scispace - formally typeset
Search or ask a question

Showing papers on "S-Nitrosylation published in 2001"


Journal ArticleDOI
TL;DR: Protein S-nitrosylation is established as a physiological signalling mechanism for neuronally generated NO in mice harbouring a genomic deletion of neuronal NO synthase (nNOS).
Abstract: Nitric oxide (NO) has been linked to numerous physiological and pathophysiological events that are not readily explained by the well established effects of NO on soluble guanylyl cyclase. Exogenous NO S-nitrosylates cysteine residues in proteins, but whether this is an important function of endogenous NO is unclear. Here, using a new proteomic approach, we identify a population of proteins that are endogenously S-nitrosylated, and demonstrate the loss of this modification in mice harbouring a genomic deletion of neuronal NO synthase (nNOS). Targets of NO include metabolic, structural and signalling proteins that may be effectors for neuronally generated NO. These findings establish protein S-nitrosylation as a physiological signalling mechanism for nNOS.

1,386 citations


Journal ArticleDOI
21 Sep 2001-Cell
TL;DR: Whereas phosphorylation clearly Spain lies at the heart of many signal transduction pathways, has been expanded re-translational modification of proteins, are conserved cently by the discovery of an enzymatic function for throughout evolution and influence most aspects of cel-hemoglobin.

1,267 citations


Journal ArticleDOI
22 Mar 2001-Nature
TL;DR: It is concluded that GSNO reductase is evolutionarily conserved from bacteria to humans, is critical for SNO homeostasis, and protects against nitrosative stress.
Abstract: Considerable evidence indicates that NO biology involves a family of NO-related molecules and that S-nitrosothiols (SNOs) are central to signal transduction and host defence. It is unknown, however, how cells switch off the signals or protect themselves from the SNOs produced for defence purposes. Here we have purified a single activity from Escherichia coli, Saccharomyces cerevisiae and mouse macrophages that metabolizes S-nitrosoglutathione (GSNO), and show that it is the glutathione-dependent formaldehyde dehydrogenase. Although the enzyme is highly specific for GSNO, it controls intracellular levels of both GSNO and S-nitrosylated proteins. Such 'GSNO reductase' activity is widely distributed in mammals. Deleting the reductase gene in yeast and mice abolishes the GSNO-consuming activity, and increases the cellular quantity of both GSNO and protein SNO. Furthermore, mutant yeast cells show increased susceptibility to a nitrosative challenge, whereas their resistance to oxidative stress is unimpaired. We conclude that GSNO reductase is evolutionarily conserved from bacteria to humans, is critical for SNO homeostasis, and protects against nitrosative stress.

849 citations


Journal ArticleDOI
TL;DR: The antiapoptotic mechanism can be understood via expression of protective genes such as heat shock proteins, Bcl-2 as well as direct inhibition of the apoptotic caspase family proteases by S-nitrosylation of the cysteine thiol.

528 citations


Journal ArticleDOI
TL;DR: Data suggest that S-nitrosylation of p50 is a physiological mechanism of NF-kappaB regulation, which is in fact regulated by S- Nitrosocysteine and cytokine-activated NOS2.
Abstract: It is not clear if redox regulation of transcription is the consequence of direct redox-related modifications of transcription factors, or if it occurs at some other redox-sensitive step. One obstacle has been the inability to demonstrate redox-related modifications of transcription factors in vivo. The redox-sensitive transcriptional activator NF-kappaB (p50-p65) is a case in point. Its activity in vitro can be inhibited by S-nitrosylation of a critical thiol in the DNA-interacting p50 subunit, but modulation of NF-kappaB activity by nitric oxide synthase (NOS) has been attributed to other mechanisms. Herein we show that cellular NF-kappaB activity is in fact regulated by S-nitrosylation. We observed that both S-nitrosocysteine and cytokine-activated NOS2 inhibited NF-kappaB in human respiratory cells or murine macrophages. This inhibition was reversed by addition of the denitrosylating agent dithiothreitol to cellular extracts, whereas NO bioactivity did not affect the TNFalpha-induced degradation of IkappaBalpha or the nuclear translocation of p65. Recapitulation of these conditions in vitro resulted in S-nitrosylation of recombinant p50, thereby inhibiting its binding to DNA, and this effect was reversed by dithiothreitol. Further, an increase in S-nitrosylated p50 was detected in cells, and the level was modulated by TNFalpha. Taken together, these data suggest that S-nitrosylation of p50 is a physiological mechanism of NF-kappaB regulation.

363 citations


Journal ArticleDOI
TL;DR: Results show that oxidative modification of H-ras can be extensive in vivo, that both S-nitrosylated and S-glutathiolated forms may be important, and that oxidation may occur on reactive cysteines that are normally targeted for lipid-modification reactions.
Abstract: The reactive cysteines in H-ras are subject to oxidative modifications that potentially alter the cellular function of this protein. In this study, purified H-ras was modified by thiol oxidants such as hydrogen peroxide (H(2)O(2)), S-nitrosoglutathione, diamide, glutathione disulphide (GSSG) and cystamine, producing as many as four charge-isomeric forms of the protein. These results suggest that all four reactive cysteines of H-ras are potential sites of regulatory modification reactions. S-nitrosylated and S-glutathiolated forms of H-ras were identified by protocols that depend on separation of alkylated proteins on electrofocusing gels. S-nitrosoglutathione could S-nitrosylate H-ras on four cysteine residues, while reduced glutathione (GSH) and H(2)O(2) mediate S-glutathiolation on at least one cysteine of H-ras. Either GSSG or diamide S-glutathiolated at least two cysteine residues of purified H-ras. Iodoacetic acid reacts with three cysteine residues. In intact NIH-3T3 cells, wild-type H-ras was S-glutathiolated by diamide. Similarly, cells expressing a C118S mutant or a C181S/C184S double mutant of H-ras were S-glutathiolated by diamide. These results suggest that H-ras can be S-glutathiolated on multiple thiols in vivo and that at least one of these thiols is normally lipid-modified. In cells treated with S-nitrosocysteine, evidence for both S-nitrosylated and S-glutathiolated H-ras was obtained and S-nitrosylation was the predominant modification. These results show that oxidative modification of H-ras can be extensive in vivo, that both S-nitrosylated and S-glutathiolated forms may be important, and that oxidation may occur on reactive cysteines that are normally targeted for lipid-modification reactions.

180 citations


Journal ArticleDOI
TL;DR: A head-to-head comparison suggests that S-nitrosylation, like O-phosphorylation, may similarly play a fundamental role in the post-translational control of protein activity and cellular function.
Abstract: Nitric oxide (NO) is a free-radical product of mammalian cell metabolism that plays diverse and important roles in the regulation of cell function. Biological actions of NO arise as a direct consequence of chemical reactions between NO or NO-derived species and protein targets. Reactions of NO with transition metals in target proteins have garnered the most attention to date as the principal mechanism of NO signaling; nonetheless, S-nitrosylation of protein Cys residues is rapidly moving to center stage in importance. In general, however, there has been a delay in adequate appreciation of the role of S-nitrosylation in biological signaling by NO. This lag is attributed to a poor understanding of the basis for selective targeting of NO to particular thiols, and methodological limitations in accurately quantifying this modification--recent breakthroughs in concepts and methods diminish these barriers. Here, we consider the wheres and whys of protein S-nitrosylation and its basis for specificity. Protein S-nitrosylation potentially represents a ubiquitous and fundamental mechanism for posttranslational control of protein activity on a par with that of O-phosphorylation.

170 citations


Journal ArticleDOI
TL;DR: Evidence that Ca(2+) regulates the S-nitrosylated and denitrosylation of tTG and thereby TGase activity is provided to suggest a novel allosteric role for Ca( 2+) in regulating the inhibition of t TG by NO and a novel function for tTG in dispensing NO bioactivity.
Abstract: Nitric oxide (NO) and related molecules play important roles in vascular biology. NO modifies proteins through nitrosylation of free cysteine residues, and such modifications are important in mediating NO's biologic activity. Tissue transglutaminase (tTG) is a sulfhydryl rich protein that is expressed by endothelial cells and secreted into the extracellular matrix (ECM) where it is bound to fibronectin. Tissue TG exhibits a Ca(2+)-dependent transglutaminase activity (TGase) that cross-links proteins involved in wound healing, tissue remodeling, and ECM stabilization. Since tTG is in proximity to sites of NO production, has 18 free cysteine residues, and utilizes a cysteine for catalysis, we investigated the factors that regulated NO binding and tTG activity. We report that TGase activity is regulated by NO through a unique Ca(2+)-dependent mechanism. Tissue TG can be poly-S-nitrosylated by the NO carrier, S-nitrosocysteine (CysNO). In the absence of Ca(2+), up to eight cysteines were nitrosylated without modifying TGase activity. In the presence of Ca(2+), up to 15 cysteines were found to be nitrosylated and this modification resulted in an inhibition of TGase activity. The addition of Ca(2+) to nitrosylated tTG was able to trigger the release of NO groups (i.e. denitrosylation). tTG nitrosylated in the absence of Ca(2+) was 6-fold more susceptible to inhibition by Mg-GTP. When endothelial cells in culture were incubated with tTG and stimulated to produce NO, the exogenous tTG was S-nitrosylated. Furthermore, S-nitrosylated tTG inhibited platelet aggregation induced by ADP. In conclusion, we provide evidence that Ca(2+) regulates the S-nitrosylation and denitrosylation of tTG and thereby TGase activity. These data suggest a novel allosteric role for Ca(2+) in regulating the inhibition of tTG by NO and a novel function for tTG in dispensing NO bioactivity.

136 citations


Journal ArticleDOI
TL;DR: The prevalence and higher reactivity of thiols over other nucleophiles account for the propensity of S-nitrosylation and mixed disulfide bridge formation, and increasing evidence suggests that parasitic cysteine proteases could represent NO targets, providing molecular bases for the parasiticidal effect of NO.

25 citations


Journal ArticleDOI
TL;DR: It is demonstrated that NO‐mediated inhibition of other targets, possibly caspases, but not AP‐1, is a crucial event responsible for protection against anti‐CD95‐stimulated apoptosis.
Abstract: Nitric oxide (NO) plays an important anti-apoptotic role by inactivating both upstream and downstream apoptotic molecules. We now report that exogenously supplied NO protected Jurkat T cells from anti-CD95-stimulated apoptosis. We have recently shown that nitrosation of the activator protein-1 (AP-1) transcriptional factor is crucial for NO-mediated inhibition of cell death triggered by etoposide or ceramide. Since the inhibition of apoptosis by NO has been reported to involve AP-1, we evaluated its involvement in in CD95-mediated cell death. Cross-linking of CD95 enhanced AP-1 DNA binding activity and AP-1-dependent CD95L transactivation, which were both significantly reduced by different NO-donors compounds. However, AP-1 induction does not seem to significantly contribute to anti-CD95-triggered apoptosis, as cell death could not be prevented by using the recombinant Fas-Fc fusion protein which inhibits the CD95/CD95L interaction. We observed that caspase 3-like activity was negatively modulated by several NO-donors in vitro and that titratable thiol groups of purified caspases 3, 7, and 9 decreased in the presence of NO-releasing compounds. In conclusion, we demonstrated that NO-mediated inhibition of other targets, possibly caspases, but not AP-1, is a crucial event responsible for protection against anti-CD95-stimulated apoptosis. Even though NO affects multiple molecular mechanisms, the relevant target for exerting the cellular effects, may vary among different models. J. Cell. Biochem. 82: 123–133, 2001. © 2001 Wiley-Liss, Inc.

12 citations