Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Strategies to increase nitric oxide signalling in cardiovascular disease

Key Points

  • Nitric oxide (NO), a diatomic free radical gas, is generated by NO synthases (NOSs) in tissues to regulate a multitude of physiological processes, most notably cardiovascular function.

  • A decrease in the formation and bioavailability of NO is a hallmark of several cardiovascular diseases.

  • Traditional ways of increasing NO levels — with nitroglycerin or other organic nitrates — have limited clinical utility, mainly owing to unfavourable pharmacokinetics and the development of tolerance.

  • Several alternative strategies to increase NO signalling in the cardiovascular system have recently emerged, with promising therapeutic potential.

  • These strategies include the identification of novel pathways for enhancing NOS activity, amplification of the nitrate–nitrite–NO pathway, novel classes of NO-donating drugs, drugs that limit NO metabolism using reactive oxygen species, and modulation of downstream phosphodiesterases and soluble guanylyl cyclases.

Abstract

Nitric oxide (NO) is a key signalling molecule in the cardiovascular, immune and central nervous systems, and crucial steps in the regulation of NO bioavailability in health and disease are well characterized. Although early approaches to therapeutically modulate NO bioavailability failed in clinical trials, an enhanced understanding of fundamental subcellular signalling has enabled a range of novel therapeutic approaches to be identified. These include the identification of: new pathways for enhancing NO synthase activity; ways to amplify the nitrate–nitrite–NO pathway; novel classes of NO-donating drugs; drugs that limit NO metabolism through effects on reactive oxygen species; and ways to modulate downstream phosphodiesterases and soluble guanylyl cyclases. In this Review, we discuss these latest developments, with a focus on cardiovascular disease.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Diseases and lifestyle factors associated with reduced bioavailability of nitric oxide.
Figure 2: Vascular nitric oxide signalling in health and disease.
Figure 3: Key reactions of nitric oxide that contribute to its diverse signalling properties.
Figure 4: Proposed mechanisms for the cardioprotective effects of nitric oxide.
Figure 5: Targets for increasing nitric oxide bioavailability in the vasculature.
Figure 6: Mechanisms by which dietary polyphenols can increase the bioavailability of nitric oxide in the cardiovascular system.

Similar content being viewed by others

References

  1. Forstermann, U. & Munzel, T. Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation 113, 1708–1714 (2006).

    PubMed  Google Scholar 

  2. Arnold, W. P., Mittal, C. K., Katsuki, S. & Murad, F. Nitric oxide activates guanylate cyclase and increases guanosine 3′:5′-cyclic monophosphate levels in various tissue preparations. Proc. Natl Acad. Sci. USA 74, 3203–3207 (1977).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Katsuki, S., Arnold, W., Mittal, C. & Murad, F. Stimulation of guanylate cyclase by sodium nitroprusside, nitroglycerin and nitric oxide in various tissue preparations and comparison to the effects of sodium azide and hydroxylamine. J. Cyclic Nucleotide Res. 3, 23–35 (1977).

    CAS  PubMed  Google Scholar 

  4. Furchgott, R. F. & Zawadzki, J. V. The obligatory role of the endothelium in the relaxation of arterial smooth muscle by acetylcholine. Nature 288, 373–376 (1980).

    CAS  PubMed  Google Scholar 

  5. Ignarro, L. J., Buga, G. M., Wood, K. S., Byrns, R. E. & Chaudhuri, G. Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc. Natl Acad. Sci. USA 84, 9265–9269 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Palmer, R. M., Ferrige, A. G. & Moncada, S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 327, 524–526 (1987). References 2–6 represent seminal papers demonstrating the discovery of NO as a signalling molecule in the cardiovascular system.

    CAS  PubMed  Google Scholar 

  7. Ghofrani, H. A., Osterloh, I. H. & Grimminger, F. Sildenafil: from angina to erectile dysfunction to pulmonary hypertension and beyond. Nat. Rev. Drug Discov. 5, 689–702 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Frostell, C., Fratacci, M. D., Wain, J. C., Jones, R. & Zapol, W. M. Inhaled nitric oxide. A selective pulmonary vasodilator reversing hypoxic pulmonary vasoconstriction. Circulation 83, 2038–2047 (1991). This is the first study to show the therapeutic effects of inhaled NO in pulmonary hypertension.

    CAS  PubMed  Google Scholar 

  9. Kinsella, J. P. et al. Early inhaled nitric oxide therapy in premature newborns with respiratory failure. N. Engl. J. Med. 355, 354–364 (2006).

    CAS  PubMed  Google Scholar 

  10. Munzel, T., Daiber, A. & Mulsch, A. Explaining the phenomenon of nitrate tolerance. Circ. Res. 97, 618–628 (2005).

    PubMed  Google Scholar 

  11. Lundberg, J. O., Feelisch, M., Bjorne, H., Jansson, E. A. & Weitzberg, E. Cardioprotective effects of vegetables: is nitrate the answer? Nitric Oxide 15, 359–362 (2006).

    CAS  PubMed  Google Scholar 

  12. Classen, H. G., Stein-Hammer, C. & Thoni, H. Hypothesis: the effect of oral nitrite on blood pressure in the spontaneously hypertensive rat. Does dietary nitrate mitigate hypertension after conversion to nitrite? J. Am. Coll. Nutr. 9, 500–502 (1990).

    CAS  PubMed  Google Scholar 

  13. Moncada, S., Palmer, R. M. J. & Higgs, E. A. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol. Rev. 43, 109–141 (1991).

    CAS  PubMed  Google Scholar 

  14. Forstermann, U. & Sessa, W. C. Nitric oxide synthases: regulation and function. Eur. Heart J. 33, 829–837 (2012).

    PubMed  Google Scholar 

  15. Dimmeler, S. et al. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature 399, 601–605 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Fulton, D. et al. Regulation of endothelium-derived nitric oxide production by the protein kinase Akt. Nature 399, 597–601 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Tayeh, M. A. & Marletta, M. A. Macrophage oxidation of L-arginine to nitric oxide, nitrite, and nitrate. Tetrahydrobiopterin is required as a cofactor. J. Biol. Chem. 264, 19654–19658 (1989).

    CAS  PubMed  Google Scholar 

  18. Landmesser, U. et al. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J. Clin. Invest. 111, 1201–1209 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Szabo, C., Ischiropoulos, H. & Radi, R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat. Rev. Drug Discov. 6, 662–680 (2007).

    CAS  PubMed  Google Scholar 

  20. Nathan, C. Inducible nitric oxide synthase: what difference does it make? J. Clin. Invest. 100, 2417–2423 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Lundberg, J. O. et al. High nitric oxide production in human paranasal sinuses. Nat. Med. 1, 370–373 (1995).

    CAS  PubMed  Google Scholar 

  22. Lowry, J. L., Brovkovych, V., Zhang, Y. & Skidgel, R. A. Endothelial nitric-oxide synthase activation generates an inducible nitric-oxide synthase-like output of nitric oxide in inflamed endothelium. J. Biol. Chem. 288, 4174–4193 (2013).

    CAS  PubMed  Google Scholar 

  23. Lundberg, J. O., Weitzberg, E. & Gladwin, M. T. The nitrate–nitrite–nitric oxide pathway in physiology and therapeutics. Nat. Rev. Drug Discov. 7, 156–167 (2008).

    CAS  PubMed  Google Scholar 

  24. Lundberg, J. O. et al. Nitrate and nitrite in biology, nutrition and therapeutics. Nat. Chem. Biol. 5, 865–869 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. van Faassen, E. E. et al. Nitrite as regulator of hypoxic signaling in mammalian physiology. Med. Res. Rev. 29, 683–741 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Kim-Shapiro, D. B. & Gladwin, M. T. Pitfalls in measuring NO bioavailability using NOx. Nitric Oxide 44, 1–2 (2014).

    PubMed  PubMed Central  Google Scholar 

  27. Weitzberg, E. & Lundberg, J. O. Novel aspects of dietary nitrate and human health. Annu. Rev. Nutr. 33, 129–159 (2013).

    CAS  PubMed  Google Scholar 

  28. Lundberg, J. O., Weitzberg, E., Cole, J. A. & Benjamin, N. Nitrate, bacteria and human health. Nat. Rev. Microbiol. 2, 593–602 (2004).

    CAS  PubMed  Google Scholar 

  29. Jansson, E. A. et al. A mammalian functional nitrate reductase that regulates nitrite and nitric oxide homeostasis. Nat. Chem. Biol. 4, 411–417 (2008).

    CAS  PubMed  Google Scholar 

  30. Spiegelhalder, B., Eisenbrand, G. & Preussman, R. Influence of dietary nitrate on nitrite content of human saliva: possible relevance to in vivo formation of N-nitroso compounds. Food Cosmet. Toxicol. 14, 545–548 (1976).

    CAS  PubMed  Google Scholar 

  31. Lundberg, J. O. Nitrate transport in salivary glands with implications for NO homeostasis. Proc. Natl Acad. Sci. USA 109, 13144–13145 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Qin, L. et al. Sialin (SLC17A5) functions as a nitrate transporter in the plasma membrane. Proc. Natl Acad. Sci. USA 109, 13434–13439 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Lundberg, J. O. & Govoni, M. Inorganic nitrate is a possible source for systemic generation of nitric oxide. Free Radic. Biol. Med. 37, 395–400 (2004).

    CAS  PubMed  Google Scholar 

  34. Carlstrom, M. et al. Cross-talk between nitrate–nitrite–NO and NO synthase pathways in control of vascular NO homeostasis. Antioxid. Redox Signal. http://dx.doi.org/10.1089/ars.2013.5481 (2014).

  35. Larsen, F. J., Ekblom, B., Sahlin, K., Lundberg, J. O. & Weitzberg, E. Effects of dietary nitrate on blood pressure in healthy volunteers. N. Engl. J. Med. 355, 2792–2793 (2006). This study demonstrates the blood pressure-lowering effects of inorganic nitrate at doses achievable through the diet.

    CAS  PubMed  Google Scholar 

  36. Omar, S. A. et al. Paradoxical normoxia-dependent selective actions of inorganic nitrite in human muscular conduit arteries and related selective actions on central blood pressures. Circulation 131, 381–389 (2015).

    CAS  PubMed  Google Scholar 

  37. Beckman, J. S., Beckman, T. W., Chen, J., Marshall, P. & Freeman, B. A. Apparent hydroxyl radical production by peroxinitrite: implications for endothelial cell injury from nitric oxide. Proc. Natl Acad. Sci. USA 87, 1620–1624 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Gow, A. J., Farkouh, C. R., Munson, D. A., Posencheg, M. A. & Ischiropoulos, H. Biological significance of nitric oxide-mediated protein modifications. Am. J. Physiol. Lung Cell. Mol. Physiol. 287, L262–L268 (2004).

    CAS  PubMed  Google Scholar 

  39. Hill, B. G., Dranka, B. P., Bailey, S. M., Lancaster, J. R. Jr & Darley-Usmar, V. M. What part of NO don't you understand? Some answers to the cardinal questions in nitric oxide biology. J. Biol. Chem. 285, 19699–19704 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu, X. et al. Diffusion-limited reaction of free nitric oxide with erythrocytes. J. Biol. Chem. 273, 18709–18713 (1998).

    CAS  PubMed  Google Scholar 

  41. Hobbs, A. J. Soluble guanylate cyclase: the forgotten sibling. Trends Pharmacol. Sci. 18, 484–491 (1997).

    CAS  PubMed  Google Scholar 

  42. Kojda, G. & Kottenberg, K. Regulation of basal myocardial function by NO. Cardiovasc. Res. 41, 514–523 (1999).

    CAS  PubMed  Google Scholar 

  43. Castro, L. R., Schittl, J. & Fischmeister, R. Feedback control through cGMP-dependent protein kinase contributes to differential regulation and compartmentation of cGMP in rat cardiac myocytes. Circ. Res. 107, 1232–1240 (2010).

    CAS  PubMed  Google Scholar 

  44. Gladwin, M. T. et al. Relative role of heme nitrosylation and β-cysteine 93 nitrosation in the transport and metabolism of nitric oxide by hemoglobin in the human circulation. Proc. Natl Acad. Sci. USA 97, 9943–9948 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Eich, R. F. et al. Mechanism of NO-induced oxidation of myoglobin and hemoglobin. Biochemistry 35, 6976–6983 (1996).

    CAS  PubMed  Google Scholar 

  46. Cleeter, M. W., Cooper, J. M., Darley-Usmar, V. M., Moncada, S. & Schapira, A. H. Reversible inhibition of cytochrome c oxidase, the terminal enzyme of the mitochondrial respiratory chain, by nitric oxide. Implications for neurodegenerative diseases. FEBS Lett. 345, 50–54 (1994).

    CAS  PubMed  Google Scholar 

  47. Brown, G. C. & Cooper, C. E. Nanomolar concentrations of nitric oxide reversibly inhibit synaptosomal respiration by competing with oxygen at cytochrome oxidase. FEBS Lett. 356, 295–298 (1994).

    CAS  PubMed  Google Scholar 

  48. Boveris, A., Costa, L. E., Poderoso, J. J., Carreras, M. C. & Cadenas, E. Regulation of mitochondrial respiration by oxygen and nitric oxide. Ann. NY Acad. Sci. 899, 121–135 (2000).

    CAS  PubMed  Google Scholar 

  49. Hess, D. T., Matsumoto, A., Nudelman, R. & Stamler, J. S. S-nitrosylation: spectrum and specificity. Nat. Cell Biol. 3, E46–E49 (2001).

    CAS  PubMed  Google Scholar 

  50. Turko, I. V. & Murad, F. Protein nitration in cardiovascular diseases. Pharmacol. Rev. 54, 619–634 (2002).

    CAS  PubMed  Google Scholar 

  51. Stamler, J. S., Lamas, S. & Fang, F. C. Nitrosylation: the prototypic redox-based signaling mechanism. Cell 106, 675–683 (2001).

    CAS  PubMed  Google Scholar 

  52. Baker, P. R. et al. Fatty acid transduction of nitric oxide signaling: multiple nitrated unsaturated fatty acid derivatives exist in human blood and urine and serve as endogenous peroxisome proliferator-activated receptor ligands. J. Biol. Chem. 280, 42464–42475 (2005).

    CAS  PubMed  Google Scholar 

  53. Sawa, T. et al. Protein S-guanylation by the biological signal 8-nitroguanosine 3′,5′-cyclic monophosphate. Nat. Chem. Biol. 3, 727–735 (2007).

    CAS  PubMed  Google Scholar 

  54. Ito, C. et al. Endogenous nitrated nucleotide is a key mediator of autophagy and innate defense against bacteria. Mol. Cell 52, 794–804 (2013).

    CAS  PubMed  Google Scholar 

  55. Fang, F. C. Mechanisms of nitric oxide-related antimicrobial activity. J. Clin. Invest. 99, 2818–2825 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Schechter, A. N. & Gladwin, M. T. Hemoglobin and the paracrine and endocrine functions of nitric oxide. N. Engl. J. Med. 348, 1483–1485 (2003).

    CAS  PubMed  Google Scholar 

  57. Azarov, I. et al. Nitric oxide scavenging by red blood cells as a function of hematocrit and oxygenation. J. Biol. Chem. 280, 39024–39032 (2005).

    CAS  PubMed  Google Scholar 

  58. Straub, A. C. et al. Endothelial cell expression of haemoglobin α regulates nitric oxide signalling. Nature 491, 473–477 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Gladwin, M. T. & Kim-Shapiro, D. B. Vascular biology: nitric oxide caught in traffic. Nature 491, 344–345 (2012).

    CAS  PubMed  Google Scholar 

  60. Fox-Robichaud, A. et al. Inhaled NO as a viable antiadhesive therapy for ischemia/reperfusion injury of distal microvascular beds. J. Clin. Invest. 101, 2497–2505 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Cannon, R. O. et al. Effects of inhaled nitric oxide on regional blood flow are consistent with intravascular nitric oxide delivery. J. Clin. Invest. 108, 279–287 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Jia, L., Bonaventura, C., Bonaventura, J. & Stamler, J. S. S-nitrosohaemoglobin: a dynamic activity of blood involved in vascular control. Nature 380, 221–226 (1996).

    CAS  PubMed  Google Scholar 

  63. Chen, C. A. et al. S-glutathionylation uncouples eNOS and regulates its cellular and vascular function. Nature 468, 1115–1118 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Wu, F. et al. Nox2-dependent glutathionylation of endothelial NOS leads to uncoupled superoxide production and endothelial barrier dysfunction in acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 307, L987–L997 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Wever, R. M., van Dam, T., van Rijn, H. J., de Groot, F. & Rabelink, T. J. Tetrahydrobiopterin regulates superoxide and nitric oxide generation by recombinant endothelial nitric oxide synthase. Biochem. Biophys. Res. Commun. 237, 340–344 (1997).

    CAS  PubMed  Google Scholar 

  66. Yang, Z. & Ming, X. F. Arginase: the emerging therapeutic target for vascular oxidative stress and inflammation. Front. Immunol. 4, 149 (2013).

    PubMed  PubMed Central  Google Scholar 

  67. Stasch, J. P., Pacher, P. & Evgenov, O. V. Soluble guanylate cyclase as an emerging therapeutic target in cardiopulmonary disease. Circulation 123, 2263–2273 (2011).

    PubMed  PubMed Central  Google Scholar 

  68. Potoka, K. P. & Gladwin, M. T. Vasculopathy and pulmonary hypertension in sickle cell disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 308, L314–L324 (2015).

    CAS  PubMed  Google Scholar 

  69. Ignarro, L. J. Nitric oxide as a unique signaling molecule in the vascular system: a historical overview. J. Physiol. Pharmacol. 53, 503–514 (2002).

    CAS  PubMed  Google Scholar 

  70. Rees, D. D., Palmer, R. M. & Moncada, S. Role of endothelium-derived nitric oxide in the regulation of blood pressure. Proc. Natl Acad. Sci. USA 86, 3375–3378 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Huang, P. L. et al. Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 377, 239–242 (1995). This study demonstrates the pivotal role of eNOS in the regulation of basal vascular tone and blood pressure.

    CAS  PubMed  Google Scholar 

  72. Moncada, S. & Higgs, A. The L-arginine–nitric oxide pathway. N. Engl. J. Med. 329, 2002–2012 (1993).

    CAS  PubMed  Google Scholar 

  73. Balligand, J. L., Kelly, R. A., Marsden, P. A., Smith, T. W. & Michel, T. Control of cardiac muscle cell function by an endogenous nitric oxide signaling system. Proc. Natl Acad. Sci. USA 90, 347–351 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Jumrussirikul, P. et al. Interaction between neuronal nitric oxide synthase and inhibitory G protein activity in heart rate regulation in conscious mice. J. Clin. Invest. 102, 1279–1285 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Takano, H. et al. Nitric oxide synthase is the mediator of late preconditioning against myocardial infarction in conscious rabbits. Circulation 98, 441–449 (1998).

    CAS  PubMed  Google Scholar 

  76. Cohen, M. V. & Downey, J. M. Ischemic postconditioning: from receptor to end-effector. Antioxid. Redox Signal. 14, 821–831 (2011).

    CAS  PubMed  Google Scholar 

  77. Seddon, M., Shah, A. M. & Casadei, B. Cardiomyocytes as effectors of nitric oxide signalling. Cardiovasc. Res. 75, 315–326 (2007).

    CAS  PubMed  Google Scholar 

  78. Tang, L., Wang, H. & Ziolo, M. T. Targeting NOS as a therapeutic approach for heart failure. Pharmacol. Ther. 142, 306–315 (2014).

    CAS  PubMed  Google Scholar 

  79. Xu, L., Eu, J. P., Meissner, G. & Stamler, J. S. Activation of the cardiac calcium release channel (ryanodine receptor) by poly-S-nitrosylation. Science 279, 234–237 (1998).

    CAS  PubMed  Google Scholar 

  80. Jones, S. P. & Bolli, R. The ubiquitous role of nitric oxide in cardioprotection. J. Mol. Cell. Cardiol. 40, 16–23 (2006).

    CAS  PubMed  Google Scholar 

  81. West, M. B. et al. Cardiac myocyte-specific expression of inducible nitric oxide synthase protects against ischemia/reperfusion injury by preventing mitochondrial permeability transition. Circulation 118, 1970–1978 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Sasaki, N., Sato, T., Ohler, A., O'Rourke, B. & Marban, E. Activation of mitochondrial ATP-dependent potassium channels by nitric oxide. Circulation 101, 439–445 (2000).

    CAS  PubMed  Google Scholar 

  83. Ertracht, O., Malka, A., Atar, S. & Binah, O. The mitochondria as a target for cardioprotection in acute myocardial ischemia. Pharmacol. Ther. 142, 33–40 (2014).

    CAS  PubMed  Google Scholar 

  84. Chouchani, E. T. et al. Cardioprotection by S-nitrosation of a cysteine switch on mitochondrial complex I. Nat. Med. 19, 753–759 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Shiva, S. et al. Nitrite augments tolerance to ischemia/reperfusion injury via the modulation of mitochondrial electron transfer. J. Exp. Med. 204, 2089–2102 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Rassaf, T. et al. Circulating nitrite contributes to cardioprotection by remote ischemic preconditioning. Circ. Res. 114, 1601–1610 (2014).

    CAS  PubMed  Google Scholar 

  87. Hataishi, R. et al. Inhaled nitric oxide decreases infarction size and improves left ventricular function in a murine model of myocardial ischemia–reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 291, H379–H384 (2006).

    CAS  PubMed  Google Scholar 

  88. Lang, J. D. Jr. et al. Inhaled NO accelerates restoration of liver function in adults following orthotopic liver transplantation. J. Clin. Invest. 117, 2583–2591 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Lang, J. D. Jr. et al. A randomized clinical trial testing the anti-inflammatory effects of preemptive inhaled nitric oxide in human liver transplantation. PLoS ONE 9, e86053 (2014).

    PubMed  PubMed Central  Google Scholar 

  90. Wallace, J. L., Ignarro, L. J. & Fiorucci, S. Potential cardioprotective actions of no-releasing aspirin. Nat. Rev. Drug Discov. 1, 375–382 (2002).

    CAS  PubMed  Google Scholar 

  91. Zacharowski, P. et al. The effects and metabolic fate of nitroflurbiprofen in healthy volunteers. Clin. Pharmacol. Ther. 76, 350–358 (2004).

    CAS  PubMed  Google Scholar 

  92. Cavet, M. E., Vittitow, J. L., Impagnatiello, F., Ongini, E. & Bastia, E. Nitric oxide (NO): an emerging target for the treatment of glaucoma. Invest. Ophthalmol. Vis. Sci. 55, 5005–5015 (2014).

    CAS  PubMed  Google Scholar 

  93. Miller, M. R. & Megson, I. L. Recent developments in nitric oxide donor drugs. Br. J. Pharmacol. 151, 305–321 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Tran, H. & Anand, S. S. Oral antiplatelet therapy in cerebrovascular disease, coronary artery disease, and peripheral arterial disease. JAMA 292, 1867–1874 (2004).

    CAS  PubMed  Google Scholar 

  95. Anderson, R. A., Bundhoo, S. & James, P. E. A new mechanism of action of thienopyridine antiplatelet drugs — a role for gastric nitrosthiol metabolism? Atherosclerosis 237, 369–373 (2014).

    CAS  PubMed  Google Scholar 

  96. Gilard, M. et al. Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole CLopidogrel Aspirin) study. J. Am. Coll. Cardiol. 51, 256–260 (2008).

    CAS  PubMed  Google Scholar 

  97. Bundhoo, S. S. et al. Direct vasoactive properties of thienopyridine-derived nitrosothiols. J. Cardiovasc. Pharmacol. 58, 550–558 (2011).

    CAS  PubMed  Google Scholar 

  98. Cosby, K. et al. Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat. Med. 9, 1498–1505 (2003). This study suggests that deoxygenated haemoglobin catalyses the reduction of nitrite to NO in the human circulation to regulate vascular tone.

    CAS  PubMed  Google Scholar 

  99. Liu, C. et al. Mechanisms of human erythrocytic bioactivation of nitrite. J. Biol. Chem. 290, 1281–1294 (2015).

    CAS  PubMed  Google Scholar 

  100. Hunter, C. J. et al. Inhaled nebulized nitrite is a hypoxia-sensitive NO-dependent selective pulmonary vasodilator. Nat. Med. 10, 1122–1127 (2004).

    CAS  PubMed  Google Scholar 

  101. Webb, A. et al. Reduction of nitrite to nitric oxide during ischemia protects against myocardial ischemia–reperfusion damage. Proc. Natl Acad. Sci. USA 101, 13683–13688 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Duranski, M. R. et al. Cytoprotective effects of nitrite during in vivo ischemia–reperfusion of the heart and liver. J. Clin. Invest. 115, 1232–1240 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Hendgen-Cotta, U. B. et al. Nitrite reductase activity of myoglobin regulates respiration and cellular viability in myocardial ischemia-reperfusion injury. Proc. Natl Acad. Sci. USA 105, 10256–10261 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Rix, P. J. et al. Pharmacokinetics, pharmacodynamics, safety, and tolerability of nebulized sodium nitrite (AIR001) following repeat-dose inhalation in healthy subjects. Clin. Pharmacokinet. 54, 261–272 (2015).

    CAS  PubMed  Google Scholar 

  105. Dejam, A. et al. Nitrite infusion in humans and nonhuman primates: endocrine effects, pharmacokinetics, and tolerance formation. Circulation 116, 1821–1831 (2007).

    CAS  PubMed  Google Scholar 

  106. Mohler, E. R. et al. Sodium nitrite in patients with peripheral artery disease and diabetes mellitus: safety, walking distance and endothelial function. Vasc. Med. 19, 9–17 (2014).

    CAS  PubMed  Google Scholar 

  107. Hunault, C. C., van Velzen, A. G., Sips, A. J., Schothorst, R. C. & Meulenbelt, J. Bioavailability of sodium nitrite from an aqueous solution in healthy adults. Toxicol. Lett. 190, 48–53 (2009).

    CAS  PubMed  Google Scholar 

  108. Pluta, R. M. et al. Safety and feasibility of long-term intravenous sodium nitrite infusion in healthy volunteers. PLoS ONE 6, e14504 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Oldfield, E. H. et al. Safety and pharmacokinetics of sodium nitrite in patients with subarachnoid hemorrhage: a Phase IIA study. J. Neurosurg. 119, 634–641 (2013).

    CAS  PubMed  Google Scholar 

  110. Siddiqi, N. et al. Intravenous sodium nitrite in acute ST-elevation myocardial infarction: a randomized controlled trial (NIAMI). Eur. Heart J. 35, 1255–1262 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Jones, D. A. et al. Randomized Phase 2 trial of intra-coronary nitrite during acute myocardial infarction. Circ. Res. 116, 437–447 (2015).

    CAS  PubMed  Google Scholar 

  112. Webb, A. J. et al. Acute blood pressure lowering, vasoprotective, and antiplatelet properties of dietary nitrate via bioconversion to nitrite. Hypertension 51, 784–790 (2008).

    CAS  PubMed  Google Scholar 

  113. Omar, S. A., Artime, E. & Webb, A. J. A comparison of organic and inorganic nitrates/nitrites. Nitric Oxide 26, 229–240 (2012).

    CAS  PubMed  Google Scholar 

  114. Carlstrom, M. et al. Dietary inorganic nitrate reverses features of metabolic syndrome in endothelial nitric oxide synthase-deficient mice. Proc. Natl Acad. Sci. USA 107, 17716–17720 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Carlstrom, M. et al. Dietary nitrate attenuates oxidative stress, prevents cardiac and renal injuries, and reduces blood pressure in salt-induced hypertension. Cardiovasc. Res. 89, 574–585 (2011).

    PubMed  Google Scholar 

  116. Jadert, C. et al. Decreased leukocyte recruitment by inorganic nitrate and nitrite in microvascular inflammation and NSAID-induced intestinal injury. Free Radic. Biol. Med. 52, 683–692 (2012).

    PubMed  Google Scholar 

  117. Bryan, N. S. et al. Dietary nitrite supplementation protects against myocardial ischemia-reperfusion injury. Proc. Natl Acad. Sci. USA 104, 19144–19149 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Hendgen-Cotta, U. B. et al. Dietary nitrate supplementation improves revascularization in chronic ischemia. Circulation 126, 1983–1992 (2012).

    CAS  PubMed  Google Scholar 

  119. Govoni, M., Jansson, E. A., Weitzberg, E. & Lundberg, J. O. The increase in plasma nitrite after a dietary nitrate load is markedly attenuated by an antibacterial mouthwash. Nitric Oxide 19, 333–337 (2008).

    CAS  PubMed  Google Scholar 

  120. Petersson, J. et al. Gastroprotective and blood pressure lowering effects of dietary nitrate are abolished by an antiseptic mouthwash. Free Radic. Biol. Med. 46, 1068–1075 (2009).

    CAS  PubMed  Google Scholar 

  121. Gao, X. et al. NADPH oxidase in the renal microvasculature is a primary target for blood pressure-lowering effects by inorganic nitrate and nitrite. Hypertension 65, 161–170 (2015).

    CAS  PubMed  Google Scholar 

  122. Lundberg, J. O., Weitzberg, E., Lundberg, J. M. & Alving, K. Intragastric nitric oxide production in humans: measurements in expelled air. Gut 35, 1543–1546 (1994). The study described in this paper demonstrates NOS-independent NO generation from inorganic nitrate and nitrite in humans.

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Pinheiro, L. C. et al. Increase in gastric pH reduces hypotensive effect of oral sodium nitrite in rats. Free Radic. Biol. Med. 53, 701–709 (2012).

    CAS  PubMed  Google Scholar 

  124. Ghebremariam, Y. T. et al. Unexpected effect of proton pump inhibitors: elevation of the cardiovascular risk factor asymmetric dimethylarginine. Circulation 128, 845–853 (2013).

    CAS  PubMed  Google Scholar 

  125. Montenegro, M. F. & Lundberg, J. O. Letter by Montenegro and Lundberg regarding article, “Unexpected effect of proton pump inhibitors: elevation of the cardiovascular risk factor asymmetric dimethylarginine”. Circulation 129, E426–E426 (2014).

    PubMed  Google Scholar 

  126. Kapil, V. et al. Physiological role for nitrate-reducing oral bacteria in blood pressure control. Free Radic. Biol. Med. 55, 93–100 (2013). This study demonstrates physiological effects of endogenous nitrate in the modulation of cardiovascular function.

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Bondonno, C. P. et al. Antibacterial mouthwash blunts oral nitrate reduction and increases blood pressure in treated hypertensive men and women. Am. J. Hypertens. 28, 572–575 (2015).

    CAS  PubMed  Google Scholar 

  128. O'Donnell, V. B. et al. Nitration of unsaturated fatty acids by nitric oxide-derived reactive nitrogen species peroxynitrite, nitrous acid, nitrogen dioxide, and nitronium ion. Chem. Res. Toxicol. 12, 83–92 (1999).

    CAS  PubMed  Google Scholar 

  129. Trostchansky, A., Bonilla, L., Gonzalez-Perilli, L. & Rubbo, H. Nitro-fatty acids: formation, redox signaling, and therapeutic potential. Antioxid. Redox Signal. 19, 1257–1265 (2013).

    CAS  PubMed  Google Scholar 

  130. Delmastro-Greenwood, M., Freeman, B. A. & Wendell, S. G. Redox-dependent anti-inflammatory signaling actions of unsaturated fatty acids. Annu. Rev. Physiol. 76, 79–105 (2014).

    CAS  PubMed  Google Scholar 

  131. Nadtochiy, S. M. et al. Nitroalkenes confer acute cardioprotection via adenine nucleotide translocase 1. J. Biol. Chem. 287, 3573–3580 (2012).

    CAS  PubMed  Google Scholar 

  132. Cole, M. P. et al. Nitro-fatty acid inhibition of neointima formation after endoluminal vessel injury. Circ. Res. 105, 965–972 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Rudolph, V. et al. Endogenous generation and protective effects of nitro-fatty acids in a murine model of focal cardiac ischaemia and reperfusion. Cardiovasc. Res. 85, 155–166 (2010).

    CAS  PubMed  Google Scholar 

  134. Lim, D. G. et al. Nitrolinoleate, a nitric oxide-derived mediator of cell function: synthesis, characterization, and vasomotor activity. Proc. Natl Acad. Sci. USA 99, 15941–15946 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Rocha, B. S. et al. Intragastric nitration by dietary nitrite: implications for modulation of protein and lipid signaling. Free Radic. Biol. Med. 52, 693–698 (2012).

    CAS  PubMed  Google Scholar 

  136. Rocha, B. S. et al. Pepsin is nitrated in the rat stomach, acquiring antiulcerogenic activity: a novel interaction between dietary nitrate and gut proteins. Free Radic. Biol. Med. 58, 26–34 (2013).

    CAS  PubMed  Google Scholar 

  137. Charles, R. L. et al. Protection from hypertension in mice by the Mediterranean diet is mediated by nitro fatty acid inhibition of soluble epoxide hydrolase. Proc. Natl Acad. Sci. USA 111, 8167–8172 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Wu, G. & Morris, S. M. Jr. Arginine metabolism: nitric oxide and beyond. Biochem. J. 336, 1–17 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Schulman, S. P. et al. L-arginine therapy in acute myocardial infarction: the Vascular Interaction with Age in Myocardial Infarction (VINTAGE MI) randomized clinical trial. JAMA 295, 58–64 (2006).

    CAS  PubMed  Google Scholar 

  140. Wilson, A. M., Harada, R., Nair, N., Balasubramanian, N. & Cooke, J. P. L-arginine supplementation in peripheral arterial disease: no benefit and possible harm. Circulation 116, 188–195 (2007).

    CAS  PubMed  Google Scholar 

  141. Dong, J. Y. et al. Effect of oral L-arginine supplementation on blood pressure: a meta-analysis of randomized, double-blind, placebo-controlled trials. Am. Heart J. 162, 959–965 (2011).

    CAS  PubMed  Google Scholar 

  142. Pernow, J. & Jung, C. Arginase as a potential target in the treatment of cardiovascular disease: reversal of arginine steal? Cardiovasc. Res. 98, 334–343 (2013).

    CAS  PubMed  Google Scholar 

  143. Boger, R. H. The pharmacodynamics of L-arginine. J. Nutr. 137, 1650S–1655S (2007).

    PubMed  Google Scholar 

  144. Solomonson, L. P., Flam, B. R., Pendleton, L. C., Goodwin, B. L. & Eichler, D. C. The caveolar nitric oxide synthase/arginine regeneration system for NO production in endothelial cells. J. Exp. Biol. 206, 2083–2087 (2003).

    CAS  PubMed  Google Scholar 

  145. Schwedhelm, E. et al. Pharmacokinetic and pharmacodynamic properties of oral L-citrulline and L-arginine: impact on nitric oxide metabolism. Br. J. Clin. Pharmacol. 65, 51–59 (2008).

    CAS  PubMed  Google Scholar 

  146. Li, H., Horke, S. & Forstermann, U. Vascular oxidative stress, nitric oxide and atherosclerosis. Atherosclerosis 237, 208–219 (2014).

    CAS  PubMed  Google Scholar 

  147. Li, H. & Forstermann, U. Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease. Curr. Opin. Pharmacol. 13, 161–167 (2013).

    PubMed  Google Scholar 

  148. Drummond, G. R., Selemidis, S., Griendling, K. K. & Sobey, C. G. Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat. Rev. Drug Discov. 10, 453–471 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Cosentino, F. et al. Chronic treatment with tetrahydrobiopterin reverses endothelial dysfunction and oxidative stress in hypercholesterolaemia. Heart 94, 487–492 (2008).

    CAS  PubMed  Google Scholar 

  150. Cunnington, C. et al. Systemic and vascular oxidation limits the efficacy of oral tetrahydrobiopterin treatment in patients with coronary artery disease. Circulation 125, 1356–1366 (2012). This study demonstrates important clinical limitations of oral BH4 treatment.

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Morris, C. R. et al. Dysregulated arginine metabolism, hemolysis-associated pulmonary hypertension, and mortality in sickle cell disease. JAMA 294, 81–90 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Xu, W. et al. Increased arginase II and decreased NO synthesis in endothelial cells of patients with pulmonary arterial hypertension. FASEB J. 18, 1746–1748 (2004).

    CAS  PubMed  Google Scholar 

  153. Tang, W. H., Wang, Z., Cho, L., Brennan, D. M. & Hazen, S. L. Diminished global arginine bioavailability and increased arginine catabolism as metabolic profile of increased cardiovascular risk. J. Am. Coll. Cardiol. 53, 2061–2067 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Tang, W. H. et al. Diminished global arginine bioavailability as a metabolic defect in chronic systolic heart failure. J. Card. Fail. 19, 87–93 (2013).

    PubMed  Google Scholar 

  155. Jung, C., Gonon, A. T., Sjoquist, P. O., Lundberg, J. O. & Pernow, J. Arginase inhibition mediates cardioprotection during ischaemia–reperfusion. Cardiovasc. Res. 85, 147–154 (2010). This study demonstrates the therapeutic effects of arginase inhibition in experimental myocardial infarction.

    CAS  PubMed  Google Scholar 

  156. Gonon, A. T. et al. Local arginase inhibition during early reperfusion mediates cardioprotection via increased nitric oxide production. PLoS ONE 7, e42038 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Gronros, J. et al. Arginase inhibition restores in vivo coronary microvascular function in type 2 diabetic rats. Am. J. Physiol. Heart Circ. Physiol. 300, H1174–H1181 (2011).

    PubMed  Google Scholar 

  158. Tratsiakovich, Y. et al. Arginase inhibition reduces infarct size via nitric oxide, protein kinase C epsilon and mitochondrial ATP-dependent K+ channels. Eur. J. Pharmacol. 712, 16–21 (2013).

    CAS  PubMed  Google Scholar 

  159. Kleinbongard, P. et al. Red blood cells express a functional endothelial nitric oxide synthase. Blood 107, 2943–2951 (2006).

    CAS  PubMed  Google Scholar 

  160. Yang, J., Gonon, A. T., Sjoquist, P. O., Lundberg, J. O. & Pernow, J. Arginase regulates red blood cell nitric oxide synthase and export of cardioprotective nitric oxide bioactivity. Proc. Natl Acad. Sci. USA 110, 15049–15054 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Shemyakin, A. et al. Arginase inhibition improves endothelial function in patients with coronary artery disease and type 2 diabetes mellitus. Circulation 126, 2943–2950 (2012).

    CAS  PubMed  Google Scholar 

  162. Ashmore, T. et al. Dietary nitrate increases arginine availability and protects mitochondrial complex I and energetics in the hypoxic rat heart. J. Physiol. 592, 4715–4731 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Witte, M. B. & Barbul, A. Arginine physiology and its implication for wound healing. Wound Repair Regen. 11, 419–423 (2003).

    PubMed  Google Scholar 

  164. Vallance, P. & Leiper, J. Cardiovascular biology of the asymmetric dimethylarginine:dimethylarginine dimethylaminohydrolase pathway. Arterioscler. Thromb. Vasc. Biol. 24, 1023–1030 (2004).

    CAS  PubMed  Google Scholar 

  165. Leiper, J. & Nandi, M. The therapeutic potential of targeting endogenous inhibitors of nitric oxide synthesis. Nat. Rev. Drug Discov. 10, 277–291 (2011).

    CAS  PubMed  Google Scholar 

  166. Ridker, P. M. LDL cholesterol: controversies and future therapeutic directions. Lancet 384, 607–617 (2014).

    CAS  PubMed  Google Scholar 

  167. Endo, A. The origin of the statins. Atheroscler. Suppl. 5, 125–130 (2004).

    CAS  PubMed  Google Scholar 

  168. Davignon, J. Beneficial cardiovascular pleiotropic effects of statins. Circulation 109, III39–III43 (2004).

    PubMed  Google Scholar 

  169. Laufs, U. & Liao, J. K. Post-transcriptional regulation of endothelial nitric oxide synthase mRNA stability by Rho GTPase. J. Biol. Chem. 273, 24266–24271 (1998).

    CAS  PubMed  Google Scholar 

  170. Kureishi, Y. et al. The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic animals. Nat. Med. 6, 1004–1010 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Kosmidou, I., Moore, J. P., Weber, M. & Searles, C. D. Statin treatment and 3′ polyadenylation of eNOS mRNA. Arterioscler. Thromb. Vasc. Biol. 27, 2642–2649 (2007).

    CAS  PubMed  Google Scholar 

  172. Feron, O., Dessy, C., Desager, J. P. & Balligand, J. L. Hydroxy-methylglutaryl-coenzyme A reductase inhibition promotes endothelial nitric oxide synthase activation through a decrease in caveolin abundance. Circulation 103, 113–118 (2001).

    CAS  PubMed  Google Scholar 

  173. Antoniades, C. et al. Rapid, direct effects of statin treatment on arterial redox state and nitric oxide bioavailability in human atherosclerosis via tetrahydrobiopterin-mediated endothelial nitric oxide synthase coupling. Circulation 124, 335–345 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Cai, H., Griendling, K. K. & Harrison, D. G. The vascular NAD(P)H oxidases as therapeutic targets in cardiovascular diseases. Trends Pharmacol. Sci. 24, 471–478 (2003).

    CAS  PubMed  Google Scholar 

  175. Ivashchenko, C. Y. et al. Regulation of the ADMA–DDAH system in endothelial cells: a novel mechanism for the sterol response element binding proteins, SREBP1c and -2. Am. J. Physiol. Heart Circ. Physiol. 298, H251–H258 (2010).

    CAS  PubMed  Google Scholar 

  176. Behrendt, D. & Ganz, P. Endothelial function: from vascular biology to clinical applications. Am. J. Cardiol. 90, 40L–48L (2002).

    CAS  PubMed  Google Scholar 

  177. Nussinovitch, U., de Carvalho, J. F., Pereira, R. M. & Shoenfeld, Y. Glucocorticoids and the cardiovascular system: state of the art. Curr. Pharm. Des. 16, 3574–3585 (2010).

    CAS  PubMed  Google Scholar 

  178. Hafezi-Moghadam, A. et al. Acute cardiovascular protective effects of corticosteroids are mediated by non-transcriptional activation of endothelial nitric oxide synthase. Nat. Med. 8, 473–479 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Perretti, M. & D'Acquisto, F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat. Rev. Immunol. 9, 62–70 (2009).

    CAS  PubMed  Google Scholar 

  180. Conti, V. et al. Adrenoreceptors and nitric oxide in the cardiovascular system. Front. Physiol. 4, 321 (2013).

    PubMed  PubMed Central  Google Scholar 

  181. Dessy, C. et al. Endothelial β3-adrenoreceptors mediate nitric oxide-dependent vasorelaxation of coronary microvessels in response to the third-generation β-blocker nebivolol. Circulation 112, 1198–1205 (2005).

    CAS  PubMed  Google Scholar 

  182. Okamoto, L. E. et al. Nebivolol, but not metoprolol, lowers blood pressure in nitric oxide-sensitive human hypertension. Hypertension 64, 1241–1247 (2014).

    CAS  PubMed  Google Scholar 

  183. Schiffrin, E. L., Park, J. B., Intengan, H. D. & Touyz, R. M. Correction of arterial structure and endothelial dysfunction in human essential hypertension by the angiotensin receptor antagonist losartan. Circulation 101, 1653–1659 (2000).

    CAS  PubMed  Google Scholar 

  184. Nguyen Dinh Cat, A., Montezano, A. C., Burger, D. & Touyz, R. M. Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid. Redox Signal. 19, 1110–1120 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Wenzel, P. et al. AT1-receptor blockade by telmisartan upregulates GTP-cyclohydrolase I and protects eNOS in diabetic rats. Free Radic. Biol. Med. 45, 619–626 (2008).

    CAS  PubMed  Google Scholar 

  186. Satoh, M. et al. Angiotensin II type 1 receptor blocker ameliorates uncoupled endothelial nitric oxide synthase in rats with experimental diabetic nephropathy. Nephrol. Dial. Transplant. 23, 3806–3813 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Oak, J. H. & Cai, H. Attenuation of angiotensin II signaling recouples eNOS and inhibits nonendothelial NOX activity in diabetic mice. Diabetes 56, 118–126 (2007).

    CAS  PubMed  Google Scholar 

  188. Knorr, M. et al. Nitroglycerin-induced endothelial dysfunction and tolerance involve adverse phosphorylation and S-glutathionylation of endothelial nitric oxide synthase: beneficial effects of therapy with the AT1 receptor blocker telmisartan. Arterioscler. Thromb. Vasc. Biol. 31, 2223–2231 (2011).

    CAS  PubMed  Google Scholar 

  189. Al Ghouleh, I. et al. Oxidases and peroxidases in cardiovascular and lung disease: new concepts in reactive oxygen species signaling. Free Radic. Biol. Med. 51, 1271–1288 (2011).

    CAS  PubMed  Google Scholar 

  190. Bedard, K. & Krause, K. H. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol. Rev. 87, 245–313 (2007).

    CAS  PubMed  Google Scholar 

  191. Gutteridge, J. M. & Halliwell, B. Antioxidants: molecules, medicines, and myths. Biochem. Biophys. Res. Commun. 393, 561–564 (2010).

    CAS  PubMed  Google Scholar 

  192. Ristow, M. et al. Antioxidants prevent health-promoting effects of physical exercise in humans. Proc. Natl Acad. Sci. USA 106, 8665–8670 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Altenhofer, S., Radermacher, K. A., Kleikers, P. W., Wingler, K. & Schmidt, H. H. Evolution of NADPH oxidase inhibitors: selectivity and mechanisms for target engagement. Antioxid. Redox Signal. http://dx.doi.org/10.1089/ars.2013.5814 (2014).

  194. Ranayhossaini, D. J. et al. Selective recapitulation of conserved and nonconserved regions of putative NOXA1 protein activation domain confers isoform-specific inhibition of Nox1 oxidase and attenuation of endothelial cell migration. J. Biol. Chem. 288, 36437–36450 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Cifuentes-Pagano, E., Meijles, D. N. & Pagano, P. J. The quest for selective nox inhibitors and therapeutics: challenges, triumphs and pitfalls. Antioxid. Redox Signal. 20, 2741–2754 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Bender, A. T. & Beavo, J. A. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol. Rev. 58, 488–520 (2006).

    CAS  PubMed  Google Scholar 

  197. Goldstein, I. et al. Oral sildenafil in the treatment of erectile dysfunction. N. Engl. J. Med. 338, 1397–1404 (1998).

    CAS  PubMed  Google Scholar 

  198. Michelakis, E. et al. Oral sildenafil is an effective and specific pulmonary vasodilator in patients with pulmonary arterial hypertension: comparison with inhaled nitric oxide. Circulation 105, 2398–2403 (2002).

    CAS  PubMed  Google Scholar 

  199. Salloum, F., Yin, C., Xi, L. & Kukreja, R. C. Sildenafil induces delayed preconditioning through inducible nitric oxide synthase-dependent pathway in mouse heart. Circ. Res. 92, 595–597 (2003).

    CAS  PubMed  Google Scholar 

  200. Thadani, U. et al. The effect of vardenafil, a potent and highly selective phosphodiesterase-5 inhibitor for the treatment of erectile dysfunction, on the cardiovascular response to exercise in patients with coronary artery disease. J. Am. Coll. Cardiol. 40, 2006–2012 (2002).

    CAS  PubMed  Google Scholar 

  201. Senthilkumar, A. et al. Sildenafil promotes ischemia-induced angiogenesis through a PKG-dependent pathway. Arterioscler. Thromb. Vasc. Biol. 27, 1947–1954 (2007).

    CAS  PubMed  Google Scholar 

  202. Fisher, P. W., Salloum, F., Das, A., Hyder, H. & Kukreja, R. C. Phosphodiesterase-5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation 111, 1601–1610 (2005).

    CAS  PubMed  Google Scholar 

  203. Roustit, M., Hellmann, M., Cracowski, C., Blaise, S. & Cracowski, J. L. Sildenafil increases digital skin blood flow during all phases of local cooling in primary Raynaud's phenomenon. Clin. Pharmacol. Ther. 91, 813–819 (2012).

    CAS  PubMed  Google Scholar 

  204. Lukowski, R., Krieg, T., Rybalkin, S. D., Beavo, J. & Hofmann, F. Turning on cGMP-dependent pathways to treat cardiac dysfunctions: boom, bust, and beyond. Trends Pharmacol. Sci. 35, 404–413 (2014).

    CAS  PubMed  Google Scholar 

  205. Ghofrani, H. A. et al. Riociguat for the treatment of pulmonary arterial hypertension. N. Engl. J. Med. 369, 330–340 (2013).

    CAS  PubMed  Google Scholar 

  206. Ghofrani, H. A. et al. Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N. Engl. J. Med. 369, 319–329 (2013). References 205 and 206 discuss studies demonstrating that an sGC stimulator improves exercise capacity and other clinical parameters in patients with pulmonary hypertension.

    CAS  PubMed  Google Scholar 

  207. Follmann, M. et al. The chemistry and biology of soluble guanylate cyclase stimulators and activators. Angew. Chem. Int. Ed Engl. 52, 9442–9462 (2013).

    CAS  PubMed  Google Scholar 

  208. Ko, F. N., Wu, C. C., Kuo, S. C., Lee, F. Y. & Teng, C. M. YC-1, a novel activator of platelet guanylate cyclase. Blood 84, 4226–4233 (1994).

    CAS  PubMed  Google Scholar 

  209. Baliga, R. S., MacAllister, R. J. & Hobbs, A. J. New perspectives for the treatment of pulmonary hypertension. Br. J. Pharmacol. 163, 125–140 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Boerrigter, G. et al. Cardiorenal and humoral properties of a novel direct soluble guanylate cyclase stimulator BAY 41–2272 in experimental congestive heart failure. Circulation 107, 686–689 (2003).

    CAS  PubMed  Google Scholar 

  211. Stasch, J. P., Dembowsky, K., Perzborn, E., Stahl, E. & Schramm, M. Cardiovascular actions of a novel NO-independent guanylyl cyclase stimulator, BAY 41-8543: in vivo studies. Br. J. Pharmacol. 135, 344–355 (2002). This study demonstrates the potential therapeutic cardiovascular effects of a NO-independent guanylyl cyclase stimulator.

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Ahluwalia, A. et al. Antiinflammatory activity of soluble guanylate cyclase: cGMP-dependent down-regulation of P-selectin expression and leukocyte recruitment. Proc. Natl Acad. Sci. USA 101, 1386–1391 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Raat, N. J. et al. Direct sGC activation bypasses NO scavenging reactions of intravascular free oxy-hemoglobin and limits vasoconstriction. Antioxid. Redox Signal. 19, 2232–2243 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Baliga, R. S., Macallister, R. J. & Hobbs, A. J. Vasoactive peptides and the pathogenesis of pulmonary hypertension: role and potential therapeutic application. Handb. Exp. Pharmacol. 218, 477–511 (2013).

    CAS  PubMed  Google Scholar 

  215. Baliga, R. S. et al. Synergy between natriuretic peptides and phosphodiesterase 5 inhibitors ameliorates pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 178, 861–869 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Scalbert, A., Johnson, I. T. & Saltmarsh, M. Polyphenols: antioxidants and beyond. Am J. Clin. Nutr. 81, 215S–217S (2005).

    CAS  PubMed  Google Scholar 

  217. Laurent, C. et al. Polyphenols decreased liver NADPH oxidase activity, increased muscle mitochondrial biogenesis and decreased gastrocnemius age-dependent autophagy in aged rats. Free Radic. Res. 46, 1140–1149 (2012).

    CAS  PubMed  Google Scholar 

  218. Schewe, T., Steffen, Y. & Sies, H. How do dietary flavanols improve vascular function? A position paper. Arch. Biochem. Biophys. 476, 102–106 (2008).

    CAS  PubMed  Google Scholar 

  219. Anter, E. et al. Activation of endothelial nitric-oxide synthase by the p38 MAPK in response to black tea polyphenols. J. Biol. Chem. 279, 46637–46643 (2004).

    CAS  PubMed  Google Scholar 

  220. Schroeter, H. et al. (−)-epicatechin mediates beneficial effects of flavanol-rich cocoa on vascular function in humans. Proc. Natl Acad. Sci. USA 103, 1024–1029 (2006). This study demonstrates NOS-dependent improvements in vascular function by flavanols in humans.

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Balzer, J. et al. Sustained benefits in vascular function through flavanol-containing cocoa in medicated diabetic patients: a double-masked, randomized, controlled trial. J. Am. Coll. Cardiol. 51, 2141–2149 (2008).

    CAS  PubMed  Google Scholar 

  222. Desideri, G. et al. Benefits in cognitive function, blood pressure, and insulin resistance through cocoa flavanol consumption in elderly subjects with mild cognitive impairment: the Cocoa, Cognition, and Aging (CoCoA) study. Hypertension 60, 794–801 (2012).

    CAS  PubMed  Google Scholar 

  223. Heiss, C., Keen, C. L. & Kelm, M. Flavanols and cardiovascular disease prevention. Eur. Heart J. 31, 2583–2592 (2010).

    CAS  PubMed  Google Scholar 

  224. Chiva-Blanch, G. et al. Dealcoholized red wine decreases systolic and diastolic blood pressure and increases plasma nitric oxide. Circ. Res. 111, 1065–1068 (2012).

    CAS  PubMed  Google Scholar 

  225. Rocha, B. S. et al. Dietary nitrite in nitric oxide biology: a redox interplay with implications for pathophysiology and therapeutics. Curr. Drug Targets 12, 1351–1363 (2011).

    CAS  PubMed  Google Scholar 

  226. Gago, B., Lundberg, J. O., Barbosa, R. M. & Laranjinha, J. Red wine-dependent reduction of nitrite to nitric oxide in the stomach. Free Radic. Biol. Med. 43, 1233–1242 (2007).

    CAS  PubMed  Google Scholar 

  227. Rodriguez-Mateos, A. et al. Interactions between cocoa flavanols and inorganic nitrate: additive effects on endothelial function at dietary achievable amounts. Free Radic. Biol. Med. 80, 121–128 (2014).

    PubMed  Google Scholar 

  228. Lee, S. Y. et al. The reaction of flavanols with nitrous acid protects against N-nitrosamine formation and leads to the formation of nitroso derivatives which inhibit cancer cell growth. Free Radic. Biol. Med. 40, 323–334 (2006).

    CAS  PubMed  Google Scholar 

  229. Lucas, D. L., Brown, R. A., Wassef, M. & Giles, T. D. Alcohol and the cardiovascular system: research challenges and opportunities. J. Am. Coll. Cardiol. 45, 1916–1924 (2005).

    CAS  PubMed  Google Scholar 

  230. Deng, X. S. & Deitrich, R. A. Ethanol metabolism and effects: nitric oxide and its interaction. Curr. Clin. Pharmacol. 2, 145–153 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  231. Kokkinos, P. & Myers, J. Exercise and physical activity: clinical outcomes and applications. Circulation 122, 1637–1648 (2010).

    PubMed  Google Scholar 

  232. Di Francescomarino, S., Sciartilli, A., Di Valerio, V., Di Baldassarre, A. & Gallina, S. The effect of physical exercise on endothelial function. Sports Med. 39, 797–812 (2009).

    PubMed  Google Scholar 

  233. Charakida, M., Masi, S., Luscher, T. F., Kastelein, J. J. & Deanfield, J. E. Assessment of atherosclerosis: the role of flow-mediated dilatation. Eur. Heart J. 31, 2854–2861 (2010).

    PubMed  Google Scholar 

  234. Hambrecht, R. et al. Regular physical activity improves endothelial function in patients with coronary artery disease by increasing phosphorylation of endothelial nitric oxide synthase. Circulation 107, 3152–3158 (2003).

    CAS  PubMed  Google Scholar 

  235. Laufs, U. et al. Physical training increases endothelial progenitor cells, inhibits neointima formation, and enhances angiogenesis. Circulation 109, 220–226 (2004).

    CAS  PubMed  Google Scholar 

  236. Iwakura, A. et al. Estrogen-mediated, endothelial nitric oxide synthase-dependent mobilization of bone marrow-derived endothelial progenitor cells contributes to reendothelialization after arterial injury. Circulation 108, 3115–3121 (2003).

    CAS  PubMed  Google Scholar 

  237. Lauer, T. et al. Age-dependent endothelial dysfunction is associated with failure to increase plasma nitrite in response to exercise. Basic Res. Cardiol. 103, 291–297 (2008).

    CAS  PubMed  Google Scholar 

  238. Jungersten, L., Ambring, A., Wall, B. & Wennmalm, A. Both physical fitness and acute exercise regulate nitric oxide formation in healthy humans. J. Appl. Physiol. 82, 760–764 (1997).

    CAS  PubMed  Google Scholar 

  239. Rassaf, T. et al. Vascular formation of nitrite after exercise is abolished in patients with cardiovascular risk factors and coronary artery disease. J. Am. Coll. Cardiol. 55, 1502–1503 (2010).

    CAS  PubMed  Google Scholar 

  240. Kelly, J. et al. Dietary nitrate supplementation reduces the oxygen cost of exercise in healthy older adults. Med. Sci. Sports Exercise 44, 443 (2012).

    Google Scholar 

  241. Bailey, S. J. et al. Dietary nitrate supplementation enhances muscle contractile efficiency during knee-extensor exercise in humans. J. Appl. Physiol. 109, 943–943 (2010).

    Google Scholar 

  242. Larsen, F. J., Weitzberg, E., Lundberg, J. O. & Ekblom, B. Effects of dietary nitrate on oxygen cost during exercise. Acta Physiol. 191, 59–66 (2007). This is the first study to demonstrate that dietary nitrate decreases oxygen consumption during exercise in humans.

    CAS  Google Scholar 

  243. Larsen, F. J., Weitzberg, E., Lundberg, J. O. & Ekblom, B. Dietary nitrate reduces maximal oxygen consumption while maintaining work performance at maximal exercise. Free Radic. Biol. Med. 48, 342–347 (2009).

    PubMed  Google Scholar 

  244. Kenjale, A. A. et al. Dietary nitrate supplementation enhances exercise performance in peripheral arterial disease. J. Appl. Physiol. 110, 1582–1591 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Larsen, F. J., Schiffer, T. A., Weitzberg, E. & Lundberg, J. O. Regulation of mitochondrial function and energetics by reactive nitrogen oxides. Free Radic. Biol. Med. 53, 1919–1928 (2012).

    CAS  PubMed  Google Scholar 

  246. Larsen, F. J. et al. Dietary inorganic nitrate improves mitochondrial efficiency in humans. Cell. Metab. 13, 149–159 (2011).

    CAS  PubMed  Google Scholar 

  247. Hernandez, A. et al. Dietary nitrate increases tetanic [Ca2+]i and contractile force in mouse fast-twitch muscle. J. Physiol. 590, 3575–3583 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Ferguson, S. K. et al. Impact of dietary nitrate supplementation via beetroot juice on exercising muscle vascular control in rats. J. Physiol. 591, 547–557 (2013).

    CAS  PubMed  Google Scholar 

  249. Zamani, P. et al. The effect of inorganic nitrate on exercise capacity in heart failure with preserved ejection fraction. Circulation 131, 371–380 (2014).

    PubMed  PubMed Central  Google Scholar 

  250. Berry, M. J. et al. Dietary nitrate supplementation improves exercise performance and decreases blood pressure in COPD patients. Nitric Oxide 48, 22–30 (2014).

    PubMed  PubMed Central  Google Scholar 

  251. Dellinger, R. P. et al. Effects of inhaled nitric oxide in patients with acute respiratory distress syndrome: results of a randomized phase II trial. Inhaled nitric oxide in ARDS study group. Crit. Care Med. 26, 15–23 (1998).

    CAS  PubMed  Google Scholar 

  252. Lopez, A. et al. Multiple-center, randomized, placebo-controlled, double-blind study of the nitric oxide synthase inhibitor 546C88: effect on survival in patients with septic shock. Crit. Care Med. 32, 21–30 (2004).

    CAS  PubMed  Google Scholar 

  253. Petros, A. et al. Effects of a nitric oxide synthase inhibitor in humans with septic shock. Cardiovasc. Res. 28, 34–39 (1994).

    CAS  PubMed  Google Scholar 

  254. Schulz, E. et al. Functional and biochemical analysis of endothelial (dys)function and NO/cGMP signaling in human blood vessels with and without nitroglycerin pretreatment. Circulation 105, 1170–1175 (2002).

    CAS  PubMed  Google Scholar 

  255. Tannenbaum, S. R. & Correa, P. Nitrate and gastric cancer risks. Nature 317, 675–676 (1985).

    CAS  PubMed  Google Scholar 

  256. Tannenbaum, S. R., Sisnkey, A. J., Weisman, M. & Bishop, W. Nitrite in human saliva. Its possible relationship to nitrosamine formation. J. Natl Cancer Inst. 53, 79–84 (1974).

    CAS  PubMed  Google Scholar 

  257. Bryan, N. S., Alexander, D. D., Coughlin, J. R., Milkowski, A. L. & Boffetta, P. Ingested nitrate and nitrite and stomach cancer risk: an updated review. Food Chem. Toxicol. 50, 3646–3665 (2012).

    CAS  PubMed  Google Scholar 

  258. Lauer, T. et al. Plasma nitrite rather than nitrate reflects regional endothelial nitric oxide synthase activity but lacks intrinsic vasodilator action. Proc. Natl Acad. Sci. USA 98, 12814–12819 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  259. Zweier, J. L., Wang, P., Samouilov, A. & Kuppusamy, P. Enzyme-independent formation of nitric oxide in biological tissues. Nat. Med. 1, 804–809 (1995).

    CAS  PubMed  Google Scholar 

  260. Benjamin, N. et al. Stomach NO synthesis. Nature 368, 502 (1994). References 259 and 260 demonstrate the NOS-independent formation of NO from inorganic nitrite in biological tissues.

    CAS  PubMed  Google Scholar 

  261. Ghosh, S. M. et al. Enhanced vasodilator activity of nitrite in hypertension: critical role for erythrocytic xanthine oxidoreductase and translational potential. Hypertension 61, 1091–1102 (2013).

    CAS  PubMed  Google Scholar 

  262. Richardson, G. et al. The ingestion of inorganic nitrate increases gastric S-nitrosothiol levels and inhibits platelet function in humans. Nitric Oxide 7, 24–29 (2002).

    CAS  PubMed  Google Scholar 

  263. Heiss, C. et al. Dietary inorganic nitrate mobilizes circulating angiogenic cells. Free Radic. Biol. Med. 52, 1767–1772 (2012).

    CAS  PubMed  Google Scholar 

  264. Kapil, V., Khambata, R. S., Robertson, A., Caulfield, M. J. & Ahluwalia, A. Dietary nitrate provides sustained blood pressure lowering in hypertensive patients: a randomized, phase 2, double-blind, placebo-controlled study. Hypertension 65, 320–327 (2015). This study shows the therapeutic effects of dietary nitrate in hypertension.

    CAS  PubMed  Google Scholar 

  265. Appel, L. J. et al. A clinical trial of the effects of dietary patterns on blood pressure. DASH Collaborative Research Group. N. Engl. J. Med. 336, 1117–1124 (1997).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors wish to sincerely thank all co-authors of the original articles from their groups, which are highlighted in this Review. A special thanks to J. Lancaster Jr for valuable comments on the manuscript. J.O.L. and E.W. receive research support from Torsten Söderbergs Foundation, Jochnick Foundation, the Swedish Research Council, the Swedish Heart and Lung Foundation and Stockholm County Council (ALF). M.T.G. receives research support from US National Institutes of Health (NIH; grants 2R01HL098032, 1R01HL125886-01, P01HL103455, T32 HL110849 and T32 HL007563), the Institute for Transfusion Medicine and the Hemophilia Center of Western Pennsylvania.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Jon O. Lundberg, Mark T. Gladwin or Eddie Weitzberg.

Ethics declarations

Competing interests

J.O.L. and E.W. are co-inventors of patent applications relating to the medical uses of inorganic nitrate and nitrite salts and co-directors of Heartbeet Ltd. M.T.G. is listed as a co-inventor on a US National Institutes of Health government patent for the use of nitrite salts in cardiovascular diseases. M.T.G. also receives grant support from Aires Pharmaceuticals (now owned by Molecular Adhesion and Sealant Technology (MAST) Therapeutics) for a Phase II proof-of-concept trial using inhaled nitrite for pulmonary arterial hypertension.

PowerPoint slides

Glossary

Organic nitrates

A group of synthetic pharmacological substances used in the treatment of cardiovascular disease. They are esters of nitric acid that contain an organic carbon residue and a nitrooxyl group (ONO2). Organic nitrates are enzymatically metabolized to generate nitric oxide and nitrite.

Vascular endothelium

A layer of cells that covers the inner surface of the vasculature and acts as a bioactive interface between the blood and the vascular wall. The endothelium is involved in many aspects of vascular biology by generating substances to uphold vascular homeostasis and in the control of vascular tone.

Phosphorylation

The addition of a phosphate group (PO43−) to a protein or other organic compound, thereby regulating its function and activity. This important mechanism is enzymatically regulated by kinases (which phosphorylate proteins) and phosphatases (which dephosphorylate proteins).

Superoxide

A reactive free radical that is constantly produced in the human body through the reduction of molecular oxygen. The main systems that generate superoxide in the body are NADPH oxidases, mitochondria and xanthine oxidoreductase. Physiological generation of superoxide is important for cell signalling, but increased production leads to oxidative stress.

Atherosclerosis

A progressive, inflammatory process characterized by the build-up of fats, cholesterol and other substances in the artery wall (plaques) that can restrict blood flow. Plaque rupture in the coronary vasculature leads to acute myocardial infarction.

Cyclic guanosine monophosphate

(cGMP). A cyclic nucleotide derived from GTP that acts as a second messenger, much like cyclic AMP.

Free radical

A molecule or ion that has an unpaired valence electron, which makes it chemically reactive with other substances. Examples relevant to this article are nitric oxide (NO) and superoxide (O2).

Transition metals

A group of elements in the periodic system. They are important in biological chemistry owing to their ability to exhibit two or more oxidation states. Examples include iron, cobalt, copper and molybdenum, of which iron is the most important because of its involvement in oxygen transport and electron transfer reactions (that is, oxidation–reduction reactions). Molybdenum-containing enzymes are involved in the reduction of nitrate to nitrite (NO3 to NO2) and nitrite to nitric oxide (NO2 to NO).

Reactive oxygen species

(ROS). Molecules that are highly reactive owing to the presence of unpaired valence-shell electrons. ROS form as a natural by-product of the normal metabolism of oxygen and have important roles in cell signalling; however, increased ROS levels can result in substantial damage to cell structures (known as oxidative stress).

Methaemoglobin

A form of the oxygen-carrying protein haemoglobin in which the iron in the haem group is in the Fe3+ state rather than the Fe2+ state of normal haemoglobin. Methaemoglobin is unable to carry oxygen.

Cytochrome c oxidase

The final component in the mitochondrial respiratory chain, which catalyses the reduction of oxygen, leading to build up of a chemiosmotic gradient used for ATP production.

Ischaemic preconditioning

A technique for producing tissue protection against the loss of blood flow to an organ by inducing short, repeated episodes of ischaemia before an ischaemic insult, which reduces subsequent tissue injury.

Inotropic

Affecting the force of muscle contractions in the heart.

Ischaemia–reperfusion injury

(I–R injury). Tissue damage caused during organ reperfusion after a period of ischaemia. Restoration of blood flow results in inflammation and oxidative stress.

Mitochondrial electron transport chain

A system that transports electrons from energy-rich electron donors and pumps protons across the inner mitochondrial membrane, building an electrochemical gradient that is used to generate ATP, the energy currency for all biochemical processes.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lundberg, J., Gladwin, M. & Weitzberg, E. Strategies to increase nitric oxide signalling in cardiovascular disease. Nat Rev Drug Discov 14, 623–641 (2015). https://doi.org/10.1038/nrd4623

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd4623

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research