Intranasal Insulin Prevents Anesthesia-Induced Cognitive Impairment and Chronic Neurobehavioral Changes

Yanxing Chen, Chun-Ling Dai, Zhe Wu, Khalid Iqbal, Fei Liu, Baorong Zhang, Cheng-Xin Gong, Yanxing Chen, Chun-Ling Dai, Zhe Wu, Khalid Iqbal, Fei Liu, Baorong Zhang, Cheng-Xin Gong

Abstract

General anesthesia increases the risk for cognitive impairment post operation, especially in the elderly and vulnerable individuals. Recent animal studies on the impact of anesthesia on postoperative cognitive impairment have provided some valuable insights, but much remains to be understood. Here, by using mice of various ages and conditions, we found that anesthesia with propofol and sevoflurane caused significant deficits in spatial learning and memory, as tested using Morris Water Maze (MWM) 2-6 days after anesthesia exposure, in aged (17-18 months old) wild-type (WT) mice and in adult (7-8 months old) 3xTg-AD mice (a triple transgenic mouse model of Alzheimer's disease (AD)), but not in adult WT mice. Anesthesia resulted in long-term neurobehavioral changes in the fear conditioning task carried out 65 days after exposure to anesthesia in 3xTg-AD mice. Importantly, daily intranasal administration of insulin (1.75 U/mouse/day) for only 3 days prior to anesthesia completely prevented the anesthesia-induced deficits in spatial learning and memory and the long-term neurobehavioral changes tested 65 days after exposure to anesthesia in 3xTg-AD mice. These results indicate that aging and AD-like brain pathology increase the vulnerability to cognitive impairment after anesthesia and that intranasal treatment with insulin can prevent anesthesia-induced cognitive impairment.

Keywords: Alzheimer’s disease; cognitive impairment; general anesthesia; insulin; intranasal administration; postoperative cognitive dysfunction; propofol; sevoflurane.

Figures

Figure 1
Figure 1
Effect of anesthesia on spatial learning and memory. Wild-type (WT) mice of the indicated ages and 7–8-month-old 3xTg-AD mice were trained in Morris Water Maze (MWM) starting 24 h after general anesthesia with propofol and sevoflurane. Control group did not receive anesthesia. (A,G,M) Training curves show the latencies to locate the platform during training sessions for four consecutive days (4 trials/day). (B–F, H–L, N–R) Probe trial was performed 24 h after the last training trial. The first latency to reach the platform location (B,H,N), the number of the platform location crossings (C,I,O), the percent time the mice stayed in the target quadrant (D,J,P), the distance the mice swam in the target quadrant (E,K,Q) and the swim speed (F,L,R) of mice during the 60 s probe tests are shown. Reversal MWM test was performed for the 3xTg-AD mice starting on the next day following the probe trial. The training curves (S), as well as the first latency to reach the platform location (T), the number of the platform location crossings (U), the percent time the mice swam in the target quadrant (V), the distance the mice swam in the target quadrant (W) and the swim speed (X) of the mice during the 60 s probe tests are shown. Data are presented as mean ± SEM (n = 10–15 mice per group).
Figure 2
Figure 2
Effect of intranasal insulin on anesthesia-induced impairment of spatial learning and memory. The 3xTg-AD mice (7–8 months old, female) received daily intranasal insulin or saline for 3 days, followed by anesthesia with propofol/sevoflurane for 3 h. The mice were then trained in the MWM starting on the following day. (A) Training curves show the latency to locate the platform during training sessions for four consecutive days (4 trials/day). (B–D) Probe trial was performed 24 h after the last training session. The number of the platform location crossing (B), the percent time the mice swam in the target quadrant (C) and the distance the mice swam in the target quadrant (D) are shown. Reversal MWM test was performed on the next day following the probe trial. The training curves (E), the number of the platform location crossing (F), the percent time the mice swam in the target quadrant (G), the distance the mice swam in the target quadrant (H) and the swim speed (I) of the mice during the 60 s probe tests are shown. Data are presented as mean ± SEM (n = 12–15 mice per group).
Figure 3
Figure 3
Effect of anesthesia and intranasal insulin on long-term behavior of 3xTg-AD mice. (A) Experimental design. The 3xTg-AD mice (7–8 months old, female) received daily intranasal insulin or saline for 3 days, followed, on the next day, by anesthesia with propofol/sevoflurane for 3 h. The mice were then tested using various behavioral tests at various time periods indicated in the diagram. MWM, Morris water maze; rMWM, reversal MWM; OF, open field; NOR, novel object recognition; FC, fear conditioning; EPM, elevated plus maze. (B) FC test schedule, which was carried out over three consecutive days. (C) Percentage of time the mice froze during the conditioning phase on day 1 of the FC test. (D) Percentage of time the mice froze in different time periods (data with 7 s bout) during the 5-min context test on day 2 of the FC test. (E) Percentage of time the mice froze during the first 3 min (data 7 s bout) in the context test. (F) Percentage of time the mice froze in different time periods (data with 7 s bout) during the cued tone test on day 3 of the FC test. (G) Percentage of time the mice froze (data with 7 s bout) during the first 3 min in the cued tone test results. Data are presented as mean ± SEM (n = 11–13 mice per group).

References

    1. Benedict C., Hallschmid M., Hatke A., Schultes B., Fehm H. L., Born J., et al. . (2004). Intranasal insulin improves memory in humans. Psychoneuroendocrinology 29, 1326–1334. 10.1016/j.psyneuen.2004.04.003
    1. Benedict C., Hallschmid M., Schmitz K., Schultes B., Ratter F., Fehm H. L., et al. . (2007). Intranasal insulin improves memory in humans: superiority of insulin aspart. Neuropsychopharmacology 32, 239–243. 10.1038/sj.npp.1301193
    1. Callaway J. K., Jones N. C., Royse A. G., Royse C. F. (2012). Sevoflurane anesthesia does not impair acquisition learning or memory in the Morris water maze in young adult and aged rats. Anesthesiology 117, 1091–1101. 10.1097/ALN.0b013e31826cb228
    1. Chen Y., Deng Y., Zhang B., Gong C.-X. (2014a). Deregulation of brain insulin signaling in Alzheimer’s disease. Neurosci. Bull. 30, 282–294. 10.1007/s12264-013-1408-x
    1. Chen Y., Run X., Liang Z., Zhao Y., Dai C.-L., Iqbal K., et al. . (2014b). Intranasal insulin prevents anesthesia-induced hyperphosphorylation of tau in 3xTg-AD mice. Front. Aging Neurosci. 6:100. 10.3389/fnagi.2014.00100
    1. Chen Y., Zhao Y., Dai C.-L., Liang Z., Run X., Iqbal K., et al. . (2014c). Intranasal insulin restores insulin signaling, increases synaptic proteins and reduces Aβ level and microglia activation in the brains of 3xTg-AD mice. Exp. Neurol. 261, 610–619. 10.1016/j.expneurol.2014.06.004
    1. Chen C.-W., Lin C.-C., Chen K.-B., Kuo Y.-C., Li C.-Y., Chung C.-J. (2014). Increased risk of dementia in people with previous exposure to general anesthesia: a nationwide population-based case-control study. Alzheimers Dement. 10, 196–204. 10.1016/j.jalz.2013.05.1766
    1. Chen P.-L., Yang C.-W., Tseng Y.-K., Sun W.-Z., Wang J.-L., Wang S.-J., et al. . (2014). Risk of dementia after anaesthesia and surgery. Br. J. Psychiatry 204, 188–193. 10.1192/bjp.bp.112.119610
    1. Chen Y., Zhang J., Zhang B., Gong C.-X. (2016). Targeting insulin signaling for the treatment of Alzheimer’s disease. Curr. Top. Med. Chem. 16, 485–492. 10.2174/1568026615666150813142423
    1. Claxton A., Baker L. D., Hanson A., Trittschuh E. H., Cholerton B., Morgan A., et al. . (2015). Long-acting intranasal insulin detemir improves cognition for adults with mild cognitive impairment or early-stage Alzheimer’s disease dementia. J. Alzheimers Dis. 44, 897–906. 10.3233/JAD-141791
    1. Clinton L. K., Billings L. M., Green K. N., Caccamo A., Ngo J., Oddo S., et al. . (2007). Age-dependent sexual dimorphism in cognition and stress response in the 3xTg-AD mice. Neurobiol. Dis. 28, 76–82. 10.1016/j.nbd.2007.06.013
    1. Craft S., Baker L. D., Montine T. J., Minoshima S., Watson G. S., Claxton A., et al. . (2012). Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch. Neurol. 69, 29–38. 10.1001/archneurol.2011.233
    1. Culley D. J., Baxter M., Yukhananov R., Crosby G. (2003). The memory effects of general anesthesia persist for weeks in young and aged rats. Anesth. Analg. 96, 1004–1009. 10.1213/01.ane.0000052712.67573.12
    1. Culley D. J., Baxter M. G., Yukhananov R., Crosby G. (2004). Long-term impairment of acquisition of a spatial memory task following isoflurane-nitrous oxide anesthesia in rats. Anesthesiology 100, 309–314. 10.1097/00000542-200402000-00020
    1. Dhuria S. V., Hanson L. R., Frey W. H. (2010). Intranasal delivery to the central nervous system: mechanisms and experimental considerations. J. Pharm. Sci. 99, 1654–1673. 10.1002/jps.21924
    1. Fanselow M. S., Poulos A. M. (2005). The neuroscience of mammalian associative learning. Annu. Rev. Psychol. 56, 207–234. 10.1146/annurev.psych.56.091103.070213
    1. Ghasemi R., Haeri A., Dargahi L., Mohamed Z., Ahmadiani A. (2013). Insulin in the brain: sources, localization and functions. Mol. Neurobiol. 47, 145–171. 10.1007/s12035-012-8339-9
    1. Giménez-Llort L., Blázquez G., Cañete T., Johansson B., Oddo S., Tobeña A., et al. . (2007). Modeling behavioral and neuronal symptoms of Alzheimer’s disease in mice: a role for intraneuronal amyloid. Neurosci. Biobehav. Rev. 31, 125–147. 10.1016/j.neubiorev.2006.07.007
    1. Hudson A. E., Hemmings H. C. (2011). Are anaesthetics toxic to the brain? Br. J. Anaesth. 107, 30–37. 10.1093/bja/aer122
    1. Le Freche H., Brouillette J., Fernandez-Gomez F.-J., Patin P., Caillierez R., Zommer N., et al. . (2012). Tau phosphorylation and sevoflurane anesthesiaan association to postoperative cognitive impairment. Anesthesiology 116, 779–787. 10.1097/ALN.0b013e31824be8c7
    1. Lee I. H., Culley D. J., Baxter M. G., Xie Z., Tanzi R. E., Crosby G. (2008). Spatial memory is intact in aged rats after propofol anesthesia. Anesth. Analg. 107, 1211–1215. 10.1213/ane.0b013e31817ee879
    1. Mao Y. F., Guo Z., Zheng T., Jiang Y., Yan Y., Yin X., et al. . (2016). Intranasal insulin alleviates cognitive deficits and amyloid pathology in young adult APPswe/PS1dE9 mice. Aging Cell 15, 893–902. 10.1111/acel.12498
    1. Marks D. R., Tucker K., Cavallin M. A., Mast T. G., Fadool D. A. (2009). Awake intranasal insulin delivery modifies protein complexes and alters memory, anxiety and olfactory behaviors. J. Neurosci. 29, 6734–6751. 10.1523/JNEUROSCI.1350-09.2009
    1. Mastrangelo M. A., Bowers W. J. (2008). Detailed immunohistochemical characterization of temporal and spatial progression of Alzheimer’s disease-related pathologies in male triple-transgenic mice. BMC Neurosci. 9:81. 10.1186/1471-2202-9-81
    1. Mena M. Á., Perucho J., Rubio I., de Yébenes J. G. (2010). Studies in animal models of the effects of anesthetics on behavior, biochemistry and neuronal cell death. J. Alzheimers Dis. 22, 43–48. 10.3233/JAD-2010-100822
    1. Morris R., Garrud P., Rawlins J., O’Keefe J. (1982). Place navigation impaired in rats with hippocampal lesions. Nature 297, 681–683. 10.1038/297681a0
    1. O’Brien H., Mohan H., O’Hare C., Reynolds J. V., Kenny R. A. (2017). Mind over matter? The hidden epidemic of cognitive dysfunction in the older surgical patient. Ann. Surg. 265, 677–691. 10.1097/SLA.0000000000001900
    1. Patel D., Lunn A., Smith A., Lehmann D., Dorrington K. (2016). Cognitive decline in the elderly after surgery and anaesthesia: results from the Oxford Project to Investigate Memory and Ageing (OPTIMA) cohort. Anaesthesia 71, 1144–1152. 10.1111/anae.13571
    1. Perucho J., Rubio I., Casarejos M. J., Gomez A., Rodriguez-Navarro J. A., Solano R. M., et al. . (2010). Anesthesia with isoflurane increases amyloid pathology in mice models of Alzheimer’s disease. J. Alzheimers Dis. 19, 1245–1257. 10.3233/JAD-2010-1318
    1. Planel E., Richter K. E., Nolan C. E., Finley J. E., Liu L., Wen Y., et al. . (2007). Anesthesia leads to tau hyperphosphorylation through inhibition of phosphatase activity by hypothermia. J. Neurosci. 27, 3090–3097. 10.1523/jneurosci.4854-06.2007
    1. Reger M. A., Watson G., Green P. S., Baker L. D., Cholerton B., Fishel M. A., et al. . (2008). Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-β in memory-impaired older adults. J. Alzheimers Dis. 13, 323–331. 10.3233/jad-2008-13309
    1. Run X., Liang Z., Gong C.-X. (2010). Anesthetics and tau protein: animal model studies. J. Alzheimers Dis. 22, 49–55. 10.3233/JAD-2010-100813
    1. Run X., Liang Z., Zhang L., Iqbal K., Grundke-Iqbal I., Gong C.-X. (2009). Anesthesia induces phosphorylation of tau. J. Alzheimers Dis. 16, 619–626. 10.3233/JAD-2009-1003
    1. Sargolini F., Roullet P., Oliverio A., Mele A. (2003). Effects of intra-accumbens focal administrations of glutamate antagonists on object recognition memory in mice. Behav. Brain Res. 138, 153–163. 10.1016/s0166-4328(02)00238-3
    1. Seitz D. P., Shah P. S., Herrmann N., Beyene J., Siddiqui N. (2011). Exposure to general anesthesia and risk of Alzheimer’s disease: a systematic review and meta-analysis. BMC Geriatr. 11:83. 10.1186/1471-2318-11-83
    1. Sprung J., Roberts R. O., Knopman D. S., Olive D. M., Gappa J. L., Sifuentes V. L., et al. . (2016). Association of mild cognitive impairment with exposure to general anesthesia for surgical and nonsurgical procedures: a population-based study. Mayo Clin. Proc. 91, 208–217. 10.1016/j.mayocp.2015.10.023
    1. Tang J. X., Mardini F., Caltagarone B. M., Garrity S. T., Li R. Q., Bianchi S. L., et al. . (2011). Anesthesia in presymptomatic Alzheimer’s disease: a study using the triple-transgenic mouse model. Alzheimers Dement. 7, 521.e1–531.e1. 10.1016/j.jalz.2010.10.003
    1. Whittington R. A., Virág L., Marcouiller F., Papon M.-A., El Khoury N. B., Julien C., et al. . (2011). Propofol directly increases tau phosphorylation. PLoS One 6:e16648. 10.1371/journal.pone.0016648
    1. Xie S., Jin N., Gu J., Shi J., Sun J., Chu D., et al. . (2016). O-GlcNAcylation of protein kinase A catalytic subunits enhances its activity: a mechanism linked to learning and memory deficits in Alzheimer’s disease. Aging Cell 15, 455–464. 10.1111/acel.12449
    1. Yang C.-W., Fuh J.-L. (2015). Exposure to general anesthesia and the risk of dementia. J. Pain Res. 8, 711–718. 10.2147/JPR.S55579
    1. Zhang Y., Dai C.-L., Chen Y., Iqbal K., Liu F., Gong C.-X. (2016). Intranasal insulin prevents anesthesia-induced spatial learning and memory deficit in mice. Sci. Rep. 6:21186. 10.1038/srep21186
    1. Zhu C., Gao J., Karlsson N., Li Q., Zhang Y., Huang Z., et al. . (2010). 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. 10.1038/jcbfm.2009.274

Source: PubMed

3
Předplatit