Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Redox Biol ; 64: 102806, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37413766

RESUMEN

The aim of this study was to examine, in biochemical detail, the functional role of the Arg152 residue in the selenoprotein Glutathione Peroxidase 4 (GPX4), whose mutation to His is involved in Sedaghatian-type Spondylometaphyseal Dysplasia (SSMD). Wild-type and mutated recombinant enzymes with selenopcysteine (Sec) at the active site, were purified and structurally characterized to investigate the impact of the R152H mutation on enzymatic function. The mutation did not affect the peroxidase reaction's catalytic mechanism, and the kinetic parameters were qualitatively similar between the wild-type enzyme and the mutant when mixed micelles and monolamellar liposomes containing phosphatidylcholine and its hydroperoxide derivatives were used as substrate. However, in monolamellar liposomes also containing cardiolipin, which binds to a cationic area near the active site of GPX4, including residue R152, the wild-type enzyme showed a non-canonical dependency of the reaction rate on the concentration of both enzyme and membrane cardiolipin. To explain this oddity, a minimal model was developed encompassing the kinetics of both the enzyme interaction with the membrane and the catalytic peroxidase reaction. Computational fitting of experimental activity recordings showed that the wild-type enzyme was surface-sensing and prone to "positive feedback" in the presence of cardiolipin, indicating a positive cooperativity. This feature was minimal, if any, in the mutant. These findings suggest that GPX4 physiology in cardiolipin containing mitochondria is unique, and emerges as a likely target of the pathological dysfunction in SSMD.


Asunto(s)
Cardiolipinas , Liposomas , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Cardiolipinas/metabolismo , Mutación
2.
Free Radic Biol Med ; 188: 117-133, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35718302

RESUMEN

The purification of a protein inhibiting lipid peroxidation led to the discovery of the selenoperoxidase GPx4 forty years ago. Thus, the evidence of the enzymatic activity was reached after identifying the biological effect and unambiguously defined the relationship between the biological function and the enzymatic activity. In the syllogism where GPx4 inhibits lipid peroxidation and its inhibition is lethal, cell death is operated by lipid peroxidation. Based on this rationale, this form of cell death emerged as regulated iron-enforced oxygen toxicity and was named ferroptosis in 2012. In the last decades, we learned that reduction of lipid hydroperoxides is indispensable and, in cooperation with prooxidant systems, controls the critical steady state of lipid peroxidation. This concept defined the GPx4 reaction as both the target for possible anti-cancer therapy and if insufficient, as cause of degenerative diseases. We know the reaction mechanism, but the details of the interaction at the membrane cytosol interface are still poorly defined. We know the gene structure, but the knowledge about expression control is still limited. The same holds true for post-transcriptional modifications. Reverse genetics indicate that GPx4 has a role in inflammation, immunity, and differentiation, but the observations emerging from these studies need a more specifically addressed biochemical evidence. Finally, the role of GPx4 in spermatogenesis disclosed an area unconnected to lipid peroxidation. In its mitochondrial and nuclear form, the peroxidase catalyzes the oxidation of protein thiols in two specific aspects of sperm maturation: stabilization of the mid-piece and chromatin compaction. Thus, although available evidence converges to the notion that GPx4 activity is vital due to the inhibition of lipid peroxidation, it is reasonable to foresee other unknown aspects of the GPx4 reaction to be disclosed.


Asunto(s)
Ferroptosis , Semen , Antioxidantes/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Humanos , Peroxidación de Lípido , Peróxidos Lipídicos/metabolismo , Masculino , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Semen/metabolismo
4.
Redox Biol ; 46: 102070, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34304108

RESUMEN

Selenoproteins are translated via animal domain-specific elongation machineries that redefine dedicated UGA opal codons from termination of translation to selenocysteine (Sec) insertion, utilizing specific tRNA species and Sec-specific elongation factors. This has made recombinant production of mammalian selenoproteins in E. coli technically challenging but recently we developed a methodology that enables such production, using recoding of UAG for Sec in an RF1-deficient host strain. Here we used that approach for production of the human glutathione peroxidases 1, 2 and 4 (GPX1, GPX2 and GPX4), with all these three enzymes being important antioxidant selenoproteins. Among these, GPX4 is the sole embryonically essential enzyme, and is also known to be essential for spermatogenesis as well as protection from cell death through ferroptosis. Enzyme kinetics, ICP-MS and mass spectrometry analyses of the purified recombinant proteins were used to characterize selenoprotein characteristics and their Sec contents. This revealed a unique phenomenon of one-codon skipping, resulting in a lack of a single amino acid at the position corresponding to the selenocysteine (Sec) residue, in about 30% of the recombinant GPX isoenzyme products. We furthermore confirmed the previously described UAG suppression with Lys or Gln as well as a minor suppression with Tyr, together resulting in about 20% Sec contents in the full-length proteins. No additional frameshifts or translational errors were detected. We subsequently found that Sec-containing GPX4 could be further purified over a bromosulfophthalein-column, yielding purified recombinant GPX4 with close to complete Sec contents. This production method for homogenously purified GPX4 should help to further advance the studies of this important selenoprotein.


Asunto(s)
Escherichia coli , Sulfobromoftaleína , Animales , Codón de Terminación , Escherichia coli/genética , Humanos , Masculino , Selenocisteína , Selenoproteínas/genética
5.
Antioxid Redox Signal ; 35(8): 595-601, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34036804

RESUMEN

Dr. Regina Brigelius-Flohé (PhD 1978) is recognized here as redox pioneer because she has published an article on redox biology, as first author, that has been cited >1000 times, plus four articles cited >500 times, and a total of 30 articles cited >100 times. She obtained her doctorate in biochemistry at the Institute of Biochemistry of the University of Münster, Germany. She held positions in both, academia (Münster, Munich, Düsseldorf, Hannover, and Potsdam, Germany) and industry (Aachen, Germany). Dr. Brigelius-Flohé is the pioneer who, as head of the department of biochemistry of micronutrients of the German Institute of Human Nutrition (DIfE; Potsdam-Rehbrücke, Germany), worked out the metabolism of tocopherols and tocotrienols ("Key Finding 1"). She was the first to sequence glutathione peroxidase 4 (GPx4) ("Key Finding 2"), and unraveled the role of selenium, in particular of GPxs, in inflammation and carcinogenesis ("Key Finding 3"). Her contributions, thus, focused on serious biomedical problems such as nutrition, inflammation, and carcinogenesis. She has been a member of the scientific advisory board of the German Society of Biochemistry and Molecular Biology for 6 years and was president of SFRR-Europe in 2005-2006. She edited several books and serves on the editorial board of journals in the fields of nutrition, free radicals, and redox regulation. Antioxid. Redox Signal. 35, 595-601.


Asunto(s)
Selenio , Bioquímica/historia , Humanos , Oxidación-Reducción , Selenio/metabolismo
6.
Free Radic Biol Med ; 167: 45-53, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33711415

RESUMEN

Ferroptosis is a non-accidental, regulated form of cell death operated by lipid peroxidation under strict control of GPx4 activity. This is consistent with the notion that lipid peroxidation is initiated by radicals produced from decomposition of traces of pre-existing lipid hydroperoxides. The question, therefore, emerges about the formation of these traces of lipid hydroperoxides interacting with Fe2+. In the most realistic option, they are produced by oxygen activated species generated during aerobic metabolism. Screening for metabolic sources of superoxide supporting ferroptosis induced by GSH depletion, we failed to detect, in our cell model, a role of respiratory chain. We observed instead that the pyruvate dehydrogenase complex -as other α keto acid dehydrogenases already known as a major source of superoxide in mitochondria- supports ferroptosis. The opposite effect on ferroptosis by silencing either the E1 or the E3 subunit of the pyruvate dehydrogenase complex pointed out the autoxidation of dihydrolipoamide as the source of superoxide. We finally observed that GSH depletion activates superoxide production, seemingly through the inhibition of the specific kinase that inhibits pyruvate dehydrogenase. In summary, this set of data is compatible with a scenario where the more electrophilic status produced by GSH depletion not only activates ferroptosis by preventing GPx4 activity, but also favors the formation of lipid hydroperoxides. In an attractive perspective of tissue homeostasis, it is the activation of energetic metabolism associated to a decreased nucleophilic tone that, besides supporting energy demanding proliferation, also sensitizes cells to a regulated form of death.


Asunto(s)
Ferroptosis , Muerte Celular , Peroxidación de Lípido , Peróxidos Lipídicos , Ácido Pirúvico
7.
Free Radic Biol Med ; 152: 175-185, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32165281

RESUMEN

Ferroptosis (FPT) is a form of cell death due to missed control of membrane lipid peroxidation (LPO). According to the axiomatic definition of non-accidental cell death, LPO takes place in a scenario of altered homeostasis. FPT, differently from apoptosis, occurs in the absence of any known specific genetically encoded death pathway or specific agonist, and thus must be rated as a regulated, although not "programmed", death pathway. It follows that LPO is under a homeostatic metabolic control and is only permitted when indispensable constraints are satisfied and the antiperoxidant machinery collapses. The activity of the selenoperoxidase Glutathione Peroxidase 4 (GPx4) is the cornerstone of the antiperoxidant defence. Converging evidence on both mechanism of LPO and GPx4 enzymology indicates that LPO is initiated by alkoxyl radicals produced by ferrous iron from the hydroperoxide derivatives of lipids (LOOH), traces of which are the unavoidable drawback of aerobic metabolism. FPT takes place when a threshold has been exceeded. This occurs when the major conditions are satisfied: i) oxygen metabolism leading to the continuous formation of traces of LOOH from phospholipid-containing polyunsaturated fatty acids; ii) missed enzymatic reduction of LOOH; iii) availability of ferrous iron from the labile iron pool. Although the effectors impacting on homeostasis and leading to FPT in physiological conditions are not known, from the available knowledge on LPO and GPx4 enzymology we propose that it is aerobic life itself that, while supporting bioenergetics, is also a critical requisite of FPT. Yet, when the homeostatic control of the steady state between LOOH formation and reduction is lost, LPO is activated and FPT is executed.


Asunto(s)
Ferroptosis , Apoptosis , Muerte Celular , Peroxidación de Lípido , Fosfolípido Hidroperóxido Glutatión Peroxidasa
8.
Free Radic Biol Med ; 147: 80-89, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31857233

RESUMEN

GPx8 is a glutathione peroxidase homolog inserted in the membranes of endoplasmic reticulum (ER), where it seemingly plays a role in controlling redox status by preventing the spill of H2O2. We addressed the impact of GPx8 silencing on the lipidome of microsomal membranes, using stably GPx8-silenced HeLa cells. The two cell lines were clearly separated by Principal Component Analysis (PCA) and Partial Least Square Discriminant analysis (PLS-DA) of lipidome. Considering in detail the individual lipid classes, we observed that unsaturated glycerophospholipids (GPL) decreased, while only in phosphatidylinositols (PI) a substitution of monounsaturated fatty acids (MUFA) for polyunsaturated fatty acids (PUFA) was observed. Among sphingolipids (SL), ceramides (CER) decreased while sphingomyelins (SM) and neutral glycophingolipids (nGSL) increased. Here, in addition, longer chains than in controls in the amide fatty acid were present. The increase up to four folds of the CER (d18:1; c24:0) containing three hexose units, was the most remarkable species increasing in the differential lipidome of siGPx8 cells. Quantitative RT-PCR complied with lipidomic analysis specifically showing an increased expression of: i) acyl-CoA synthetase 5 (ACSL5); ii) CER synthase 2 and 4; iii) CER transporter (CERT); iv) UDP-glucosyl transferase (UDP-GlcT), associated to a decreased expression of UDP-galactosyl transferase (UDP-GalT). A role of the unfolded protein response (UPR) and the spliced form of the transcription factor XBP1 on the transcriptional changes of GPx8 silenced cells was ruled-out. Similarly, also the involvement of Nrf2 and NF-κB. Altogether our results indicate that GPx8-silencing of HeLa yields a membrane depleted by about 24% of polyunsaturated GPL and a corresponding increase of saturated or monounsaturated SM and specific nGSL. This is tentatively interpreted as an adaptive mechanism leading to an increased resistance to radical oxidations. Moreover, the marked shift of fatty acid composition of PI emerges as a possibly relevant issue in respect to the impact of GPx8 on signaling pathways.


Asunto(s)
Retículo Endoplásmico , Peróxido de Hidrógeno , Ceramidas , Glutatión Peroxidasa/genética , Células HeLa , Humanos , Peroxidasas
9.
Redox Biol ; 28: 101328, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31574461

RESUMEN

Ferroptosis is a form of cell death primed by iron and lipid hydroperoxides and prevented by GPx4. Ferrostatin-1 (fer-1) inhibits ferroptosis much more efficiently than phenolic antioxidants. Previous studies on the antioxidant efficiency of fer-1 adopted kinetic tests where a diazo compound generates the hydroperoxyl radical scavenged by the antioxidant. However, this reaction, accounting for a chain breaking effect, is only minimally useful for the description of the inhibition of ferrous iron and lipid hydroperoxide dependent peroxidation. Scavenging lipid hydroperoxyl radicals, indeed, generates lipid hydroperoxides from which ferrous iron initiates a new peroxidative chain reaction. We show that when fer-1 inhibits peroxidation, initiated by iron and traces of lipid hydroperoxides in liposomes, the pattern of oxidized species produced from traces of pre-existing hydroperoxides is practically identical to that observed following exhaustive peroxidation in the absence of the antioxidant. This supported the notion that the anti-ferroptotic activity of fer-1 is actually due to the scavenging of initiating alkoxyl radicals produced, together with other rearrangement products, by ferrous iron from lipid hydroperoxides. Notably, fer-1 is not consumed while inhibiting iron dependent lipid peroxidation. The emerging concept is that it is ferrous iron itself that reduces fer-1 radical. This was supported by electroanalytical evidence that fer-1 forms a complex with iron and further confirmed in cells by fluorescence of calcein, indicating a decrease of labile iron in the presence of fer-1. The notion of such as pseudo-catalytic cycle of the ferrostatin-iron complex was also investigated by means of quantum mechanics calculations, which confirmed the reduction of an alkoxyl radical model by fer-1 and the reduction of fer-1 radical by ferrous iron. In summary, GPx4 and fer-1 in the presence of ferrous iron, produces, by distinct mechanism, the most relevant anti-ferroptotic effect, i.e the disappearance of initiating lipid hydroperoxides.


Asunto(s)
Ciclohexilaminas/farmacología , Ferroptosis/efectos de los fármacos , Fenilendiaminas/farmacología , Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Cromatografía Liquida , Ciclohexilaminas/química , Teoría Funcional de la Densidad , Relación Dosis-Respuesta a Droga , Ferroptosis/genética , Hidrógeno/química , Peroxidación de Lípido/efectos de los fármacos , Peróxidos Lipídicos/metabolismo , Lipidómica/métodos , Lípidos/química , Modelos Moleculares , Estructura Molecular , Oxidación-Reducción , Fenilendiaminas/química , Espectrometría de Masas en Tándem
10.
FEBS Lett ; 594(4): 611-624, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31581313

RESUMEN

Ras-selective lethal small molecule 3 (RSL3), a drug candidate prototype for cancer chemotherapy, triggers ferroptosis by inactivating the glutathione peroxidase glutathione peroxidase 4 (GPx4). Here, we report the purification of the protein indispensable for GPx4 inactivation by RSL3. Mass spectrometric analysis identified 14-3-3 isoforms as candidates, and recombinant human 14-3-3ε confirms the identification. The function of 14-3-3ε is redox-regulated. Moreover, overexpression or silencing of the gene coding for 14-3-3ε consistently controls the inactivation of GPx4 by RSL3. The interaction of GPx4 with a redox-regulated adaptor protein operating in cell signaling further contributes to frame it within redox-regulated pathways of cell survival and death and opens new therapeutic perspectives.


Asunto(s)
Proteínas 14-3-3/metabolismo , Carbolinas/farmacología , Ferroptosis/efectos de los fármacos , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Animales , Citosol/efectos de los fármacos , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Ratas
11.
Antioxid Redox Signal ; 29(1): 61-74, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28462584

RESUMEN

SIGNIFICANCE: Iron-dependent lipid peroxidation is a complex oxidative process where phospholipid hydroperoxides (PLOOH) are produced in membranes and finally transformed into a series of decomposition products, some of which are endowed with biological activity. It is specifically prevented by glutathione peroxidase 4 (GPx4), the selenoenzyme that reduces PLOOH by glutathione (GSH). PLOOH is both a product and the major initiator of peroxidative chain reactions, as well as an activator of lipoxygenases. α-Tocopherol both specifically breaks peroxidative chain propagation and inhibits lipoxygenases. Thus, GPx4, GSH, and α-tocopherol are integrated in a concerted anti-peroxidant mechanism. Recent Advances: Ferroptosis has been recently identified as a cell death subroutine that is specifically activated by missing GPx4 activity and inhibited by iron chelation or α-tocopherol supplementation. Ferroptosis induction may underlie spontaneous human diseases, such as major neurodegeneration and neuroinflammation, causing an excessive cell death. The basic mechanism of ferroptosis, therefore, fits the features of activation of lipid peroxidation. CRITICAL ISSUES: Still lacking are convincing proofs that lipoxygenases are involved in ferroptosis. Also, unknown are the molecules eventually killing cells and the mechanisms underlying the drop of the cellular anti-peroxidant capacity. FUTURE DIRECTIONS: Molecular events and mechanisms of ferroptosis to be unraveled and validated on animal models are GPx4 inactivation, role of GSH concentration, increased iron availability, and membrane structure and composition. This is expected to drive drug discovery that is aimed at halting cell death in degenerative diseases or boosting it in cancer cells. Antioxid. Redox Signal. 29, 61-74.


Asunto(s)
Muerte Celular , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido , Animales , Muerte Celular/efectos de los fármacos , Glutatión Peroxidasa/antagonistas & inhibidores , Humanos , Quelantes del Hierro/farmacología , Peroxidación de Lípido/efectos de los fármacos , Fosfolípido Hidroperóxido Glutatión Peroxidasa , alfa-Tocoferol/farmacología
12.
Geroscience ; 39(5-6): 499-550, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29270905

RESUMEN

A paradox is a seemingly absurd or impossible concept, proposition, or theory that is often difficult to understand or explain, sometimes apparently self-contradictory, and yet ultimately correct or true. How is it possible, for example, that oxygen "a toxic environmental poison" could be also indispensable for life (Beckman and Ames Physiol Rev 78(2):547-81, 1998; Stadtman and Berlett Chem Res Toxicol 10(5):485-94, 1997)?: the so-called Oxygen Paradox (Davies and Ursini 1995; Davies Biochem Soc Symp 61:1-31, 1995). How can French people apparently disregard the rule that high dietary intakes of cholesterol and saturated fats (e.g., cheese and paté) will result in an early death from cardiovascular diseases (Renaud and de Lorgeril Lancet 339(8808):1523-6, 1992; Catalgol et al. Front Pharmacol 3:141, 2012; Eisenberg et al. Nat Med 22(12):1428-1438, 2016)?: the so-called, French Paradox. Doubtless, the truth is not a duality and epistemological bias probably generates apparently self-contradictory conclusions. Perhaps nowhere in biology are there so many apparently contradictory views, and even experimental results, affecting human physiology and pathology as in the fields of free radicals and oxidative stress, antioxidants, foods and drinks, and dietary recommendations; this is particularly true when issues such as disease-susceptibility or avoidance, "healthspan," "lifespan," and ageing are involved. Consider, for example, the apparently paradoxical observation that treatment with low doses of a substance that is toxic at high concentrations may actually induce transient adaptations that protect against a subsequent exposure to the same (or similar) toxin. This particular paradox is now mechanistically explained as "Adaptive Homeostasis" (Davies Mol Asp Med 49:1-7, 2016; Pomatto et al. 2017a; Lomeli et al. Clin Sci (Lond) 131(21):2573-2599, 2017; Pomatto and Davies 2017); the non-damaging process by which an apparent toxicant can activate biological signal transduction pathways to increase expression of protective genes, by mechanisms that are completely different from those by which the same agent induces toxicity at high concentrations. In this review, we explore the influences and effects of paradoxes such as the Oxygen Paradox and the French Paradox on the etiology, progression, and outcomes of many of the major human age-related diseases, as well as the basic biological phenomenon of ageing itself.


Asunto(s)
Adaptación Fisiológica , Envejecimiento/genética , Dieta Rica en Proteínas/estadística & datos numéricos , Hipercolesterolemia/epidemiología , Estrés Oxidativo/fisiología , Oxígeno/metabolismo , Anciano , Anciano de 80 o más Años , Envejecimiento/fisiología , Femenino , Francia , Radicales Libres/metabolismo , Evaluación Geriátrica , Humanos , Masculino , Persona de Mediana Edad , Medición de Riesgo
13.
Free Radic Biol Med ; 112: 1-11, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28709976

RESUMEN

GPx4 is a monomeric glutathione peroxidase, unique in reducing the hydroperoxide group (-OOH) of fatty acids esterified in membrane phospholipids. This reaction inhibits lipid peroxidation and accounts for enzyme's vital role. Here we investigated the interaction of GPx4 with membrane phospholipids. A cationic surface near the GPx4 catalytic center interacts with phospholipid polar heads. Accordingly, SPR analysis indicates cardiolipin as the phospholipid with maximal affinity to GPx4. Consistent with the electrostatic nature of the interaction, KCl increases the KD. Molecular dynamic (MD) simulation shows that a -OOH posed in the core of the membrane as 13 - or 9 -OOH of tetra-linoleoyl cardiolipin or 15 -OOH stearoyl-arachidonoyl-phosphaphatidylcholine moves to the lipid-water interface. Thereby, the -OOH groups are addressed toward the GPx4 redox center. In this pose, however, the catalytic site facing the membrane would be inaccessible to GSH, but the consecutive redox processes facilitate access of GSH, which further primes undocking of the enzyme, because GSH competes for the binding residues implicated in docking. During the final phase of the catalytic cycle, while GSSG is produced, GPx4 is disconnected from the membrane. The observation that GSH depletion in cells induces GPx4 translocation to the membrane, is in agreement with this concept.


Asunto(s)
Cardiolipinas/química , Glutatión Peroxidasa/química , Peróxidos Lipídicos/química , Liposomas/química , Fosfatidilcolinas/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Biocatálisis , Cardiolipinas/metabolismo , Expresión Génica , Glutatión Peroxidasa/aislamiento & purificación , Glutatión Peroxidasa/metabolismo , Células HEK293 , Humanos , Cinética , Peróxidos Lipídicos/metabolismo , Liposomas/metabolismo , Masculino , Simulación del Acoplamiento Molecular , Oxidación-Reducción , Fosfatidilcolinas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Electricidad Estática , Especificidad por Sustrato , Testículo/química , Testículo/enzimología
14.
Arch Biochem Biophys ; 617: 120-128, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27638050

RESUMEN

Reversible oxidation of Cys residues is a crucial element of redox homeostasis and signaling. According to a popular concept in oxidative stress signaling, the oxidation of targets of signals can only take place following an overwhelming of the cellular antioxidant capacity. This concept, however, ignores the activation of feedback mechanisms possibly leading to a paradoxical effect. In a model of cancer stem cells (CSC), stably overexpressing the TAZ oncogene, we observed that the increased formation of oxidants is associated with a globally more reduced state of proteins. Redox proteomics revealed that several proteins, capable of undergoing reversible redox transitions, are indeed more reduced while just few are more oxidized. Among the proteins more oxidized, G6PDH emerges as both more expressed and activated by oxidation. This accounts for the observed more reduced state of the NADPH/NADP+ couple. The dynamic redox flux generating this apparently paradoxical effect is rationalized in a computational system biology model highlighting the crucial role of G6PDH activity on the rate of redox transitions eventually leading to the reduction of reversible redox switches.


Asunto(s)
Células Madre Neoplásicas/citología , Oxidación-Reducción , Línea Celular Transformada , Línea Celular Tumoral , Glucosafosfato Deshidrogenasa/metabolismo , Glutarredoxinas/metabolismo , Humanos , Mutación , Nucleótidos/genética , Estrés Oxidativo , Oxígeno/química , Proteómica , Piridinas/química , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/metabolismo
15.
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
16.
Redox Biol ; 8: 205-15, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26820564

RESUMEN

The notion that electrophiles serve as messengers in cell signaling is now widely accepted. Nonetheless, major issues restrain acceptance of redox homeostasis and redox signaling as components of maintenance of a normal physiological steady state. The first is that redox signaling requires sudden switching on of oxidant production and bypassing of antioxidant mechanisms rather than a continuous process that, like other signaling mechanisms, can be smoothly turned up or down. The second is the misperception that reactions in redox signaling involve "reactive oxygen species" rather than reaction of specific electrophiles with specific protein thiolates. The third is that hormesis provides protection against oxidants by increasing cellular defense or repair mechanisms rather than by specifically addressing the offset of redox homeostasis. Instead, we propose that both oxidant and antioxidant signaling are main features of redox homeostasis. As the redox shift is rapidly reversed by feedback reactions, homeostasis is maintained by continuous signaling for production and elimination of electrophiles and nucleophiles. Redox homeostasis, which is the maintenance of nucleophilic tone, accounts for a healthy physiological steady state. Electrophiles and nucleophiles are not intrinsically harmful or protective, and redox homeostasis is an essential feature of both the response to challenges and subsequent feedback. While the balance between oxidants and nucleophiles is preserved in redox homeostasis, oxidative stress provokes the establishment of a new radically altered redox steady state. The popular belief that scavenging free radicals by antioxidants has a beneficial effect is wishful thinking. We propose, instead, that continuous feedback preserves nucleophilic tone and that this is supported by redox active nutritional phytochemicals. These nonessential compounds, by activating Nrf2, mimic the effect of endogenously produced electrophiles (parahormesis). In summary, while hormesis, although globally protective, results in setting up of a new phenotype, parahormesis contributes to health by favoring maintenance of homeostasis.


Asunto(s)
Antioxidantes/metabolismo , Fitoquímicos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Antioxidantes/uso terapéutico , Homeostasis/efectos de los fármacos , Hormesis/genética , Humanos , Cinética , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Estrés Oxidativo/genética , Fitoquímicos/uso terapéutico , Transducción de Señal/genética
17.
Free Radic Biol Med ; 87: 1-14, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26163004

RESUMEN

Glutathione peroxidases (GPxs) are enzymes working with either selenium or sulfur catalysis. They adopted diverse functions ranging from detoxification of H(2)O(2) to redox signaling and differentiation. The relative stability of the selenoenzymes, however, remained enigmatic in view of the postulated involvement of a highly unstable selenenic acid form during catalysis. Nevertheless, density functional theory calculations obtained with a representative active site model verify the mechanistic concept of GPx catalysis and underscore its efficiency. However, they also allow that the selenenic acid, in the absence of the reducing substrate, reacts with a nitrogen in the active site. MS/MS analysis of oxidized rat GPx4 complies with the predicted structure, an 8-membered ring, in which selenium is bound as selenenylamide to the protein backbone. The intermediate can be re-integrated into the canonical GPx cycle by glutathione, whereas, under denaturing conditions, its selenium moiety undergoes ß-cleavage with formation of a dehydro-alanine residue. The selenenylamide bypass prevents destruction of the redox center due to over-oxidation of the selenium or its elimination and likely allows fine-tuning of GPx activity or alternate substrate reactions for regulatory purposes.


Asunto(s)
Glutatión Peroxidasa/química , Glutatión/química , Oxidación-Reducción , Selenocisteína/química , Animales , Catálisis , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Peróxido de Hidrógeno/química , Cinética , Teoría Cuántica , Ratas , Selenio/química , Selenocisteína/metabolismo , Azufre/química , Espectrometría de Masas en Tándem
18.
Free Radic Biol Med ; 83: 352-60, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25724691

RESUMEN

The glutathione peroxidase homologs (GPxs) efficiently reduce hydroperoxides using electrons from glutathione (GSH), thioredoxin (Trx), or protein disulfide isomerase (PDI). Trx is preferentially used by the GPxs of the majority of bacteria, invertebrates, plants, and fungi. GSH or PDI, instead, is preferentially used by vertebrate GPxs that operate by Sec or Cys catalysis, respectively. Mammalian GPx7 and GPx8 are unique homologs that contain a peroxidatic Cys (CP). Being reduced by PDI and located within the endoplasmic reticulum (ER), these enzymes have been involved in oxidative protein folding. Kinetic analysis indicates that oxidation of PDI by recombinant GPx7 occurs at a much faster rate than that of GSH. Nonetheless, activity measurement suggests that, at physiological concentrations, a competition between these two substrates takes place, with the rate of PDI oxidation by GPx7 controlled by the concentration of GSH, whereas the GSSG produced in the competing reaction contributes to the ER redox buffer. A mechanism has been proposed for GPx7 involving two Cys residues, in which an intramolecular disulfide of the CP is formed with an alleged resolving Cys (CR) located in the strongly conserved FPCNQ motif (C86 in humans), a noncanonical position in GPxs. Kinetic measurements and comparison with the other thiol peroxidases containing a functional CR suggest that a resolving function of C86 in the catalytic cycle is very unlikely. We propose that GPx7 is catalytically active as a 1-Cys-GPx, in which CP both reduces H2O2 and oxidizes PDI, and that the CP-C86 disulfide has instead the role of stabilizing the oxidized peroxidase in the absence of the reducing substrate.


Asunto(s)
Proteínas Portadoras/metabolismo , Glutatión/metabolismo , Peroxidasas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Catálisis , Glutatión Peroxidasa , Humanos , Datos de Secuencia Molecular , Oxidación-Reducción , Peroxidasas/química , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
19.
Free Radic Biol Med ; 81: 58-68, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25557012

RESUMEN

GPx8 is a mammalian Cys-glutathione peroxidase of the endoplasmic reticulum membrane, involved in protein folding. Its regulation is mostly unknown. We addressed both the functionality of two hypoxia-response elements (HREs) within the promoter, GPx8 HRE1 and GPx8 HRE2, and the GPx8 physiological role. In HeLa cells, treatment with HIFα stabilizers, such as diethyl succinate (DES) or 2-2'-bipyridyl (BP), induces GPx8 expression at both mRNA and protein level. Luciferase activity of pGL3(GPx8wt), containing a fragment of the GPx8 promoter including the two HREs, is also induced by DES/BP or by overexpressing either individual HIFα subunit. Mutating GPx8 HRE1 within pGL3(GPx8wt) resulted in a significantly higher inhibition of luciferase activity than mutating GPx8 HRE2. Electrophoretic mobility-shift assay showed that both HREs exhibit enhanced binding to a nuclear extract from DES/BP-treated cells, with stronger binding by GPx8 HRE1. In DES-treated cells transfected with pGL3(GPx8wt) or mutants thereof, silencing of HIF2α, but not HIF1α, abolishes luciferase activity. Thus GPx8 is a novel HIF target preferentially responding to HIF2α binding at its two novel functional GPx8 HREs, with GPx8 HRE1 playing the major role. Fibroblast growth factor (FGF) treatment increases GPx8 mRNA expression, and reporter gene experiments indicate that induction occurs via HIF. Comparing the effects of depleting GPx8 on the downstream effectors of FGF or insulin signaling revealed that absence of GPx8 results in a 16- or 12-fold increase in phosphorylated ERK1/2 by FGF or insulin treatment, respectively. Furthermore, in GPx8-depleted cells, phosphorylation of AKT by insulin treatment increases 2.5-fold. We suggest that induction of GPx8 expression by HIF slows down proliferative signaling during hypoxia and/or growth stimulation through receptor tyrosine kinases.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Retículo Endoplásmico/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Peroxidasas/genética , 2,2'-Dipiridil/farmacología , Secuencia de Aminoácidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia de la Célula , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Insulina/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Peroxidasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Elementos de Respuesta , Alineación de Secuencia , Transducción de Señal , Succinatos/farmacología , Transcripción Genética
20.
Free Radic Biol Med ; 71: 90-98, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24642086

RESUMEN

Reversible oxidation of cysteine residues is a relevant posttranslational modification of proteins. However, the low activation energy and transitory nature of the redox switch and the intrinsic complexity of the analysis render quite challenging the aim of a rigorous high-throughput screening of the redox status of redox-sensitive cysteine residues. We describe here a quantitative workflow for redox proteomics, where the ratio between the oxidized forms of proteins in the control vs treated samples is determined by a robust label-free approach. We critically present the convenience of the procedure by specifically addressing the following aspects: (i) the accurate ratio, calculated from the whole set of identified peptides rather than just isotope-tagged fragments; (ii) the application of a robust analytical pipeline to frame the most consistent data averaged over the biological variability; (iii) the relevance of using stringent criteria of analysis, even at the cost of losing potentially interesting but statistically uncertain data. The pipeline has been assessed on red blood cell membrane challenged with diamide as a model of a mild oxidative condition. The cluster of identified proteins encompassed components of the cytoskeleton more oxidized. Indirectly, our analysis confirmed the previous observation that oxidized hemoglobin binds to membranes while oxidized peroxiredoxin 2 loses affinity.


Asunto(s)
Cisteína/química , Membrana Eritrocítica/química , Proteínas de la Membrana/química , Procesamiento Proteico-Postraduccional , Proteómica/métodos , Cisteína/metabolismo , Citoesqueleto/química , Diamida/química , Diamida/farmacología , Disulfuros/química , Membrana Eritrocítica/efectos de los fármacos , Membrana Eritrocítica/metabolismo , Hemoglobinas/metabolismo , Humanos , Oxidación-Reducción , Estrés Oxidativo , Peroxirredoxinas/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA