Cholinergic deficiency hypothesis in delirium: a synthesis of current evidence

Tammy T Hshieh, Tamara G Fong, Edward R Marcantonio, Sharon K Inouye, Tammy T Hshieh, Tamara G Fong, Edward R Marcantonio, Sharon K Inouye

Abstract

Deficits in cholinergic function have been postulated to cause delirium and cognitive decline. This review examines current understanding of the cholinergic deficiency hypothesis in delirium by synthesizing evidence on potential pathophysiological pathways. Acetylcholine synthesis involves various precursors, enzymes, and receptors, and dysfunction in these components can lead to delirium. Insults to the brain, like ischemia and immunological stressors, can precipitously alter acetylcholine levels. Imbalances between cholinergic and other neurotransmitter pathways may result in delirium. Furthermore, genetic, enzymatic, and immunological overlaps exist between delirium and dementia related to the cholinergic pathway. Important areas for future research include identifying biomarkers, determining genetic contributions, and evaluating response to cholinergic drugs in delirium. Understanding how the cholinergic pathway relates to delirium may yield innovative approaches in the diagnosis, prevention, and treatment of this common, costly, and morbid condition.

Figures

Figure 1
Figure 1
Central cholinergic pathways overlap with locations of neuroimaging abnormalities in delirium brain studies. Positron emission tomography (PET) and single photon emission computed tomography (SPECT) studies on patients with delirium from hepatic encephalopathy, cardiotomy, and traumatic brain injury demonstrate abnormal perfusion in the same cortical, subcortical regions as cholinergic pathways. Cholinergic pathways are indicated by black arrows, locations of abnormal neuroimaging are shaded, brain regions involved in attention are textured.
Figure 2
Figure 2
Potential pathophysiological mechanisms for delirium. AD = Alzheimer’s disease; ApoE = apolipoprotein E; βA = β-amyloid; APP = amyloid precursor protein; Ach = acetylcholine; ChAT = choline acetyltransferase; AChE = acetylcholinesterase; IL-1 = interleukin-1; TNF-α = tumor necrosis factor-α; IGF-1 = insulin-like growth factor-1; CoA = coenzyme A; NAD+= nicotinamide adenine dinucleotide; NADP = nicotinamide adenine dinucleotide phosphate; CAC = citric acid cycle; GPCR = G-protein coupled receptor; Ca+2 = calcium; 5-HT = serotonin.
Figure 3
Figure 3
Relationship of stroke, traumatic brain injury, and diverse stressors to delirium. Schematic demonstration of potential pathophysiologic pathways linking stroke, traumatic brain injury and environmental, medical stressors with delirium. Ach = acetylcholine; ChAT = choline acetyltransferase; IGF-1 = insulin-like growth factor 1; CNS = central nervous system. *Represents one potential pathophysiologic pathway to delirium; other mechanisms exist.
Figure 4
Figure 4
Interactions between acetylcholine and other neurotransmitters in delirium. Schematic demonstration of how decreased acetylcholine levels in delirium alter levels of other neurotransmitters.

References

    1. Burns AG, Byrne J. Delirium. J Neurol Neurosurg Psychiatry. 2005;75:362–367.
    1. Inouye SK. Delirium in older persons. N Engl J Med. 2006;354:1157–1165.
    1. Inouye SK, Bogardus ST, Jr, Charpentier PA, et al. A multicomponent intervention to prevent delirium in hospitalized older patients. N Engl J Med. 1999;340:669–676.
    1. Pitkala KH, Laurila JV, Strandberg TE, Tilvis RS. Multicomponent geriatric intervention for elderly inpatients with delirium: a randomized, controlled trial. J Gerontol A Biol Sci Med Sci. 2006;61:176–181.
    1. Bellelli G, Speciale S, Barisione E, Trabucchi M. Delirium subtypes and 1-year mortality among elderly patients discharged from a post-acute rehabilitation facility. J Gerontol A Biol Sci Med Sci. 2007;62:1182–1183.
    1. Inouye SK, Ferrucci L. Elucidating the pathophysiology of delirium and the interrelationship of delirium and dementia. J Gerontol A Biol Sci Med Sci. 2006;61:1277–1280.
    1. Gottlieb GL, Johnson J, Wanich C, Sullivan E. Delirium in the medically ill elderly: operationalizing the DSM-III criteria. Int Psychogeriatr. 1991;3:181–196.
    1. Alsop DC, Fearing MA, Johnson K, Sperling R, Fong TG, Inouye SK. The role of neuroimaging in elucidating delirium pathophysiology. J Gerontol A Biol Sci Med Sci. 2006;61:1287–1293.
    1. Fong TG, Bogardus ST, Jr, Daftary A, et al. Cerebral perfusion changes in older delirious patients using 99mTc HMPAO SPECT. J Gerontol A Biol Sci Med Sci. 2006;61:1294–1299.
    1. Bartus RT, Dean RL, 3rd, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217:408–414.
    1. Tune LE, Damlouji NF, Holland A, Gardner TJ, Folstein MF, Coyle JT. Association of postoperative delirium with raised serum levels of anticholinergic drugs. Lancet. 1981;2:651–653.
    1. Han L, McCusker J, Cole M, Abrahamowicz M, Primeau F, Elie M. Use of medications with anticholinergic effect predicts clinical severity of delirium symptoms in older medical inpatients. Arch Intern Med. 2001;161:1099–1105.
    1. Flacker JM, Cummings V, Mach JR, Jr, Bettin K, Kiely DK, Wei J. The association of serum anticholinergic activity with delirium in elderly medical patients. Am J Geriatr Psychiatry. 1998;6:31–41.
    1. Cummings JL, Gorman DG, Shapira J. Physostigmine ameliorates the delusions of Alzheimer’s disease. Biol Psychiatry. 1993;33:536–541.
    1. Blass JP, Gibson GE. Cerebrometabolic aspects of delirium in relationship to dementia. Dement Geriatr Cogn Disord. 1999;10:335–338.
    1. Ghajar JB, Gibson GE, Duffy TE. Regional acetylcholine metabolism in brain during acute hypoglycemia and recovery. J Neurochem. 1985;44:94–98.
    1. Nakagawasai O. Behavioral and neurochemical alterations following thiamine deficiency in rodents: relationship to functions of cholinergic neurons. Yakugaku Zasshi. 2005;125:549–554.
    1. Pratico C, Quattrone D, Lucanto T, et al. Drugs of anesthesia acting on central cholinergic system may cause post-operative cognitive dysfunction and delirium. Med Hypotheses. 2005;65:972–982.
    1. Coffman JA, Dilsaver SC. Cholinergic mechanisms in delirium. Am J Psychiatry. 1988;145:382–383.
    1. Sala C, Rudolph-Correia S, Sheng M. Developmentally regulated NMDA receptor-dependent dephosphorylation of cAMP response element-binding protein (CREB) in hippocampal neurons. J Neurosci. 2000;20:3529–3536.
    1. Arendt T. Impairment in memory function and neurodegenerative changes in the cholinergic basal forebrain system induced by chronic intake of ethanol. J Neural Transm Suppl. 1994;44:173–187.
    1. Trzepacz P, editor. The Neuropathophysiology of Delirium. Oxford: Oxford University Press; 2002.
    1. Dixon CE, Bao J, Bergmann JS, Johnson KM. Traumatic brain injury reduces hippocampal high-affinity [3H]choline uptake but not extra-cellular choline levels in rats. Neurosci Lett. 1994;180:127–130.
    1. Dixon CE, Hamm RJ, Taft WC, Hayes RL. Increased anticholinergic sensitivity following closed skull impact and controlled cortical impact traumatic brain injury in the rat. J Neurotrauma. 1994;11:275–287.
    1. Eikelenboom P, Hoogendijk WJ. Do delirium and Alzheimer’s dementia share specific pathogenetic mechanisms? Dement Geriatr Cogn Disord. 1999;10:319–324.
    1. Eikelenboom P, Hoogendijk WJ, Jonker C, van Tilburg W. Immunological mechanisms and the spectrum of psychiatric syndromes in Alzheimer’s disease. J Psychiatr Res. 2002;36:269–280.
    1. Kronfol Z, Remick DG. Cytokines and the brain: implications for clinical psychiatry. Am J Psychiatry. 2000;157:683–694.
    1. Willard LB, Hauss-Wegrzyniak B, Wenk GL. Pathological and biochemical consequences of acute and chronic neuroinflammation within the basal forebrain cholinergic system of rats. Neuroscience. 1999;88:193–200.
    1. Broadhurst C, Wilson K. Immunology of delirium: new opportunities for treatment and research. Br J Psychiatry. 2001;179:288–289.
    1. Stefano GB, Bilfinger TV, Fricchione GL. The immune-neuro-link and the macrophage: postcardiotomy delirium, HIV-associated dementia and psychiatry. Prog Neurobiol. 1994;42:475–488.
    1. Venters HD, Tang Q, Liu Q, VanHoy RW, Dantzer R, Kelley KW. A new mechanism of neurodegeneration: a proinflammatory cytokine inhibits receptor signaling by a survival peptide. Proc Natl Acad Sci U S A. 1999;96:9879–9884.
    1. Lindesay J. The concept of delirium. Dement Geriatr Cogn Disord. 1999;10:310–314.
    1. Seamans JK, Floresco SB, Phillips AG. D1 receptor modulation of hippocampal-prefrontal cortical circuits integrating spatial memory with executive functions in the rat. J Neurosci. 1998;18:1613–1621.
    1. Wilkinson LS. The nature of interactions involving prefrontal and striatal dopamine systems. J Psychopharmacol. 1997;11:143–150.
    1. Platt MM, Breitbart W, Smith M, Marotta R, Weisman H, Jacobsen PB. Efficacy of neuroleptics for hypoactive delirium. J Neuropsychiatry Clin Neurosci. 1994;6:66–67.
    1. Mrzljak L, Goldman-Rakic PS. Acetylcholinesterase reactivity in the frontal cortex of human and monkey: contribution of AChE-rich pyramidal neurons. J Comp Neurol. 1992;324:261–281.
    1. Broderick PA, Gibson GE. Dopamine and serotonin in rat striatum during in vivo hypoxic-hypoxia. Metab Brain Dis. 1989;4:143–153.
    1. Breitbart W, Strout D. Delirium in the terminally ill. Clin Geriatr Med. 2000;16:357–372.
    1. Hirano H, Day J, Fibiger HC. Serotonergic regulation of acetylcholine release in rat frontal cortex. J Neurochem. 1995;65:1139–1145.
    1. Tanda G, Carboni E, Frau R, Di Chiara G. Increase of extracellular dopamine in the prefrontal cortex: a trait of drugs with antidepressant potential? Psychopharmacology (Berl) 1994;115:285–288.
    1. Chan CH, Liu HC, Huang MC. Delirium associated with concomitant use of low-dose bupropion sustained release and fluoxetine. J Clin Psychopharmacol. 2006;26:677–679.
    1. Tassin JP. Norepinephrine-dopamine interactions in the prefrontal cortex and the ventral tegmental area: relevance to mental diseases. Adv Pharmacol. 1998;42:712–716.
    1. Trzepacz PT. The neuropathophysiology of delirium. In: Lindesay JRK, Macdonald A, editors. Delirium in Old Age. Oxford: Oxford University Press; 2002. pp. 51–90.
    1. Neef D, Walling AD. Dementia with Lewy bodies: an emerging disease. Am Fam Physician. 2006;73:1223–1229.
    1. Levkoff SE, Evans DA, Liptzin B, et al. Delirium. The occurrence and persistence of symptoms among elderly hospitalized patients. Arch Intern Med. 1992;152:334–340.
    1. Jackson JC, Gordon SM, Hart RP, Hopkins RO, Ely EW. The association between delirium and cognitive decline: a review of the empirical literature. Neuropsychol Rev. 2004;14:87–98.
    1. Kiely DK, Jones RN, Bergmann MA, Murphy KM, Orav EJ, Marcantonio ER. Association between delirium resolution and functional recovery among newly admitted postacute facility patients. J Gerontol A Biol Sci Med Sci. 2006;61:204–208.
    1. Yokota H, Ogawa S, Kurokawa A, Yamamoto Y. Regional cerebral blood flow in delirium patients. Psychiatry Clin Neurosci. 2003;57:337–339.
    1. Rakonczay Z, Horvath Z, Juhasz A, Kalman J. Peripheral cholinergic disturbances in Alzheimer’s disease. Chem Biol Interact. 2005;157–158:233–238.
    1. Di Lazzaro V, Pilato F, Dileone M, et al. In vivo cholinergic circuit evaluation in frontotemporal and Alzheimer dementias. Neurology. 2006;66:1111–1113.
    1. Oddo S, LaFerla FM. The role of nicotinic acetylcholine receptors in Alzheimer’s disease. J Physiol (Paris) 2006;99:172–179.
    1. Ely EW, Girard TD, Shintani AK, et al. Apolipoprotein E4 polymorphism as a genetic predisposition to delirium in critically ill patients. Crit Care Med. 2007;35:112–117.
    1. Lehtovirta M, Soininen H, Helisalmi S, et al. Clinical and neuro-psychological characteristics in familial and sporadic Alzheimer’s disease: relation to apolipoprotein E polymorphism. Neurology. 1996;46:413–419.
    1. Lehtovirta M, Partanen J, Kononen M, et al. Spectral analysis of EEG in Alzheimer’s disease: relation to apolipoprotein E polymorphism. Neurobiol Aging. 1996;17:523–526.
    1. Kowall NW, Beal MF, Busciglio J, Duffy LK, Yankner BA. An in vivo model for the neurodegenerative effects of beta amyloid and protection by substance P. Proc Natl Acad Sci U S A. 1991;88:7247–7251.
    1. Ozturk A, DeKosky ST, Kamboh MI. Genetic variation in the choline acetyltransferase (CHAT) gene may be associated with the risk of Alzheimer’s disease. Neurobiol Aging. 2006;27:1440–1444.
    1. Soininen H, Lehtovirta M, Helisalmi S, Linnaranta K, Heinonen O, Riekkinen P. Increased acetylcholinesterase activity in the CSF of Alzheimer patients carrying apolipoprotein epsilon4 allele. Neuroreport. 1995;6:2518–2520.
    1. Liptzin B, Laki A, Garb JL, Fingeroth R, Krushell R. Donepezil in the prevention and treatment of post-surgical delirium. Am J Geriatr Psychiatry. 2005;13:1100–1106.
    1. Sampson EL, Raven PR, Ndhlovu PN, et al. A randomized, double-blind, placebo-controlled trial of donepezil hydrochloride (Aricept) for reducing the incidence of postoperative delirium after elective total hip replacement. Int J Geriatr Psychiatry. 2007;22:343–349.
    1. Adamis D, Treloar A, Martin FC, Gregson N, Hamilton G, Macdonald AJ. APOE and cytokines as biological markers for recovery of prevalent delirium in elderly medical inpatients. Int J Geriatr Psychiatry. 2007;22:688–694.
    1. Marcantonio ER, Rudolph JL, Culley D, Crosby G, Alsop D, Inouye SK. Serum biomarkers for delirium. J Gerontol A Biol Sci Med Sci. 2006;61:1281–1286.
    1. Volpicelli LA, Levey AI. Muscarinic acetylcholine receptor subtypes in cerebral cortex and hippocampus. Prog Brain Res. 2004;145:59–66.
    1. Singh VK, Guthikonda P. Circulating cytokines in Alzheimer’s disease. J Psychiatr Res. 1997;31:657–660.
    1. Hong M, Lee VM. Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J Biol Chem. 1997;272:19547–19553.
    1. Nordberg A, Wahlstrom G. Cholinergic mechanisms in physical dependence on barbiturates, ethanol and benzodiazepines. J Neural Transm Gen Sect. 1992;88:199–221.
    1. Kikuchi T, Wang Y, Shinbori H, Sato K, Okumura F. Effects of ketamine and pentobarbitone on acetylcholine release from the rat frontal cortex in vivo. Br J Anaesth. 1997;79:128–130.
    1. Meltzer HY. Serotonin receptors and antipsychotic drug action. Psychopharmacol Ser. 1993;10:70–81.
    1. Papassotiropoulos A, Bagli M, Jessen F, et al. A genetic variation of the inflammatory cytokine interleukin-6 delays the initial onset and reduces the risk for sporadic Alzheimer’s disease. Ann Neurol. 1999;45:666–668.
    1. Nicoll JA, Mrak RE, Graham DI, et al. Association of interleukin-1 gene polymorphisms with Alzheimer’s disease. Ann Neurol. 2000;47:365–368.
    1. Dunn AJ, Wang J, Ando T. Effects of cytokines on cerebral neurotransmission. Comparison with the effects of stress. Adv Exp Med Biol. 1999;461:117–127.

Source: PubMed

3
Subskrybuj