Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-pftt2 Total loading time: 0 Render date: 2024-04-30T13:10:35.013Z Has data issue: false hasContentIssue false

Section 3 - Symptomatology and Diagnosis for the Perioperative Neurocognitive Disorders

Published online by Cambridge University Press:  11 April 2019

Roderic G. Eckenhoff
Affiliation:
University of Pennsylvania
Niccolò Terrando
Affiliation:
Duke University, North Carolina
Get access
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2019

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Murkin, JM, Newman, SP, Stump, DA, Blumenthal, JA. Statement of consensus on assessment of neurobehavioral outcomes after cardiac surgery. Ann Thorac Surg. 1995;59(5):1289–95.Google Scholar
Folstein, MF, Robins, LN, Helzer, JE. The Mini-Mental State Examination. Arch Gen Psychiatry. 1983;40:812.CrossRefGoogle ScholarPubMed
Ismail, Z, Rajji, TK, Shulman, KI. Brief cognitive screening instruments: an update. Int J Geriatr Psychiatry. 2010;25(2):111–20.CrossRefGoogle ScholarPubMed
Silbert, BS, Maruff, P, Evered, LA, Scott, DA, Kalpokas, M, Martin, KJ, et al. Detection of cognitive decline after coronary surgery: a comparison of computerized and conventional tests. Br J Anaesth. 2004;92(6):814–20.Google Scholar
Hawkins, KA, Dean, D, Pearlson, GD. Alternative forms of the Rey Auditory Verbal Learning Test: a review. Behav Neurol. 2004;15(3–4):99107.CrossRefGoogle ScholarPubMed
Salthouse, TA. Influence of age on practice effects in longitudinal neurocognitive change. Neuropsychology. 2010;24(5):563–72.CrossRefGoogle ScholarPubMed
Duff, K. Evidence-based indicators of neuropsychological change in the individual patient: relevant concepts and methods. Arch Clin Neuropsychol. 2012;27(3):248–61.Google Scholar
Silbert, B, Evered, L, Scott, DA, McMahon, S, Choong, P, Ames, D, et al. Preexisting cognitive impairment is associated with postoperative cognitive dysfunction after hip joint replacement surgery. Anesthesiology. 2015;122(6):1224–34.Google Scholar
Evered, L, Scott, DA, Silbert, B, Maruff, P. Postoperative cognitive dysfunction is independent of type of surgery and anesthetic. Anesth Analg. 2011;112(5):1179–85.Google Scholar
Newman, MF, Kirchner, JL, Phillips-Bute, B, Gaver, V, Grocott, H, Jones, RH, et al. Longitudinal assessment of neurocognitive function after coronary-artery bypass surgery. N Engl J Med. 2001;344(6):395402.CrossRefGoogle ScholarPubMed
Silbert, BS, Scott, DA, Evered, LA, Lewis, MS, Kalpokas, M, Maruff, P, et al. A comparison of the effect of high- and low-dose fentanyl on the incidence of postoperative cognitive dysfunction after coronary artery bypass surgery in the elderly. Anesthesiology. 2006;104(6):1137–45.CrossRefGoogle ScholarPubMed
Jacobson, NS, Truax, P. Clinical significance: a statistical approach to defining meaningful change in psychotherapy research. J Consult Clin Psychol. 1991;59(1):12–19.Google Scholar
Collie, ADD, Falleti, MG, Silbert, BS, Maruff, P. Determining the extent of cognitive change after coronary surgery: a review of statistical procedures. Ann Thorac Surg. 2002;73:2005–11.Google Scholar
Kneebone, AC, Andrew, MJ, Baker, RA, Knight, JL. Neuropsychologic changes after coronary artery bypass grafting: use of reliable change indices. Ann Thorac Surg. 1998;65(5):1320–5.CrossRefGoogle ScholarPubMed
Rasmussen, LS, Larsen, K, Houx, P, Skovgaard, LT, Hanning, CD, Moller, JT. The assessment of postoperative cognitive function. Acta Anaesthesiol Scand. 2001;45(3):275–89.Google Scholar
Ingraham, L, Aiken, C. An empirical approach to determine criteria for abnormality in test batteries with multiple results. Neuropsychology. 1996;10:120–4.CrossRefGoogle Scholar
Keizer, AM, Hijman, R, Kalkman, CJ, Kahn, RS, van Dijk, D. The incidence of cognitive decline after (not) undergoing coronary artery bypass grafting: the impact of a controlled definition. Acta Anaesthesiol Scand. 2005;49(9):1232–5.CrossRefGoogle ScholarPubMed

References

Berger, M, Nadler, JW, Browndyke, J, et al. Postoperative cognitive dysfunction: minding the gaps in our knowledge of a common postoperative complication in the elderly. Anesthesiology Clinics. 2015;33(3):517550.Google Scholar
Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clinical Pharmacology and Therapeutics. 2001;69(3):8995.Google Scholar
Chen, Y, Bidwell, LC, Norton, D. Trait vs. state markers for schizophrenia: identification and characterization through visual processes. Current Psychiatry Reviews. 2006;2(4):431438.CrossRefGoogle ScholarPubMed
Cheng, Q, Wang, J, Wu, A, Zhang, R, Li, L, Yue, Y. Can urinary excretion rate of 8-isoprostrane and malonaldehyde predict postoperative cognitive dysfunction in aging? Neurological Sciences. 2013;34(9):16651669.Google Scholar
Zhang, Y-H, Guo, X-H, Zhang, Q-M, Yan, G-T, Wang, T-L. Serum CRP and urinary trypsin inhibitor implicate postoperative cognitive dysfunction especially in elderly patients. International Journal of Neuroscience. 2015;125(7):501506.Google Scholar
Wu, Y, Wang, J, Wu, A, Yue, Y. Do fluctuations in endogenous melatonin levels predict the occurrence of postoperative cognitive dysfunction (POCD)? International Journal of Neuroscience. 2014;124(11):787791.Google Scholar
Balan, S, Leibovitz, A, Zila, SO, et al. The relation between the clinical subtypes of delirium and the urinary level of 6-SMT. Journal of Neuropsychiatry and Clinical Neuroscience. 2003;15(3):363366.Google Scholar
de Jonghe, A, van Munster, BC, Goslings, JC, et al. Effect of melatonin on incidence of delirium among patients with hip fracture: a multicentre, double-blind randomized controlled trial. CMAJ. 2014;186(14):E547–E556.Google Scholar
Linstedt, U, Meyer, O, Kropp, P, Berkau, A, Tapp, E, Zenz, M. Serum concentration of S-100 protein in assessment of cognitive dysfunction after general anesthesia in different types of surgery. Acta Anaesthesiologica Scandinavica. 2002;46(4):384389.Google Scholar
Li, YC, Xi, CH, An, YF, Dong, WH, Zhou, M. Perioperative inflammatory response and protein S-100beta concentrations – relationship with post-operative cognitive dysfunction in elderly patients. Acta Anaesthesiologica Scandinavica. 2012;56(5):595600.CrossRefGoogle ScholarPubMed
Rohan, D, Buggy, DJ, Crowley, S, et al. Increased incidence of postoperative cognitive dysfunction 24 hr after minor surgery in the elderly. Canadian Journal of Anaesthesia/Journal canadian d’anesthesie. 2005;52(2):137142.Google Scholar
Iohom, G, Szarvas, S, Larney, V, et al. Perioperative plasma concentrations of stable nitric oxide products are predictive of cognitive dysfunction after laparoscopic cholecystectomy. Anesthesia & Analgesia. 2004;99(4):12451252.Google Scholar
Evered, L, Silbert, B, Scott, DA, Zetterberg, H, Blennow, K. Association of changes in plasma neurofilament light and tau levels with anesthesia and surgery: results from the CAPACITY and ARCADIAN studies. JAMA Neurology. 2018;75(5):542547.doi:10.1001/jamaneurol.2017.4913.Google Scholar
Vasunilashorn, SM, Ngo, L, Inouye, SK, et al. Cytokines and postoperative delirium in older patients undergoing major elective surgery. Journals of Gerontology, Series A. 2015;70(10):12891295.CrossRefGoogle ScholarPubMed
Lin, GX, Wang, T, Chen, MH, Hu, ZH, Ouyang, W. Serum high-mobility group box 1 protein correlates with cognitive decline after gastrointestinal surgery. Acta Anaesthesiologica Scandinavica. 2014;58(6):668674.Google Scholar
Li, X, Wen, D-X, Zhao, Y-H, Hang, Y-N, Mandell, MS. Increase of beta-amyloid and C-reactive protein in liver transplant recipients with postoperative cognitive dysfunction. Hepatobiliary and Pancreatic Diseases International. 2013;12(4):370376.CrossRefGoogle ScholarPubMed
Li, Y, He, R, Chen, S, Qu, Y. Effect of dexmedetomidine on early postoperative cognitive dysfunction and peri-operative inflammation in elderly patients undergoing laparoscopic cholecystectomy. Experimental and Therapeutic Medicine. 2015;10(5):16351642.Google Scholar
Zhang, Q, Li, S, Feng, C, et al. Serum proteomics of early postoperative cognitive dysfunction in elderly patients. Chinese Medical Journal. 2012;125(14):24552461.Google Scholar
Harmon, D, Eustace, N, Ghori, K, et al. Plasma concentrations of nitric oxide products and cognitive dysfunction following coronary artery bypass surgery. European Journal of Anaesthesiology. 2005;22(4):269276.Google Scholar
Twomey, C, Corrigan, M, Burlacu, C, Butler, M, Iohom, G, Shorten, G. Nitric oxide index is not a predictor of cognitive dysfunction following laparotomy. Journal of Clinical Anesthesia. 2010;22(1):2228.Google Scholar
Nakamura, A, Kaneko, N, Villemagne, VL, et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature. 2018;554(7691):249254.Google Scholar
Simon, MJ, Iliff, JJ. Regulation of cerebrospinal fluid (CSF) flow in neurodegenerative, neurovascular and neuroinflammatory disease. Biochimica et Biophysica Acta. 2016;1862(3):442451.Google Scholar
Zetterberg, H, Hietala, MA, Jonsson, M, et al. Neurochemical aftermath of amateur boxing. Archives of Neurology. 2006;63(9):12771280.Google Scholar
Berger, M. The effect of propofol versus isoflurane anesthesia on human CSF markers of Alzheimer’s disease: results of a randomized trial. Journal of Alzheimers Disease. 2016;52:12991310.CrossRefGoogle ScholarPubMed
Tang, JX, Baranov, D, Hammond, M, Shaw, LM, Eckenhoff, MF, Eckenhoff, RG. Human Alzheimer and inflammation biomarkers after anesthesia and surgery. Anesthesiology. 2011;115(4):727732.Google Scholar
Anckarsater, R, Anckarsater, H, Bromander, S, Blennow, K, Wass, C, Zetterberg, H. Non-neurological surgery and cerebrospinal fluid biomarkers for neuronal and astroglial integrity. Journal of Neural Transmission. 2014;121(6):649653.Google Scholar
Palotas, A, Reis, HJ, Bogats, G, et al. Coronary artery bypass surgery provokes Alzheimer’s disease-like changes in the cerebrospinal fluid. Journal of Alzheimers Disease. 2010;21(4):11531164.CrossRefGoogle ScholarPubMed
Ji, MH, Yuan, HM, Zhang, GF, et al. Changes in plasma and cerebrospinal fluid biomarkers in aged patients with early postoperative cognitive dysfunction following total hip-replacement surgery. Journal of Anesthesia. 2013;27(2):236242.Google Scholar
Xie, Z, McAuliffe, S, Swain, CA, et al. Cerebrospinal fluid aβ to tau ratio and postoperative cognitive change. Annals of Surgery. 2013;258(2):364369.Google Scholar
Xie, Z, Swain, CA, Ward, SA, et al. Preoperative cerebrospinal fluid beta-Amyloid/Tau ratio and postoperative delirium. Annals of Clinical and Translational Neurology. 2014;1(5):319328.Google Scholar
Evered, L, Silbert, B, Scott, DA, Ames, D, Maruff, P, Blennow, K. Cerebrospinal fluid biomarker for Alzheimer disease predicts postoperative cognitive dysfunction. Anesthesiology. 2016;124(2):353361.Google Scholar
Buvanendran, A, Kroin, JS, Berger, RA, et al. Upregulation of prostaglandin E2 and interleukins in the central nervous system and peripheral tissue during and after surgery in humans. Anesthesiology. 2006;104(3):403410.Google Scholar
Bromander, S, Anckarsater, R, Kristiansson, M, et al. Changes in serum and cerebrospinal fluid cytokines in response to non-neurological surgery: an observational study. Journal of Neuroinflammation. 2012;9:242.CrossRefGoogle ScholarPubMed
Yeager, MP, Lunt, P, Arruda, J, Whalen, K, Rose, R, DeLeo, JA. Cerebrospinal fluid cytokine levels after surgery with spinal or general anesthesia. Regional Anesthesia and Pain Medicine. 1999;24(6):557562.Google Scholar
Mathew, JP, Podgoreanu, MV, Grocott, HP, et al. Genetic variants in P-selectin and C-reactive protein influence susceptibility to cognitive decline after cardiac surgery. Journal of the American College of Cardiology. 2007;49(19):19341942.Google Scholar
McDonagh, DL, Mathew, JP, White, WD, et al. Cognitive function after major noncardiac surgery, apolipoprotein E4 genotype, and biomarkers of brain injury. Anesthesiology. 2010;112(4):852859.Google Scholar
Laskowitz, DT, Vitek, MP. Apolipoprotein E and neurological disease: therapeutic potential and pharmacogenomic interactions. Pharmacogenomics. 2007;8(8):959969.CrossRefGoogle ScholarPubMed
Schenning, KJ, Murchison, CF, Mattek, NC, Silbert, LC, Kaye, JA, Quinn, JF. Surgery is associated with ventricular enlargement as well as cognitive and functional decline. Alzheimer’s and Dementia. 2015;5:590597.Google Scholar
Berger, M, Burke, J, Eckenhoff, R, Mathew, J. Alzheimer’s disease, anesthesia, and surgery: a clinically focused review. Journal of Cardiothoracic and Vascular Anesthesia. 2014;28:16091623.Google Scholar
Radtke, FM, Franck, M, Lendner, J, Kruger, S, Wernecke, KD, Spies, CD. Monitoring depth of anaesthesia in a randomized trial decreases the rate of postoperative delirium but not postoperative cognitive dysfunction. British Journal of Anaesthesia. 2013;110 Suppl 1:98105.Google Scholar
Chan, MT, Cheng, BC, Lee, TM, Gin, T, CODA Trial, Group. BIS-guided anesthesia decreases postoperative delirium and cognitive decline. Journal of Neurosurgical Anesthesiology. 2013;25(1):3342.Google Scholar
Sieber, FE, Zakriya, KJ, Gottschalk, A, et al. Sedation depth during spinal anesthesia and the development of postoperative delirium in elderly patients undergoing hip fracture repair. Mayo Clinic Proceedings. 2010;85(1):1826.Google Scholar
Andresen, JM, Girard, TD, Pandharipande, PP, Davidson, MA, Ely, EW, Watson, PL. Burst suppression on processed electroencephalography as a predictor of postcoma delirium in mechanically ventilated ICU patients. Critical Care Medicine. 2014;42(10):22442251.Google Scholar
Soehle, M, Dittmann, A, Ellerkmann, RK, Baumgarten, G, Putensen, C, Guenther, U. Intraoperative burst suppression is associated with postoperative delirium following cardiac surgery: a prospective, observational study. BMC Anesthesiology. 2015;15:61.Google Scholar
Deiner, S, Luo, X, Silverstein, JH, Sano, M. Can intraoperative processed EEG predict postoperative cognitive dysfunction in the elderly? Clinical Therapeutics. 2015;37(12):27002705.Google Scholar
Purdon, PL, Sampson, A, Pavone, KJ, Brown, EN. Clinical electroencephalography for anesthesiologists: Part I: Background and basic signatures. Anesthesiology. 2015;123(4):937960.Google Scholar
Purdon, PL, Pavone, KJ, Akeju, O, et al. The ageing brain: age-dependent changes in the electroencephalogram during propofol and sevoflurane general anaesthesia. British Journal of Anaesthesia. 2015;115 Suppl 1:4657.Google Scholar
McDonagh, DL, Berger, M, Mathew, JP, Graffagnino, C, Milano, CA, Newman, MF. Neurological complications of cardiac surgery. Lancet Neurology. 2014;13(5):490502.Google Scholar
Price, CC, Tanner, JJ, Schmalfuss, I, et al. A pilot study evaluating presurgery neuroanatomical biomarkers for postoperative cognitive decline after total knee arthroplasty in older adults. Anesthesiology. 2014;120(3):601613.Google Scholar
Browndyke, J. Postoperative changes in resting-state functional connectivity and cognition following major cardiac surgery in older adults: preliminary findings. Journal of the American Geriatrics Society. 2017;65:612.Google Scholar
Farag, E, Chelune, GJ, Schubert, A, Mascha, EJ. Is depth of anesthesia, as assessed by the Bispectral Index, related to postoperative cognitive dysfunction and recovery? Anesthesia & Analgesia. 2006;103(3):633640.Google Scholar
Gerriets, T, Schwarz, N, Bachmann, G, et al. Evaluation of methods to predict early long-term neurobehavioral outcome after coronary artery bypass grafting. American Journal of Cardiology. 2010;105(8):10951101.CrossRefGoogle ScholarPubMed

References

Glover, GH. Overview of functional magnetic resonance imaging. Neurosurg Clin N Am. 2011;22(2):133–9, vii.Google Scholar
Ibinson, JW, Vogt, KM. Pain does not follow the boxcar model: temporal dynamics of the BOLD fMRI signal during constant current painful electric nerve stimulation. J Pain. 2013;14(12):1611–19.Google Scholar
Biswal, B, Yetkin, FZ, Haughton, VM, Hyde, JS. Functional connectivity in the motor cortex of resting human brain using echo-planar MRI. Magn Reson Med. 1995;34(4):537–41.Google Scholar
van den Heuvel, MP, Hulshoff Pol, HE. Exploring the brain network: a review on resting-state fMRI functional connectivity. Eur Neuropsychopharmacol. 2010;20(8):519–34.Google Scholar
Drysdale, AT, Grosenick, L, Downar, J, Dunlop, K, Mansouri, F, Meng, Y, et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med. 2017;23(1):2838.Google Scholar
Sheline, YI, Raichle, ME. Resting state functional connectivity in preclinical Alzheimer’s disease. Biol Psychiatry. 2013;74(5):340–7.Google Scholar
Klunk, WE, Engler, H, Nordberg, A, Wang, Y, Blomqvist, G, Holt, DP, et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol. 2004;55(3):306–19.Google Scholar
Ishii, K. PET approaches for diagnosis of dementia. AJNR Am J Neuroradiol. 2014;35(11):2030–8.Google Scholar
Zimmer, ER, Leuzy, A, Benedet, AL, Breitner, J, Gauthier, S, Rosa-Neto, P. Tracking neuroinflammation in Alzheimer’s disease: the role of positron emission tomography imaging. J Neuroinflammation. 2014;11:120.Google Scholar
Chen, CW, Lin, CC, Chen, KB, Kuo, YC, Li, CY, Chung, CJ. Increased risk of dementia in people with previous exposure to general anesthesia: a nationwide population-based case-control study. Alzheimers Dement. 2014;10(2):196204.Google Scholar
Chen, PL, Yang, CW, Tseng, YK, Sun, WZ, Wang, JL, Wang, SJ, et al. Risk of dementia after anaesthesia and surgery. Br J Psychiatry. 2014;204(3):188–93.Google Scholar
Berger, M, Burke, J, Eckenhoff, R, Mathew, J. Alzheimer’s disease, anesthesia, and surgery: a clinically focused review. J Cardiothorac Vasc Anesth. 2014;28(6):1609–23.CrossRefGoogle ScholarPubMed
Evered, L, Silbert, B, Scott, DA, Ames, D, Maruff, P, Blennow, K. Cerebrospinal fluid biomarker for Alzheimer disease predicts postoperative cognitive dysfunction. Anesthesiology. 2016;124(2):353–61.CrossRefGoogle ScholarPubMed
Coimbra, A, Williams, DS, Hostetler, ED. The role of MRI and PET/SPECT in Alzheimer’s disease. Curr Top Med Chem. 2006;6(6):629–47.Google Scholar
Mosconi, L, Berti, V, Glodzik, L, Pupi, A, De Santi, S, de Leon, MJ. Pre-clinical detection of Alzheimer’s disease using FDG-PET, with or without amyloid imaging. J Alzheimers Dis. 2010;20(3):843–54.Google Scholar
Rabinovici, GD, Jagust, WJ. Amyloid imaging in aging and dementia: testing the amyloid hypothesis in vivo. Behav Neurol. 2009;21(1):117–28.Google Scholar
Small, GW, Bookheimer, SY, Thompson, PM, Cole, GM, Huang, SC, Kepe, V, et al. Current and future uses of neuroimaging for cognitively impaired patients. Lancet Neurol. 2008;7(2):161–72.CrossRefGoogle ScholarPubMed
Rowe, CC, Ellis, KA, Rimajova, M, Bourgeat, P, Pike, KE, Jones, G, et al. Amyloid imaging results from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging. Neurobiol Aging. 2010;31(8):1275–83.Google Scholar
Hamelin, L, Lagarde, J, Dorothee, G, Leroy, C, Labit, M, Comley, RA, et al. Early and protective microglial activation in Alzheimer’s disease: a prospective study using 18F-DPA-714 PET imaging. Brain. 2016;139(Pt 4):1252–64.Google Scholar
Saidlitz, P, Voisin, T, Vellas, B, Payoux, P, Gabelle, A, Formaglio, M, et al. Amyloid imaging in Alzheimer’s disease: a literature review. J Nutr Health Aging. 2014;18(7):723–40.Google Scholar
Wang, L, Benzinger, TL, Su, Y, Christensen, J, Friedrichsen, K, Aldea, P, et al. Evaluation of tau imaging in staging Alzheimer disease and revealing interactions between beta-amyloid and tauopathy. JAMA Neurol. 2016;73(9):1070–7.Google Scholar
Hoenig, MC, Bischof, GN, Seemiller, J, Hammes, J, Kukolja, J, Onur, OA, et al. Networks of tau distribution in Alzheimer’s disease. Brain. 2018;141(2):568–81.Google Scholar
Kang, JM, Lee, SY, Seo, S, Jeong, HJ, Woo, SH, Lee, H, et al. Tau positron emission tomography using [(18)F]THK5351 and cerebral glucose hypometabolism in Alzheimer’s disease. Neurobiol Aging. 2017;59:210–19.Google Scholar
Adlard, PA, Tran, BA, Finkelstein, DI, Desmond, PM, Johnston, LA, Bush, AI, et al. A review of beta-amyloid neuroimaging in Alzheimer’s disease. Front Neurosci. 2014;8:327.Google Scholar
Glodzik-Sobanska, L, Rusinek, H, Mosconi, L, Li, Y, Zhan, J, de Santi, S, et al. The role of quantitative structural imaging in the early diagnosis of Alzheimer’s disease. Neuroimag Clin N Am. 2005;15(4):803–26, x.Google Scholar
Ramani, A, Jensen, JH, Helpern, JA. Quantitative MR imaging in Alzheimer disease. Radiology. 2006;241(1):2644.Google Scholar
Vemuri, P, Jack, CR Jr. Role of structural MRI in Alzheimer’s disease. Alzheimers Res Ther. 2010;2(4):23.Google Scholar
Frisoni, GB, Fox, NC, Jack, CR Jr, Scheltens, P, Thompson, PM. The clinical use of structural MRI in Alzheimer disease. Nat Rev Neurol. 2010;6(2):6777.Google Scholar
Lehericy, S, Delmaire, C, Galanaud, D, Dormont, D. Neuroimaging in dementia. Presse Med. 2007;36(10 Pt 2):1453–63.Google Scholar
Huijbers, W, Mormino, EC, Schultz, AP, Wigman, S, Ward, AM, Larvie, M, et al. Amyloid-β deposition in mild cognitive impairment is associated with increased hippocampal activity, atrophy and clinical progression. Brain. 2015;138(4):1023–35.Google Scholar
Allen, G, Barnard, H, McColl, R, Hester, AL, Fields, JA, Weiner, MF, et al. Reduced hippocampal functional connectivity in Alzheimer disease. Arch Neurol. 2007;64(10):1482–7.Google Scholar
Sheline, YI, Raichle, ME, Snyder, AZ, Morris, JC, Head, D, Wang, S, et al. Amyloid plaques disrupt resting state default mode network connectivity in cognitively normal elderly. Biol Psychiatry. 2010;67(6):584–7.Google Scholar
Onoda, K, Yada, N, Ozasa, K, Hara, S, Yamamoto, Y, Kitagaki, H, et al. Can a resting-state functional connectivity index identify patients with Alzheimer’s disease and mild cognitive impairment across multiple sites? Brain Connect. 2017;Jun 30.Google Scholar
Eikelenboom, P, van Gool, WA. Neuroinflammatory perspectives on the two faces of Alzheimer’s disease. J Neural Transm (Vienna). 2004;111(3):281–94.Google Scholar
Griffin, WS, Stanley, LC, Ling, C, White, L, MacLeod, V, Perrot, LJ, et al. Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci USA. 1989;86(19):7611–15.Google Scholar
Alam, MM, Lee, J, Lee, SY. Recent progress in the development of TSPO PET ligands for neuroinflammation imaging in neurological diseases. Nucl Med Mol Imaging. 2017;51(4):283–96.Google Scholar
Gerhard, A, Pavese, N, Hotton, G, Turkheimer, F, Es, M, Hammers, A, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol Dis. 2006;21(2):404–12.Google Scholar
Jucaite, A, Svenningsson, P, Rinne, JO, Cselenyi, Z, Varnas, K, Johnstrom, P, et al. Effect of the myeloperoxidase inhibitor AZD3241 on microglia: a PET study in Parkinson’s disease. Brain. 2015;138(Pt 9):2687–700.CrossRefGoogle ScholarPubMed
Iaccarino, L, Sala, A, Caminiti, SP, Perani, D. The emerging role of PET imaging in dementia. F1000Res. 2017;6:1830.Google Scholar
Zhang, X, Paule, MG, Newport, GD, Liu, F, Callicott, R, Liu, S, et al. MicroPET/CT imaging of [18F]-FEPPA in the nonhuman primate: a potential biomarker of pathogenic processes associated with anesthetic-induced neurotoxicity. ISRN Anesthesiol. 2012;2012:11.Google Scholar
Forsberg, A, Cervenka, S, Jonsson Fagerlund, M, Rasmussen, LS, Zetterberg, H, Erlandsson Harris, H, et al. The immune response of the human brain to abdominal surgery. Ann Neurol. 2017;81(4):572–82.Google Scholar
Vik, A, Brubakk, AO, Rinck, PA, Sande, E, Levang, OW, Sellevold, O. MRI: a method to detect minor brain damage following coronary bypass surgery? Neuroradiology. 1991;33(5):396–8.Google Scholar
Sun, X, Lindsay, J, Monsein, LH, Hill, PC, Corso, PJ. Silent brain injury after cardiac surgery: a review: cognitive dysfunction and magnetic resonance imaging diffusion-weighted imaging findings. J Am Coll Cardiol. 2012;60(9):791–7.Google Scholar
Gerriets, T, Schwarz, N, Bachmann, G, Kaps, M, Kloevekorn, WP, Sammer, G, et al. Evaluation of methods to predict early long-term neurobehavioral outcome after coronary artery bypass grafting. Am J Cardiol. 2010;105(8):1095–101.Google Scholar
Knipp, SC, Matatko, N, Schlamann, M, Wilhelm, H, Thielmann, M, Forsting, M, et al. Small ischemic brain lesions after cardiac valve replacement detected by diffusion-weighted magnetic resonance imaging: relation to neurocognitive function. Eur J Cardiothorac Surg. 2005;28(1):8896.Google Scholar
Knipp, SC, Matatko, N, Wilhelm, H, Schlamann, M, Thielmann, M, Losch, C, et al. Cognitive outcomes three years after coronary artery bypass surgery: relation to diffusion-weighted magnetic resonance imaging. Ann Thorac Surg. 2008;85(3):872–9.Google Scholar
Cook, DJ, Huston, J 3rd, Trenerry, MR, Brown, RD Jr, Zehr, KJ, Sundt, TM 3rd. Postcardiac surgical cognitive impairment in the aged using diffusion-weighted magnetic resonance imaging. Ann Thorac Surg. 2007;83(4):1389–95.Google Scholar
Kahlert, P, Knipp, SC, Schlamann, M, Thielmann, M, Al-Rashid, F, Weber, M, et al. Silent and apparent cerebral ischemia after percutaneous transfemoral aortic valve implantation: a diffusion-weighted magnetic resonance imaging study. Circulation. 2010;121(7):870–8.CrossRefGoogle ScholarPubMed
Scott, DA, Evered, LA, Gerraty, RP, MacIsaac, A, Lai-Kwon, J, Silbert, BS. Cognitive dysfunction follows left heart catheterisation but is not related to microembolic count. Int J Cardiol. 2014;175(1):6771.Google Scholar
Kruis, RW, Vlasveld, FA, Van Dijk, D. The (un)importance of cerebral microemboli. Semin Cardiothorac Vasc Anesth. 2010;14(2):111–18.Google Scholar
Xie, P, Yu, T, Fu, X, Tu, Y, Zou, Y, Lui, S, et al. Altered functional connectivity in an aged rat model of postoperative cognitive dysfunction: a study using resting-state functional MRI. PLoS One. 2013;8(5):e64820.Google Scholar
Browndyke, JN, Berger, M, Harshbarger, TB, Smith, PJ, White, W, Bisanar, TL, et al. Resting-state functional connectivity and cognition after major cardiac surgery in older adults without preoperative cognitive impairment: preliminary findings. J Am Geriatr Soc. 2017;65(1):e6e12.Google Scholar
Browndyke, JN, Berger, M, Smith, PJ, Harshbarger, TB, Monge, ZA, Panchal, V, et al. Task-related changes in degree centrality and local coherence of the posterior cingulate cortex after major cardiac surgery in older adults. Hum Brain Mapp. 2017;39:9851003.Google Scholar
Huang, H, Tanner, J, Parvataneni, H, Rice, M, Horgas, A, Ding, M, et al. Impact of total knee arthroplasty with general anesthesia on brain networks: cognitive efficiency and ventricular volume predict functional connectivity decline in older adults. J Alzheimers Dis. 2018;62:31933.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×