Saturday, March 9, 2019

Anesthetic drug & AMPK activator Propofol shown for the first time to excite neurons before loss & return of consciousness: Hypothesis substantiated

Source: By Lt.Cmdr. Jesse Ehrenfeld - https://www.army.mil/article/141145/anesthesiologists_keep_soldiers_safe_in_afghanistan, Public Domain, https://commons.wikimedia.org/w/index.php?curid=71554756

 A recently published paper in December of 2018 in the journal Frontiers in Molecular Neuroscience demonstrated for the first time that the popular anesthetic drug and AMPK activator propofol (the anti-diabetic drug Metformin also acts via AMPK) paradoxically activated neurons in the brain of rats just before loss of consciousness and return of consciousness [3]. These results provide direct support and substantiate my previous hypotheses in which I proposed for the first time that low doses of anesthetic drugs including propofol promote paradoxical excitation in neurons before loss of consciousness and may potentially facilitate restoration of consciousness via cellular stress induction and AMPK activation (i.e. increases in calcium [Ca2+], reactive oxygen species [ROS]), etc.) [1,2].

As seen in the figure below, the authors showed that propofol induced an abrupt increase in Ca2+ levels and activated parabrachial nucleus (PBN) neurons in vivo just before loss of righting reflex (LORR, analogous to loss of consciousness in humans) and recovery of righting reflex (RORR, analogous to return of consciousness in humans) in rats [3]. Such evidence indicates that propofol is excitatory at low doses and promotes paradoxical excitation and possible facilitation of return of consciousness via cellular stress induction (i.e. Ca2+ and/or ROS), after the concentration of propofol decreases in the brain to a low, stimulatory level upon anesthetic removal [1,2].


Source: See Reference #3

During the initial period of general anesthesia, known as induction, a bolus dose of an anesthetic drug is administered that leads to loss of consciousness as evidenced by a lack of response to an oral command [1]. However, an intriguing phenomenon known as paradoxical excitation may also occur after initial administration of an anesthetic drug [1]. When administered at a low dose, nearly every anesthetic induces behavioral signs of neuronal activation such as eccentric body movements and a transient increase in beta activity (13–25 Hz) on the electroencephalogram (EEG) [1]. Consequently, many anesthetics appear to paradoxically excite the brain before inducing unconsciousness [1]. Anesthesiologists and neuroscientists are currently unable to explain how anesthetics are able to induce paradoxical excitation.

However, as noted in the figure above, the increase in Ca2+ levels in PBN neurons just before LORR in rats is analogous to paradoxical excitation in humans demonstrated by several anesthetics, including propofol [4]. PBN neurons have also been shown to play critical role in maintaining consciousness [9].  In addition to behavioral signs of neuronal activation, an increase in EEG beta power has also been observed during paradoxical excitation and just before return of consciousness in humans [4,5]. Because propofol increased Ca2+ levels and activated PBN neurons just before both LORR and RORR, propofol is likely excitatory at low doses and promotes paradoxical excitation and possible facilitation of return of consciousness via cellular stress induction. This notion is in line with the recent findings that lower concentrations of anesthetics are present in the brain during emergence from general anesthesia compared to the initial induction phase [6,7]. Additionally, activation of glutamatergic neurons in the PBN accelerates emergence from sevoflurane anesthesia in mice and lesions in the PBN leads to a coma-like state in rats, indicating that low doses of propofol and other anesthetics may also paradoxically facilitate return of consciousness in disorders of consciousness (e.g. coma) [8,9].

Preconditioning refers to the exposure of a cell or an organism to a mild or sublethal stressor that leads to an adaptive response and protection against a subsequent and potentially lethal application of the same or a similar stressor [1]. Interestingly, propofol and many other anesthetics used clinically act as preconditioning agents at low doses and transient increases in intracellular ROS, Ca2+, and AMPK activation exert preconditioning effects in various cell types (e.g. neurons), suggesting that paradoxical excitation is analogous to both the increase in Ca2+ in PBN neurons just before RORR and preconditioning [10-15].

Anesthetic-induced paradoxical excitation has also been demonstrated in non-mammalian organisms, with exposure of the nematode C. elegans to volatile anesthetics initially resulting in a paradoxical increase in movement, later followed by a progressive lack of coordination, immobility, and ultimately unresponsiveness [16,17]. Loss of neural AMPK (aak-2 in C. elegans) inhibits movement whereas isoflurane acts as a preconditioning agent in C. elegans [18,19]. Additionally, the anesthetic drug diethyl ether was recently shown to induce a “sedation-like” effect in plants, epitomized by a lack of response to a stimulus that normally induces movement in the Venus flytrap [20]. Preliminary data however demonstrated that the production of ROS by cold (i.e. room-temperature) plasma induced activation and trap closing of the Venus flytrap [21], suggesting that a common mechanism of cellular stress-induced AMPK activation crosses species boundaries and underlies the phenomenon of anesthetic-induced paradoxical excitation.

Anesthetics and increases in ROS have also been shown to promote seed germination (analogous to paradoxical excitation) and AMPK (SnRK1 in plants), ROS, and Ca2+ promotes pollen germination and fertilization in Arabidopsis thaliana [22-25]. Also, although they do not have a nervous system, plants produce nearly all neurotransmitters (i.e. glutamate, acetylcholine, histamine, dopamine, serotonin and norepinephrine) that are critical for maintaining consciousness in humans and biotic and abiotic stressors have been well-described to increase the production and activity of these neurotransmitters in plants [26-28]. As several neurotransmitters that play key roles in human consciousness also act as preconditioning agents (i.e. glutamate, acetylcholine, histamine, dopamine, and norepinephrine), a common mechanism of cellular stress-induced AMPK activation by neurotransmitters may have been evolutionarily conserved to promote neuronal activation in the human brain [29-33].

AMPK, known as the master regulator of cellular metabolism, increases lifespan and healthspan in several model organisms, is present throughout the mammalian brain (e.g. neurons of the thalamus, hypothalamus, striatum, hippocampus, and cortex), and is activated by cellular stress (i.e. increases in ROS, Ca2+, AMP/ATP ratio, etc.) [34-37]. AMPK is also activated by nearly all neurotransmitters that play a critical role in maintaining consciousness (glutamate, acetylcholine, histamine, orexin-A, dopamine, and norepinephrine) as well as by several anesthetic drugs that are commonly used to induce and/or maintain loss of consciousness in humans (e.g. propofol, sevoflurane, isoflurane, ketamine, dexmedetomidine, and midazolam) [38-50]. Indeed, propofol has been shown to inhibit mitochondrial electron transport chain function and increase ROS levels in human neuroblastoma SH-SY5Y cells, effects that were enhanced via the addition of the AMPK activator metformin [51]. Metformin also promotes neurogenesis in both the subventricular zone and the dentate gyrus in vitro and in vivo, potentially enhancing brain repair and recovery from disorders of consciousness (e.g. coma) [52-55].

Interestingly, as long-term potentiation (LTP) is considered the cellular correlate for learning and memory, ROS inhibition prevents LTP in vitro and AMPK knockdown severely impairs hippocampal CA1 LTP and blocks long-term memory formation in mice (as I first hypothesized), indicating that cellular stress-induced AMPK activation is not only required for learning and memory, but may also play an indispensable role in the neural correlates of consciousness [56-58]. Indeed, AMPK has recently been shown to improve postoperative cognitive dysfunction in sevoflurane-anesthetized rats [59]. Overexpression of AMPKα1 upregulated the expression of phosphorylated/activated AMPK in the hippocampus of rats anesthetized with sevoflurane and also decreased escape latencies, increased target quadrant swimming times, swimming distances, and platform crossing times during Morris water maze tests. The AMPK inhibitor compound C however abolished AMPKα1-mediated improvement of postoperative cognitive dysfunction [59].

Lastly, as noted in my most recent publication, cellular stress and AMPK activation may link human consciousness with seemingly disparate physiological and pathophysiological phenomena, including aging (metformin and AMPK alleviate accelerated aging), human reproduction (stress and AMPK are critical for oocyte maturation and sperm acrosome reaction), gene regulation (e.g. transposable elements, stress beneficially activates “jumping genes” in human cells), plasma medicine (cold plasma induces beneficial effects in cells by increasing ROS), meditation (meditation increases genes in the AMPK signaling pathway in humans), parabiosis (i.e. young blood, young plasma activates AMPK), planarian regeneration (stress and AMPK play crucial roles in regeneration of worm body parts), and stress-induced CRISPR-Cas activation in bacteria (e.g. gene editing technology, various stressors including nutrient starvation and temperature stress activate CRISPR-Cas systems in bacteria) [60-70].

References
  1.  Finley J. Cellular stress and AMPK links metformin and diverse compounds with accelerated emergence from anesthesia and potential recovery from disorders of consciousness. Med Hypotheses. 2019 Mar;124:42-52.
  2. Finley J. Transposable elements, placental development, and oocyte activation: Cellular stress and AMPK links jumping genes with the creation of human life. Med Hypotheses. 2018 Sep;118:44-54.
  3. Luo T, Yu S, Cai S, et al. Parabrachial neurons promote behavior and electroencephalographic arousal from general anesthesia. Front Mol Neurosci 2018;4(11):420.
  4. McCarthy MM, Brown EN, Kopell N. Potential network mechanisms mediating electroencephalographic beta rhythm changes during propofol-induced paradoxical excitation. J Neurosci 2008;28(50):13488–504.
  5. Gugino LD, Chabot RJ, Prichep LS, John ER, Formanek V, Aglio LS. Quantitative EEG changes associated with loss and return of consciousness in healthy adult volunteers anaesthetized with propofol or sevoflurane. Br J Anaesth 2001;87(3):421–8.
  6. Friedman EB, Sun Y, Moore JT, et al. A conserved behavioral state barrier impedes transitions between anesthetic-induced unconsciousness and wakefulness: evidence for neural inertia. PLoS One 2010;5(7):e11903.
  7. Ferreira AL, Correia R, Vide S, et al. Patterns of hysteresis between induction and emergence of neuroanesthesia are present in spinal and intracranial surgeries. J Neurosurg Anesthesiol. 2018 Oct 26. doi: 10.1097/ANA.0000000000000559. [Epub ahead of print].
  8. Wang TX, Xiong B, Xu W, et al. Activation of parabrachial nucleus glutamatergic neurons accelerates reanimation from sevoflurane anesthesia in mice. Anesthesiology. 2019 Jan;130(1):106-118.
  9. Fuller PM, Sherman D, Pedersen NP, Saper CB, Lu J. Reassessment of the structural basis of the ascending arousal system. J Comp Neurol. 2011 Apr 1;519(5):933-56.
  10. Zhang Y, Chen Z, Feng N, et al. Protective effect of propofol preconditioning on ischemia-reperfusion injury in human hepatocyte. J Thorac Dis 2017;9(3):702–10.
  11. Li L, Saiyin H, Xie J, et al. Sevoflurane preconditioning induced endogenous neurogenesis against ischemic brain injury by promoting microglial activation. Oncotarget 2017;8(17):28544–57.
  12. Wei H, Liang G, Yang H. Isoflurane preconditioning inhibited isoflurane-induced neurotoxicity. Neurosci Lett 2007;425(1):59–62.
  13. De Barros S, Dehez S, Arnaud E, et al. Aging-related decrease of human ASC angiogenic potential is reversed by hypoxia preconditioning through ROS production. Mol Ther 2013;21(2):399–408.
  14. Cain BS, Meldrum DR, Meng X, Shames BD, Banerjee A, Harken AH. Calcium preconditioning in human myocardium. Ann Thorac Surg 1998;65(4):1065–70.
  15. Shen P, Hou S, Zhu M, Zhao M, Ouyang Y, Feng J. Cortical spreading depression preconditioning mediates neuroprotection against ischemic stroke by inducing AMP-activated protein kinase-dependent autophagy in a rat cerebral ischemic/reperfusion injury model. J Neurochem 2017;140(5):799–813.
  16. Morgan PG, Kayser EB, Sedensky MM. C. elegans and volatile anesthetics. WormBook 2007;3:1–11.
  17. Morgan PG, Cascorbi HF. Effect of anesthetics and a convulsant on normal and mutant Caenorhabditis elegans. Anesthesiology 1985;62(6):738–44.
  18. Cunningham KA, Bouagnon AD, Barros AG, et al. Loss of a neural AMP-activated kinase mimics the effects of elevated serotonin on fat, movement, and hormonal secretions. PLoS Genet 2014;10(6):e1004394.
  19. Jia B, Crowder CM. Volatile anesthetic preconditioning present in the invertebrate Caenorhabditis elegans. Anesthesiology 2008;108(3):426–33.
  20. Yokawa K, Kagenishi T, Pavlovic A, et al. Anaesthetics stop diverse plant organ movements, affect endocytic vesicle recycling and ROS homeostasis, and block action potentials in Venus flytraps. Ann Bot. 2018 Nov 3;122(5):747-756.
  21. http://meetings.aps.org/Meeting/GEC18/Session/PR2.9, last accessed 02/17/19.
  22. Taylorson RB, Hendricks SB. Overcoming dormancy in seeds with ethanol and other anesthetics. Planta 1979;145(5):507–10.
  23. Leymarie J, Vitkauskaité G, Hoang HH, et al. Role of reactive oxygen species in the regulation of Arabidopsis seed dormancy. Plant Cell Physiol 2012;53(1):96–106.
  24. Gao XQ, Liu CZ, Li DD, et al. The arabidopsis KINβγ Subunit of the SnRK1 complex regulates pollen hydration on the stigma by mediating the level of reactive oxygen species in pollen. PLoS Genet 2016;12(7):e1006228.
  25. Duan Q, Kita D, Johnson EA, et al. Reactive oxygen species mediate pollen tube rupture to release sperm for fertilization in Arabidopsis. Nat Commun 2014;5:3129.
  26. Roshchina VV. New trends and perspectives in the evolution of neurotransmitters in microbial, plant, and animal cells. Adv Exp Med Biol 2016;874:25–77.
  27. Kulma A, Szopa J. Catecholamines are active compounds in plants. Plant Sci 2007;172(3):433–40.
  28. Toyota M, Spencer D, Sawai-Toyota S, et al. Glutamate triggers long-distance, calcium-based plant defense signaling. Science. 2018 Sep 14;361(6407):1112-1115.
  29. Lin CH, Chen PS, Gean PW. Glutamate preconditioning prevents neuronal death induced by combined oxygen-glucose deprivation in cultured cortical neurons. Eur J Pharmacol 2008;589(1–3):85–93.
  30. Qian YZ, Levasseur JE, Yoshida K, Kukreja RC. KATP channels in rat heart: blockade of ischemic and acetylcholine-mediated preconditioning by glibenclamide. Am J Physiol 1996;271(1 Pt 2):H23–8.
  31. Gupta V, Goyal R, Sharma PL. Preconditioning offers cardioprotection in hyperlipidemic rat hearts: possible role of Dopamine (D2) signaling. BMC Cardiovasc Disord 2015;28(15):77.
  32. Fan YY, Hu WW, Dai HB, et al. Activation of the central histaminergic system is involved in hypoxia-induced stroke tolerance in adult mice. J Cereb Blood Flow Metab 2011;31(1):305–14.
  33. Parikh V, Singh M. Possible role of cardiac mast cells in norepinephrine-induced myocardial preconditioning. Methods Find Exp Clin Pharmacol 1999;21(4):269–74.
  34. Lage R, Diéguez C, Vidal-Puig A, López M. AMPK: a metabolic gauge regulating whole-body energy homeostasis. Trends Mol Med 2008;14(12):539–49.
  35. Salminen A, Kaarniranta K. AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res Rev 2012;11(2):230–41.
  36. Sook SH, Lee HJ, Kim JH, et al. Reactive oxygen species-mediated activation of AMP-activated protein kinase and c-Jun N-terminal kinase plays a critical role in beta-sitosterol-induced apoptosis in multiple myeloma U266 cells. Phytother Res 2014;28(3):387–94.
  37. Culmsee C, Monnig J, Kemp BE, Mattson MP. AMP-activated protein kinase is highly expressed in neurons in the developing rat brain and promotes neuronal survival following glucose deprivation. J Mol Neurosci 2001;17(1):45–58.
  38. Terunuma M, Vargas KJ, Wilkins ME, et al. Prolonged activation of NMDA receptors promotes dephosphorylation and alters postendocytic sorting of GABAB receptors. Proc Natl Acad Sci U S A 2010;107(31):13918–23.
  39. Zhao M, Sun L, Yu XJ, et al. Acetylcholine mediates AMPK-dependent autophagic cytoprotection in H9c2 cells during hypoxia/reoxygenation injury. Cell Physiol Biochem 2013;32(3):601–13.
  40. Wu WN, Wu PF, Zhou J, et al. Orexin-A activates hypothalamic AMP-activated protein kinase signaling through a Ca2+-dependent mechanism involving voltage gated L-type calcium channel. Mol Pharmacol 2013;84(6):876–87.
  41. Thors B, Halldórsson H, Thorgeirsson G. eNOS activation mediated by AMPK after stimulation of endothelial cells with histamine or thrombin is dependent on LKB1. Biochim Biophys Acta 2011;1813(2):322–31.
  42. Hutchinson DS, Chernogubova E, Dallner OS, Cannon B, Bengtsson T. Beta-adrenoceptors, but not alpha-adrenoceptors, stimulate AMP-activated protein kinase in brown adipocytes independently of uncoupling protein-1. Diabetologia 2005;48(11):2386–95.
  43. Bone NB, Liu Z, Pittet JF, Zmijewski JW. Frontline Science: D1 dopaminergic receptor signaling activates the AMPK-bioenergetic pathway in macrophages and alveolar epithelial cells and reduces endotoxin-induced ALI. J Leukoc Biol 2017;101(2):357–65.
  44. Chen X, Li LY, Jiang JL, et al. Propofol elicits autophagy via endoplasmic reticulum stress and calcium exchange in C2C12 myoblast cell line. PLoS One 2018;13(5):e0197934.
  45. Chen X, Li K, Zhao G. Propofol inhibits HeLa cells by impairing autophagic flux via AMP-activated protein kinase (AMPK) activation and endoplasmic reticulum stress regulated by calcium. Med Sci Monit 2018;18(24):2339–49.
  46. Lamberts RR, Onderwater G, Hamdani N, et al. Reactive oxygen species-induced stimulation of 5'AMP-activated protein kinase mediates sevoflurane-induced cardioprotection. Circulation 2009;120(11 Suppl):S10–5.
  47. Rao Z, Pan X, Zhang H, et al. Isoflurane preconditioning alleviated murine liver ischemia and reperfusion injury by restoring AMPK/mTOR-mediated autophagy. Anesth Analg 2017;125(4):1355–63.
  48. Xu SX, Zhou ZQ, Li XM, et al. The activation of adenosine monophosphate-activated protein kinase in rat hippocampus contributes to the rapid antidepressant effect of ketamine. Behav Brain Res 2013;15(253):305–9.
  49. Wang Z, Zhou W, Dong H, Ma X, He Z. Dexmedetomidine pretreatment inhibits cerebral ischemia/reperfusion induced neuroinflammation via activation of AMPK. Mol Med Rep 2018;18(4):3957–64.
  50. Shindo S, Numazawa S, Yoshida T. A physiological role of AMP-activated protein kinase in phenobarbital-mediated constitutive androstane receptor activation and CYP2B induction. Biochem J 2007;401(3):735–41.
  51. Sumi C, Okamoto A, Tanaka H, et al. Propofol induces a metabolic switch to glycolysis and cell death in a mitochondrial electron transport chain-dependent manner. PLoS One 2018;13(2):e0192796.
  52. Fatt M, Hsu K, He L, et al. Metformin acts on two different molecular pathways to enhance adult neural precursor proliferation/self-renewal and differentiation. Stem Cell Rep 2015;5(6):988–95.
  53. Wang J, Gallagher D, DeVito LM, et al. Metformin activates an atypical PKC-CBP pathway to promote neurogenesis and enhance spatial memory formation. Cell Stem Cell 2012;11(1):23–35.
  54. Liu Y, Tang G, Zhang Z, Wang Y, Yang GY. Metformin promotes focal angiogenesis and neurogenesis in mice following middle cerebral artery occlusion. Neurosci Lett 2014;5(579):46–51.
  55. Dadwal P, Mahmud N, Sinai L, et al. Activating endogenous neural precursor cells using metformin leads to neural repair and functional recovery in a model of childhood brain injury. Stem Cell Rep 2015;5(2):166–73.
  56. Finley J. Facilitation of hippocampal long-term potentiation and reactivation of latent HIV-1 via AMPK activation: common mechanism of action linking learning, memory, and the potential eradication of HIV-1. Med Hypotheses 2018;116:61–73.
  57. Klann E. Cell-permeable scavengers of superoxide prevent long-term potentiation in hippocampal area CA1. J Neurophysiol 1998;80(1):452–7.
  58. Marinangeli C, Didier S, Ahmed T, et al. AMP-activated protein kinase is essential for the maintenance of energy levels during synaptic activation. iScience 2018;12(9):1–13.
  59. Yan WJ, Wang DB, Ren DQ, et al. AMPKα1 overexpression improves postoperative cognitive dysfunction in aged rats through AMPK-Sirt1 and autophagy signaling. J Cell Biochem. 2019 Feb 18. doi: 10.1002/jcb.28443. [Epub ahead of print].
  60. Finley J. Alteration of splice site selection in the LMNA gene and inhibition of progerin production via AMPK activation. Med Hypotheses. 2014 Nov;83(5):580-7.
  61. Finley J. Cellular stress and AMPK activation as a common mechanism of action linking the effects of metformin and diverse compounds that alleviate accelerated aging defects in Hutchinson-Gilford progeria syndrome. Med Hypotheses. 2018 Sep;118:151-162.
  62. Finley J. Oocyte activation and latent HIV-1 reactivation: AMPK as a common mechanism of action linking the beginnings of life and the potential eradication of HIV-1. Med Hypotheses. 2016 Aug;93:34-47.
  63. Calle-Guisado V, de Llera AH, Martin-Hidalgo D, et al. AMP-activated kinase in human spermatozoa: identification, intracellular localization, and key function in the regulation of sperm motility. Asian J Androl. 2017 Nov-Dec;19(6):707-714.
  64. Finley J. Transposable elements, placental development, and oocyte activation: Cellular stress and AMPK links jumping genes with the creation of human life. Med Hypotheses. 2018 Sep;118:44-54.
  65. Schmidt A, Bekeschus S. Redox for Repair: Cold Physical Plasmas and Nrf2 Signaling Promoting Wound Healing. Antioxidants (Basel). 2018 Oct 19;7(10). pii: E146.
  66. Bhasin MK, Denninger JW, Huffman JC, et al. Specific Transcriptome Changes Associated with Blood Pressure Reduction in Hypertensive Patients After Relaxation Response Training. J Altern Complement Med. 2018 May;24(5):486-504.
  67. Liu A, Yang J, Hu Q, et al. Young plasma attenuates age-dependent liver ischemia reperfusion injury. FASEB J. 2018 Nov 1:fj201801234R. doi: 10.1096/fj.201801234R. [Epub ahead of print].
  68. Pirotte N, Stevens AS, Fraguas, et al. Reactive Oxygen Species in Planarian Regeneration: An Upstream Necessity for Correct Patterning and Brain Formation. Oxid Med Cell Longev. 2015;2015:392476.
  69. Lei K, Thi-Kim Vu H, Mohan RD, et al. Egf Signaling Directs Neoblast Repopulation by Regulating Asymmetric Cell Division in Planarians. Dev Cell. 2016 Aug 22;38(4):413-29.
  70. Ratner HK, Sampson TR, Weiss DS. I can see CRISPR now, even when phage are gone: a view on alternative CRISPR-Cas functions from the prokaryotic envelope. Curr Opin Infect Dis. 2015 Jun;28(3):267-74.