Trends in Molecular Medicine
Volume 15, Issue 9, September 2009, Pages 391-404
Journal home page for Trends in Molecular Medicine

Review
Feature Review
Protein S-nitrosylation in health and disease: a current perspective

https://doi.org/10.1016/j.molmed.2009.06.007Get rights and content

Protein S-nitrosylation constitutes a large part of the ubiquitous influence of nitric oxide on cellular signal transduction and accumulating evidence indicates important roles for S-nitrosylation both in normal physiology and in a broad spectrum of human diseases. Here we review recent findings that implicate S-nitrosylation in cardiovascular, pulmonary, musculoskeletal and neurological (dys)function, as well as in cancer. The emerging picture shows that, in many cases, pathophysiology correlates with hypo- or hyper-S-nitrosylation of specific protein targets rather than a general cellular insult due to loss of or enhanced nitric oxide synthase activity. In addition, it is increasingly evident that dysregulated S-nitrosylation can not only result from alterations in the expression, compartmentalization and/or activity of nitric oxide synthases, but can also reflect a contribution from denitrosylases, including prominently the S-nitrosoglutathione (GSNO)-metabolizing enzyme GSNO reductase. Finally, because exogenous mediators of protein S-nitrosylation or denitrosylation can substantially affect the development or progression of disease, potential therapeutic agents that modulate S-nitrosylation could well have broad clinical utility.

Introduction

In mammalian cells, the L-Arg-dependent nitric oxide (NO) synthases – neuronal NOS (nNOS, NOS1), inducible NOS (iNOS, NOS2) and endothelial NOS (eNOS, NOS3) – are the major sources of endogenous NO, and stimulus-coupled activation or induction of NO synthases has been shown to mediate or modulate a broad range of cellular signaling pathways. The physiological influence of NO is exerted predominantly through the posttranslational modification and functional regulation of proteins. It was first established that nitrosylation of heme iron within soluble guanylate cyclase activates the enzyme to generate cyclic GMP and thereby subserves NO-based vasoactivity. However, hemes do not generally elicit cellular signaling involving posttranslational modification of proteins and thus an explanation for most NO-based bioactivity was not apparent. Subsequently, a large body of experimental evidence has demonstrated that S-nitrosylation of Cys residues within a broad functional spectrum of proteins constitute a large part of the ubiquitous influence of NO on cellular function [1].

Expression of iNOS is induced in many mammalian cell types by a variety of stressors or injury and the cytotoxic action of NO, generated in particular by phagocytic cells, raised the possibility that NO generated at relatively high and sustained levels by iNOS could compromise cellular function through generalized nitrosative stress. However, the emerging recognition that NO is involved in a multiplicity of cellular signal transduction pathways through protein S-nitrosylation pointed to the possibility that dysregulated S-nitrosylation could contribute to pathophysiologies characteristic of a wide range of disease states [2]. The relatively recent development of improved methods for analysis of protein S-nitrosylation 3, 4 has facilitated the identification of numerous S-nitrosylated proteins (SNO-proteins) for which levels of S-nitrosylation can be altered in disease. The emerging picture shows that hypo- or hyper-S-nitrosylation of these specific protein targets (which result in alterations in protein function) are directly implicated in the etiology and symptomatology of an increasing number of human diseases, prominently including disorders of the cardiovascular, musculoskeletal and nervous systems (Table 1). In a number of cases, specific Cys residues that are the loci of (patho)physiological regulation by S-nitrosylation have been identified.

The molecular mechanisms underlying (dys)regulation of S-nitrosylation and possible approaches to therapeutic alteration of SNO-protein levels are now the focus of increasing attention. Although NOS expression and activity are obvious governors of S-nitrosylation, the co-localization of NOS enzymes with target proteins, including direct interactions, seems in many cases to be an important determinant of S-nitrosylation under physiological conditions; accordingly, aberrant NOS localization seems to be involved in at least several diseases (Figure 1). Such deficits can have a genetic basis. For example, in a variant of long QT syndrome, a mutation in an nNOS scaffold protein results in disinhibition of nNOS and aberrant S-nitrosylation of a cardiac ion channel [5]. In addition to localization, it has been established that transfer of NO groups between proteins and glutathione governs a cellular equilibrium between low-molecular-weight and protein S-nitrosothiols (SNOs) (Figure 2). In mouse models, genetic ablation of S-nitrosoglutathione reductase (GSNOR), the enzyme principally responsible for GSNO metabolism, results in enhanced levels of SNO-proteins and significantly attenuates experimental asthma and heart failure 6, 7, but increases the severity of endotoxic shock [8]. Finally, the therapeutic potential of agents that affect S-nitrosylation is being explored with promising results (Table 2). These agents have the potential to restore deficient SNO-proteins to physiological levels or to otherwise influence cellular signaling pathways that are mediated or modulated by S-nitrosylation. For example, SNO-repleting agents are highly efficacious in the setting of inflammation, including experimental models of lung injury, stroke and multiple sclerosis, in which S-nitrosylation seems to play a major role in expression of the innate immune response 9, 10, 11, 12. This review focuses on the numerous proteins and signaling pathways that are regulated by S-nitrosylation in the context of diseases, in which aberrant S-nitrosylation has recently been implicated.

Section snippets

Modulation of SNO production: NO synthases and organic nitrates

In tissues and extracellular fluids, SNO levels are likely to reflect NOS activity and, accordingly, can be modulated through altered expression or activity of enzymes that control the availability of endogenous NOS substrates (e.g. L-Arg) or endogenous NOS inhibitors (e.g. asymmetric dimethylarginine, ADMA). In addition, a number of enzymes in the L-Arg/NO pathway are also targets of regulatory S-nitrosylation [1], including arginase, which catabolizes L-Arg. Arginase 1 (Arg1; cytosolic

GSNO reductase and its regulation by NO/SNO: implications for asthma

A major mechanism for protein S-nitrosylation in vivo is the reversible transnitrosylation of protein thiols by GSNO, the predominant low-molecular-weight SNO (Figure 2) [2]. GSNO is the preferred physiological substrate for glutathione-dependent formaldehyde dehydrogenase (class III alcohol dehydrogenase, ADH III, which in methylotropic bacteria can also metabolize formaldehyde), whereas alcohols are apparently not physiological substrates of ADH III. Thus, the enzyme has been renamed GSNO

Ras S-nitrosylation: adaptive immunity and tumor maintenance

Many GTPases within the Ras superfamily contain redox-sensitive Cys residues that are susceptible to S-nitrosylation [34]. In the cases of H-, K- and N-Ras, NO promotes the conversion of inactive GDP-bound Ras to its active GTP-bound form by guanine nucleotide exchange. Ras activation is coincident with S-nitrosylation of C118, which resides within the nucleotide-binding domain; mutation of C118 abolishes NO-induced Ras activation [34]. Although S-nitrosylation of Ras is closely coupled to NOS

Aberrant hemoglobin S-nitrosylation and the human respiratory cycle: sickle cell anemia, banked blood and pulmonary arterial hypertension

Accumulating evidence supports a role for red blood cell (RBC) SNOs, which originate from S-nitroso-(βCys93)hemoglobin (SNO-Hb), in mediating oxygen tension (PO2)-dependent vasodilatory activity [43] in the human respiratory cycle. In the three-gas (NO, O2, CO2) model of the respiratory cycle, RBCs liberate NO-based bioactivity to enable efficient O2 delivery (which is primarily a function of blood flow). This activity of RBCs seems to require the transfer of SNO from Hb to RBC

Control of hypoxic signaling: tumor radiotherapy, protection from myocardial ischemia, and development of pulmonary arterial hypertension

HIF-1, a master transcriptional regulator, is activated at low oxygen tension through stabilization of its α subunit (HIF-1α). Under normoxia, hydroxylation of HIF-1α by an O2-dependent prolyl hydroxylase promotes its binding with and ubiquitylation by the E3 ligase complex containing Von Hippel-Lindau disease tumor suppressor (pVHL) and subsequent proteasomal degradation. Accumulating evidence suggests that, under normoxia, NO/SNO stabilizes HIF-1α (thus mimicking hypoxia) [1].

S

(Dys)regulated S-nitrosylation in the heart through differential NOS expression and localization: contractility, ischemia and long QT

The importance of NOS subcellular compartmentalization is well exemplified in the heart, where eNOS and nNOS are differentially localized and exhibit opposite effects on myocardial contractility 25, 59, 60. eNOS, associated primarily with sarcolemmal caveolae, attenuates β-AR-dependent myocardial contractility [61]; inhibition of the L-type Ca2+ channel (LTCC), at least in part by S-nitrosylation [26], seems to underlie this effect. By contrast, nNOS is primarily localized to the sarcoplasmic

S-nitrosylation in disorders of skeletal muscle

The skeletal muscle ryanodine receptor (RyR) isoform RyR1 is activated by S-nitrosylation of C3635, which reduces the inhibitory effect of Ca2+–CaM on the channel [74]. A number of mutations in RyR1 (e.g. Y522S) are associated with malignant hyperthermia and related diseases that are characterized by involuntary muscle contractures, tissue lysis (rhabdomyolysis) and sudden death in response to elevated environmental temperatures 75, 76. Heterozygous RyR1Y522S/wt mutant mice, which have a

iNOS-induced S-nitrosylation in diabetes

Accumulating evidence links insulin resistance in type 2 diabetes to NO production and protein S-nitrosylation [83]. iNOS expression is increased in mouse models of diabetes and iNOS knockout mice fed a high-fat diet develop obesity but have improved glucose tolerance and normal insulin sensitivity (in vivo) and insulin-stimulated glucose uptake (ex vivo) compared with diet-induced obese wild-type mice [84]. iNOS-dependent S-nitrosylation of protein kinase B (PKB/Akt), the insulin receptor

Nitrosative stress in neurodegenerative diseases

Dysregulated protein S-nitrosylation (which can result from overactivation of NMDA receptors) seems to be prevalent in neurodegenerative disorders characterized, in particular, by the accumulation of misfolded proteins 2, 93. Hyper-S-nitrosylation of parkin, protein disulfide isomerase (PDI), peroxiredoxin 2 (Pdx2), X-linked inhibitor of apoptosis (XIAP) and dynamin-related protein 1 (Drp1) is observed in brains from patients with neurological disorders (including Alzheimer's, sporadic

SNO-GAPDH and cell death: therapeutic implications in neurodegeneration and cancer

Nuclear accumulation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a hallmark of stress-induced cell death [100]. Stimulation of nNOS in neurons or of iNOS in macrophages (by NMDA and cytokines, respectively) triggers GAPDH S-nitrosylation at its active site, C145, which promotes GAPDH association with the E3 ubiquitin ligase Siah1 and results in nuclear translocation of the SNO-GAPDHSiah1 complex [101]. Within the nucleus, SNO-GAPDH stabilizes Siah1, which facilitates the

Regulation of prostaglandin synthesis: preconditioning in ischemia–reperfusion injury

Prostaglandin synthesis by cyclo-oxygenase (COX) is coupled to iNOS activation in cells and tissues and S-nitrosylation is implicated in this effect. iNOS binds directly to and activates COX-2 by S-nitrosylation of a single cysteine (C526) proximal to the substrate (arachidonic acid, AA) binding site [112] and COX-2 S-nitrosylation and prostaglandin E2 formation are attenuated by blocking iNOSCOX-2 interaction [112]. Similarly, binding and S-nitrosylation of COX-2 by nNOS seem to mediate NMDA

Anti-inflammatory activities of endogenous and exogenous S-nitrosothiols

Protein S-nitrosylation by iNOS-derived NO occurs downstream of TLR activation and can be proinflammatory; targets of TLR-dependent S-nitrosylation include surfactant protein D, which is increasingly implicated in lung inflammation 122, 123. However, numerous additional studies point to a role for S-nitrosylation in the feedback inhibition of TLR-mediated signaling. NOS and/or low-molecular-weight SNOs inhibit the expression both of iNOS 124, 125, 126 and of multiple cytokines {interleukin-1β

Future directions

The broad purview of protein S-nitrosylation in normal and disturbed cell function presents, in principle, novel therapeutic opportunities in a wide range of human diseases. These opportunities remain largely untapped. In addition, the improvement and dissemination of new methodologies for analysis of protein S-nitrosylation – which have facilitated many of the recent discoveries detailed here and have been reviewed recently 137, 138 – will allow the role of aberrant S-nitrosylation to be

Disclosure statement

JSS holds equity in LifeHealth, N30 Pharma and Vindica, companies developing assays and uses for NO-based molecules.

Acknowledgements

This work was supported by grants U19-ES012496, HL075443 and HL059130 from the NIH.

References (153)

  • S. Ibiza

    Endothelial nitric oxide synthase regulates T cell receptor signaling at the immunological synapse

    Immunity

    (2006)
  • M.P. Moya

    Inhaled ethyl nitrite gas for persistent pulmonary hypertension of the newborn

    Lancet

    (2002)
  • F. Li

    Regulation of HIF-1α stability through S-nitrosylation

    Mol. Cell

    (2007)
  • H. Yasuda

    Solid tumor physiology and hypoxia-induced chemo/radio-resistance: novel strategy for cancer therapy: nitric oxide donor as a therapeutic enhancer

    Nitric Oxide

    (2008)
  • P.B. Massion

    Regulation of the mammalian heart function by nitric oxide

    Comp. Biochem. Physiol. A Mol. Integr. Physiol.

    (2005)
  • R.C. Siow

    Cardiovascular targets for estrogens and phytoestrogens: transcriptional regulation of nitric oxide synthase and antioxidant defense genes

    Free Radic. Biol. Med.

    (2007)
  • T. Damy

    Increased neuronal nitric oxide synthase-derived NO production in the failing human heart

    Lancet

    (2004)
  • K. Asada

    Redox- and calmodulin-dependent S-nitrosylation of the KCNQ1 channel

    J. Biol. Chem.

    (2009)
  • W.J. Durham

    RyR1 S-nitrosylation underlies environmental heat stroke and sudden death in Y522S RyR1 knockin mice

    Cell

    (2008)
  • J.S. Stamler

    A SNO storm in skeletal muscle

    Cell

    (2008)
  • T. Yasukawa

    S-Nitrosylation-dependent inactivation of Akt/protein kinase B in insulin resistance

    J. Biol. Chem.

    (2005)
  • H. Sugita

    Inducible nitric-oxide synthase and NO donor induce insulin receptor substrate-1 degradation in skeletal muscle cells

    J. Biol. Chem.

    (2005)
  • D.T. Hess

    Protein S-nitrosylation: purview and parameters

    Nat. Rev. Mol. Cell Biol.

    (2005)
  • S.R. Jaffrey

    Protein S-nitrosylation: a physiological signal for neuronal nitric oxide

    Nat. Cell Biol.

    (2001)
  • K. Ueda

    Syntrophin mutation associated with long QT syndrome through activation of the nNOS–SCN5A macromolecular complex

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • L.G. Que

    Protection from experimental asthma by an endogenous bronchodilator

    Science

    (2005)
  • B. Lima

    Endogenous S-nitrosothiols protect against myocardial injury

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • R.L. Auten

    Inhaled ethyl nitrite prevents hyperoxia-impaired postnatal alveolar development in newborn rats

    Am. J. Respir. Crit. Care Med.

    (2007)
  • H.E. Marshall

    Protection from LPS-induced lung injury by augmentation of airway S-nitrosothiols

    Am. J. Respir. Crit. Care Med.

    (2009)
  • R. Prasad

    GSNO attenuates EAE disease by S-nitrosylation-mediated modulation of endothelial-monocyte interactions

    Glia

    (2007)
  • L. Santhanam

    Inducible NO synthase dependent S-nitrosylation and activation of arginase1 contribute to age-related endothelial dysfunction

    Circ. Res.

    (2007)
  • L. Santhanam

    Arginase and vascular aging

    J. Appl. Physiol.

    (2008)
  • K. Ckless

    Inhibition of arginase activity enhances inflammation in mice with allergic airway disease, in association with increases in protein S-nitrosylation and tyrosine nitration

    J. Immunol.

    (2008)
  • M.L. North

    Functionally important role for arginase 1 in the airway hyperresponsiveness of asthma

    Am. J. Physiol. Lung Cell. Mol. Physiol.

    (2009)
  • M.T. Salam

    Roles of arginase variants, atopy, and ozone in childhood asthma

    J. Allergy Clin. Immunol.

    (2009)
  • H. Maarsingh

    Arginase inhibition protects against allergen-induced airway obstruction, hyperresponsiveness, and inflammation

    Am. J. Respir. Crit. Care Med.

    (2008)
  • J.S. Stamler

    Nitroglycerin-mediated S-nitrosylation of proteins: a field comes full cycle

    Circ. Res.

    (2008)
  • N. Sayed

    Desensitization of soluble guanylyl cyclase, the NO receptor, by S-nitrosylation

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • N. Sayed

    Nitroglycerin-induced S-nitrosylation and desensitization of soluble guanylyl cyclase contribute to nitrate tolerance

    Circ. Res.

    (2008)
  • A.L. Taylor

    Combination of isosorbide dinitrate and hydralazine in blacks with heart failure

    N. Engl. J. Med.

    (2004)
  • J.M. Hare et al.

    NO/redox disequilibrium in the failing heart and cardiovascular system

    J. Clin. Invest.

    (2005)
  • J. Sun

    Hypercontractile female hearts exhibit increased S-nitrosylation of the L-type Ca2+ channel α1 subunit and reduced ischemia/reperfusion injury

    Circ. Res.

    (2006)
  • D.R. Gonzalez

    Deficient ryanodine receptor S-nitrosylation increases sarcoplasmic reticulum calcium leak and arrhythmogenesis in cardiomyocytes

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • L. Que

    S-Nitrosoglutathione reductase: an important regulator in asthma

    Am. J. Respir. Crit. Care Med.

    (2009)
  • V. Bhandari

    Essential role of nitric oxide in VEGF-induced, asthma-like angiogenic, inflammatory, mucus, and physiologic responses in the lung

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • K. Zaman

    Concentration-dependent effects of endogenous S-nitrosoglutathione on gene regulation by specificity proteins Sp3 and Sp1

    Biochem. J.

    (2004)
  • K.W. Raines

    Nitric oxide cell signaling: S-nitrosation of Ras superfamily GTPases

    Cardiovasc. Res.

    (2007)
  • J. Heo et al.

    Ras regulation by reactive oxygen and nitrogen species

    Biochemistry

    (2006)
  • S. Ibiza

    Endothelial nitric oxide synthase regulates N-Ras activation on the Golgi complex of antigen-stimulated T cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • Y. Iwakiri

    Nitric oxide synthase generates nitric oxide locally to regulate compartmentalized protein S-nitrosylation and protein trafficking

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • Cited by (623)

    View all citing articles on Scopus
    View full text