Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Glia ; 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39360557

RESUMEN

Histidine dipeptides (HDs) are synthesized in brain oligodendrocytes by carnosine synthase (carns1), but their role is unknown. Using metabolomics and in vivo experiments with both constitutive and oligodendrocyte-selective carns1-KO mouse models, we found that HDs are critical for oligodendrocyte survival and protect against oxidative stress. Carns1-KO mouse models had lower numbers of mature oligodendrocytes, increased lipid peroxidation, and behavioral changes. Cuprizone administration, which increases reactive oxygen species in vivo, resulted in higher oligodendrocyte death, demyelination, axonal alterations, and oxidative damage in the corpus callosum of carns1-KO mice. Gliosis and oxidative damage by cuprizone were prevented by pretreatment with the antioxidant N-acetylcysteine. NADPH levels were increased threefold in the brains of carns1-KO mice as an antioxidant response to oxidative stress through acceleration of the pentose phosphate pathway (PPP). This was due to overexpression of glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Likewise, expression of NAD kinase, the biosynthetic enzyme for NADP+, and NAMPT, which replenishes the NAD+ pool, was higher in carns1-KO mice brains than in controls. Our observations suggest that HDs cell-autonomously protect oligodendrocytes from oxidative stress, with implications for demyelinating diseases.

2.
J Biol Chem ; 295(11): 3590-3600, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32041780

RESUMEN

Hydrogen sulfide has been implicated in a large number of physiological processes including cell survival and death, encouraging research into its mechanisms of action and therapeutic potential. Results from recent studies suggest that the cellular effects of hydrogen sulfide are mediated in part by sulfane sulfur species, including persulfides and polysulfides. In the present study, we investigated the apoptosis-modulating effects of polysulfides, especially on the caspase cascade, which mediates the intrinsic apoptotic pathway. Biochemical analyses revealed that organic or synthetic polysulfides strongly and rapidly inhibit the enzymatic activity of caspase-3, a major effector protease in apoptosis. We attributed the caspase-3 inhibition to persulfidation of its catalytic cysteine. In apoptotically stimulated HeLa cells, short-term exposure to polysulfides triggered the persulfidation and deactivation of cleaved caspase-3. These effects were antagonized by the thioredoxin/thioredoxin reductase system (Trx/TrxR). Trx/TrxR restored the activity of polysulfide-inactivated caspase-3 in vitro, and TrxR inhibition potentiated polysulfide-mediated suppression of caspase-3 activity in situ We further found that under conditions of low TrxR activity, early cell exposure to polysulfides leads to enhanced persulfidation of initiator caspase-9 and decreases apoptosis. Notably, we show that the proenzymes procaspase-3 and -9 are basally persulfidated in resting (unstimulated) cells and become depersulfidated during their processing and activation. Inhibition of TrxR attenuated the depersulfidation and activation of caspase-9. Taken together, our results reveal that polysulfides target the caspase-9/3 cascade and thereby suppress cancer cell apoptosis, and highlight the role of Trx/TrxR-mediated depersulfidation in enabling caspase activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Sulfuros/metabolismo , Sulfuros/farmacología , Tiorredoxinas/farmacología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Inhibidores de Caspasas/farmacología , Activación Enzimática/efectos de los fármacos , Células HeLa , Humanos , Transducción de Señal/efectos de los fármacos , Reductasa de Tiorredoxina-Disulfuro/metabolismo
3.
Nat Rev Mol Cell Biol ; 10(10): 721-32, 2009 10.
Artículo en Inglés | MEDLINE | ID: mdl-19738628

RESUMEN

S-Nitrosylation, the redox-based modification of Cys thiol side chains by nitric oxide, is a common mechanism in signal transduction. Dysregulated S-nitrosylation contributes to a range of human pathologies. New roles for protein denitrosylation in regulating S-nitrosylation are being revealed. Recently, several denitrosylases - the enzymes that mediate Cys denitrosylation - have been discovered, of which two enzyme systems in particular, the S-nitrosoglutathione reductase and thioredoxin systems, have been shown to be physiologically relevant. These highly conserved enzymes regulate signalling through multiple classes of receptors and influence diverse cellular responses. In addition, they protect from nitrosative stress in microorganisms, mammals and plants, thereby exerting profound effects on host-microbe interactions and innate immunity.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Proteínas/metabolismo , Tiorredoxinas/metabolismo , Humanos , Óxido Nítrico/metabolismo , Nitrosación , Oxidación-Reducción , S-Nitrosoglutatión/metabolismo , S-Nitrosotioles/metabolismo , Transducción de Señal
4.
Biochim Biophys Acta ; 1850(12): 2476-84, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26388496

RESUMEN

BACKGROUND: The free radical nitric oxide (NO) and the thiol oxidoreductase thioredoxin (Trx) play essential roles in cellular redox regulation. Recent biochemical and cellular studies have revealed a complex thiol-dependent crosstalk between NO and Trx that modulates multiple redox-dependent pathways. SCOPE OF REVIEW: This review aims to discuss recent progress, as well as the remaining questions, regarding the interaction and cross regulation between NO and Trx in cellular function and dysfunction. MAJOR CONCLUSIONS: The importance and ubiquity of NO-mediated S-nitrosylation of protein thiols as a signaling mechanism is increasingly recognized as is the central role of Trx in regulating S-nitrosylation processes. By denitrosylating diverse protein substrates, Trx plays an active role in attenuating NO signaling as well as in ameliorating nitrosative stress. Yet, at the same time, Trx can also support the activity of NO synthases, thus promoting NO production and its downstream effects. Finally, NO can reciprocally modulate the redox activity of Trx and Trx reductase. GENERAL SIGNIFICANCE: Further elucidation of the crosstalk between NO and Trx will be important for an improved understanding of the effects of reactive oxygen and nitrogen species on cellular signaling and function.


Asunto(s)
Óxido Nítrico/fisiología , Tiorredoxinas/fisiología , Animales , Humanos , Oxidación-Reducción
5.
Mol Cell Proteomics ; 13(10): 2573-83, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24973421

RESUMEN

Protein S-nitrosylation, the nitric oxide-mediated posttranslational modification of cysteine residues, has emerged as an important regulatory mechanism in diverse cellular processes. Yet, knowledge about the S-nitrosoproteome in different cell types and cellular contexts is still limited and many questions remain regarding the precise roles of protein S-nitrosylation and denitrosylation. Here we present a novel strategy to identify reversibly nitrosylated proteins. Our approach is based on nitrosothiol capture and enrichment using a thioredoxin trap mutant, followed by protein identification by mass spectrometry. Employing this approach, we identified more than 400 putative nitroso-proteins in S-nitrosocysteine-treated human monocytes and about 200 nitrosylation substrates in endotoxin and cytokine-stimulated mouse macrophages. The large majority of these represent novel nitrosylation targets and they include many proteins with key functions in cellular homeostasis and signaling. Biochemical and functional experiments in vitro and in cells validated the proteomic results and further suggested a role for thioredoxin in the denitrosylation and activation of inducible nitric oxide synthase and the protein kinase MEK1. Our findings contribute to a better understanding of the macrophage S-nitrosoproteome and the role of thioredoxin-mediated denitrosylation in nitric oxide signaling. The approach described here may prove generally useful for the identification and exploration of nitroso-proteomes under various physiological and pathophysiological conditions.


Asunto(s)
Macrófagos/metabolismo , Monocitos/metabolismo , Óxido Nítrico/farmacología , Proteínas/metabolismo , Proteómica/métodos , Animales , Línea Celular , Cisteína/análogos & derivados , Cisteína/farmacología , Células HEK293 , Humanos , Interferón gamma/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Monocitos/citología , Monocitos/efectos de los fármacos , Mutación , Procesamiento Proteico-Postraduccional , Proteínas/química , S-Nitrosotioles/farmacología , Transducción de Señal/efectos de los fármacos , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
6.
Biochim Biophys Acta ; 1840(10): 3153-61, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25065288

RESUMEN

BACKGROUND: The thioredoxin/thioredoxin reductase system, which is best known for its essential role in antioxidant defense and redox homeostasis, is increasingly implicated in the regulation of multiple cellular signaling pathways. In the present study, we asked if the thioredoxin system in macrophages might regulate toll-like receptor 4 (TLR4)-dependent gene expression and consequent responses. METHODS: Using microarray analysis we analyzed the effect of auranofin, a highly potent and specific inhibitor of thioredoxin reductase, on the transcriptional program activated in J774 macrophages by the TLR4 agonist, lipopolysaccharide (LPS). We used quantitative real-time PCR (qPCR), Western blotting, ELISA and cytotoxicity assays to confirm and extend the microarray results. RESULTS: Global transcriptional profiling revealed that macrophage treatment with auranofin exerted a selective effect on LPS-induced gene expression, suppressing the induction of a small number of genes. Interestingly, among these suppressed genes were three members of the interleukin-1 (IL-1) family of genes, among which IL-1ß was most affected. qPCR analyses confirmed the repressive effects of auranofin on IL-1 genes. In addition, qPCR and Western blot analyses showed that auranofin impaired TLR4-dependent induction of the inflammasome receptor NLRP3, which plays a critical role in IL-1ß processing. Consistent with these findings, inflammasome-dependent release of IL-1ß from stimulated macrophages was suppressed by auranofin as was inflammasome-mediated cell death. CONCLUSIONS: Our findings suggest a regulatory role for the thioredoxin system in macrophage inflammatory signaling. Inhibition of the thioredoxin system in macrophages exerts an anti-inflammatory effect by repressing the activation of the NLRP3/IL-1ß pathway.


Asunto(s)
Antirreumáticos/farmacología , Auranofina/farmacología , Proteínas Portadoras/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/metabolismo , Transducción de Señal/efectos de los fármacos , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Inflamasomas/metabolismo , Lipopolisacáridos/farmacología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/metabolismo , Transcripción Genética/efectos de los fármacos
7.
J Biol Chem ; 288(16): 11312-24, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23479738

RESUMEN

S-nitrosothiols (SNOs), formed by nitric oxide (NO)-mediated S-nitrosylation, and hydrogen peroxide (H2O2), a prominent reactive oxygen species, are implicated in diverse physiological and pathological processes. Recent research has shown that the cellular action and metabolism of SNOs and H2O2 involve overlapping, thiol-based mechanisms, but how these reactive species may affect each other's fate and function is not well understood. In this study we investigated how NO/SNO may affect the redox cycle of mammalian peroxiredoxin-1 (Prx1), a representative of the 2-Cys Prxs, a group of thioredoxin (Trx)-dependent peroxidases. We found that, both in a cell-free system and in cells, NO/SNO donors such as S-nitrosocysteine and S-nitrosoglutathione readily induced the S-nitrosylation of Prx1, causing structural and functional alterations. In particular, nitrosylation promoted disulfide formation involving the pair of catalytic cysteines (Cys-52 and Cys-173) and disrupted the oligomeric structure of Prx1, leading to loss of peroxidase activity. A highly potent inhibition of the peroxidase catalytic reaction by NO/SNO was seen in assays employing the coupled Prx-Trx system. In this setting, S-nitrosocysteine (10 µM) effectively blocked the Trx-mediated regeneration of oxidized Prx1. This effect appeared to be due to both competition between S-nitrosocysteine and Prx1 for the Trx system and direct modulation by S-nitrosocysteine of Trx reductase activity. Our findings that NO/SNO target both Prx and Trx reductase may have implications for understanding the impact of nitrosylation on cellular redox homeostasis.


Asunto(s)
Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Animales , Catálisis , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Células HeLa , Proteínas de Homeodominio/genética , Humanos , Ratones , Óxido Nítrico/genética , Oxidación-Reducción , Reductasa de Tiorredoxina-Disulfuro/química , Reductasa de Tiorredoxina-Disulfuro/genética , Reductasa de Tiorredoxina-Disulfuro/metabolismo
8.
Redox Biol ; 72: 103125, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38574432

RESUMEN

Acute inflammatory responses often involve the production of reactive oxygen and nitrogen species by innate immune cells, particularly macrophages. How activated macrophages protect themselves in the face of oxidative-inflammatory stress remains a long-standing question. Recent evidence implicates reactive sulfur species (RSS) in inflammatory responses; however, how endogenous RSS affect macrophage function and response to oxidative and inflammatory insults remains poorly understood. In this study, we investigated the endogenous pathways of RSS biogenesis and clearance in macrophages, with a particular focus on exploring how hydrogen sulfide (H2S)-mediated S-persulfidation influences macrophage responses to oxidative-inflammatory stress. We show that classical activation of mouse or human macrophages using lipopolysaccharide and interferon-γ (LPS/IFN-γ) triggers substantial production of H2S/RSS, leading to widespread protein persulfidation. Biochemical and proteomic analyses revealed that this surge in cellular S-persulfidation engaged ∼2% of total thiols and modified over 800 functionally diverse proteins. S-persulfidation was found to be largely dependent on the cystine importer xCT and the H2S-generating enzyme cystathionine γ-lyase and was independent of changes in the global proteome. We further investigated the role of the sulfide-oxidizing enzyme sulfide quinone oxidoreductase (SQOR), and found that it acts as a negative regulator of S-persulfidation. Elevated S-persulfidation following LPS/IFN-γ stimulation or SQOR inhibition was associated with increased resistance to oxidative stress. Upregulation of persulfides also inhibited the activation of the macrophage NLRP3 inflammasome and provided protection against inflammatory cell death. Collectively, our findings shed light on the metabolism and effects of RSS in macrophages and highlight the crucial role of persulfides in enabling macrophages to withstand and alleviate oxidative-inflammatory stress.


Asunto(s)
Sulfuro de Hidrógeno , Activación de Macrófagos , Macrófagos , Estrés Oxidativo , Estrés Oxidativo/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Animales , Ratones , Humanos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Activación de Macrófagos/efectos de los fármacos , Lipopolisacáridos , Inflamación/metabolismo , Cistationina gamma-Liasa/metabolismo , Sulfuros/farmacología , Interferón gamma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Oxidación-Reducción , Proteómica/métodos
9.
Antioxidants (Basel) ; 13(2)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38397843

RESUMEN

Reactive sulfur species (RSS) like hydrogen sulfide (H2S) and cysteine persulfide (Cys-SSH) emerged as key signaling molecules with diverse physiological roles in the body, depending on their concentration and the cellular environment. While it is known that H2S and Cys-SSH are produced by both colonocytes and by the gut microbiota through sulfur metabolism, it remains unknown how these RSS affect amebiasis caused by Entamoeba histolytica, a parasitic protozoan that can be present in the human gastrointestinal tract. This study investigates H2S and Cys-SSH's impact on E. histolytica physiology and explores potential therapeutic implications. Exposing trophozoites to the H2S donor, sodium sulfide (Na2S), or to Cys-SSH led to rapid cytotoxicity. A proteomic analysis of Cys-SSH-challenged trophozoites resulted in the identification of >500 S-sulfurated proteins, which are involved in diverse cellular processes. Functional assessments revealed inhibited protein synthesis, altered cytoskeletal dynamics, and reduced motility in trophozoites treated with Cys-SSH. Notably, cysteine proteases (CPs) were significantly inhibited by S-sulfuration, affecting their bacterial biofilm degradation capacity. Immunofluorescence microscopy confirmed alterations in actin dynamics, corroborating the proteomic findings. Thus, our study reveals how RSS perturbs critical cellular functions in E. histolytica, potentially influencing its pathogenicity and interactions within the gut microbiota. Understanding these molecular mechanisms offers novel insights into amebiasis pathogenesis and unveils potential therapeutic avenues targeting RSS-mediated modifications in parasitic infections.

10.
Antioxid Redox Signal ; 38(4-6): 388-402, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35979894

RESUMEN

Aims: Oxidative modifications of cysteine (Cys) thiols regulate various physiological processes, including inflammatory responses. The thioredoxin (Trx) system plays a key role in thiol redox control. The aim of this study was to characterize the dynamic cysteine proteome of human macrophages upon activation by the prototypical proinflammatory agent, bacterial lipopolysaccharide (LPS), and/or perturbation of the Trx system. Results: In this study, we profiled the cellular and redox proteome of human THP-1-derived macrophages during the early phase of LPS activation and/or inhibition of Trx system activity by auranofin (AF) by employing a peptide-centric, resin-assisted capture, redox proteomic workflow. Among 4200 identified cysteines, oxidation of nearly 10% was selectively affected by LPS or AF treatments. Notably, the proteomic analysis uncovered a subset of ∼100 thiols, mapped to proteins involved in diverse processes, whose oxidation is antagonistically regulated by LPS and Trx. Compared with the redox proteome, the cellular proteome was largely unchanged, highlighting the importance of redox modification as a mechanism that allows for rapid modulation of macrophage activities in response to a proinflammatory or pro-oxidant insult. Structural-functional analyses provided mechanistic insights into redox regulation of selected proteins, including the glutathione-synthesizing enzyme, glutamate-cysteine ligase, and the autophagy adaptor, SQSTM1/p62, suggesting mechanisms by which macrophages adapt and fine-tune their responses according to a changing inflammatory and redox environment. Innovation: This study provides a rich resource for further characterization of redox mechanisms that regulate macrophage inflammatory activities. Conclusion: The dynamic thiol redox proteome allows macrophages to efficiently respond and adapt to redox and inflammatory challenges. Antioxid. Redox Signal. 38, 388-402.


Asunto(s)
Cisteína , Compuestos de Sulfhidrilo , Humanos , Compuestos de Sulfhidrilo/metabolismo , Cisteína/metabolismo , Proteoma/metabolismo , Proteómica , Lipopolisacáridos/farmacología , Tiorredoxinas/metabolismo , Oxidación-Reducción , Macrófagos/metabolismo
11.
J Biol Chem ; 286(35): 30433-30443, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21724851

RESUMEN

Protein S-nitrosylation is a reversible protein modification implicated in both physiological and pathophysiological regulation of protein function. In obesity, skeletal muscle insulin resistance is associated with increased S-nitrosylation of insulin-signaling proteins. However, whether adipose tissue is similarly affected in obesity and, if so, what are the causes and functional consequences of increased S-nitrosylation in this tissue are unknown. Total protein S-nitrosylation was increased in intra-abdominal adipose tissue of obese humans and in high fat-fed or leptin-deficient ob/ob mice. Both the insulin receptor ß-subunit and Akt were S-nitrosylated, correlating with body weight. Elevated protein and mRNA expression of inducible NO synthase and decreased protein levels of thioredoxin reductase were associated with increased adipose tissue S-nitrosylation. Cultured differentiated pre-adipocyte cell lines exposed to the NO donors S-nitrosoglutathione (GSNO) or S-nitroso-N-acetylpenicillamine exhibited diminished insulin-stimulated phosphorylation of Akt but not of GSK3 nor of insulin-stimulated glucose uptake. Yet the anti-lipolytic action of insulin was markedly impaired in both cultured adipocytes and in mice injected with GSNO prior to administration of insulin. In cells, impaired ability of insulin to diminish phosphorylated PKA substrates in response to isoproterenol suggested impaired insulin-induced activation of PDE3B. Consistently, increased S-nitrosylation of PDE3B was detected in adipose tissue of high fat-fed obese mice. Site-directed mutagenesis revealed that Cys-768 and Cys-1040, two putative sites for S-nitrosylation adjacent to the substrate-binding site of PDE3B, accounted for ∼50% of its GSNO-induced S-nitrosylation. Collectively, PDE3B and the anti-lipolytic action of insulin may constitute novel targets for increased S-nitrosylation of adipose tissue in obesity.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/metabolismo , Insulina/metabolismo , Nitrógeno/química , Obesidad/metabolismo , Animales , Cisteína/química , Femenino , Humanos , Resistencia a la Insulina/fisiología , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Estrés Oxidativo , Fosforilación
12.
Biochemistry ; 49(32): 6963-9, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20695533

RESUMEN

Reversible protein cysteine nitrosylation (S-nitrosylation) is a common mechanism utilized in signal transduction and other diverse cellular processes. Protein denitrosylation is largely mediated by cysteine denitrosylases, but the functional scope and significance of these enzymes are incompletely defined, in part due to limited information on their cognate substrates. Here, using Jurkat cells, we employed stable isotope labeling by amino acids in cell culture (SILAC), coupled to the biotin switch technique and mass spectrometry, to identify 46 new substrates of one denitrosylase, thioredoxin 1. These substrates are involved in a wide range of cellular functions including cytoskeletal organization, cellular metabolism, signal transduction, and redox homeostasis. We also identified multiple S-nitrosylated proteins that are not substrates of thioredoxin 1. A verification of our principal findings was made in a second cell type (RAW264.7 cells). Our results point to thioredoxin 1 as a major protein denitrosylase in mammalian cells and demonstrate the utility of quantitative proteomics for large-scale identification of denitrosylase substrates.


Asunto(s)
Proteómica , Tiorredoxinas/metabolismo , Aminoácidos/metabolismo , Animales , Línea Celular , Cromatografía Liquida , Humanos , Marcaje Isotópico , Células Jurkat , Ratones , Modelos Teóricos , S-Nitrosotioles/metabolismo , Espectrometría de Masas en Tándem
13.
J Biol Chem ; 284(52): 36160-36166, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19847012

RESUMEN

Nitric oxide exerts a plethora of biological effects via protein S-nitrosylation, a redox-based reaction that converts a protein Cys thiol to a S-nitrosothiol. However, although the regulation of protein S-nitrosylation has been the subject of extensive study, much less is known about the systems governing protein denitrosylation. Most recently, thioredoxin/thioredoxin reductases were shown to mediate both basal and stimulus-coupled protein denitrosylation. We now demonstrate that protein denitrosylation by thioredoxin is regulated dynamically by thioredoxin-interacting protein (Txnip), a thioredoxin inhibitor. Endogenously synthesized nitric oxide represses Txnip, thereby facilitating thioredoxin-mediated denitrosylation. Autoregulation of denitrosylation thus allows cells to survive nitrosative stress. Our findings reveal that denitrosylation of proteins is dynamically regulated, establish a physiological role for thioredoxin in protection from nitrosative stress, and suggest new approaches to manipulate cellular S-nitrosylation.


Asunto(s)
Proteínas Portadoras/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/fisiología , Proteínas Portadoras/genética , Línea Celular , Supervivencia Celular/fisiología , Humanos , Óxido Nítrico/genética , Oxidación-Reducción , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/genética , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
14.
Antioxidants (Basel) ; 9(4)2020 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-32290499

RESUMEN

It is well appreciated that biological reactive oxygen and nitrogen species such as hydrogen peroxide, superoxide and nitric oxide, as well as endogenous antioxidant systems, are important modulators of cell survival and death in diverse organisms and cell types. In addition, oxidative stress, nitrosative stress and dysregulated cell death are implicated in a wide variety of pathological conditions, including cancer, cardiovascular and neurological diseases. Therefore, much effort is devoted to elucidate the molecular mechanisms linking oxidant/antioxidant systems and cell death pathways. This review is focused on thiol redox modifications as a major mechanism by which oxidants and antioxidants influence specific regulated cell death pathways in mammalian cells. Growing evidence indicates that redox modifications of cysteine residues in proteins are involved in the regulation of multiple cell death modalities, including apoptosis, necroptosis and pyroptosis. In addition, recent research suggests that thiol redox switches play a role in the crosstalk between apoptotic and necrotic forms of regulated cell death. Thus, thiol-based redox circuits provide an additional layer of control that determines when and how cells die.

15.
Free Radic Biol Med ; 160: 566-574, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-32898624

RESUMEN

Nitric oxide (NO)-dependent signaling and cytotoxic effects are mediated in part via protein S-nitrosylation. The magnitude and duration of S-nitrosylation are governed by the two main thiol reducing systems, the glutathione (GSH) and thioredoxin (Trx) antioxidant systems. In recent years, approaches have been developed to harness the cytotoxic potential of NO/nitrosylation to inhibit tumor cell growth. However, progress in this area has been hindered by insufficient understanding of the balance and interplay between cellular nitrosylation, other oxidative processes and the GSH/Trx systems. In addition, the mechanistic relationship between thiol redox imbalance and cancer cell death is not fully understood. Herein, we explored the redox and cellular effects induced by the S-nitrosylating agent, S-nitrosocysteine (CysNO), in GSH-sufficient and -deficient human tumor cells. We used l-buthionine-sulfoximine (BSO) to induce GSH deficiency, and employed redox, biochemical and cellular assays to interrogate molecular mechanisms. We found that, under GSH-sufficient conditions, a CysNO challenge (100-500 µM) results in a marked yet reversible increase in protein S-nitrosylation in the absence of appreciable S-oxidation. In contrast, under GSH-deficient conditions, CysNO induces elevated and sustained levels of both S-nitrosylation and S-oxidation. Experiments in various cancer cell lines showed that administration of CysNO or BSO alone commonly induce minimal cytotoxicity whereas BSO/CysNO combination therapy leads to extensive cell death. Studies in HeLa cancer cells revealed that treatment with BSO/CysNO results in dual inhibition of the GSH and Trx systems, thereby amplifying redox stress and causing cellular dysfunction. In particular, BSO/CysNO induced rapid oxidation and collapse of the actin cytoskeletal network, followed by loss of mitochondrial function, leading to profound and irreversible decrease in ATP levels. Further observations indicated that BSO/CysNO-induced cell death occurs via a caspase-independent mechanism that involves multiple stress-induced pathways. The present findings provide new insights into the relationship between cellular nitrosylation/oxidation, thiol antioxidant defenses and cell death. These results may aid future efforts to develop NO/redox-based anticancer approaches.


Asunto(s)
Glutatión , S-Nitrosotioles , Butionina Sulfoximina/farmacología , Muerte Celular , Cisteína/análogos & derivados , Glutatión/metabolismo , Humanos , Oxidación-Reducción
16.
Front Immunol ; 10: 590, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31001247

RESUMEN

Background: Human α1-antitrypsin (hAAT) is a circulating anti-inflammatory serine-protease inhibitor that rises during acute phase responses. in vivo, hAAT reduces bacterial load, without directly inhibiting bacterial growth. In conditions of excess nitric-oxide (NO), hAAT undergoes S-nitrosylation (S-NO-hAAT) and gains antibacterial capacity. The impact of S-NO-hAAT on immune cells has yet to be explored. Aim: Study the effects of S-NO-hAAT on immune cells during bacterial infection. Methods: Clinical-grade hAAT was S-nitrosylated and then compared to unmodified hAAT, functionally, and structurally. Intracellular bacterial clearance by THP-1 macrophages was assessed using live Salmonella typhi. Murine peritoneal macrophages were examined, and signaling pathways were evaluated. S-NO-hAAT was also investigated after blocking free mambranal cysteine residues on cells. Results: S-NO-hAAT (27.5 uM) enhances intracellular bacteria elimination by immunocytes (up to 1-log reduction). S-NO-hAAT causes resting macrophages to exhibit a pro-inflammatory and antibacterial phenotype, including release of inflammatory cytokines and induction of inducible nitric oxide synthase (iNOS) and TLR2. These pro-inflammatory effects are dependent upon cell surface thiols and activation of MAPK pathways. Conclusions: hAAT duality appears to be context-specific, involving S-nitrosylation in a nitric oxide rich environment. Our results suggest that S-nitrosylation facilitates the antibacterial activity of hAAT by promoting its ability to activate innate immune cells. This pro-inflammatory effect may involve transferring of nitric oxide from S-NO-hAAT to a free cysteine residue on cellular targets.


Asunto(s)
Inmunidad Innata , Macrófagos Peritoneales/inmunología , Óxido Nítrico/inmunología , Salmonella typhi/inmunología , alfa 1-Antitripsina/inmunología , Animales , Femenino , Macrófagos Peritoneales/microbiología , Ratones , alfa 1-Antitripsina/genética
17.
Cell Metab ; 30(6): 1152-1170.e13, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31735592

RESUMEN

Life on Earth emerged in a hydrogen sulfide (H2S)-rich environment eons ago and with it protein persulfidation mediated by H2S evolved as a signaling mechanism. Protein persulfidation (S-sulfhydration) is a post-translational modification of reactive cysteine residues, which modulate protein structure and/or function. Persulfides are difficult to label and study due to their reactivity and similarity with cysteine. Here, we report a facile strategy for chemoselective persulfide bioconjugation using dimedone-based probes, to achieve highly selective, rapid, and robust persulfide labeling in biological samples with broad utility. Using this method, we show persulfidation is an evolutionarily conserved modification and waves of persulfidation are employed by cells to resolve sulfenylation and prevent irreversible cysteine overoxidation preserving protein function. We report an age-associated decline in persulfidation that is conserved across evolutionary boundaries. Accordingly, dietary or pharmacological interventions to increase persulfidation associate with increased longevity and improved capacity to cope with stress stimuli.


Asunto(s)
Envejecimiento/metabolismo , Sulfuro de Hidrógeno/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Sulfuros/metabolismo , Animales , Caenorhabditis elegans , Línea Celular , Ciclohexanonas/química , Cisteína/química , Cisteína/metabolismo , Drosophila melanogaster , Escherichia coli , Fibroblastos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar , Saccharomyces cerevisiae , Coloración y Etiquetado
18.
Free Radic Biol Med ; 127: 160-164, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378334

RESUMEN

Mammalian cells employ elaborate antioxidant systems to effectively handle reactive oxygen and nitrogen species (ROS and RNS). At the heart of these systems operate two selenoprotein families consisting of glutathione peroxidase (GPx) and thioredoxin reductase (TrxR) enzymes. Although mostly studied in the context of oxidative stress, considerable evidence has amassed to indicate that these selenoenzymes also play important roles in nitrosative stress responses. GPx and TrxR, together with their redox partners, metabolize nitrosothiols and peroxynitrite, two major RNS. As such, these enzymes play active roles in the cellular defense against nitrosative stress. However, under certain conditions, these enzymes are inactivated by nitrosothiols or peroxynitrite, which may exacerbate oxidative and nitrosative stress in cells. The selenol groups in the active sites of GPx and TrxR enzymes are critically involved in these beneficial and detrimental processes. Further elucidation of the biochemical interactions between distinct RNS and GPx/TrxR will lead to a better understanding of the roles of these selenoenzymes in cellular homeostasis and disease.


Asunto(s)
Glutatión Peroxidasa/metabolismo , Estrés Nitrosativo/fisiología , Selenoproteínas/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Animales , Humanos
20.
PLoS One ; 12(6): e0179803, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28604816

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0169862.].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA