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:

Lasting impact of general anaesthesia on the brain: mechanisms and relevance

Key Points

  • Every year, millions of patients receive general anaesthesia for surgery or diagnostic procedures, and there is a growing concern that anaesthetic drugs might exert long-term effects on the CNS, especially at the extremes of age.

  • There is currently mixed epidemiological evidence in humans for an association between early-life anaesthesia exposure and an increased risk of subsequent sustained neurobehavioural deficits. Postoperative cognitive dysfunction in elderly individuals is of multifactorial origin, and there is no evidence for a direct causal link between anaesthesia exposure and Alzheimer disease in humans.

  • In both neonatal and ageing animals, exposure to general anaesthetics can induce lasting behavioural and cognitive deficits. These effects are dose, exposure length and sex dependent.

  • In the neonatal brain of experimental animals, administration of general anaesthetics induces impaired growth factor signalling and mitochondrial dysfunction, which in turn result in apoptosis, impaired neurogenesis or altered synaptogenesis. How these events are related to altered cognitive function remains unclear.

  • In the mature brain of rodents, general anaesthetics can induce dose- and exposure length-dependent amyloid-β aggregation and tau hyperphosphorylation, as well as neuroinflammation.

  • There are important gaps related to the translational relevance of currently available experimental models in anaesthesia-neurotoxicity research. Among them, determining how concurrent surgical and other perioperative stimuli modify the effect of general anaesthetics on the brain is of primary importance.

Abstract

General anaesthesia is usually considered to safely induce a reversible brain state allowing the performance of surgery under optimal conditions. An increasing number of clinical and experimental observations, however, suggest that anaesthetic drugs, especially when they are administered at the extremes of age, can trigger long-term morphological and functional alterations in the brain. Here, we review available mechanistic data linking general-anaesthesia exposure to impaired cognitive performance in both young and mature nervous systems. We also provide a critical appraisal of the translational value of animal models and highlight the important challenges that need to be addressed to strengthen the link between laboratory work and clinical investigations in the field of anaesthesia-neurotoxicity research.

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: General anaesthetic-induced dendritic spinogenesis in the hippocampus.
Figure 2: Molecular mechanisms involved in developmental anaesthesia neurotoxicity.
Figure 3: The hypothesized pathway of anaesthesia-induced neurotoxicity associated with Alzheimer disease neuropathogenesis.

Similar content being viewed by others

References

  1. Weiser, T. G. et al. An estimation of the global volume of surgery: a modelling strategy based on available data. Lancet 372, 139–144 (2008).

    PubMed  Google Scholar 

  2. Eckenhoff, J. E. Relationship of anesthesia to postoperative personality changes in children. Am. J. Dis. Child. 86, 587–591 (1953).

    CAS  Google Scholar 

  3. Bedford, P. D. Adverse cerebral effects of anaesthesia on old people. Lancet 6, 259–263 (1955).

    Google Scholar 

  4. Servick, K. Researchers struggle to gauge risks of childhood anesthesia. Science 346, 1161–1162 (2014).

    CAS  PubMed  Google Scholar 

  5. Rappaport, B. A., Suresh, S., Hertz, S., Evers, A. S. & Orser, B. A. Anesthetic neurotoxicity — clinical implications of animal models. N. Engl. J. Med. 372, 796–797 (2015).

    CAS  PubMed  Google Scholar 

  6. Boney, O. et al. Identifying research priorities in anaesthesia and perioperative care: final report of the joint National Institute of Academic Anaesthesia/James Lind Alliance Research Priority Setting Partnership. BMJ Open 5, e010006 (2015).

    PubMed  PubMed Central  Google Scholar 

  7. Hensch, T. K. Critical period plasticity in local cortical circuits. Nat. Rev. Neurosci. 6, 877–888 (2005).

    CAS  PubMed  Google Scholar 

  8. Burke, S. N. & Barnes, C. A. Neural plasticity in the ageing brain. Nat. Rev. Neurosci. 7, 30–40 (2006).

    CAS  PubMed  Google Scholar 

  9. Jevtovic-Todorovic, V. et al. Early exposure to common anesthetic agents causes widespread neurodegeneration in the developing rat brain and persistent learning deficits. J. Neurosci. 23, 876–882 (2003). This paper is the first to demonstrate that exposure to anaesthesia in the early postnatal period can induce histological, functional and behavioural alterations in the developing brain.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Stratmann, G., Sall, J. W., May, L. D., Loepke, A. W. & Lee, M. T. Beyond anesthetic properties: the effects of isoflurane on brain cell death, neurogenesis, and long-term neurocognitive function. Anesth. Analg. 110, 431–437 (2010).

    CAS  PubMed  Google Scholar 

  11. Loepke, A. W. & Soriano, S. G. An assessment of the effects of general anesthetics on developing brain structure and neurocognitive function. Anesth. Analg. 106, 1681–1707 (2008).

    PubMed  Google Scholar 

  12. Lin, E. P., Soriano, S. G. & Loepke, A. W. Anesthetic neurotoxicity. Anesthesiol. Clin. 32, 133–155 (2014).

    PubMed  Google Scholar 

  13. Paule, M. G. et al. Ketamine anesthesia during the first week of life can cause long-lasting cognitive deficits in rhesus monkeys. Neurotoxicol. Teratol. 33, 220–230 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Raper, J., Alvarado, M. C., Murphy, K. L. & Baxter, M. G. Multiple anesthetic exposure in infant monkeys alters emotional reactivity to an acute stressor. Anesthesiology 123, 1084–1092 (2015). This publication demonstrates that exposure of NHP infants to the commonly used anaesthetic agent sevoflurane can induce lasting alterations in emotional reactivity.

    CAS  PubMed  Google Scholar 

  15. Raper, J., Bush, A., Murphy, K. L., Baxter, M. G. & Alvarado, M. C. Multiple sevoflurane exposures in infant monkeys do not impact the mother-infant bond. Neurotoxicol. Teratol. 54, 46–51 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Sanders, R. D. et al. Dexmedetomidine attenuates isoflurane-induced neurocognitive impairment in neonatal rats. Anesthesiology 110, 1077–1085 (2009).

    CAS  PubMed  Google Scholar 

  17. Fredriksson, A., Archer, T., Alm, H., Gordh, T. & Eriksson, P. Neurofunctional deficits and potentiated apoptosis by neonatal NMDA antagonist administration. Behav. Brain Res. 153, 367–376 (2004).

    CAS  PubMed  Google Scholar 

  18. Fredriksson, A., Ponten, E., Gordh, T. & Eriksson, P. Neonatal exposure to a combination of N-methyl-d-aspartate and γ-aminobutyric acid type A receptor anesthetic agents potentiates apoptotic neurodegeneration and persistent behavioral deficits. Anesthesiology 107, 427–436 (2007).

    CAS  PubMed  Google Scholar 

  19. Yu, D., Jiang, Y., Gao, J., Liu, B. & Chen, P. Repeated exposure to propofol potentiates neuroapoptosis and long-term behavioral deficits in neonatal rats. Neurosci. Lett. 534, 41–46 (2013).

    CAS  PubMed  Google Scholar 

  20. Qiu, L. et al. Acute and long-term effects of brief sevoflurane anesthesia during the early postnatal period in rats. Toxicol. Sci. 149, 121–133 (2016).

    CAS  PubMed  Google Scholar 

  21. Shen, X. et al. Early life exposure to sevoflurane impairs adulthood spatial memory in the rat. Neurotoxicology 39, 45–56 (2013).

    CAS  PubMed  Google Scholar 

  22. Shen, X. et al. Selective anesthesia-induced neuroinflammation in developing mouse brain and cognitive impairment. Anesthesiology 118, 502–515 (2013).

    CAS  PubMed  Google Scholar 

  23. Stratmann, G. et al. Isoflurane differentially affects neurogenesis and long-term neurocognitive function in 60-day-old and 7-day-old rats. Anesthesiology 110, 834–848 (2009).

    CAS  PubMed  Google Scholar 

  24. Ponten, E., Fredriksson, A., Gordh, T., Eriksson, P. & Viberg, H. Neonatal exposure to propofol affects BDNF but not CaMKII, GAP-43, synaptophysin and tau in the neonatal brain and causes an altered behavioural response to diazepam in the adult mouse brain. Behav. Brain Res. 223, 75–80 (2011).

    CAS  PubMed  Google Scholar 

  25. Zhu, C. et al. Isoflurane anesthesia induced persistent, progressive memory impairment, caused a loss of neural stem cells, and reduced neurogenesis in young, but not adult, rodents. J. Cereb. Blood Flow Metab. 30, 1017–1030 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Lee, B. H., Chan, J. T., Kraeva, E., Peterson, K. & Sall, J. W. Isoflurane exposure in newborn rats induces long-term cognitive dysfunction in males but not females. Neuropharmacology 83, 9–17 (2014). This publication convincingly shows that the neurocognitive effect of early-life anaesthesia exposure is sex dependent and that the extent of anaesthesia-induced neuroapoptosis does not determine cognitive function.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ikonomidou, C. et al. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science 283, 70–74 (1999).

    CAS  PubMed  Google Scholar 

  28. Ikonomidou, C. et al. Ethanol-induced apoptotic neurodegeneration and fetal alcohol syndrome. Science 287, 1056–1060 (2000). This work can be considered as the one that provided the biological rational to evaluate the effect of anaesthesia on the developing brain.

    CAS  PubMed  Google Scholar 

  29. Gentry, K. R., Steele, L. M., Sedensky, M. M. & Morgan, P. G. Early developmental exposure to volatile anesthetics causes behavioral defects in Caenorhabditis elegans. Anesth. Analg. 116, 185–189 (2013).

    CAS  PubMed  Google Scholar 

  30. Brambrink, A. M. et al. Isoflurane-induced neuroapoptosis in the neonatal rhesus macaque brain. Anesthesiology 112, 834–841 (2010).

    CAS  PubMed  Google Scholar 

  31. Lu, L. X., Yon, J. H., Carter, L. B. & Jevtovic-Todorovic, V. General anesthesia activates BDNF-dependent neuroapoptosis in the developing rat brain. Apoptosis 11, 1603–1615 (2006). This publication provides valuable insights into the molecular mechanisms underlying anaesthesia-induced neuroapoptosis.

    CAS  PubMed  Google Scholar 

  32. Brambrink, A. M. et al. Isoflurane-induced apoptosis of oligodendrocytes in the neonatal primate brain. Ann. Neurol. 72, 525–535 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Istaphanous, G. K. et al. Characterization and quantification of isoflurane-induced developmental apoptotic cell death in mouse cerebral cortex. Anesth. Analg. 116, 845–854 (2013).

    CAS  PubMed  Google Scholar 

  34. Creeley, C. et al. Propofol-induced apoptosis of neurones and oligodendrocytes in fetal and neonatal rhesus macaque brain. Br. J. Anaesth. 110 (Suppl. 1), i29–i38 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Young, C. et al. Potential of ketamine and midazolam, individually or in combination, to induce apoptotic neurodegeneration in the infant mouse brain. Br. J. Pharmacol. 146, 189–197 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Istaphanous, G. K. et al. Comparison of the neuroapoptotic properties of equipotent anesthetic concentrations of desflurane, isoflurane, or sevoflurane in neonatal mice. Anesthesiology 114, 578–587 (2011).

    CAS  PubMed  Google Scholar 

  37. Osterop, S. F. et al. Developmental stage-dependent impact of midazolam on calbindin, calretinin and parvalbumin expression in the immature rat medial prefrontal cortex during the brain growth spurt. Int. J. Dev. Neurosci. 45, 19–28 (2015).

    CAS  PubMed  Google Scholar 

  38. Ma, D., Wilhelm, S., Maze, M. & Franks, N. P. Neuroprotective and neurotoxic properties of the 'inert' gas, xenon. Br. J. Anaesth. 89, 739–746 (2002).

    CAS  PubMed  Google Scholar 

  39. Massa, H., Lacoh, C. & Vutskits, L. Effects of morphine on the differentiation and survival of developing pyramidal neurons during the brain growth spurt. Toxicol. Sci. 130, 168–179 (2012).

    CAS  PubMed  Google Scholar 

  40. Deng, W., Aimone, J. B. & Gage, F. H. New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat. Rev. Neurosci. 11, 339–350 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Tashiro, A., Sandler, V. M., Toni, N., Zhao, C. & Gage, F. H. NMDA-receptor-mediated, cell-specific integration of new neurons in adult dentate gyrus. Nature 442, 929–933 (2006).

    CAS  PubMed  Google Scholar 

  42. Ge, S. et al. GABA regulates synaptic integration of newly generated neurons in the adult brain. Nature 439, 589–593 (2006).

    CAS  PubMed  Google Scholar 

  43. Stratmann, G. et al. Isoflurane does not affect brain cell death, hippocampal neurogenesis, or long-term neurocognitive outcome in aged rats. Anesthesiology 112, 305–315 (2010).

    CAS  PubMed  Google Scholar 

  44. Krzisch, M. et al. Propofol anesthesia impairs the maturation and survival of adult-born hippocampal neurons. Anesthesiology 118, 602–610 (2013).

    CAS  PubMed  Google Scholar 

  45. Hofacer, R. D. et al. Cell age-specific vulnerability of neurons to anesthetic toxicity. Ann. Neurol. 73, 695–704 (2013). This paper and reference 44 demonstrate that the neuronal vulnerability to anaesthetics depends on the developmental stage of individual neurons.

    PubMed  PubMed Central  Google Scholar 

  46. De Felipe, J., Marco, P., Fairen, A. & Jones, E. G. Inhibitory synaptogenesis in mouse somatosensory cortex. Cereb. Cortex 7, 619–634 (1997).

    CAS  PubMed  Google Scholar 

  47. Briner, A. et al. Developmental stage-dependent persistent impact of propofol anesthesia on dendritic spines in the rat medial prefrontal cortex. Anesthesiology 115, 282–293 (2011). This work shows that the administration of GAs in the early postnatal period can induce lasting changes in the number of dendritic spines and synapses, and demonstrates that the direction of these changes depends on the developmental stage at drug exposure.

    CAS  PubMed  Google Scholar 

  48. Head, B. P. et al. Inhibition of p75 neurotrophin receptor attenuates isoflurane-mediated neuronal apoptosis in the neonatal central nervous system. Anesthesiology 110, 813–825 (2009). This publication describes a molecular pathway linking anaesthesia exposure to synapse loss and cell death.

    CAS  PubMed  Google Scholar 

  49. Lunardi, N., Ori, C., Erisir, A. & Jevtovic-Todorovic, V. General anesthesia causes long-lasting disturbances in the ultrastructural properties of developing synapses in young rats. Neurotox. Res. 17, 179–188 (2010).

    CAS  PubMed  Google Scholar 

  50. Lunardi, N., Oklopcic, A., Prillaman, M., Erisir, A. & Jevtovic-Todorovic, V. Early exposure to general anesthesia disrupts spatial organization of presynaptic vesicles in nerve terminals of the developing rat subiculum. Mol. Neurobiol. 52, 942–951 (2015).

    CAS  PubMed  Google Scholar 

  51. Amrock, L. G., Starner, M. L., Murphy, K. L. & Baxter, M. G. Long-term effects of single or multiple neonatal sevoflurane exposures on rat hippocampal ultrastructure. Anesthesiology 122, 87–95 (2015).

    CAS  PubMed  Google Scholar 

  52. DiGruccio, M. R. et al. Hyperexcitability of rat thalamocortical networks after exposure to general anesthesia during brain development. J. Neurosci. 35, 1481–1492 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Joksovic, P. M., Lunardi, N., Jevtovic-Todorovic, V. & Todorovic, S. M. Early exposure to general anesthesia with isoflurane downregulates inhibitory synaptic neurotransmission in the rat thalamus. Mol. Neurobiol. 52, 952–958 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Briner, A. et al. Volatile anesthetics rapidly increase dendritic spine density in the rat medial prefrontal cortex during synaptogenesis. Anesthesiology 112, 546–556 (2010).

    PubMed  Google Scholar 

  55. De Roo, M. et al. Anesthetics rapidly promote synaptogenesis during a critical period of brain development. PLoS ONE 4, e7043 (2009). This paper describes dendritic spine dynamics upon anaesthesia exposure.

    PubMed  PubMed Central  Google Scholar 

  56. Vutskits, L. General anesthesia: a gateway to modulate synapse formation and neural plasticity? Anesth. Analg. 115, 1174–1182 (2012).

    CAS  PubMed  Google Scholar 

  57. Mintz, C. D., Barrett, K. M., Smith, S. C., Benson, D. L. & Harrison, N. L. Anesthetics interfere with axon guidance in developing mouse neocortical neurons in vitro via a γ-aminobutyric acid type A receptor mechanism. Anesthesiology 118, 825–833 (2013).

    CAS  PubMed  Google Scholar 

  58. Rudolph, U. & Antkowiak, B. Molecular and neuronal substrates for general anaesthetics. Nat. Rev. Neurosci. 5, 709–720 (2004).

    CAS  PubMed  Google Scholar 

  59. Yon, J. H., Daniel-Johnson, J., Carter, L. B. & Jevtovic-Todorovic, V. Anesthesia induces neuronal cell death in the developing rat brain via the intrinsic and extrinsic apoptotic pathways. Neuroscience 135, 815–827 (2005).

    CAS  PubMed  Google Scholar 

  60. Popic, J. et al. Propofol-induced changes in neurotrophic signaling in the developing nervous system in vivo. PLoS ONE 7, e34396 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Lemkuil, B. P. et al. Isoflurane neurotoxicity is mediated by p75NTR-RhoA activation and actin depolymerization. Anesthesiology 114, 49–57 (2011).

    CAS  PubMed  Google Scholar 

  62. Pearn, M. L. et al. Propofol neurotoxicity is mediated by p75 neurotrophin receptor activation. Anesthesiology 116, 352–361 (2012).

    CAS  PubMed  Google Scholar 

  63. Sanchez, V. et al. General anesthesia causes long-term impairment of mitochondrial morphogenesis and synaptic transmission in developing rat brain. Anesthesiology 115, 992–1002 (2011).

    PubMed  Google Scholar 

  64. Boscolo, A. et al. Early exposure to general anesthesia disturbs mitochondrial fission and fusion in the developing rat brain. Anesthesiology 118, 1086–1097 (2013). This publication describes anaesthesia exposure-induced mitochondrial dysfunction as a plausible cause of anaesthesia neurotoxicity.

    CAS  PubMed  Google Scholar 

  65. Boscolo, A. et al. The abolishment of anesthesia-induced cognitive impairment by timely protection of mitochondria in the developing rat brain: the importance of free oxygen radicals and mitochondrial integrity. Neurobiol. Dis. 45, 1031–1041 (2012).

    CAS  PubMed  Google Scholar 

  66. Kargaran, P. et al. Impact of propofol anaesthesia on cytokine expression profiles in the developing rat brain: a randomised placebo-controlled experimental in-vivo study. Eur. J. Anaesthesiol. 32, 336–345 (2015).

    CAS  PubMed  Google Scholar 

  67. Zhang, L. et al. The potential dual effects of sevoflurane on AKT/GSK3β signaling pathway. Med. Gas Res. 4, 5 (2014).

    PubMed  PubMed Central  Google Scholar 

  68. Takesian, A. E. & Hensch, T. K. Balancing plasticity/stability across brain development. Prog. Brain Res. 207, 3–34 (2013).

    PubMed  Google Scholar 

  69. Tao, G. et al. Sevoflurane induces tau phosphorylation and glycogen synthase kinase 3β activation in young mice. Anesthesiology 121, 510–527 (2014).

    CAS  PubMed  Google Scholar 

  70. Eckenhoff, R. G. & Laudansky, K. F. Anesthesia, surgery, illness and Alzheimer's disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 47, 162–166 (2012).

    PubMed  Google Scholar 

  71. Silverstein, J. H. Influence of anesthetics on Alzheimer's disease: biophysical, animal model, and clinical reports. J. Alzheimers Dis. 40, 839–848 (2014).

    CAS  PubMed  Google Scholar 

  72. Inouye, S. K., Westendorp, R. G. & Saczynski, J. S. Delirium in elderly people. Lancet 383, 911–922 (2014).

    PubMed  Google Scholar 

  73. Culley, D. J., Baxter, M., Yukhananov, R. & Crosby, G. The memory effects of general anesthesia persist for weeks in young and aged rats. Anesth. Analg. 96, 1004–1009 (2003).

    CAS  PubMed  Google Scholar 

  74. Culley, D. J., Baxter, M. G., Yukhananov, R. & Crosby, G. Long-term impairment of acquisition of a spatial memory task following isoflurane-nitrous oxide anesthesia in rats. Anesthesiology 100, 309–314 (2004).

    CAS  PubMed  Google Scholar 

  75. Callaway, J. K., Jones, N. C., Royse, A. G. & Royse, C. F. Sevoflurane anesthesia does not impair acquisition learning or memory in the Morris water maze in young adult and aged rats. Anesthesiology 117, 1091–1101 (2012).

    CAS  PubMed  Google Scholar 

  76. Li, C., Liu, S., Xing, Y. & Tao, F. The role of hippocampal tau protein phosphorylation in isoflurane-induced cognitive dysfunction in transgenic APP695 mice. Anesth. Analg. 119, 413–419 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Le Freche, H. et al. Tau phosphorylation and sevoflurane anesthesia: an association to postoperative cognitive impairment. Anesthesiology 116, 779–787 (2012).

    CAS  PubMed  Google Scholar 

  78. Callaway, J. K., Jones, N. C., Royse, A. G. & Royse, C. F. Memory impairment in rats after desflurane anesthesia is age and dose dependent. J. Alzheimers Dis. 44, 995–1005 (2015).

    CAS  PubMed  Google Scholar 

  79. Lee, I. H. et al. Spatial memory is intact in aged rats after propofol anesthesia. Anesth. Analg. 107, 1211–1215 (2008).

    PubMed  Google Scholar 

  80. Xie, G., Zhang, W., Chang, Y. & Chu, Q. Relationship between perioperative inflammatory response and postoperative cognitive dysfunction in the elderly. Med. Hypotheses 73, 402–403 (2009).

    PubMed  Google Scholar 

  81. Baranov, D. et al. Consensus statement: first international workshop on anesthetics and Alzheimer's disease. Anesth. Analg. 108, 1627–1630 (2009).

    PubMed  PubMed Central  Google Scholar 

  82. Hsiao, K. et al. Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice. Science 274, 99–102 (1996).

    CAS  PubMed  Google Scholar 

  83. Bianchi, S. L. et al. Brain and behavior changes in 12-month-old Tg2576 and nontransgenic mice exposed to anesthetics. Neurobiol. Aging 29, 1002–1010 (2008).

    CAS  PubMed  Google Scholar 

  84. Perucho, J. et al. Anesthesia with isoflurane increases amyloid pathology in mice models of Alzheimer's disease. J. Alzheimers Dis. 19, 1245–1257 (2010).

    CAS  PubMed  Google Scholar 

  85. Games, D. et al. Alzheimer-type neuropathology in transgenic mice overexpressing V717F β-amyloid precursor protein. Nature 373, 523–527 (1995).

    CAS  PubMed  Google Scholar 

  86. Sturchler-Pierrat, C. et al. Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proc. Natl Acad. Sci. USA 94, 13287–13292 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Eckel, B. et al. Effects of isoflurane-induced anaesthesia on cognitive performance in a mouse model of Alzheimer's disease: a randomised trial in transgenic APP23 mice. Eur. J. Anaesthesiol. 30, 605–611 (2013).

    CAS  PubMed  Google Scholar 

  88. Oddo, S. et al. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron 39, 409–421 (2003).

    CAS  PubMed  Google Scholar 

  89. Tang, J. X. & Eckenhoff, M. F. Anesthetic effects in Alzheimer transgenic mouse models. Prog. Neuropsychopharmacol. Biol. Psychiatry 47, 167–171 (2012).

    PubMed  Google Scholar 

  90. Xie, Z. et al. The common inhalation anesthetic isoflurane induces caspase activation and increases amyloid β-protein level in vivo. Ann. Neurol. 64, 618–627 (2008). This is the first publication to show that anaesthesia exposure in vivo can induce pathological features of AD in experimental animals.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Valentim, A. M. et al. Lower isoflurane concentration affects spatial learning and neurodegeneration in adult mice compared with higher concentrations. Anesthesiology 113, 1099–1108 (2010).

    CAS  PubMed  Google Scholar 

  92. Lin, D. & Zuo, Z. Isoflurane induces hippocampal cell injury and cognitive impairments in adult rats. Neuropharmacology 61, 1354–1359 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Eckenhoff, R. G. et al. Inhaled anesthetic enhancement of amyloid-β oligomerization and cytotoxicity. Anesthesiology 101, 703–709 (2004). This in vitro work is the first to demonstrate that anaesthetics can increase pathological events that are associated with AD.

    CAS  PubMed  Google Scholar 

  94. Xie, Z. et al. The common inhalation anesthetic isoflurane induces apoptosis and increases amyloid β protein levels. Anesthesiology 104, 988–994 (2006).

    CAS  PubMed  Google Scholar 

  95. Xie, Z. et al. The inhalation anesthetic isoflurane induces a vicious cycle of apoptosis and amyloid β-protein accumulation. J. Neurosci. 27, 1247–1254 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhen, Y. et al. Nitrous oxide plus isoflurane induces apoptosis and increases β-amyloid protein levels. Anesthesiology 111, 741–752 (2009).

    CAS  PubMed  Google Scholar 

  97. Dong, Y. et al. The common inhalational anesthetic sevoflurane induces apoptosis and increases β-amyloid protein levels. Arch. Neurol. 66, 620–631 (2009).

    PubMed  PubMed Central  Google Scholar 

  98. Tanzi, R. E. & Bertram, L. Twenty years of the Alzheimer's disease amyloid hypothesis: a genetic perspective. Cell 120, 545–555 (2005).

    CAS  PubMed  Google Scholar 

  99. Planel, E. et al. Anesthesia leads to tau hyperphosphorylation through inhibition of phosphatase activity by hypothermia. J. Neurosci. 27, 3090–3097 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Planel, E. et al. Anesthesia-induced hyperphosphorylation detaches 3-repeat tau from microtubules without affecting their stability in vivo. J. Neurosci. 28, 12798–12807 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Planel, E. et al. Acceleration and persistence of neurofibrillary pathology in a mouse model of tauopathy following anesthesia. FASEB J. 23, 2595–2604 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Whittington, R. A. et al. Propofol directly increases tau phosphorylation. PLoS ONE 6, e16648 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Whittington, R. A. et al. Dexmedetomidine increases tau phosphorylation under normothermic conditions in vivo and in vitro. Neurobiol. Aging 36, 2414–2428 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Tang, J. X. et al. Anesthesia in presymptomatic Alzheimer's disease: a study using the triple-transgenic mouse model. Alzheimers Dement. 7, 521–531.e1 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Dong, Y., Wu, X., Xu, Z., Zhang, Y. & Xie, Z. Anesthetic isoflurane increases phosphorylated tau levels mediated by caspase activation and Aβ generation. PLoS ONE 7, e39386 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Palotás, M. et al. Coronary artery bypass surgery provokes Alzheimer's disease-like changes in the cerebrospinal fluid. J. Alzheimers Dis. 21, 1153–1164 (2010).

    PubMed  Google Scholar 

  107. Tang, J. X. et al. Human Alzheimer and inflammation biomarkers after anesthesia and surgery. Anesthesiology 115, 727–732 (2011).

    CAS  PubMed  Google Scholar 

  108. Zhang, B. et al. Effects of anesthetic isoflurane and desflurane on human cerebrospinal fluid Aβ and τ level. Anesthesiology 119, 52–60 (2013).

    CAS  PubMed  Google Scholar 

  109. Wu, X. et al. The inhalation anesthetic isoflurane increases levels of proinflammatory TNF-α, IL-6, and IL-1β. Neurobiol. Aging 33, 1364–1378 (2012). This publication shows that the exposure of ageing animals to anaesthetics, without surgery, can increase the level of pro-inflammatory cytokines in the brain.

    CAS  PubMed  Google Scholar 

  110. Cao, L., Li, L., Lin, D. & Zuo, Z. Isoflurane induces learning impairment that is mediated by interleukin 1β in rodents. PLoS ONE 7, e51431 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Li, Z. Q. et al. Activation of the canonical nuclear factor-κB pathway is involved in isoflurane-induced hippocampal interleukin-1β elevation and the resultant cognitive deficits in aged rats. Biochem. Biophys. Res. Commun. 438, 628–634 (2013).

    CAS  PubMed  Google Scholar 

  112. Ye, X., Lian, Q., Eckenhoff, M. F., Eckenhoff, R. G. & Pan, J. Z. Differential general anesthetic effects on microglial cytokine expression. PLoS ONE 8, e52887 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Tian, Y., Guo, S., Guo, Y. & Jian, L. Anesthetic propofol attenuates apoptosis, Aβ accumulation, and inflammation induced by sevoflurane through NF-κB pathway in human neuroglioma cells. Cell. Mol. Neurobiol. 35, 891–898 (2015).

    CAS  PubMed  Google Scholar 

  114. Avramescu, S. et al. Inflammation increases neuronal sensitivity to general anesthetics. Anesthesiology 124, 417–427 (2016).

    CAS  PubMed  Google Scholar 

  115. Hemmings, H. C. et al. Emerging molecular mechanisms of general anesthetic action. Trends Pharmacol. Sci. 26, 503–510 (2005).

    CAS  PubMed  Google Scholar 

  116. Cheng, V. Y. et al. α5GABAA receptors mediate the amnestic but not sedative-hypnotic effects of the general anesthetic etomidate. J. Neurosci. 26, 3713–3720 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Saab, B. J. et al. Short-term memory impairment after isoflurane in mice is prevented by the α5 γ-aminobutyric acid type A receptor inverse agonist L-655,708. Anesthesiology 113, 1061–1071 (2010).

    CAS  PubMed  Google Scholar 

  118. Zurek, A. A. et al. Sustained increase in α5GABAA receptor function impairs memory after anesthesia. J. Clin. Invest. 124, 5437–5441 (2014). This study demonstrates the role of α5GABA A R signalling in the pathogenesis of cognitive deficits.

    PubMed  PubMed Central  Google Scholar 

  119. Davidson, A. J. et al. Anesthesia and the developing brain: a way forward for clinical research. Paediatr. Anaesth. 25, 447–452 (2015).

    PubMed  Google Scholar 

  120. Vutskits, L. General anesthetics in brain injury: friends or foes. Curr. Pharm. Des. 20, 4203–4210 (2014).

    CAS  PubMed  Google Scholar 

  121. Cheng, B., Zhang, Y., Wang, A., Dong, Y. & Xie, Z. Vitamin C attenuates isoflurane-induced caspase-3 activation and cognitive impairment. Mol. Neurobiol. 52, 1580–1589 (2015).

    CAS  PubMed  Google Scholar 

  122. Shih, J. et al. Delayed environmental enrichment reverses sevoflurane-induced memory impairment in rats. Anesthesiology 116, 586–602 (2012).

    CAS  PubMed  Google Scholar 

  123. Cibelli, M. et al. Role of interleukin-1β in postoperative cognitive dysfunction. Ann. Neurol. 68, 360–368 (2010). This seminal work demonstrates a role for systemic inflammation in the pathogenesis of postoperative cognitive dysfunction.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Terrando, N. et al. Tumor necrosis factor-α triggers a cytokine cascade yielding postoperative cognitive decline. Proc. Natl Acad. Sci. USA 107, 20518–20522 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Terrando, N. et al. Resolving postoperative neuroinflammation and cognitive decline. Ann. Neurol. 70, 986–995 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Shu, Y. et al. Nociceptive stimuli enhance anesthetic-induced neuroapoptosis in the rat developing brain. Neurobiol. Dis. 45, 743–750 (2012).

    CAS  PubMed  Google Scholar 

  127. Anand, K. J. et al. Ketamine reduces the cell death following inflammatory pain in newborn rat brain. Pediatr. Res. 62, 283–290 (2007).

    CAS  PubMed  Google Scholar 

  128. Zhang, X. et al. Surgical incision-induced nociception causes cognitive impairment and reduction in synaptic NMDA receptor 2B in mice. J. Neurosci. 33, 17737–17748 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Berger, M. et al. Postoperative cognitive dysfunction: minding the gaps in our knowledge of a common postoperative complication in the elderly. Anesthesiol. Clin. 33, 517–550 (2015).

    PubMed  PubMed Central  Google Scholar 

  130. Liimatainen, J. et al. Improved cognitive flexibility after aortic valve replacement surgery. Interact. Cardiovasc. Thorac. Surg. http://dx.doi.org/10.1093/icvts/ivw170 (2016).

  131. Wilder, R. T. et al. Early exposure to anesthesia and learning disabilities in a population-based birth cohort. Anesthesiology 110, 796–804 (2009).

    PubMed  Google Scholar 

  132. Flick, R. P. et al. Cognitive and behavioral outcomes after early exposure to anesthesia and surgery. Pediatrics 128, e1053–e1061 (2011).

    PubMed  PubMed Central  Google Scholar 

  133. DiMaggio, C., Sun, L. S., Kakavouli, A., Byrne, M. W. & Li, G. A retrospective cohort study of the association of anesthesia and hernia repair surgery with behavioral and developmental disorders in young children. J. Neurosurg. Anesthesiol. 21, 286–291 (2009).

    PubMed  PubMed Central  Google Scholar 

  134. DiMaggio, C., Sun, L. S. & Li, G. Early childhood exposure to anesthesia and risk of developmental and behavioral disorders in a sibling birth cohort. Anesth. Analg. 113, 1143–1151 (2011).

    PubMed  PubMed Central  Google Scholar 

  135. Ing, C. et al. Long-term differences in language and cognitive function after childhood exposure to anesthesia. Pediatrics 130, e476–e485 (2012).

    PubMed  Google Scholar 

  136. Bartels, M., Althoff, R. R. & Boomsma, D. I. Anesthesia and cognitive performance in children: no evidence for a causal relationship. Twin Res. Hum. Genet. 12, 246–253 (2009).

    PubMed  Google Scholar 

  137. Hansen, T. G. et al. Academic performance in adolescence after inguinal hernia repair in infancy: a nationwide cohort study. Anesthesiology 114, 1076–1085 (2011).

    PubMed  Google Scholar 

  138. McCann, M. E. et al. Infantile postoperative encephalopathy: perioperative factors as a cause for concern. Pediatrics 133, e751–e757 (2014).

    PubMed  Google Scholar 

  139. Vutskits, L., Davis, P. J., Hansen, T. G. & Davidson, A. Anesthetics and the developing brain: time for a change in practice? A pro/con debate. Paediatr. Anaesth. 22, 973–980 (2012).

    PubMed  Google Scholar 

  140. Gleich, S. J. et al. Neurodevelopment of children exposed to anesthesia: design of the Mayo Anesthesia Safety in Kids (MASK) study. Contemp. Clin. Trials 41, 45–54 (2015).

    PubMed  Google Scholar 

  141. Ing, C. H. et al. Comparative analysis of outcome measures used in examining neurodevelopmental effects of early childhood anesthesia exposure. Anesthesiology 120, 1319–1332 (2014).

    CAS  PubMed  Google Scholar 

  142. Sun, L. S. et al. Feasibility and pilot study of the Pediatric Anesthesia NeuroDevelopment Assessment (PANDA) project. J. Neurosurg. Anesthesiol. 24, 382–388 (2012).

    PubMed  PubMed Central  Google Scholar 

  143. Davidson, A. J. et al. Neurodevelopmental outcome at 2 years of age after general anaesthesia and awake-regional anaesthesia in infancy (GAS): an international multicentre, randomised controlled trial. Lancet 387, 239–250 (2016).

    PubMed  Google Scholar 

  144. Sun, L. S. et al. Association between a single general anesthesia exposure before age 36 months and neurocognitive outcomes in later childhood. JAMA 315, 2312–2320 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Anand, K. J. et al. Can the human neonate mount an endocrine and metabolic response to surgery. J. Pediatr. Surg. 20, 41–48 (1985).

    CAS  PubMed  Google Scholar 

  146. Taddio, A., Katz, J., Ilersich, A. L. & Koren, G. Effect of neonatal circumcision on pain response during subsequent routine vaccination. Lancet 349, 599–603 (1997).

    CAS  PubMed  Google Scholar 

  147. Müller, A. et al. Peri- and postoperative cognitive and consecutive functional problems of elderly patients. Curr. Opin. Crit. Care 22, 406–411 (2016).

    PubMed  Google Scholar 

  148. Phillips-Bute, B. et al. Association of neurocognitive function and quality of life 1 year after coronary artery bypass graft (CABG) surgery. Psychosom. Med. 68, 369–375 (2006).

    PubMed  Google Scholar 

  149. Steinmetz, J., Christensen, K. B., Lund, T., Lohse, N. & Rasmussen, L. S. Long-term consequences of postoperative cognitive dysfunction. Anesthesiology 110, 548–555 (2009).

    PubMed  Google Scholar 

  150. Newman, M. F. et al. Report of the substudy assessing the impact of neurocognitive function on quality of life 5 years after cardiac surgery. Stroke 32, 2874–2881 (2001).

    CAS  PubMed  Google Scholar 

  151. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders 5th edn (American Psychiatric Association, 2013).

  152. Monk, T. G. & Price, C. C. Postoperative cognitive disorders. Curr. Opin. Crit. Care 17, 376–381 (2011).

    PubMed  Google Scholar 

  153. Robinson, T. N. et al. Postoperative delirium in the elderly: risk factors and outcomes. Ann. Surg. 249, 173–178 (2009).

    PubMed  Google Scholar 

  154. Androsova, G., Krause, R., Winterer, G. & Schneider, R. Biomarkers of postoperative delirium and cognitive dysfunction. Front. Aging Neurosci. 7, 112 (2015).

    PubMed  PubMed Central  Google Scholar 

  155. Moller, J. T. et al. Long-term postoperative cognitive dysfunction in the elderly: ISPOCD1 study. Lancet 351, 857–861 (1998).

    CAS  PubMed  Google Scholar 

  156. Abildstrom, H. et al. Cognitive dysfunction 1–2 years after non-cardiac surgery in the elderly. Acta Anaesthesiol. Scand. 44, 1246–1251 (2000).

    CAS  PubMed  Google Scholar 

  157. Avidan, M. S. & Evers, A. S. The fallacy of persistent postoperative cognitive decline. Anesthesiology 124, 255–258 (2016).

    PubMed  Google Scholar 

  158. Liu, L. L. & Leung, J. M. Predicting adverse postoperative outcomes in patients aged 80 years or older. J. Am. Geriatr. Soc. 48, 405–412 (2000).

    CAS  PubMed  Google Scholar 

  159. Monk, T. G. et al. Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology 108, 18–30 (2008).

    PubMed  Google Scholar 

  160. Koch, S. et al. Cerebral fat microembolism and cognitive decline after hip and knee replacement. Stroke 38, 1079–1081 (2007).

    PubMed  Google Scholar 

  161. Puskas, F. et al. Intraoperative hyperglycemia and cognitive decline after CABG. Ann. Thorac. Surg. 84, 1467–1473 (2007).

    PubMed  Google Scholar 

  162. Evered, L., Scott, D. A., Silbert, B. & Maruff, P. Postoperative cognitive dysfunction is independent of type of surgery and anesthetic. Anesth. Analg. 112, 1179–1185 (2011).

    PubMed  Google Scholar 

  163. Williams-Russo, P., Sharrock, N. E., Mattis, S., Szatrowski, T. P. & Charlson, M. E. Cognitive effects after epidural versus general anesthesia in older adults. A randomized trial. JAMA 274, 44–50 (1995).

    CAS  PubMed  Google Scholar 

  164. Rasmussen, L. S. et al. Does anaesthesia cause postoperative cognitive dysfunction? A randomised study of regional versus general anaesthesia in 438 elderly patients. Acta Anaesthesiol. Scand. 47, 260–266 (2003).

    CAS  PubMed  Google Scholar 

  165. Todd, M. M. Anesthetic neurotoxicity: the collision between laboratory neuroscience and clinical medicine. Anesthesiology 101, 272–273 (2004).

    CAS  PubMed  Google Scholar 

  166. Anand, K. J. & Soriano, S. G. Anesthetic agents and the immature brain: are these toxic or therapeutic? Anesthesiology 101, 527–530 (2004).

    CAS  PubMed  Google Scholar 

  167. Soriano, S. G., Anand, K. J., Rovnaghi, C. R. & Hickey, P. R. Of mice and men: should we extrapolate rodent experimental data to the care of human neonates? Anesthesiology 102, 866–868 (2005).

    PubMed  Google Scholar 

  168. Vutskits, L. & Patel, P. Pushing the standards forward: in-depth monitoring of physiological parameters in anesthetized neonatal mice. Anesth. Analg. 119, 1029–1031 (2014).

    PubMed  Google Scholar 

  169. Wu, B. et al. Physiological disturbance may contribute to neurodegeneration induced by isoflurane or sevoflurane in 14 day old rats. PLoS ONE 9, e84622 (2014).

    PubMed  PubMed Central  Google Scholar 

  170. Zhang, B. et al. The inhalation anesthetic desflurane induces caspase activation and increases amyloid β-protein levels under hypoxic conditions. J. Biol. Chem. 283, 11866–11875 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Jevtovic-Todorovic, V. & Olney, J. W. PRO: anesthesia-induced developmental neuroapoptosis: status of the evidence. Anesth. Analg. 106, 1659–1663 (2008).

    PubMed  Google Scholar 

  172. Fitzgerald, M. The development of nociceptive circuits. Nat. Rev. Neurosci. 6, 507–520 (2005).

    CAS  PubMed  Google Scholar 

  173. Clancy, B., Finlay, B. L., Darlington, R. B. & Anand, K. J. Extrapolating brain development from experimental species to humans. Neurotoxicology 28, 931–937 (2007).

    PubMed  Google Scholar 

  174. Workman, A. D., Charvet, C. J., Clancy, B., Darlington, R. B. & Finlay, B. L. Modeling transformations of neurodevelopmental sequences across mammalian species. J. Neurosci. 33, 7368–7383 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Bohnen, N. I., Warner, M. A., Kokmen, E. & Kurland, L. T. Early and midlife exposure to anesthesia and age of onset of Alzheimer's disease. Int. J. Neurosci. 77, 181–185 (1994).

    CAS  PubMed  Google Scholar 

  176. Bohnen, N. I., Warner, M. A., Kokmen, E., Beard, C. M. & Kurland, L. T. Alzheimer's disease and cumulative exposure to anesthesia: a case-control study. J. Am. Geriatr. Soc. 42, 198–201 (1994).

    CAS  PubMed  Google Scholar 

  177. Lee, T. A., Wolozin, B., Weiss, K. B. & Bednar, M. M. Assessment of the emergence of Alzheimer's disease following coronary artery bypass graft surgery or percutaneous transluminal coronary angioplasty. J. Alzheimers Dis. 7, 319–324 (2005).

    PubMed  Google Scholar 

  178. Chen, C. W. et al. Increased risk of dementia in people with previous exposure to general anesthesia: a nationwide population-based case-control study. Alzheimers Dement. 10, 196–204 (2014).

    PubMed  Google Scholar 

  179. Chen, P. L. et al. Risk of dementia after anaesthesia and surgery. Br. J. Psychiatry 204, 188–193 (2014).

    PubMed  PubMed Central  Google Scholar 

  180. Gasparini, M. et al. A case-control study on Alzheimer's disease and exposure to anesthesia. Neurol. Sci. 23, 11–14 (2002).

    CAS  PubMed  Google Scholar 

  181. Avidan, M. S. et al. Long-term cognitive decline in older subjects was not attributable to noncardiac surgery or major illness. Anesthesiology 111, 964–970 (2009).

    PubMed  Google Scholar 

  182. Aiello Bowles, E. J. et al. Anesthesia exposure and risk of dementia and Alzheimer's disease: a prospective study. J. Am. Geriatr. Soc. 64, 602–607 (2016).

    PubMed  PubMed Central  Google Scholar 

  183. Seitz, D. P., Reimer, C. L. & Siddiqui, N. A review of epidemiological evidence for general anesthesia as a risk factor for Alzheimer's disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 47, 122–127 (2013).

    CAS  PubMed  Google Scholar 

  184. Terrando, N., Eriksson, L. I. & Eckenhoff, R. G. Perioperative neurotoxicity in the elderly: summary of the 4th International Workshop. Anesth. Analg. 120, 649–652 (2015).

    PubMed  Google Scholar 

  185. Zhang, Y. et al. Anesthetics isoflurane and desflurane differently affect mitochondrial function, learning, and memory. Ann. Neurol. 71, 687–698 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Zhang, Y. et al. The mitochondrial pathway of anesthetic isoflurane-induced apoptosis. J. Biol. Chem. 285, 4025–4037 (2010).

    CAS  PubMed  Google Scholar 

  187. Wang, H. et al. Isoflurane induces endoplasmic reticulum stress and caspase activation through ryanodine receptors. Br. J. Anaesth. 113, 695–707 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Zhang, G. et al. Isoflurane-induced caspase-3 activation is dependent on cytosolic calcium and can be attenuated by memantine. J. Neurosci. 28, 4551–4560 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Wei, H. et al. The common inhalational anesthetic isoflurane induces apoptosis via activation of inositol 1,4,5-trisphosphate receptors. Anesthesiology 108, 251–260 (2008).

    CAS  PubMed  Google Scholar 

  190. Wei, H. & Xie, Z. Anesthesia, calcium homeostasis and Alzheimer's disease. Curr. Alzheimer Res. 6, 30–35 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Yang, H. et al. Inhalational anesthetics induce cell damage by disruption of intracellular calcium homeostasis with different potencies. Anesthesiology 109, 243–250 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank M. de Roo for helpful discussions during the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Laszlo Vutskits or Zhongcong Xie.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Delirium

A syndrome characterized by an acute disturbance of consciousness that usually occurs in association with impairment in attention and cognitive functions.

Midazolam

A member of the benzodiazepine family of sedative drugs, which act as positive allosteric modulators of type A GABA receptors.

Anaesthetic equipotency

The comparative dose or concentration of anaesthetic drugs that provides equal ability or strength to induce general anaesthesia.

Volatile anaesthetic

A chemical compound that can be volatilized to vapour, which, when inhaled, can induce general anaesthesia.

Brain growth spurt

This term refers to the rapid growth of the developing brain that takes place between the last trimester of pregnancy and the first few years of life in humans and during the first postnatal month in rodents.

Mitochondrial fission–fusion balance

Dynamic fluctuations in the architecture of mitochondria that aim to allow cells to adapt to environmental demands; impairment of these dynamics can result in reduced oxidative phosphorylation, cell death and thereby disease.

Amyloid precursor protein

(APP). A transmembrane protein that is thought to have a central role in the pathogenesis of Alzheimer disease.

Amyloid-β

A peptide of varying length (between 36 and 43 amino acids) that is generated by the proteolytic cleavage of β-amyloid precursor protein and that constitutes the main component of the amyloid plaques that are found in the brains of patients with Alzheimer disease.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vutskits, L., Xie, Z. Lasting impact of general anaesthesia on the brain: mechanisms and relevance. Nat Rev Neurosci 17, 705–717 (2016). https://doi.org/10.1038/nrn.2016.128

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2016.128

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing