Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 146(1): 53-66, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21729780

RESUMO

Malignant gliomas are aggressive brain tumors with limited therapeutic options, and improvements in treatment require a deeper molecular understanding of this disease. As in other cancers, recent studies have identified highly tumorigenic subpopulations within malignant gliomas, known generally as cancer stem cells. Here, we demonstrate that glioma stem cells (GSCs) produce nitric oxide via elevated nitric oxide synthase-2 (NOS2) expression. GSCs depend on NOS2 activity for growth and tumorigenicity, distinguishing them from non-GSCs and normal neural progenitors. Gene expression profiling identified many NOS2-regulated genes, including the cell-cycle inhibitor cell division autoantigen-1 (CDA1). Further, high NOS2 expression correlates with decreased survival in human glioma patients, and NOS2 inhibition slows glioma growth in a murine intracranial model. These data provide insight into how GSCs are mechanistically distinct from their less tumorigenic counterparts and suggest that NOS2 inhibition may be an efficacious approach to treating this devastating disease.


Assuntos
Proliferação de Células , Glioma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Animais , Autoantígenos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Óxido Nítrico/metabolismo , Células Tumorais Cultivadas
2.
Nat Rev Mol Cell Biol ; 10(10): 721-32, 2009 10.
Artigo em Inglês | MEDLINE | ID: mdl-19738628

RESUMO

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.


Assuntos
Aldeído Oxirredutases/metabolismo , Proteínas/metabolismo , Tiorredoxinas/metabolismo , Humanos , Óxido Nítrico/metabolismo , Nitrosação , Oxirredução , S-Nitrosoglutationa/metabolismo , S-Nitrosotióis/metabolismo , Transdução de Sinais
3.
J Biol Chem ; 294(36): 13336-13343, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31320475

RESUMO

Dynamic control of thioredoxin (Trx) oxidoreductase activity is essential for balancing the need of cells to rapidly respond to oxidative/nitrosative stress and to temporally regulate thiol-based redox signaling. We have previously shown that cytokine stimulation of the respiratory epithelium induces a precipitous decline in cell S-nitrosothiol, which depends upon enhanced Trx activity and proteasome-mediated degradation of Txnip (thioredoxin-interacting protein). We now show that tumor necrosis factor-α-induced Txnip degradation in A549 respiratory epithelial cells is regulated by the extracellular signal-regulated protein kinase (ERK) mitogen-activated protein kinase pathway and that ERK inhibition augments both intracellular reactive oxygen species and S-nitrosothiol. ERK-dependent Txnip ubiquitination and proteasome degradation depended upon phosphorylation of a PXTP motif threonine (Thr349) located within the C-terminal α-arrestin domain and proximal to a previously characterized E3 ubiquitin ligase-binding site. Collectively, these findings demonstrate the ERK mitogen-activated protein kinase pathway to be integrally involved in regulating Trx oxidoreductase activity and that the regulation of Txnip lifetime via ERK-dependent phosphorylation is an important mediator of this effect.


Assuntos
Proteínas de Transporte/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo , Células A549 , Humanos , Espectrometria de Massas , Células Tumorais Cultivadas
4.
J Biol Chem ; 289(5): 3066-72, 2014 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-24338024

RESUMO

S-nitrosylation of nuclear factor κB (NF-κB) on the p65 subunit of the p50/p65 heterodimer inhibits NF-κB DNA binding activity. We have recently shown that p65 is constitutively S-nitrosylated in the lung and that LPS-induced injury elicits a decrease in SNO-p65 levels concomitant with NF-κB activation in the respiratory epithelium and initiation of the inflammatory response. Here, we demonstrate that TNFα-mediated activation of NF-κB in the respiratory epithelium similarly induces p65 denitrosylation. This process is mediated by the denitrosylase thioredoxin (Trx), which becomes activated upon cytokine-induced degradation of thioredoxin-interacting protein (Txnip). Similarly, inhibition of Trx activity in the lung attenuates LPS-induced SNO-p65 denitrosylation, NF-κB activation, and airway inflammation, supporting a pathophysiological role for this mechanism in lung injury. These data thus link stimulus-coupled activation of NF-κB to a specific, protein-targeted denitrosylation mechanism and further highlight the importance of S-nitrosylation in the regulation of the immune response.


Assuntos
Lesão Pulmonar/metabolismo , Transdução de Sinais/imunologia , Tiorredoxinas/metabolismo , Fator de Transcrição RelA/metabolismo , Adenocarcinoma , Animais , Linhagem Celular Tumoral , Citocinas/metabolismo , Modelos Animais de Doenças , Células HEK293 , Humanos , Lipopolissacarídeos/toxicidade , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Neoplasias Pulmonares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Subunidade p50 de NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Tiorredoxinas/genética , Tiorredoxinas/imunologia
5.
Methods ; 62(2): 130-7, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23064468

RESUMO

The proteomic analysis of S-nitrosylated protein (SNO-proteins) has long depended on the biotin switch technique (BST), which requires blocking of free thiols, ascorbate-based denitrosylation of SNO-Cys, biotinylation of nascent thiol and avidin-based affinity isolation. A more recent development is resin assisted-capture of SNO-proteins (SNO-RAC), which substitutes thiopropyl Sepharose (TPS) for biotin-avidin, thus reducing the number of steps required for enrichment of S-nitrosylated proteins. In addition, SNO-RAC facilitates on-resin proteolytic digestion following SNO-protein capture, greatly simplifying the purification of peptides containing sites of S-nitrosylation ("SNO-sites"). This resin-based approach has also now been applied to detection of alternative Cys-based modifications, including S-palmitoylation (Acyl-RAC) and S-oxidation (Ox-RAC). Here, we review the important steps to minimize false-positive identification of SNO-proteins, give detailed methods for processing of protein-bound TPS for mass spectrometry (MS) based analysis, and discuss the various quantitative MS methods that are compatible with SNO-RAC. We also discuss strategies to overcome the current limitations surrounding MS-based SNO-site localization in peptides containing more than one potential target Cys residue. This article therefore serves as a starting point and guide for the MS-focused exploration of SNO-proteomes by SNO-RAC.


Assuntos
Proteoma/química , S-Nitrosotióis/química , Animais , Cromatografia de Afinidade , Humanos , Processamento de Proteína Pós-Traducional , Proteoma/isolamento & purificação , Proteoma/metabolismo , S-Nitrosotióis/isolamento & purificação , S-Nitrosotióis/metabolismo , Extração em Fase Sólida , Coloração e Rotulagem , Espectrometria de Massas em Tandem/métodos
6.
bioRxiv ; 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38585928

RESUMO

Proteins undergo reversible S -acylation via a thioester linkage in vivo. S -palmitoylation, modification by C16:0 fatty acid, is a common S -acylation that mediates critical protein-membrane and protein-protein interactions. The most widely used S -acylation assays, including acyl-biotin exchange and acyl resin-assisted capture, utilize blocking of free Cys thiols, hydroxylamine-dependent cleavage of the thioester and subsequent labeling of nascent thiol. These assays generally require >500 micrograms of protein input material per sample and numerous reagent removal and washing steps, making them laborious and ill-suited for high throughput and low input applications. To overcome these limitations, we devised Acyl-Trap, a suspension trap-based assay that utilizes a thiol-reactive quartz to enable buffer exchange and hydroxylamine-mediated S -acyl enrichment. We show that the method is compatible with protein-level detection of S -acylated proteins (e.g. H-Ras) as well as S -acyl site identification and quantification using on-trap isobaric labeling and LC-MS/MS from as little as 20 micrograms of protein input. In mouse brain, Acyl-Trap identified 279 reported sites of S -acylation and 1298 previously unreported putative sites. Also described are conditions for long-term hydroxylamine storage, which streamlines the assay. More generally, Acyl-Trap serves as a proof-of-concept for PTM-tailored suspension traps suitable for both traditional protein detection and chemoproteomic workflows.

7.
Nitric Oxide ; 34: 37-46, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23438482

RESUMO

The cytokine-inducible isoform of nitric oxide synthase (NOS2) is constitutively expressed in human respiratory epithelia and is upregulated in inflammatory lung disease. Here, we sought to better define the protein interactions that may be important for NOS2 activity and stability, as well as to identify potential targets of NOS2-derived NO, in the respiratory epithelium. We overexpressed Flag-tagged, catalytically-inactive NOS2 in A549 cells and used mass spectrometry to qualitatively identify NOS2 co-immunoprecipitating proteins. Stable isotope labeling of amino acids in cell culture (SILAC) was used to quantify the coordinate effects of cytokine stimulation on NOS2-protein interactions. Multi-protein networks dominated the NOS2 interactome, and cytokine-inducible interactions with allosteric activators and with the ubiquitin-proteasome system were correlated with cytokine-dependent increases in NO metabolites and in NOS2 ubiquitination. The ubiquitin ligase scaffolding protein, FBXO45, was identified as a novel, direct NOS2 interactor. Similar to the SPRY domain-containing SOCS box (SPSB) proteins, FBXO45 requires Asn27 in the (23)DINNN(27) motif of NOS2 for its interaction. However, FBXO45 is unique from the SPSBs in that it recruits a distinct E3 ligase complex containing MYCBP2 and SKP1. Collectively, these findings demonstrate the general utility of interaction proteomics for defining new aspects of NOS2 physiology.


Assuntos
Óxido Nítrico Sintase Tipo II/metabolismo , Proteoma/análise , Proteoma/metabolismo , Mucosa Respiratória/citologia , Sequência de Aminoácidos , Linhagem Celular , Citocinas/metabolismo , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Óxido Nítrico Sintase/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteômica/métodos , Alinhamento de Sequência , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
8.
Circ Res ; 106(4): 633-46, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20203313

RESUMO

Well over 2 decades have passed since the endothelium-derived relaxation factor was reported to be the gaseous molecule nitric oxide (NO). Although soluble guanylyl cyclase (which generates cyclic guanosine monophosphate, cGMP) was the first identified receptor for NO, it has become increasingly clear that NO exerts a ubiquitous influence in a cGMP-independent manner. In particular, many, if not most, effects of NO are mediated by S-nitrosylation, the covalent modification of a protein cysteine thiol by an NO group to generate an S-nitrosothiol (SNO). Moreover, within the current framework of NO biology, endothelium-derived relaxation factor activity (ie, G protein-coupled receptor-mediated, or shear-induced endothelium-derived NO bioactivity) is understood to involve a central role for SNOs, acting both as second messengers and signal effectors. Furthermore, essential roles for S-nitrosylation have been implicated in virtually all major functions of NO in the cardiovascular system. Here, we review the basic biochemistry of S-nitrosylation (and denitrosylation), discuss the role of S-nitrosylation in the vascular and cardiac functions of NO, and identify current and potential clinical applications.


Assuntos
Doenças Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Óxido Nítrico/metabolismo , Processamento de Proteína Pós-Traducional , S-Nitrosotióis/metabolismo , Transdução de Sinais , Animais , Apoptose , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/patologia , Sistema Cardiovascular/fisiopatologia , GMP Cíclico/metabolismo , Endotélio Vascular/metabolismo , Fatores Relaxantes Dependentes do Endotélio/metabolismo , Humanos , Inflamação/metabolismo , Miocárdio/metabolismo , Neovascularização Fisiológica , Resistência Vascular
9.
Proc Natl Acad Sci U S A ; 106(45): 18948-53, 2009 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-19864628

RESUMO

The ubiquitous cellular influence of nitric oxide (NO) is exerted substantially through protein S-nitrosylation. Whereas NO is highly promiscuous, physiological S-nitrosylation is typically restricted to one or very few Cys residue(s) in target proteins. The molecular basis for this specificity may derive from properties of the target protein, the S-nitrosylating species, or both. Here, we describe a protein microarray-based approach to investigate determinants of S-nitrosylation by biologically relevant low-mass S-nitrosothiols (SNOs). We identify large sets of yeast and human target proteins, among which those with active-site Cys thiols residing at N termini of alpha-helices or within catalytic loops were particularly prominent. However, S-nitrosylation varied substantially even within these families of proteins (e.g., papain-related Cys-dependent hydrolases and rhodanese/Cdc25 phosphatases), suggesting that neither secondary structure nor intrinsic nucleophilicity of Cys thiols was sufficient to explain specificity. Further analyses revealed a substantial influence of NO-donor stereochemistry and structure on efficiency of S-nitrosylation as well as an unanticipated and important role for allosteric effectors. Thus, high-throughput screening and unbiased proteome coverage reveal multifactorial determinants of S-nitrosylation (which may be overlooked in alternative proteomic analyses), and support the idea that target specificity can be achieved through rational design of S-nitrosothiols.


Assuntos
Cisteína/metabolismo , Óxido Nítrico/metabolismo , Proteínas/metabolismo , S-Nitrosotióis/metabolismo , Amida Sintases/metabolismo , Humanos , Análise Serial de Proteínas/métodos , Proteínas Tirosina Fosfatases/metabolismo , Proteômica/métodos , Especificidade por Substrato , Tiossulfato Sulfurtransferase/metabolismo , Ubiquitina Tiolesterase/metabolismo , Leveduras
10.
J Lipid Res ; 52(2): 393-8, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21044946

RESUMO

Protein S-acylation is a major posttranslational modification whereby a cysteine thiol is converted to a thioester. A prototype is S-palmitoylation (fatty acylation), in which a protein undergoes acylation with a hydrophobic 16 carbon lipid chain. Although this modification is a well-recognized determinant of protein function and localization, current techniques to study cellular S-acylation are cumbersome and/or technically demanding. We recently described a simple and robust methodology to rapidly identify S-nitrosylation sites in proteins via resin-assisted capture (RAC) and provided an initial description of the applicability of the technique to S-acylated proteins (acyl-RAC). Here we expand on the acyl-RAC assay, coupled with mass spectrometry-based proteomics, to characterize both previously reported and novel sites of endogenous S-acylation. Acyl-RAC should therefore find general applicability in studies of both global and individual protein S-acylation in mammalian cells.


Assuntos
Cisteína/metabolismo , Lipoilação , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Acilação , Eletroforese em Gel de Poliacrilamida/métodos , Sefarose/análogos & derivados , Sefarose/química , Proteínas ras/análise
11.
J Biol Chem ; 284(52): 36160-36166, 2009 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-19847012

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo/fisiologia , Proteínas de Transporte/genética , Linhagem Celular , Sobrevivência Celular/fisiologia , Humanos , Óxido Nítrico/genética , Oxirredução , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores , Tiorredoxina Dissulfeto Redutase/genética , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
12.
Free Radic Biol Med ; 52(9): 1620-33, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22343413

RESUMO

Nitric oxide (NO) is an inevitable product of life in an oxygen- and nitrogen-rich environment. This reactive diatomic molecule exhibits microbial cytotoxicity, in large part by facilitating nitrosative stress and inhibiting heme-containing proteins within the aerobic respiratory chain. Metabolism of NO is therefore essential for microbial life. In many bacteria, fungi, and protozoa, the evolutionarily ancient flavohemoglobin (flavoHb) converts NO and O(2) to inert nitrate (NO(3)(-)) and undergoes catalytic regeneration via flavin-dependent reduction. Since its identification, widespread efforts have characterized roles for flavoHb in microbial nitrosative stress protection. Subsequent genomic studies focused on flavoHb have elucidated the transcriptional machinery necessary for inducible NO protection, such as NsrR in Escherichia coli, as well as additional proteins that constitute a nitrosative stress protection program. As an alternative strategy, flavoHb has been heterologously employed in higher eukaryotic organisms such as plants and human tumors to probe the function(s) of endogenous NO signaling. Such an approach may also provide a therapeutic route to in vivo NO depletion. Here we focus on the molecular features of flavoHb, the hitherto characterized NO-sensitive transcriptional machinery responsible for its induction, the roles of flavoHb in resisting mammalian host defense systems, and heterologous applications of flavoHb in plant/mammalian systems (including human tumors), as well as unresolved questions surrounding this paradigmatic NO-consuming enzyme.


Assuntos
Di-Hidropteridina Redutase/fisiologia , Proteínas de Escherichia coli/fisiologia , Hemeproteínas/fisiologia , NADH NADPH Oxirredutases/fisiologia , Nitrosação , Estresse Oxidativo , Aerobiose , Sequência de Aminoácidos , Anaerobiose , Di-Hidropteridina Redutase/química , Di-Hidropteridina Redutase/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Hemeproteínas/química , Hemeproteínas/genética , Humanos , Dados de Sequência Molecular , NADH NADPH Oxirredutases/química , NADH NADPH Oxirredutases/genética , Óxido Nítrico/fisiologia , Conformação Proteica , Homologia de Sequência de Aminoácidos , Transcrição Gênica
13.
Biotechniques ; 50(1): 41-5, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21231921

RESUMO

A wide range of mammalian signaling and stress pathways are mediated by nitric oxide (NO), which is synthesized in vivo by the nitric oxide synthase (NOS) family of enzymes. Experimental manipulations of NO are frequently achieved by either inhibition or activation of endogenous NOS or via providing exogenous NO sources. On the contrary, many microbes consume NO via flavohemoglobin (FlavoHb), a highly efficient NO-dioxygenase that protects from nitrosative stress. Here we report a novel resource for studying NO in mammalian cells by heterologously expressing Escherichia coli FlavoHb within a lentiviral delivery system. This technique boosts endogenous cellular consumption of NO, thus providing a simple and efficacious approach to studying mammalian NO biology that can be employed as both a primary experimental and confirmatory tool.


Assuntos
Di-Hidropteridina Redutase/química , Proteínas de Escherichia coli/química , Hemeproteínas/química , NADH NADPH Oxirredutases/química , Óxido Nítrico/metabolismo , Di-Hidropteridina Redutase/análise , Di-Hidropteridina Redutase/genética , Escherichia coli/genética , Proteínas de Escherichia coli/análise , Proteínas de Escherichia coli/genética , Flavina-Adenina Dinucleotídeo/química , Células HEK293 , Hemeproteínas/análise , Hemeproteínas/genética , Humanos , Lentivirus/genética , NADH NADPH Oxirredutases/análise , NADH NADPH Oxirredutases/genética , Óxido Nítrico/química , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Oxirredução , Transdução de Sinais , Transfecção/métodos
14.
Free Radic Biol Med ; 46(2): 119-26, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18977293

RESUMO

Protein S-nitrosylation, the posttranslational modification of cysteine thiols to form S-nitrosothiols, is a principle mechanism of nitric oxide-based signaling. Studies have demonstrated myriad roles for S-nitrosylation in organisms from bacteria to humans, and recent efforts have greatly advanced our scientific understanding of how this redox-based modification is dynamically regulated during physiological and pathophysiological conditions. The focus of this review is the biotin-switch technique (BST), which has become a mainstay assay for detecting S-nitrosylated proteins in complex biological systems. Potential pitfalls and modern adaptations of the BST are discussed, as are future directions for this assay in the burgeoning field of protein S-nitrosylation.


Assuntos
Biotina/química , Técnicas de Química Analítica/métodos , Óxido Nítrico/metabolismo , Processamento de Proteína Pós-Traducional , S-Nitrosotióis/química , Animais , Biotina/metabolismo , Extratos Celulares/química , Técnicas de Química Analítica/instrumentação , Técnicas de Química Analítica/tendências , Cisteína/química , Cisteína/metabolismo , Humanos , Oxirredução , Processamento de Proteína Pós-Traducional/fisiologia , Transdução de Sinais , Compostos de Sulfidrila/química
15.
Nat Biotechnol ; 27(6): 557-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19483679

RESUMO

We have modified the biotin switch assay for protein S-nitrosothiols (SNOs), using resin-assisted capture (SNO-RAC). Compared with existing methodologies, SNO-RAC requires fewer steps, detects high-mass S-nitrosylated proteins more efficiently, and facilitates identification and quantification of S-nitrosylated sites by mass spectrometry. When combined with iTRAQ labeling, SNO-RAC revealed that intracellular proteins may undergo rapid denitrosylation on a global scale. This methodology is readily adapted to analyzing diverse cysteine-based protein modifications, including S-acylation.


Assuntos
Proteoma/análise , Proteômica/métodos , S-Nitrosotióis/metabolismo , Compostos de Sulfidrila/metabolismo , Biotina/metabolismo , Linhagem Celular , Cisteína/análogos & derivados , Cisteína/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Espectrometria de Massas , Sensibilidade e Especificidade
16.
Sci Signal ; 2(78): ra33, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19584355

RESUMO

Agonist-induced ubiquitylation and degradation of heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) play an essential role in surface receptor homeostasis, thereby tuning many physiological processes. Although beta-arrestin and affiliated E3 ligases mediate agonist-stimulated lysosomal degradation of the beta(2)-adrenergic receptor (beta(2)AR), a prototypic GPCR, the molecular cues that mark receptors for ubiquitylation and the regulation of receptor degradation by the proteasome remain poorly understood. We show that the von Hippel-Lindau tumor suppressor protein (pVHL)-E3 ligase complex, known for its regulation of hypoxia-inducible factor (HIF) proteins, interacts with and ubiquitylates the beta(2)AR, thereby decreasing receptor abundance. We further show that the interaction of pVHL with beta(2)AR is dependent on proline hydroxylation (proline-382 and -395) and that the dioxygenase EGLN3 interacts directly with the beta(2)AR to serve as an endogenous beta(2)AR prolyl hydroxylase. Under hypoxic conditions, receptor hydroxylation and subsequent ubiquitylation decrease dramatically, thus attenuating receptor degradation and down-regulation. Notably, in both cells and tissue, the abundance of endogenous beta(2)AR is shown to reflect constitutive turnover by EGLN3 and pVHL. Our findings provide insight into GPCR regulation, broaden the functional scope of prolyl hydroxylation, and expand our understanding of the cellular response to hypoxia.


Assuntos
Dioxigenases/fisiologia , Oxigênio/fisiologia , Receptores Adrenérgicos beta 2/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/fisiologia , Linhagem Celular , Regulação para Baixo , Humanos , Hidroxilação , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia , Oxigênio/farmacologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolina/metabolismo , Ligação Proteica , Ubiquitinação
17.
Science ; 320(5879): 1050-4, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18497292

RESUMO

Nitric oxide acts substantially in cellular signal transduction through stimulus-coupled S-nitrosylation of cysteine residues. The mechanisms that might subserve protein denitrosylation in cellular signaling remain uncharacterized. Our search for denitrosylase activities focused on caspase-3, an exemplar of stimulus-dependent denitrosylation, and identified thioredoxin and thioredoxin reductase in a biochemical screen. In resting human lymphocytes, thioredoxin-1 actively denitrosylated cytosolic caspase-3 and thereby maintained a low steady-state amount of S-nitrosylation. Upon stimulation of Fas, thioredoxin-2 mediated denitrosylation of mitochondria-associated caspase-3, a process required for caspase-3 activation, and promoted apoptosis. Inhibition of thioredoxin-thioredoxin reductases enabled identification of additional substrates subject to endogenous S-nitrosylation. Thus, specific enzymatic mechanisms may regulate basal and stimulus-induced denitrosylation in mammalian cells.


Assuntos
Citosol/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Óxido Nítrico/metabolismo , S-Nitrosotióis/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxinas/metabolismo , Animais , Apoptose , Auranofina/farmacologia , Sítios de Ligação , Caspase 3/metabolismo , Inibidores de Caspase , Linhagem Celular , Dinitroclorobenzeno/farmacologia , Células HeLa , Humanos , Células Jurkat , Macrófagos/metabolismo , Mitocôndrias/enzimologia , Ratos , Proteínas Recombinantes/metabolismo , Linfócitos T/metabolismo , Receptor fas/metabolismo
18.
J Biol Chem ; 282(19): 13977-83, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17376775

RESUMO

Protein S-nitrosylation has emerged as a principal mechanism by which nitric oxide exerts biological effects. Among methods for studying protein S-nitrosylation, the biotin switch technique (BST) has rapidly gained popularity because of the ease with which it can detect individual S-nitrosylated (SNO) proteins in biological samples. The identification of SNO sites by the BST relies on the ability of ascorbate to generate a thiol from an S-nitrosothiol, but not from alternatively S-oxidized thiols (e.g. disulfides, sulfenic acids). However, the specificity of this reaction has recently been challenged, prompting several claims that the BST may produce false-positive results and raising concerns about the application of the BST under oxidizing conditions. Here we perform a comparative analysis of the BST using differentially S-oxidized and S-nitrosylated forms of protein tyrosine phosphatase 1B, as well as intact and lysed human embryonic kidney 293 cells treated with S-oxidizing and S-nitrosylating agents, and verify that the assay is highly specific for SNO. Strikingly, exposure of samples to indirect sunlight from a laboratory window resulted in artifactual ascorbate-dependent signals that are likely promoted by the semidehydroascorbate radical; protection from sunlight eliminated the artifact. In contrast, exposure of SNO proteins to a strong ultraviolet light source (SNO photolysis) prior to the BST provided independent verification of assay specificity. By combining BST with photolysis, we have shown that anti-cancer drug-induced oxidative stress facilitates the S-nitrosylation of the major apoptotic effector glyceraldehyde-3-phosphate dehydrogenase. Collectively, these experiments demonstrate that SNO-dependent signaling pathways can be modulated by oxidative conditions and suggest a potential role for S-nitrosylation in antineoplastic drug action.


Assuntos
Biotina/metabolismo , Cisteína/metabolismo , Nitrogênio/metabolismo , Estresse Oxidativo , Proteínas Tirosina Fosfatases/metabolismo , Compostos de Sulfidrila/metabolismo , Ácido Ascórbico/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Células Cultivadas/efeitos da radiação , Glutationa/metabolismo , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Nitrogênio/química , Oxirredução , Fotólise , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/química , Compostos de Sulfidrila/química , Luz Solar
19.
ACS Chem Biol ; 1(6): 355-8, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-17163772

RESUMO

S-Nitrosylation, the covalent addition of a nitrogen monoxide group to a cysteine thiol, has been shown to modify the function of a broad spectrum of mammalian, plant, and microbial proteins and thereby to convey the ubiquitous influence of nitric oxide on cellular signal transduction and host defense. Accumulating evidence indicates that dysregulated, diminished, or excessive S-nitrosylation may be implicated in a wide range of pathophysiological conditions. A recent study establishes a functional relationship between inhibitory S-nitrosylation of the redox enzyme protein disulfide isomerase (PDI), defects in regulation of protein folding within the endoplasmic reticulum (ER), and neurodegeneration. Further, an examination of human brains afflicted with Parkinson's or Alzheimer's disease supports a causal role for the S-nitrosylation of PDI and consequent ER stress in these prevalent neurodegenerative disorders.


Assuntos
Retículo Endoplasmático/metabolismo , Doenças Neurodegenerativas/metabolismo , Óxido Nítrico/química , Óxido Nítrico/fisiologia , Animais , Retículo Endoplasmático/química , Humanos , Nitrosação , Dobramento de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA