Skip to main content
Log in

Lachgas

Sinn oder Unsinn für die heutige Narkoseführung

Nitrous oxide

Sense or nonsense for todays anaesthesia

  • Leitthema
  • Published:
Der Anaesthesist Aims and scope Submit manuscript

Zusammenfassung

Aufgrund seiner günstigen physikalischen Eigenschaften und geringen kardiovaskulären Nebenwirkungen galt Lachgas (N2O) lange Zeit als ideales Narkosegas; es wird seit 150 Jahren zur inhalativen Analgesie und Hypnose bei der Allgemeinanästhesie in Kombination mit volatilen Anästhetika, aber auch zur kurzfristigen Analgosedierung eingesetzt. Bis heute ist der genaue Wirkmechanismus nicht geklärt, relevante Komplikationen beim N2O-Einsatz sind jedoch bekannt. Die Einführung neuer Narkosegase sowie kurz wirksamer Opiate hat zu einer zunehmend kritischen Bewertung der N2O-Anwendung geführt. Lachgas hat jedoch nach wie vor einen Stellenwert im Rahmen der Kinderanästhesie zur Maskeneinleitung sowie in der Geburtshilfe. Neuere Untersuchungen zeigen, dass es durch den Verzicht auf N2O zu einer erhöhten Rate an intraoperativer Wachheit („awareness“) kommen kann. Unter sorgfältiger Berücksichtigung der dargestellten Kontraindikationen und Nebenwirkungen sowie nach Abwägung der verfügbaren Alternativen stellt N2O auch heute noch eine Option im Rahmen einer Allgemeinanästhesie dar.

Abstract

Nitrous oxide has been used in addition to other volatile anaesthetics to provide general anaesthesia and short time sedation for more than 150 years. However, the exact mechanisms of action remain unclear. For decades nitrous oxide was considered to be the ideal anaesthetic because of his favourable physical properties and low cardiovascular side effects. However, the known side effects of nitrous oxide as well as the implementation of new anaesthetic agents and short acting opiates led to more and more criticism about the administration of this gas. Nitrous oxide is still frequently used for mask induction primarily in paediatric anaesthesia and gynaecology. However, recent studies have shown that omitting nitrous oxide can also be a risk factor because of an increased susceptibility to intraoperative awareness. Careful consideration of the illustrated contraindications and side effects as well as the available alternatives, shows that nitrous oxide is still an option in general anaesthesia.

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.

Abb. 1
Abb. 2

Literatur

  1. Abballe C, Camaioni D, Mascaro A, Boccardi M, Evangelista M (1993) Anesthesia for laparoscopic cholecystectomy: the use of nitrous oxide in the anesthetic mixture. G Chir 14:493–495

    CAS  PubMed  Google Scholar 

  2. Apfel CC, Roewer N (2000) Einflußfaktoren von Übelkeit und Erbrechen nach Narkosen. Anaesthesist 49:629–642

    Article  CAS  PubMed  Google Scholar 

  3. Apfel CC, Roewer N (2004) Postoperative Übelkeit und Erbrechen. Anaesthesist 53:377–391

    Article  CAS  PubMed  Google Scholar 

  4. Apfel CC, Läärä E, Koivuranta M, Greim CA, Roewer N (1999) A simplified risk score for predicting postoperative nausea and vomiting: conclusions from cross-validations between two centers. Anesthesiology 91:693–700

    Article  CAS  PubMed  Google Scholar 

  5. Apfel CC, Korttila K, Abdalla M et al. (2004) A factorial trial of six interventions for the prevention of postoperative nausea and vomiting. N Engl J Med 350:2441–2451

    Article  CAS  PubMed  Google Scholar 

  6. Astrom S, Kjellgren D, Monestam E, Backlund U (2003) Nitrous oxide anesthesia and intravitreal gastamponade. Acta Anaesthesiol Scand 47:361–362

    CAS  PubMed  Google Scholar 

  7. Badner NH, Drader K, Freeman D, Spence JD (1998) The use of intraoperative nitrous oxide leads to postoperative increases in plasma homocysteine. Anesth Analg 87:711–713

    Article  CAS  PubMed  Google Scholar 

  8. Badner NH, Beattie WS, Freeman D, Spence JD (2000) Nitrous oxide-induced increased homocysteine concentrations are associated with increased postoperative myocardial ischemia in patients undergoing carotid endarterectomy. Anesth Analg 91:1073–1079

    Article  CAS  PubMed  Google Scholar 

  9. Badner NH, Freeman D, Spence JD (2001) Preoperative oral B vitamins prevent nitrous oxide-induced postoperative plasma homocysteine increases. Anesth Analg 93:1507–1510

    Article  CAS  PubMed  Google Scholar 

  10. Balasaraswathi K, Kumar P, Rao TL, El-Etr AA (1981) Left ventricular end-diastolic pressure (LVEDP) as an index for nitrous oxide use during coronary artery surgery. Anesthesiology 55:708–709

    CAS  PubMed  Google Scholar 

  11. Baskett PJ (1994) Nitrous oxide in pre-hospital care. Acta Anaesthesiol Scand 38:775–776

    Google Scholar 

  12. Baum J, Sievert B, Stanke H-G, Brauer K, Sachs G (2000) Lachgasfreie Niedrigflußnarkosen. Anaesthesiol Reanimat 25:60–67

    CAS  Google Scholar 

  13. Baur CP, Klingler W, Jurkat-Rott K et al. (2000) Xenon does not induce contracture in human malignant hyperthermia muscle. Br J Anaesth 85:712–716

    Article  CAS  PubMed  Google Scholar 

  14. Berkowitz BA, Finck AD, Ngai SH (1977) Nitrous oxide analgesia: reversal by naloxone and development of tolerance. J Pharmacol Exp Ther 203:539–547

    CAS  PubMed  Google Scholar 

  15. Bindslev L, Cannon D, Sykes MK (1986) Reversal of nitrous oxide-induced depression of hypoxic pulmonary vasoconstriction by lignocaine hydrochloride during collapse and ventilation hypoxia of the left lower lobe. Br J Anaesth 58:451–456

    CAS  PubMed  Google Scholar 

  16. Blackstock D, Gettes MA (1986) Negative pressure in the middle ear in children after nitrous oxide anaesthesia. Can Anaesth Soc J 33:32–35

    CAS  PubMed  Google Scholar 

  17. Burkhardt-His M (1873) Erfahrungen über die Anwendung des Stickoxydulgases als Anaestheticum. Correspondenzblatt für Schweizer Ärzte 11:283–285

  18. Cahalan MK, Weiskopf RB, Eger EI 2nd et al. (1991) Hemodynamic effects of desflurane/nitrous oxide anesthesia in volunteers. Anesth Analg 73:157–164

    CAS  PubMed  Google Scholar 

  19. Carton EG, Housmans PR (1992) Role of transsarcolemmal Ca2+ entry in the negative inotropic effect of nitrous oxide in isolated ferret myocardium. Anesth Analg 74:575–579

    CAS  PubMed  Google Scholar 

  20. Chambers JC, McGregor A, Jean-Marie J, Kooner JS (1998) Acute hyperhomocysteinaemia and endothelial dysfunction. Lancet 351:36–37

    CAS  Google Scholar 

  21. Chambers JC, McGregor A, Jean-Marie J, Obeid OA, Kooner JS (1999) Demonstration of rapid onset vascular endothelial dysfunction after homocysteinemia: an effect reversible with vitamin C therapy. Circulation 99:1156–1160

    CAS  PubMed  Google Scholar 

  22. Chapman CR, Benedetti C (1979) Nitrous oxide effects on cerebral evoked potential to pain: partial reversal with a narcotic antagonist. Anesthesiology 51:135–138

    CAS  PubMed  Google Scholar 

  23. Christensen B, Refsum H, Garras A, Ueland PM (1992) Homocysteine remethylation during nitrous oxide exposure of cells cultured in media containing various concentration of folates. J Pharmacol Exp Ther 261:1096–1105

    CAS  PubMed  Google Scholar 

  24. Christensen B, Guttormsen AB, Schneede J, Riedel B, Refsum H, Svardal A, Ueland PM (1994) Preoperative methionine loading enhances restoration of the cobalamin-dependent enzyme methionine synthase after nitrous oxide anesthesia. Anesthesiology 80:1046–1056

    Google Scholar 

  25. Cohen EN, Bellville JB, Brown BW (1971) Anesthesia, pregnancy, and miscarriage: a study of operating room nurses and anesthetists. Anesthesiology 35:343–347

    CAS  PubMed  Google Scholar 

  26. Cohen EN, Brown BW, Bruce DL (1975) A survey of anesthetic health hazards among dentists. J Am Dent Assoc 90:1291–1296

    CAS  PubMed  Google Scholar 

  27. Constant I, Abbas M, Boucheseiche S, Laude D, Murat I (2002) Non-invasive assessment of cardiovascular autonomic activity induced by brief exposure to 50% nitrous oxide in children. Br J Anaesth 88:637–643

    Article  CAS  PubMed  Google Scholar 

  28. Deutsch G, Samra SK (1990) Effects of nitrous oxide on global and regional cortical blood flow. Stroke 21:1293–1298

    CAS  PubMed  Google Scholar 

  29. Deutsch S, Bastron RD, Pierce EC Jr, Vandam LD (1969) The effects of anaesthesia with thiopentone, nitrous oxide, narcotics and neuromuscular blocking drugs on renal function in normal man. Br J Anaesth 41:807–815

    CAS  PubMed  Google Scholar 

  30. Divatia VJ, Vaidya JS, Badwe RA, Hawaldar RW (1996) Omission of nitrous oxide during anesthesia reduces the incidence of postoperative nausea and vomiting. Anesthesiology 85:1055–1062

    Article  CAS  PubMed  Google Scholar 

  31. Ebert TJ (1990) Differential effects of nitrous oxide on baroreflex control of heart rate and peripheral sympathetic nerve activity in humans. Anesthesiology 72:16–22

    CAS  PubMed  Google Scholar 

  32. Eger EI 2nd (1963) The effect of inspired concentration on the rate of rise of alveolar concentration. Anesthesiology 24:153–157

    Google Scholar 

  33. Eger EI 2nd, Saidman LJ (1965) Hazards of nitrous oxide anesthesia in bowel obstruction and pneumothorax. Anesthesiology 26:61–66

    PubMed  Google Scholar 

  34. Eisele JH Jr, Milstein JM, Goetzman BW (1986) Pulmonary vascular responses to nitrous oxide in newborn lambs. Anesth Analg 65:62–64

    CAS  PubMed  Google Scholar 

  35. Epstein RM, Rackow H, Salanitre E, Wolf GL (1964) Influence of the concentration effect on the uptake of anesthetic mixtures: the second gas effect. Anesthesiology 25:364–371

    CAS  PubMed  Google Scholar 

  36. Erkola O (1994) Nitrous oxide: laparoscopic surgery, bowel function, and PONV. Acta Anaesthesiol Scand 38:767–768

    Google Scholar 

  37. Fang F, Guo T-Z, Davies MF, Maze M (1997) Opiate receptors in the periaqueductal gray mediate analgesic effect of nitrous oxide in rats. Eur J Pharmacol 336:137–141

    Article  CAS  PubMed  Google Scholar 

  38. Fauroux B, Onody P, Gall O, Tourniaire B, Koscielny S, Clement A (2004) The efficacy of premixed nitrous oxide and oxygen for fiberoptic bronchoscopy in pediatric patients: a randomised, double-blind controlled study. Chest 125:13–14

    Article  PubMed  Google Scholar 

  39. Felts JA, Poler SM, Spitznagel EL (1990) Nitrous oxide, nausea, and vomiting after outpatient gynaecologic surgery. J Clin Anesth 2:168–171

    Article  CAS  PubMed  Google Scholar 

  40. Field LM, Dorrance DE, Krzeminska KE, Barsoum LZ (1993) Effect of nitrous oxide on cerebral blood flow in normal humans. Br J Anesth 70:154–159

    CAS  Google Scholar 

  41. Finck DA, Samaniego E, Ngai SH (1995) Nitrous oxide selectively releases met5-enkephalin and met5-enkephalin-arg6-phe7 into canine third ventricular cerebrospinal fluid. Anesth Analg 80:664–670

    Article  CAS  PubMed  Google Scholar 

  42. Fleisher LA, Nelson AK, Rosenbaum SH (1995) Post-operative myocardial ischemia: etiology of cardiac morbidity or manifestation of underlying disease? J Clin Anesth 7:97–102

    Article  CAS  PubMed  Google Scholar 

  43. Flippo TS, Holder WD (1993) Neurologic degeneration associated with nitrous oxide anesthesia in patients with vitamin B12 deficiency. Arch Surg 128:1391–1395

    CAS  PubMed  Google Scholar 

  44. Foschi D, Rizzi A, Zighetti ML et al. (2001) Effects of surgical stress and nitrous oxide anaesthesia on peri-operative plasma levels of total homocysteine. Anaesthesia 56:670–689

    Article  PubMed  Google Scholar 

  45. Freund FG, Martin WE, Wong KC, Horbein TF (1973) Abdominal muscle rigidity induced by morphine and nitrous oxide. Anesthesiology 38:358–362

    CAS  PubMed  Google Scholar 

  46. Fröhlich D, Rothe G, Wittmann S, Schmitz G, Schmid P, Taeger K, Hobbhahn J (1998) Nitrous oxide impairs the neutrophil oxidative response. Anesthesiology 88:1281–1290

    PubMed  Google Scholar 

  47. Fu AD, McDonald HR, Eliott D et al. (2002) Complications of general anesthesia using nitrous oxide in eye surgery with preexisting gas bubbles. Retina 22:569–574

    Article  PubMed  Google Scholar 

  48. Fujinaga M, Doone R, Davies MF, Maze M (2000) Nitrous oxide lacks the antinociceptive effect on the tail flick test in newborn rats. Anesth Analg 91:6–10

    Article  CAS  PubMed  Google Scholar 

  49. Giuffre M, Gross JB (1986) The effects of nitrous oxide on postoperative bowel motility. Anesthesiology 65:699–700

    CAS  PubMed  Google Scholar 

  50. Goerig M, Schulte am Esch J (2002) Frühe Beiträge zur Entwicklung der Lachgas-Sauerstoff-Anästhesie in Mitteleuropa. Anaesthesiol Reanimat 27:42–53

    CAS  Google Scholar 

  51. Goldman LJ (2003) Anesthetic uptake of sevoflurane and nitrous oxide during an inhaled induction in children. Anesth Analg 96:400–406

    Article  CAS  PubMed  Google Scholar 

  52. Graham IM, Daly LE, Refsum H et al. (1997) Plasma homocysteine as a risk factor for vascular disease: the European Concerted Action Project. JAMA 277:1775–1781

    Article  CAS  PubMed  Google Scholar 

  53. Gruss M, Bushell JT, Bright DP, Lieb WR, Mathie A, Franks PN (2004) Two-pore-domain K+ channels are a novel target for the anesthetic gases xenon, nitrous oxide, and cyclopropane. Mol Pharmacol 65:443–452

    Article  CAS  PubMed  Google Scholar 

  54. Guo T-Z, Poree L, Golden W, Stein J, Fujinaga M, Maze M (1996) Antinociceptive response to nitrous oxide is mediated by supraspinal opiate and spinal α2 adrenergic receptors in the rat. Anesthesiology 85:846–852

    Article  CAS  PubMed  Google Scholar 

  55. Guo T-Z, Davies MF, Kingery WS, Patterson AJ, Limbird LE, Maze M (1999) Nitrous oxide produces antinociceptive response via α2B and/or α2C adrenoceptor subtypes in mice. Anesthesiology 90:470–476

    Article  CAS  PubMed  Google Scholar 

  56. Guttormsen AB, Refsum H, Ueland PM (1994) The interaction between nitrous oxide and cobalamin. Biochemical effects and clinical consequences. Acta Anaesthesiol Scand 38:753–756

    Google Scholar 

  57. Hackam DG, Peterson JC, Spence JD (2000) What level of plasma homocyst(e)ine should be treated? Effects of vitamin therapy on progression of carotid atherosclerosis in patients with homocyst(e)ine levels above 14 µmol/L. Am J Hypertens 13:105–108

    Article  CAS  PubMed  Google Scholar 

  58. Hadzic A, Krysztof G, Sanborn KV, Thys DM (1995) Severe neurologic deficit after nitrous oxide anesthesia. Anesthesiology 83:863–866

    CAS  PubMed  Google Scholar 

  59. Halliday NJ (2003) Malignant hyperthermia. J Craniofac Surg 14:800–802

    Article  PubMed  Google Scholar 

  60. Hapfelmeier G, Zieglgänsberger W, Haseneder R, Schneck H, Kochs E (2000) Nitrous oxide and xenon increase the efficacy of GABA at recombinant mammalian GABA(A) receptors. Anesth Analg 91:1542–1549

    CAS  PubMed  Google Scholar 

  61. Harmon D, Rozario C, Lowe D (2003) Nitrous oxide/oxygen mixture and the prevention of pain during injection of propofol. Eur J Anaesthesiol 20:158–161

    CAS  PubMed  Google Scholar 

  62. Hashimoto T, Maze M, Ohashi Y, Fujinaga M (2001) Nitrous oxide activates GABAergic neurons in the spinal cord in fischer rats. Anesthesiology 95:463–469

    Article  CAS  PubMed  Google Scholar 

  63. Hecker KE (2001) Einsatz von Lachgas und Krankenhausplanung. Anaesthesist 50:463–464

    Article  CAS  PubMed  Google Scholar 

  64. Hee HI, Goy RW, As NG (2003) Effective reduction of anxiety and pain during venous cannulation in children: a comparison of analgesic efficacy conferred by nitrous oxide, EMLA and combination. Paediatr Anaesth 13:210–216

    PubMed  Google Scholar 

  65. Hellenthal A, Lauven PM (2001) Klinische Pharmakologie von Stickoxydul. Anaesthesiol Intensivmed Notfallmed Schmerzther 36:642–644

    Article  CAS  Google Scholar 

  66. Hemelrijck J van, Smith I, White PF (1991) Use of desflurane for outpatient anesthesia. Anesthesiology 75:197–203

    PubMed  Google Scholar 

  67. Hendrickx JFA, Coddens J, Callebaut F, Artico H, Deloof T, Demeyer I, De Wolf MA (2002) Effect of N2O on sevoflurane vaporizer settings during minimal- and low-flow anesthesia. Anesthesiology 97:400–404

    Article  CAS  PubMed  Google Scholar 

  68. Henriksen HT, Jørgensen PB (1973) The effect of nitrous oxide on intracranial pressure in patients with intracranial disorders. Br J Anaesth 45:486–492

    CAS  PubMed  Google Scholar 

  69. Herbert MK, Raile S (2001) Brauchen wir noch Lachgas?. Anaesthesiol Intensivmed Notfallmed Schmerzther 36:380–381

    Article  CAS  Google Scholar 

  70. Hermann L (1866) Notiz ueber die Empfehlung des Stickoxyduls als Anaestheticum. Bln Klein Wochenschr 3:115–116

    Google Scholar 

  71. Hobbhahn J (2001) Toxizität von Stickoxydul. Anaesthesiol Intensivmed Notfallmed Schmerzther 36:641–642

    Article  CAS  Google Scholar 

  72. Hodges BL, Gagnon MJ, Gillespie TR, Breneisen JR, O’Leary DF, Hara S, Quock RM (1994) Antagonism of nitrous oxide antinociception in the rat hot plate test by site-specific mu and epsilon opioid receptor blockage. J Pharmacol Exp Ther 269:596–600

    Google Scholar 

  73. Hohner P, Reiz S (1994) Nitrous oxide and the cardiovascular system. Acta Anaesthesiol Scand 38:763–766

    Google Scholar 

  74. Jensen AG, Prevedoros H, Kullman E, Anderberg B, Lennmarken C (1993) Perioperative nitrous oxide does not influence recovery after laparoscopic cholecystectomy. Acta Anaesthesiol Scand 37:683–686

    CAS  PubMed  Google Scholar 

  75. Jenstrup M, Fruergaard KO, Mortensen CR (1999) Pollution with nitrous oxide using laryngeal mask or face mask. Acta Anaesthesiol Scand 43:663–666

    Article  CAS  PubMed  Google Scholar 

  76. Jevtovic-Todorovic V, Todorovic SM, Mennerick S et al. (1998) Nitrous oxide (laughing gas) is an NMDA antagonist, neuroprotectant and neurotoxin. Nat Med 4:40–464

    Google Scholar 

  77. Jevtovic-Todorovic V, Beals J, Benshoff N, Olney JW (2003) Prolonged exposure to inhalational anesthetic nitrous oxide kills neurons in adult rat brain. Neuroscience 122:609–616

    Article  CAS  PubMed  Google Scholar 

  78. Kingery WS, Sawamura S, Agashe GS, Davies MF, Clark JD, Zimmer A (2001) Enkephalin release and opioid receptor activation does not mediate the antinociceptive or sedative/hypnotic effects of nitrous oxide. Eur J Pharmacol 427:27–35

    Article  CAS  PubMed  Google Scholar 

  79. Klikowitsch S (1881) Ueber das Stickstoffoxydul als Anaestheticum bei Geburten. Arch Gynaekol 18:81–108

    Google Scholar 

  80. Knill-Jones RP, Rodrigues LV, Moir DD, Spence AA (1972) Anaesthetic practice and pregnancy: controlled survey of women anaesthetists in the United Kingdom. Lancet 1:1326–1328

    Article  CAS  PubMed  Google Scholar 

  81. Koblin DD, Waskel L, Watson JE (1982) Nitrous oxide inactivates methionine synthase in human liver. Anesth Analg 61:75–78

    CAS  PubMed  Google Scholar 

  82. Kondo H, Osborne ML, Kolhouse JF et al. (1981) Nitrous oxide has multiple deleterious effects on cobalamin metabolism and causes decreases in activities of both mammalian cobalamin-dependant enzymes in rats. J Clin Invest 67:1270–1283

    CAS  PubMed  Google Scholar 

  83. Korman B, Mapleson WW (1997) Concentration and second gas effects: can the accepted explanation be improved? Br J Anaesth 78:618–625

    CAS  PubMed  Google Scholar 

  84. Krogh B, Jensen J, Henneberg SW, Hole P, Kronborg O (1994) Nitrous oxide does not influence operating conditions or postoperative course in colonic surgery. Br J Anaesth 72:55–57

    Google Scholar 

  85. Kugel G, Letelier C, Zive MA, King JC (1990) Nitrous oxide and infertility. Anesth Prog 37:176–180

    CAS  PubMed  Google Scholar 

  86. Lahoud GY, Averley PA (2002) Comparison of sevoflurane and nitrous oxide mixture with nitrous oxide alone for inhalation conscious sedation in children having dental treatment: a randomised controlled trial. Anaesthesia 57:446–450

    Article  CAS  PubMed  Google Scholar 

  87. Landon MJ, Creagh-Barry P, McArthur S, Charlett A (1992) Influence of vitamin B12 status on the inactivation of methionine synthase by nitrous oxide. Br J Anaesth 69:81–86

    CAS  PubMed  Google Scholar 

  88. Landsberg G, Luria MH, Cotev S et al. (1993) Importance of long duration ST-segment depression in cardiac morbidity after vascular surgery. Lancet 341:715–719

    Article  PubMed  Google Scholar 

  89. Langbein T, Sonntag H, Trapp D et al. (1999) Volatile anaesthetics and the atmosphere: atmospheric effects of halothane, enflurane, isoflurane, desflurane and sevoflurane. Br J Anaesth 82:66–73

    CAS  PubMed  Google Scholar 

  90. Larsen R (2001) Inhalationsanästhesie. In: Larsen R (Hrsg) Anästhesie. Urban & Fischer, München, Jena, S 22

  91. Lassen HCA, Henriksen E, Neukirch F, Kristensen H (1956) Treatment of tetanus. Severe bone-marrow depression after prolonged nitrous oxide anaesthesia. Lancet 1:527–530

    Article  Google Scholar 

  92. Layzer RB (1978) Myeloneuropathy after prolonged exposure to nitrous oxide. Lancet 2:1227–1230

    Article  CAS  PubMed  Google Scholar 

  93. Lebenbom-Mansour MH, Pandit SK, Kothary SP, Randel GI, Levy L (1993) Desflurane versus propofol anesthesia: a comparative analysis in outpatients. Anesth Analg 76:936–941

    CAS  PubMed  Google Scholar 

  94. Li SH, Li SN, Shih HY, Yi HD, Chiang CY (2002) Personnel exposure to waste sevoflurane and nitrous oxide during general anesthesia with cuffed endotracheal tube. Acta Anaesthesiol Sin 40:185–190

    PubMed  Google Scholar 

  95. Logan M, Farmer JG (1989) Anaesthesia and the ozone layer. Br J Anaesth 63:654–647

    Google Scholar 

  96. Lunn JK, Stanley TH, Eisele J (1979) High-dose fentanyl anesthesia for coronary artery surgery: plasma fentanyl concentrations and influence of nitrous oxide on cardiovascular responses. Anesth Analg 58:390–395

    CAS  PubMed  Google Scholar 

  97. Ma D, Wilhelm S, Maze M, Franks NP (2002) Neuroprotective and neurotoxic properties of the „inert“ gas, xenon. Br J Anaesth 89:739–746

    Article  CAS  PubMed  Google Scholar 

  98. Mapleson WW, Korman B (1998) Concentration and second-gas effects in the water analogue. Br J Anaesth 81:837–843

    CAS  PubMed  Google Scholar 

  99. Marx T (1997) Belastung des Arbeitsplatzes mit volatilen Anästhetika und Lachgas. Anaesthesiol Intensivmed Notfallmed Schmerzther 32:532–540

    CAS  Google Scholar 

  100. Masood J, Shah N, Lane T, Andrews H, Simpson P, Barua JM (2002) Nitrous oxide (Entonox) inhalation and tolerance of transrectal ultrasound guided prostate biopsy: a double-blind randomised controlled study. J Urol 168:116–120

    Google Scholar 

  101. Mayer EL, Jakobsen DW, Robinson K (1996) Homocysteine and coronary atherosclerosis. J Am Coll Cardiol 27:517–527

    Article  CAS  PubMed  Google Scholar 

  102. Maze M, Fujinaga M (2000) Recent advances in understanding the actions and toxicity of nitrous oxide. Anaesthesia 55:311–314

    Article  CAS  PubMed  Google Scholar 

  103. McGlothlin JD, Jensen PA, Todd WF, Fischbach TJ (1988) Study protocol: control of anesthetic gases in dental opertories. Cincinnati: National Institute for Occupational Safety and Health

  104. Mennerick S, Jevtovic-Todorovic V, Todorovic SM, Shen W, Olney JW, Zorumski CF (1998) Effect of nitrous oxide on excitatory and inhibitory synaptic transmission in hippocampal cultures. J Neurosci 18:9716–9726

    CAS  PubMed  Google Scholar 

  105. Meretoja OA, Takkunen O, Heikkilä H, Wegelius U (1985) Hemodynamic response to nitrous oxide during high-dose fentanyl pancuronium anaesthesia. Acta Anaesthesiol Scand 29:137–141

    CAS  PubMed  Google Scholar 

  106. Milocco I, Schlossman D, William-Olsson G, Appelgren LK (1985) Fentanyl-droperidol-nitrous oxide anaesthesia in patients with ischemic heart disease and various degrees of left ventricular functional impairment. Acta Anaesthesiol Scand 29:683–692

    CAS  PubMed  Google Scholar 

  107. Moudgil GC, Gordon J, Forrest JB (1984) Comparative effects of volatile anaesthetic agents and nitrous oxide on human leukocyte migration in vitro. Can Anaesth Soc J 31:631–637

    CAS  PubMed  Google Scholar 

  108. Muir JJ, Warner MA, Offord KP, Buck CF, Harper JV, Kunkel SE (1987) Role of nitrous oxide and other factors in postoperative nausea and vomiting: a randomised and blinded prospective study. Anesthesiology 66:513–518

    CAS  PubMed  Google Scholar 

  109. Murakawa M, Adachi T, Nakao S, Seo N, Shingu K, Mori K (1994) Activation of the cortical and medullary dopaminergic systems by nitrous oxide in rats: a possible neurochemical basis for psychotropic effects and postanesthetic nausea and vomiting. Anesth Analg 78:376–381

    Google Scholar 

  110. Nagata A, Nakao S, Nishizawa N, Masuzawa M, Inada T, Murao K, Miyamoto E, Shingu K (2001) Xenon inhibits but N(2)O enhances ketamine-induced c-Fos expression in the rat posterior cingulated and retrosplenial cortices. Anesth Analg 92:362–368

    Article  CAS  PubMed  Google Scholar 

  111. Nakayama H, Takahashi H, Okubo N, Miyabe M, Toyooka H (2002) Xenon and nitrous oxide do not depress cardiac function in an isolated rat heart model. Can J Anaesth 49:375–379

    PubMed  Google Scholar 

  112. Nishikawa K, Kunimoto F, Isa Y, Miyoshi S, Takahashi K, Morita T, Arii H, Goto F (2000) Second gas effect of N2O on oxygen uptake. Can J Anaesth 47:506–510

    CAS  PubMed  Google Scholar 

  113. Nunn JF (1987) Clinical aspects of the interaction between nitrous oxide and vitamin B12. Br J Anaesth 59:3–13

    CAS  PubMed  Google Scholar 

  114. Nunn JF, O’Morain C (1982) Nitrous oxide decreases motility of human neutrophils in vitro. Anesthesiology 56:45–48

    CAS  PubMed  Google Scholar 

  115. Nygard O, Nordrehaug JE, Refsum H et al. (1997) Plasma homocysteine levels and mortality in patients with coronary artery disease. N Engl J Med 337:230–236

    Article  CAS  PubMed  Google Scholar 

  116. Olney JW, Labruyere J, Price MT (1989) Pathological changes induced in cerebrocortical neurones by phencyclidine and related drugs. Science 244:1360–1362

    CAS  PubMed  Google Scholar 

  117. Olney JW, Labruyere J, Wang G (1991) NMDA antagonist neurotoxicity: mechanism and prevention. Science 254:1515–1518

    CAS  PubMed  Google Scholar 

  118. O’Shea, Moultrie S, Drummond GB (2001) Influence of nitrous oxide on induction of anaesthesia with sevoflurane. Br J Anaesth 87:286–288

    Article  PubMed  Google Scholar 

  119. Pac-Soo CK, Wang C, Chakrabarti MK, Whitwam JG (2000) Comparison of the effects of inhalational anaesthetic agents on sympathetic activity in rabbits. Eur J Anaesthesiol 17:311–318

    Article  CAS  PubMed  Google Scholar 

  120. Parbrook GD, Kennedy BR (1964) Values of pre-mixed nitrous oxide and oxygen mixtures in relief of post-operative pain. BMJ 2:1303

    Google Scholar 

  121. Paut O, Calméjane C, Delorme J, Lacroix F, Camboulives J (2001) EMLA versus nitrous oxide for venous cannulation in children. Anesth Analg 93:590–593

    Article  CAS  PubMed  Google Scholar 

  122. Pedersen FM, Wilken-Jensen C, Knudsen F, Lindekaer AL, Svare E (1993) The influence of nitrous oxide on recovery of bowel function after abdominal hysterectomy. Acta Anaesthesiol Scand 37:692–696

    CAS  PubMed  Google Scholar 

  123. Pelligrino DA, Miletich DJ, Hoffman WE, Albrecht RF (1984) Nitrous oxide markedly increases cerebral cortical metabolic rate and blood flow in the goat. Anesthesiology 60:405–412

    CAS  PubMed  Google Scholar 

  124. Perreault L, Normandin N, Plamondon L (1982) Middle ear pressure variations during nitrous oxide anaesthesia. Can Anaesth Soc J 29:428–434

    CAS  PubMed  Google Scholar 

  125. Pypendop BH, Ilkiw JE, Imai A, Bolich JA (2003) Hemodynamic effects of nitrous oxide in isoflurane-anesthetized cats. Am J Vet Res 64:273–278

    CAS  PubMed  Google Scholar 

  126. Quock RM, Graczak LM (1988) Influence of narcotic drugs upon nitrous oxide analgesia in mice. Brain Res 440:35–41

    Article  CAS  PubMed  Google Scholar 

  127. Quock RM, Curtis BA, Reynolds BJ, Mueller JL (1993) Dose-dependent antagonism and potentiation of nitrous oxide antinociception by naloxone in mice. J Pharmacol Exp Ther 267:117–122

    CAS  PubMed  Google Scholar 

  128. Rampil IJ, Kim JS, Lenhardt R, Negishi C, Sessler DI (1998) Bispectral EEG index during nitrous oxide administration. Anesthesiology 89:671–677

    Article  CAS  PubMed  Google Scholar 

  129. Reinstrup P, Messeter K (1994) Cerebrovascular response to nitrous oxide. Acta Anaesthesiol Scand 38:761–762

    Google Scholar 

  130. Reinstrup P, Ryding E, Algotsson L, Berntman L, Uski T (1994) Effects of nitrous oxide on human regional cerebral blood flow and isolated pial arteries. Anesthesiology 81:396–402

    CAS  PubMed  Google Scholar 

  131. Reinstrup P, Ryding E, Ohlsson T, Dahm PL, Uski T (2001) Cerebral blood volume (cbv) in humans during normo- and hypocapnia; influence of nitrous oxide (N2O). Anesthesiology 95:1079–1082

    Article  CAS  PubMed  Google Scholar 

  132. Rowland AS, Baird DD, Weinberg CA, Shore DL, Shy CM, Wilcox AJ (1992) Reduced fertility among women employed as dental assistants exposed to high levels of nitrous oxide. N Engl J Med 327:993–997

    CAS  PubMed  Google Scholar 

  133. Rupreht J, Dworacek B, Bonke B, Dzoljic MR, Eijndhoven MR van, Vlieger M de (1985) Tolerance to nitrous oxide in volunteers. Acta Anaesthesiol Scand 29:635–638

    CAS  PubMed  Google Scholar 

  134. Russell GB, Snider MT, Richard RB, Loomis JL (1990) Hyperbaric nitrous oxide as a sole anesthetic agent in humans. Anesth Analg 70:289–295

    CAS  PubMed  Google Scholar 

  135. Salanitre E, Rackow H (1976) N2O volumes absorbed and excreted during N2O anesthesia in children. Anesth Analg 55:95–99

    CAS  PubMed  Google Scholar 

  136. Sarner JB, Levine M, Davis PJ, Lerman J, Cook DR, Motoyama EK (1995) Clinical characteristics of sevoflurane in children; a comparison with halothane. Anesthesiology 82:38–46

    Article  CAS  PubMed  Google Scholar 

  137. Sawamura S, Kingery WS, Davies MF et al. (2000) Antinociceptive action of nitrous oxide is mediated by stimulation of noradrenergic neurons in the brainstem and activation of α2B adrenoceptors. J Neurosci 20:9242–9251

    CAS  PubMed  Google Scholar 

  138. Scheinin B, Lindgren L, Scheinin TM (1990) Perioperative nitrous oxide delays bowel function after colonic surgery. Br J Anaesth 64:154–158

    CAS  PubMed  Google Scholar 

  139. Schirmer U (1998) Lachgas: Entwicklung und heutiger Stellenwert. Anaesthesist 47:245–255

    Article  CAS  PubMed  Google Scholar 

  140. Schulte-Sasse U, Hess W, Tarnow J (1982) Pulmonary vascular responses to nitrous oxide in patients with normal and high pulmonary vascular resistance. Anesthesiology 57:9–13

    CAS  PubMed  Google Scholar 

  141. Sellgren J, Pontén J, Wallin BG (1990) Percutaneus recordings of muscle nerve sympathetic activity during propofol, nitrous oxide and isoflurane anesthesia in humans. Anesthesiology 73:20–27

    CAS  PubMed  Google Scholar 

  142. Sherman SJ, Cullen BF (1999) Nitrous oxide and the greenhouse effect. Anesthesiology 68:817

    Google Scholar 

  143. Shiga T, Wajima Z, Inoue T, Ogawa R (2003) Nitrous oxide produces minimal hemodynamic changes in patients receiving a propofol-based anesthetic: an esophageal Doppler ultrasound study. Can J Anaesth 50:649–652

    PubMed  Google Scholar 

  144. Splinter WM, Roberts DJ, Rgine EJ, MacNeill HB, Komocar L (1995) Nitrous oxide does not increase vomiting in children after myringotomy. Can J Anaesth 42:274–276

    CAS  PubMed  Google Scholar 

  145. Stamler JS, Osborne JA, Jaraki O, Rabbani LE, Mullins M, Singel D, Loscalzo J (1993) Adverse vascular effects of homocysteine are modulated by endothelium-derived relaxing factor and related oxides of nitrogen. J Clin Invest 91:308–318

    CAS  PubMed  Google Scholar 

  146. Stenqvist O, Husum B, Dale O (2001) Nitrous oxide: an ageing gentleman. Acta Anaesthesiol Scand 45:135–137

    Article  CAS  PubMed  Google Scholar 

  147. Stoelting RK, Eger EI 2nd (1969) An additional explanation for the second gas effect: a concentrating effect. Anesthesiology 30:273–277

    CAS  PubMed  Google Scholar 

  148. Stubbs PJ, Al-Obaidi MK, Conroy RM, Collinson PO, Graham IM, Noble MI (2000) Effect of plasma homocysteine concentration on early and late events in patients with acute coronary syndromes. Circulation 102:605–610

    CAS  PubMed  Google Scholar 

  149. Sun X-G, Su F, Shi YQ, Lee C (1999) The „second gas effect“ is not a valid concept. Anesth Analg 88:188–192

    Article  PubMed  Google Scholar 

  150. Swan HD, Crawford MW, Pua HL, Stephens D, Lerman J (1999) Additive contribution of nitrous oxide to sevoflurane minimum alveolar concentration for tracheal intubation in children. Anesthesiology 91:667–671

    Article  CAS  PubMed  Google Scholar 

  151. Taheri S, Eger EI 2nd (1999) A demonstration of the concentration and second gas effects in humans anesthetized with nitrous oxide and desflurane. Anesth Analg 89:774–780

    Article  CAS  PubMed  Google Scholar 

  152. Takàcs J (1996) N2O-induzierte akute funikuläre Myelose bei latentem Vitamin B12-Mangel. Anaesthesiol Intensivmed Notfallmed Schmerzther 31:525–528

    Google Scholar 

  153. Taylor E, Feinstein R, White PF, Soper N (1992) Anesthesia for laparoscopic cholecystectomy. Anesthesiology 76:541–543

    CAS  PubMed  Google Scholar 

  154. Thomson IA, Hughes RL, Fitch W, Campbell D (1982) Effects of nitrous oxide on liver haemodynamics and oxygen consumption in the greyhound. Anaesthesia 37:548–553

    CAS  PubMed  Google Scholar 

  155. Todorovic SM, Jevtovic-Todorovic V, Mennerick S, Perez-Reyes E, Zorumski CF (2001) Cav3.2 channel is a molecular substrate for inhibition of T-type calcium currents in rat sensory neurons by nitrous oxide. Mol Pharmacol 60:603–610

    CAS  PubMed  Google Scholar 

  156. Todorovic SM, Jevtovic-Todorovic V, Meyenburg A et al. (2001) Redox modulation of T-type calcium channels in rat peripheral nociceptors. Neuron 31:75–85

    Article  CAS  PubMed  Google Scholar 

  157. Tramer M, Moore A, McQuay H (1996) Omitting nitrous oxide in general anaesthesia: meta-analysis of intraoperative awareness and postoperative emesis in randomised controlled trials. Br J Anaesth 76:186–193

    CAS  PubMed  Google Scholar 

  158. Tunstall ME (1961) Obstetric analgesia. Lancet 2:964

    Article  CAS  PubMed  Google Scholar 

  159. Tunstall ME, Hawksworth GM (1981) Halothane uptake and nitrous oxide concentration: arterial halothane levels during caesarian section. Anesthesia 36:177–182

    CAS  Google Scholar 

  160. Vaisman AI (1967) Working conditions in the operating room and their effect on the health of anesthetists. Eksp Khir Anestheziol 12:44–49

    CAS  Google Scholar 

  161. Vessey MP (1978) Epidemiological studies of the occupational hazards of anaesthesia—a review. Anaesthesia 33:430–438

    CAS  PubMed  Google Scholar 

  162. Vieira E, Cleaton-Jones P, Moyes D (1983) Effects of low intermittent concentrations of nitrous oxide on the developing rat fetus. Br J Anaesth 55:67–69

    CAS  PubMed  Google Scholar 

  163. Vollmar B, Conzen P, Habazettl H, Adili F, Peter K (1995) Does nitrous oxide affect coronary microcirculation? An intravital microscopic study in the canine heart. Anesth Analg 80:249–255

    Article  CAS  PubMed  Google Scholar 

  164. Watanabe S, Asakura N, Taguchi N (1993) Supramaximal second gas effect: more rapid rise of alveolar halothane concentration during ipsilateral lung N2O administration compared to bilateral administration. Anesth Analg 76:69–76

    PubMed  Google Scholar 

  165. Watcha MF, Simeon RM, White PF, Stevens JL (1991) Effect of propofol on the incidence of postoperative vomiting after strabismus surgery in pediatric outpatients. Anesthesiology 75:204–209

    CAS  PubMed  Google Scholar 

  166. Weimann J (2003) Toxicity of nitrous oxide. Best Pract Res Clin Anaesthesiol 17:47–61

    Article  CAS  PubMed  Google Scholar 

  167. Welch WD (1984) Effect of enflurane, isoflurane, and nitrous oxide on the microbicidal activity of human polymorphonuclear leukocytes. Anesthesiology 61:188–192

    CAS  PubMed  Google Scholar 

  168. Whitcher CE, Zimmermann DC, Tonn EM, Piziali RL (1977) Control of occupational exposure to nitrous oxide in the dental operatory. J Am Dent Assoc 95:736–776

    Google Scholar 

  169. Wilson IG, Fell D (1993) Nitrous oxide and postoperative vomiting in children undergoing myringotomy as a day case. Paediatr Anaesth 3:283–285

    CAS  Google Scholar 

  170. Wrigley SR, Fairfield JE, Jones RM, Black AE (1991) Induction and recovery characteristics of desflurane in day case patients: a comparison with propofol. Anaesthesia 46:615–622

    CAS  PubMed  Google Scholar 

  171. Wynne J, Mann T, Alpert S (1980) Hemodynamic effects of nitrous oxide administered during cardiac catheterisation. JAMA 243:1440–1442

    Article  CAS  PubMed  Google Scholar 

  172. Yacoub O, Doell D, Kryger MH, Anthonisen NR (1975) Depression of hypoxic ventilatory response by nitrous oxide. Anesthesiology 45:385–389

    Google Scholar 

  173. Yang JC, Clark WC, Ngai SH (1980) Antagonism of nitrous oxide analgesia by naloxone in man. Anesthesiology 52:414–417

    CAS  PubMed  Google Scholar 

  174. Yang YF, Herbert L, Rüschen H, Cooling RJ (2002) Nitrous oxide anaesthesia in the presence of intraocular gas can cause irreversible blindness. BMJ 325:532–533

    Article  CAS  PubMed  Google Scholar 

  175. Yoo KY, Jeong ST, Ha IH, Lee J (2003) Nitrous oxide attenuates pressor but augments norepinephrine response to laryngoscopy and endotracheal intubation. Anesth Analg 96:1516–1521

    CAS  PubMed  Google Scholar 

  176. Zacny JP, Hurst RJ, Graham L, Janiszewski DJ (2002) Preoperative dental anxiety and mood changes during nitrous oxide inhalation. J Am Dent Assoc 133:82–88

    PubMed  Google Scholar 

  177. Zhang C, Davies MF, Guo T-Z, Maze M (1999) The analgesic action of nitrous oxide is dependent on the release of norepinephrine in the dorsal horn of the spinal cord. Anesthesiology 9:1401–1407

    Article  Google Scholar 

  178. Zuniga J, Joseph S, Knigge K (1987) Nitrous oxide analgesia: partial antagonism by naloxone and total reversal after periaqueductal gray lesions in the rat. Eur J Pharmacol 142:51–60

    Article  CAS  PubMed  Google Scholar 

  179. Zwass MS, Fisher DM, Welborn LG et al. (1992) Induction and maintenance characteristics of anesthesia with desflurane and nitrous oxide in infants and children. Anesthesiology 76:373–378

    CAS  PubMed  Google Scholar 

Download references

Interessenkonflikt:

Der korrespondierende Autor versichert, dass keine Verbindungen mit einer Firma, deren Produkt in dem Artikel genannt ist, oder einer Firma, die ein Konkurrenzprodukt vertreibt, bestehen.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to M. W. Hollmann.

Additional information

Dr. Hollmann wird unterstützt durch ein Stipendium der Deutschen Forschungsgemeinschaft (DFG HO 2199/4-1), Bonn, über den BenCovino Award 2000 und 2002 der Firma AstraZeneca Pain Control, Sweden, sowie durch ein Forschungs-Förderungsprogramm der Medizinischen Fakultät Heidelberg.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schönherr, M.E., Hollmann, M.W. & Graf, B. Lachgas. Anaesthesist 53, 796–812 (2004). https://doi.org/10.1007/s00101-004-0742-9

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00101-004-0742-9

Schlüsselwörter

Keywords

Navigation