Skip to main content
Log in

Attenuation of nitric oxide-stimulated soluble guanylyl cyclase from the rat brain by halogenated volatile anesthetics

  • Original Articles
  • Published:
Journal of Anesthesia Aims and scope Submit manuscript

Abstract

Purpose. The present study was undertaken to examine whether interaction between halogenated volatile anesthetics and nitric oxide (NO) at soluble guanylyl cyclase (sGC) would occur in rat brain.

Methods. A soluble brain fraction was prepared from extensively perfused Sprague-Dawley rat brains by centrifugation and used as the source of sGC. sGC was incubated with NO and halogenated volatile anesthetics, and cGMP production was determined by enzyme immunoassay in aliquots of the supernatant.

Results. Halothane and sevoflurane produced significant (P<0.01) and dose-dependent inhibition of NO-stimulated sGC activity over a range of NO concentrations (2×10−9 to 2×10−5 M). Among the anesthetics, halothane tended to have a large inhibitory effect on NO-stimulated sGC activity, which was, however, not significant. sGC activity was also inhibited by both anesthetics (P<0.05) in the absence of NO stimulation. GTP dose-dependently increased both NO-stimulated and-nonstimulated sGC activities. Halothane and sevoflurane decreased these activities (P<0.01), but the inhibition by these anesthetics was not significant at higher GTP concentrations.

Conclusion. These results suggest that halogenated volatile anesthetics can attenuate the activity of NO-stimulated sGC by competing with NO for the NO binding site on the enzyme.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Palmer RMJ, Ashton DS, Moncada S (1988) Vascular endothelial cells synthesize nitric oxide froml-arginine. Nature (Lond) 333:664–666

    Article  CAS  Google Scholar 

  2. Azuma H, Ishikawa M, Sekizawa S (1986) Endothelium-dependent inhibition of platelet aggregation. Br J Pharmacol 88:411–415

    PubMed  CAS  Google Scholar 

  3. Bredt DS, Snyder SH (1992) Nitric oxide, a novel neuronal messenger. Neuron 8:3–11

    Article  PubMed  CAS  Google Scholar 

  4. Olstein EH, Wood KS, Ignarro LJ (1982) Purification and properties of heme-deficient soluble guanylate cyclase: effects of heme and other factors on enzyme activation of NO, NO-heme and protoporphyrin. Arch Biochem Biophys 218:187–198

    Article  Google Scholar 

  5. Vincent SR, Hope BT (1992) Neurons that say NO. Trends Neurosci 15:108–117

    Article  PubMed  CAS  Google Scholar 

  6. Garthweite J, Charles SL, Chess-Williams R (1988) Endothelium-derived relaxing factor release on activation of NMDA receptor suggests role as intracellular messenger in the brain. Nature (Lond) 336:385–388

    Article  Google Scholar 

  7. Garthweite J, Garthweite G, Palmer RMJ, Moncada S (1989) NMDA receptor activation induces nitric oxide synthesis from arginine in rat brain slices. Eur J Pharmacol 172:413–416

    Article  Google Scholar 

  8. Bredt DS, Snyder SH (1989) Nitric oxide mediates glutamate-linked enhancement of cGMP levels in the cerebellum. Proc Natl Acad Sci USA 86:9030–9033

    Article  PubMed  CAS  Google Scholar 

  9. Kant GJ, Muller TW, Lenox RH, Meyerhoff JL (1980) In vivo effects of pentobarbital and halothane anesthesia on levels of adenosine 3′,5′-monophosphate and guanosine 3′,5′-monophosphate in rat brain regions and pituitary. Biochem Pharmacol 29:1891–1896

    Article  PubMed  CAS  Google Scholar 

  10. Vulliemoz Y, Verosky M, Alpert M, Triner L (1983) Effects of enflurane on cerebellar cGMP and on motor activity in the mouse. Br J Anesth 55:79–84

    CAS  Google Scholar 

  11. Gonzales JM, Loeb AL, Reichard PS, Irvine S (1995) Ketamine inhibits glutamateN-methyl-d-aspartate and quisqualate-stimulated cGMP production in cultured cerebral neurons. Anesthesiology 82:205–213

    Article  PubMed  CAS  Google Scholar 

  12. Johns RA, Moscicki JC, DiFazio CA (1992) Nitric oxide synthase inhibitor dose-dependently and reversibly reduces the threshold for halothane anesthesia. Anesthesiology 77:779–784

    PubMed  CAS  Google Scholar 

  13. Ichinose F, Huang PL, Zapol WM (1995) Effects of targeted neuronal nitric oxide synthase gene disruption and nitroG-l-arginine methylester on the threshold for isoflurane anesthesia. Anesthesiology 83:101–108

    Article  PubMed  CAS  Google Scholar 

  14. Pajewski TN, DiFazio CA, Moscicki JC, Johns RA (1996) Nitric oxide synthase inhibitors 7-nitro indazole and nitroG-l-arginine methyl ester, dose dependently reduced the threshold for isoflurane anesthesia. Anesthesiology 85:1111–1119

    Article  PubMed  CAS  Google Scholar 

  15. Kawabata A, Umeda N, Takagi H (1993)l-Arginine exerts a dual role in niciceptive processing in the brain: involvement of the kyotorphin-Met-enkephalin pathway and NO-cyclic GMP pathway. Br J Pharmacol 109:73–79

    PubMed  CAS  Google Scholar 

  16. Ahr HJ, King LJ, Nastainczyk W, Ullrich V (1982) The mechanism of reductive dehalogenation of halothane by liver cytochrome P-450. Biochem Pharmacol 41:383–390

    Article  Google Scholar 

  17. Baker MT, Nelson RM, Van Dyke RA (1983) The release of inorganic fluoride form halothane and halothane metabolites by cytochrome P-450, hemin and hemoglobin. Drug Metab Dispos 11:308–311

    PubMed  CAS  Google Scholar 

  18. Masaki E, Van Dyke RA (1992) Effect of nitric oxide on the anaerobic metabolism of halothane by rat hepatic cytochrome P-450. FASEB J 6:A1568 (abstract)

    Google Scholar 

  19. Hart JL, Jing M, Bina S, Freas W, Van Dyke RA, Muldoon SM (1993) Effects of halothane on EDRF/cGMP-mediated vascular smooth muscle relaxation. Anesthesiology 79:323–331

    PubMed  CAS  Google Scholar 

  20. Jing M, Hart JL, Masaki E, Van Dyke RA, Bina S, Muldoon SM (1995) Vascular effects of halothane and isoflurane: cGMP dependent and independent actions. Life Sci 56:19–29

    Article  PubMed  CAS  Google Scholar 

  21. Zuo Z, Johns RA (1995) Halothane, Enflurane, and isoflurane do not affect the basal or agonist-stimulated activity of partially isolated soluble and particulate guanylyl cyclases of rat brain. Anesthesiology 83:395–404

    Article  PubMed  CAS  Google Scholar 

  22. Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein binding. Anal Biochem 72:248–254

    Article  PubMed  CAS  Google Scholar 

  23. Moy JA, Bates JN, Fisher RA (1991) Effects of nitric oxide on platelet-activation factor and α-adrenergic-stimulated vasoconstriction and glycogenolysis in perfused rat liver. J Biol Chem 266:8092–8096

    PubMed  CAS  Google Scholar 

  24. Myers PR, Minor RJ Jr, Bates JN, Harrison DG (1990) Vasorelaxant properties of the endothelium-derived relaxing factor more closely resembles-nitrosocysteine than nitric oxide. Nature (Lond) 345:161–163

    Article  CAS  Google Scholar 

  25. Ignarro LJ, Wood KS, Wolin MS (1982) Activation of purified soluble guanylate cyclase by protoporphyrin IX. Proc Natl Acad Sci USA 79:2870–2873

    Article  PubMed  CAS  Google Scholar 

  26. Brume B, Ullrich V (1987) Inhibition of platelet aggregation by carbon monoxide is mediated by activation of guanylate cyclase. Mol Pharmacol 32:497–504

    Google Scholar 

  27. Marks GS, Brien JF, Nakatsu K, McLaughlin BE (1991) Does carbon monoxide have a physiological function? Trends Pharmacol Sci 12:185–188

    Article  PubMed  CAS  Google Scholar 

  28. Palmer RMJ, Ferrige AG, Moncada S (1987) Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature (London) 327:524–526

    Article  CAS  Google Scholar 

  29. Ignarro LJ, Adams JB, Horwitz PM, Wood KS (1986) Activation of soluble guanylate cyclase by NO-hemeproteins involves NO-heme change. Comparison of heme-containing and heme-deficient enzyme forms. J Biol Chem 261:4997–5002

    PubMed  CAS  Google Scholar 

  30. Baker MT, Van Dyke RA (1992) Biochemical and toxicological aspects of the volatile anesthetics. In: Barash PG, Cullen BF, Stoelting RK (eds) Clinical anesthesia, 2nd edn JB Lippincott, Philadelphia, pp 467–480

    Google Scholar 

  31. Van Dyke RA, Wineman CG (1971) Enzymatic dechlorination: dechlorination of chloroethanes and propanes in vitro. Biochem Pharmacol 20:463–470

    Article  PubMed  Google Scholar 

  32. Jing M, Bina S, Verma A, Hart JL, Muldoon SM (1996) Effects of halothane and isoflurane on carbon monoxide-induced relaxations in the rat aorta. Anesthesiology 85:347–354

    Article  PubMed  CAS  Google Scholar 

  33. Blaise G, To Q, Parent M, Lagarde B, Asenjo F, Sauve R (1994) Does halothane interfere with the release action or stability of endothelium-derived relaxing factor/nitric oxide? Anesthesiology 80:417–426

    PubMed  CAS  Google Scholar 

  34. Yoshida K, Okabe E (1992) Selective impairment of endothelium-dependent relaxation by sevoflurane: oxygen free radicals participation. Anesthesiology 76:440–447

    PubMed  CAS  Google Scholar 

  35. Terasako K, Nakamura K, Miyawaki I, Toda H, Karuyama M, Mori K (1994) Inhibitory effects of anesthetics on cyclic guanosine monophosphate (cGMP) accumulation in rat cerebellar slices. Anesth Analg 79:91–96

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

About this article

Cite this article

Masaki, E., Kondo, I. Attenuation of nitric oxide-stimulated soluble guanylyl cyclase from the rat brain by halogenated volatile anesthetics. J Anesth 12, 81–86 (1998). https://doi.org/10.1007/BF02480777

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1007/BF02480777

Key words

Navigation