regulation and cell signaling via temporal inhibition of its protein-disulfide reductase activity for the transmission of oxidative signals under oxidative stress.


Asunto(s)
Glutarredoxinas/metabolismo , Estrés Oxidativo/fisiología , Peroxirredoxinas/metabolismo , Transducción de Señal/fisiología , Tiorredoxinas/metabolismo , Glutarredoxinas/genética , Glutatión/genética , Glutatión/metabolismo , Glutatión Reductasa/genética , Glutatión Reductasa/metabolismo , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , NADP/genética , NADP/metabolismo , Oxidantes/farmacología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Peroxirredoxinas/genética , Transducción de Señal/efectos de los fármacos , Tiorredoxinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA