Sie sind auf Seite 1von 6

Available online at www.sciencedirect.

com

Brain damage in cardiac surgery patients


Wojciech Dabrowski1, Ziemowit Rzecki1, Jacek Pilat2 and Marek Czajkowski3
Neuropsychological disorders and brain injury are still a serious problem in cardiac surgery patients. Owing to multifactorial mechanism of brain injury during extracorporeal circulation, the effective and safe protection is extremely difcult. Despite several studies, the ideal neuroprotective treatment has not been found. Based on literature we analysed the main mechanisms of brain injury and new methods of brain protection.
Addresses 1 Department of Anaesthesiology Intensive Therapy, Medical University of Lublin, Poland 2 Department of General Surgery, Transplantology and Clinical Nutrition, Medical University of Lublin, Poland 3 Department of Cardiac Surgery, Medical University of Lublin, Poland Corresponding author: Dabrowski, Wojciech (wojciechdabrowski@interia.pl, w.dabrowski5@yahoo.com)

intellectual deterioration and memory decits [10]. All these disorders were also divided into focal, multifocal or global. The brain injury after cardiac surgery is multifactorial. Global or focal ischaemia appears to be the main factor leading to brain injury, and may result from cerebral hypoperfusion, macroembolisation or microembolisation, massive inammatory responses, perioperative disorders in oxygen delivery resulting from prolonged anaemia and postoperative cerebral hyperthermia, cardiac arrhythmias and genetic predisposition [1,1117]. Several clinicians and researchers suggest that intraoperative and postoperative cerebral embolisms connected with global or focal hypoperfusion are the crucial factors leading to postoperative neurocognitive disorders [11,12,15,18]. About 3050% of postoperative strokes result from cerebral macroembolism, whereas encephalopathy and neuropsychological disorders develop owing to microemboli [11,12,19,20]. There is strong evidence that a signicant number of cerebral embolisms result from the disrupted atherosclerotic aorta, opened left-side cardiac chamber or extracorporeal machine [12,1820]. Indeed, atherosclerosis of the ascending aorta is the predictive factor of brain injury in cardiac surgery patients [12,21]. A fragment of atherosclerotic plaque liberated by surgical manipulation of the aorta (clamping or cannulation) and translocated into the brain vessel induces ischaemia and causes transient or permanent cellular dysfunction in the vital centres of the brain. Such cerebral emboli may contribute to the postoperative morbidity or mortality. Importantly, similar disorders are observed after fat embolism associated with extracorporeal circulation and uncritical cardiotomy suction. Fat embolism causes brain oedema with haemorrhage [22]. Unfortunately, the arterial and venous lters routinely used inadequately protect patients from fat microemboli [23]. Disorders in cerebral blood ow are another important factor resulting in postoperative neuropsychological dysfunction. Physiologically, the cerebral blood ow ranges from 40 to 60 mL/100 g brain tissue per min. It is mainly regulated by the mean artery pressure. A traditional extracorporeal machine with roller pumps produces the pulsatile blood ow with the index of cardiac output between 2 and 2.4 L min1 m2. Despite this, the cerebral blood ow decreases to 2060 mL/100 g of brain tissue per min [24]. Additionally, this low brain perfusion may be reduced by carotid artery stenosis, which strongly
Current Opinion in Pharmacology 2012, 12:189194

Current Opinion in Pharmacology 2012, 12:189194 This review comes from a themed issue on Cardiovascular and renal Edited by Jamie Y Jeremy, Kai Zacharowski, Nilima Shukla and Song Wan Available online 9th February 2012 1471-4892/$ see front matter # 2012 Elsevier Ltd. All rights reserved. DOI 10.1016/j.coph.2012.01.013

Postoperative brain damage and neuropsychological disorders are frequent complications of cardiac surgery and elevate mortality, morbidity and hospital costs, as well as signicantly impair the quality of life [13]. They occur in 3080% of patients undergoing extracorporeal circulation (ECC) and in 3050% of patients undergoing cardiac surgery without ECC [4,58]. In many cases, they subside within three postoperative months; however, in 17 35% of cardiac surgery patients, the disorders can persist for even one year [2,5,8]. Their incidences are age-related and are signicantly higher in patients aged 65 years or more [9]. For instance, the incidence rate of the most serious brain damage stroke, is less than 1% in younger patients compared to 65.5% in patients aged 6575, and more than 7% in patients over the age of 75. The American College of Cardiology/American Heart Association classied the brain injury following cardiac surgery into two main categories: type 1 severe neurological dysfunction, stupor and coma, and type 2
www.sciencedirect.com

190 Cardiovascular and renal

predicts the postoperative brain injury. The moderate carotid artery diseases are noted in 1722% and severe stenosis (higher than 80%) is observed in 612% of cardiac surgery patients [25,26]. In patients over the age of 65, past transient ischaemic attack (TIA), smoking, peripheral artery diseases and female sex are associated with severe carotid stenosis [26]. For this reason many clinicians perform carotid endarterectomy preoperatively or synchronously with cardiac surgery [25,27]. Others suggest that the use of centrifugal pumps signicantly reduces adverse outcomes after cardiac surgery [28,29]. Moreover, some other factors such as anaesthetic agents or low carbon dioxide tension reduce the possibility of brain vessels autoregulation and have a strong impact on the cerebral blood ow. Therefore, the method of anaesthesia and correction of arterial carbon dioxide tension according to body temperature play a crucial role in intraoperative brain protection. The role of venous outow from the brain seems interesting, yet not well documented. Several experimental and clinical studies present the adverse effects of raised venous pressure in brain circulation [30,31]. Decreased venous outow from the brain impairs cerebral circulation, declines cerebral perfusion pressure (CPP) and increases brain blood barrier (BBB) permeability, which raises vascular water shift into the extracellular space resulting in cytotoxic and vasogenic oedema and extensive haemorrhagic cerebral infarction [28]. Moreover, increased brain venous pressure may cause transient ischaemia, which manifests as a transient ischaemic attack [30]. Interestingly, the elevated central venous pressure during cardiac surgery increases the risk of postoperative neurological complications [31]; however, the effect of brain venous hypertension has not been described and requires further studies. Postoperative hyperthermia is another risk factor of brain injury. Many clinicians focus on intra-operative hypothermia and disregard the re-warming period at the end of extracorporeal circulation. Fast re-warming of the brain and postoperative hyperthermia predispose to total cerebral ischaemia and stroke [13]. Therefore, the avoidance of full re-warming at the end of extracorporeal circulation is the main strategy to reduce the number of hypothermia events [2]. Systemic inammatory responses are also serious problems after extracorporeal circulation. Several authors suggest that an inammatory response following cardiac surgery with extracorporeal circulation plays a crucial role in the development of brain injury [16,32,33]. It has been well documented that cardiac surgery increases the plasma and cerebrospinal levels of cytokines, which strongly correlates with severe postoperative encephalopathy and other adverse neuropsychological disorders [3234]. The prolonged contact with an articial material surface of the
Current Opinion in Pharmacology 2012, 12:189194

extracorporeal circuit and ischaemia-reperfusion injury of organs activate the immunological reaction leading to disorders of vascular permeability and cellular structures [3336]. Interestingly, the use of anti-inammatory agents, such as corticosteroids, signicantly reduces postoperative neuropsychological disorders and brain injury [36]. Different cardiac arrhythmias, particularly atrial brillation, are a serious problem in patients after cardiac surgery. Atrial brillation episodes occur in 1040% of such patients, their frequency increases with age and the peak of its incidence is on the rst postoperative day. It potentially leads to many serious complications such as thromboembolism with its worst forms neurological disorders, haemodynamic disturbances, shock and higher early and late postoperative mortality [37,38]. There is strong evidence that atrial brillation precedes stroke in 36.5% of patients with cerebral ischaemic events [39]. Importantly, atrial brillation is not associated with the risk of postoperative stroke but it is the major independent predictor of brain injury during the postoperative period. Recently, several authors have studied the genetic predisposition to postoperative brain injury in cardiac surgery patients [4042]. The presence of the apolipoprotein E e4 allele is the highest genetic risk factor of postoperative stroke [40]. Additionally, minor alleles of C-reactive protein, a variant of the platelet glycoprotein IIb/III receptor and interleukin-6 are postulated as independent risk factors [41,42]. Nevertheless, our knowledge of genetic predisposition to postoperative brain injury is limited and further studies are needed to determine the mechanism and risk stratication for cardiac surgery. The main aims of brain protection are to reduce the number of sources leading to brain injury and to increase the brain tolerance to ischaemic events. Generally, there are two kinds of brain protection: non-pharmacologic and pharmacologic. The shortened duration of articial circuit and time spent in the operating theatre, reduced cardiac suction and avoidance of hyperthermia may strongly reduce adverse neurological disorders in the non-pharmacologic way. Several experimental studies documented that mild hypothermia reduced the risk of brain injury; however, clinical trials did not conrm that nding [43,44]. Nevertheless, many clinicians use mild hypothermia during extracorporeal circulation. A decrease in body temperature reduces cellular metabolism and energy consumption, inhibits the release of neurotransmitters and decreases the calcium inux into brain cells [45 47]. Moreover, deep hypothermia signicantly decreases the cerebral blood ow and number of microemboli [48,49,50]. On the contrary, patients re-warming favours brain injury and is associated with worse outcomes [50]. The maintenance of arterial carbon dioxide tension (alpha-stat acidbase management) at the level of
www.sciencedirect.com

Brain injury and heart surgery Dabrowski et al. 191

3538 mmHg prevents spasms of cerebral arteries and reduces postoperative neuropsychological disorders [51]. Similar effects are achieved by maintaining the adequate mean artery pressure of 5060 mmHg [52,53]. The pressure higher than 80 mmHg and lower than 50 should be avoided, irrespective of the disease and age. This pressure should be maintained at the undisturbed level, because the cerebral perfusion pressure strongly depends on the mean artery pressure. Rapid changes in mean artery pressure strongly predispose to brain injury, particularly during the disconnection of the extracorporeal machine. During the recent years, the cerebral oxygen saturation monitored continuously using near infrared spectroscopy is frequently used to maintain the adequate cerebral oxygen delivery during cardiac surgery. Increasing evidence reveals that decreased oxygen saturation below 50% associates with worse neurological outcomes [54,55]. This simple measurement gives much information about the brain status, especially its oxygen metabolism during extracorporeal circulation. Moreover, the continuous measurement of brain saturation allows fast correction of disturbances in brain oxygen delivery. Despite the non-pharmacologic protection, some anaesthetic agents should be used for brain protection. The volatile anaesthetics are one of them. The proposed mechanisms of volatile anaesthetics related neuroprotection are based on an increase in cerebral blood ow, slight increase in intracellular calcium, upregulation of bioenergy metabolism in brain, nitric oxide synthase and antiapoptotic factors as well as reduction in oxidative stress [56]. The use of volatile anaesthetics increases mitochondrial adenosine triphosphate-sensitive potassium channels (mitoKATP) activity leading to mild hyperpolarisation [5759]. The opened mitoKATP channel decreases mitochondrial energy consumption by ATP sparing, enlarging mitochondrial energy. Moreover, moderately increased intracellular calcium prevents large intracellular calcium inux and plays a crucial role in neuronal survivability after hypoxia/ ischaemia events [58]. Volatile anaesthetics cause the membrane depolarisation in the presynaptic mitochondria, also after the removal of extracellular calcium [57]. This increase in intracellular calcium concentration results from a decrease in ischaemic activity of calcium-calmodulin receptors, inhibition of N-methylD-aspartate (NMDA) and a decrease in glutamate release [5760]. Moreover, the use of volatile anaesthetics signicantly reduces the concentration of brain injury markers [61]. Similar effects are observed in patients receiving magnesium infusions. Magnesium plays an important role in the central nervous system. Its decline affects a number of secondary brain injury factors, including neurotransmitter release, ion changes, oxidative reaction and energy metabolism. Numerous studies have
www.sciencedirect.com

shown that magnesium is essential in CNS injuries [62]. Indeed, magnesium decreases after brain trauma, which is correlated with neuronal cell dysfunction. The moderate brain injury results in a decrease in intracellular brain free magnesium up to 60% [63]. Recently, several magnesium disorders in brain circulation have been documented during extracorporeal circulation [64]. Importantly, continuous supplementation of magnesium reduces substantially the plasma S100b protein concentration [65]. Moreover, intravenous infusions of magnesium reduce the incidence of atrial brillation, improve cardiac function and postoperative neuropsychological outcomes [64,66]. Nevertheless, the neuroprotective effect of magnesium is still open to debate, particularly in cardiac surgery patients. Moreover, the neuroprotective effect of lidocaine is still controversial. Indeed, infusion of antiarrhythmic dose of lidocaine before ischaemia onset increases the number of surviving neurons in the CA1 region of the hippocampus and preserves cognitive functions in experimental studies [67]. Some randomised, double-blind prospective clinical studies conrm neuroprotective effects of intra-operative infused lidocaine in cardiac surgery patients [68]. Some other studies document lack of them [69]. Therefore, the neuroprotective effect of lidocaine requires further clinical studies. Propofol is another interesting agent used for neuroprotection in cardiac surgery patients. It strongly inhibits the NMDA receptor, activates gamma amino-butyric acid (GABA) receptors and slightly modulates the intracellular calcium inux. By decreasing the internalisation of Dacid amino-3-hydroxy-5-methyl-4-isoxazole-propionic (AMPA) receptor glutamine R2 (GluR2) subunit, it reduces infarct size, promotes neurogenesis and improves spatial learning and memory in rats [70]. Likewise, clinical studies demonstrate its neuroprotective effect [71]. Contrarily, there is experimental evidence that propofol activates the mechanisms leading to cell death in the cortex and thalamus of the developing brain [72]. From this reason, propofol is recommended as an anaesthetic of choice in adult patients undergoing cardiac surgery. Several studies document the neuroprotective effect of barbiturates [73]. Barbiturates depress the brain metabolism, inhibit mitoKATP channel, facilitate protein synthesis, activate GABA receptors, and present antioxidative activity. Similarly to propofol, barbiturates cause a dosedependent neurodegeneration in the developing brain. The use of pentobarbital alters synaptic plasticity and leads to a long-lasting spatial learning decit [74]. Interestingly, the perioperative use of b-blockers improves postoperative neurological outcomes in cardiac surgery patients. b-blockers are known to have many
Current Opinion in Pharmacology 2012, 12:189194

192 Cardiovascular and renal

benets during non-cardiac and cardiac surgery. The neuroprotective action of this drug has been not fully known. However, several authors reported a signicant reduction of postoperative neuropsychological disorders in patients receiving b-blockers during the perioperative period [75,76]. It is recommended to continue b-blockers therapy until surgery with the last dose included as a part of oral premedication. Moreover, this effect is intensied by preoperative statin administration [75]. The analysis of 6813 patients undergoing cardiac surgery showed that such a combination has a strong neuroprotective effect. Therefore, it can be suggested to include statins to bblockers therapy, however their neuroprotective effect requires future studies. Several other drugs have been studies as potential neuroprotective agents in cardiac surgery patients. Amantadine and memantine, the antagonists of NMDA receptor, appear to have promising neuroprotective effects. They bind to NMDA with a higher afnity than magnesium and thus decrease the prolonged inux of calcium into the cellular space. Moreover, they increase dopamine release and block dopamine reuptake. Memantine moderately decreases clinical deterioration and protably affects the mood, behaviour and cognitive function [77]. Therefore, they can improve neuropsychological outcome, however, their neuroprotective effect has been completely undocumented in cardiac surgery patients. The ideal brain protection has been not specied in cardiac surgery patients. Despite several adverse effects of extracorporeal circulation, cardiac surgery is the only method of treatment of many cardiac diseases. Many neuroprotective techniques and pharmacologic methods are used to improve the quality of life; however, a signicant reduction of brain injury is still the subject of many experimental and clinical studies. Therefore, neuropsychological disorders and brain injury remain the main problem of cardiac surgery patients.

4. Ahonen J, Salmenpera M: Brain injury after adult cardiac surgery. Acta Anaesthesiol Scand 2004, 48:4-19.  In this important up-to-date review, the authors characterised and systematised types, incidences and consequences of postoperative neuropsychological damage, and analysed the main mechanisms of these pathologies in cardiac surgery patients. 5. van Dijk D, Moons KG, Keizer AM, Jansen EW, Hijman R, Diephuis JC, Borst C, de Jaegere PP, Grobbee DE, Kalkman CJ, Octopus Study Group: Association between early and three month cognitive outcome after off-pump and on-pump coronary artery bypass. Heart 2004, 90:431-434. Hernandez F, Brown JR, Likosky D, Clough RA, Hess AL, Roth RM, Ross CS, Whited CM, OConnor GT, Klemperer JD: Neurocognitive outcomes of off-pump versus on-pump coronary artery bypass: a prospective randomized controlled trial. Ann Thorac Surg 2007, 84:1897-1903. Grocott HP: Characteristics, incidence and pathomechanisms of neurological damage during cardiac surgery. Appl Cardiopulm Pathophysiol 2009, 13:20-26. Yin Y, Luo A, Gio XY, Li LH, Huang YG: Postoperative neuropsychological change and its underlying mechanism in patients undergoing coronary artery bypass grafting. Chin Med J 2007, 120:1951-1957. Murkin JM: Central nervous system dysfunction after cardiopulmonary bypass. In Cardiac Anaesthesia. Edited by 1259-1279. Kaplan JA. Philadelphia: W.B. Saunders Company; 1999:

6.

7.

8.

9.

10. Eagle KA, Guyton RA, Davidoff R, Edwards FH, Ewy GA,  Gardner TJ, Hart JC, Herrmann HC, Hillis LD, Hutter AM Jr et al.: ACC/AHA guidelines update for coronary artery bypass graft surgery: a report of the American College of Cardiology/ American Heart Association task force on practice guidelines (Committee to update the 1999 guidelines for coronary artery bypass graft surgery). Circulation 2004, 110:e340-e437. In this review article, the authors described the incidence of coronary artery diseases and current recommendation of treatment. They presented advantages and risk factors of surgical revascularisation of atherosclerotic heart disease as well as outcomes and quality of life after this surgery. Moreover, they characterised morbidity associated with coronary artery bypass graft surgery, classied the brain injuries following cardiac surgery and presented the current recommendation in preoperative prediction of postoperative complication. 11. Pugsey W, Klinger L, Paschalis C, Treasure T, Harrisin M, Newman S: The impact of microemboli during cardiopulmonary bypass on neuropsychological functioning. Stroke 1994, 25:1393-1399. kken SK, Russel D, Brucher R, Abdelnoor M, Svennevig JL: 12. Brc Cerebral microembolic signals during cardiopulmonary bypass surgery. Frequency, time of occurrence and association with patient and surgical characteristics. Stroke 1997, 28:1988-1992. 13. Grocott HP, Macensen GB, Grigore AM, Mathew J, Reves JG, Phillips-Bute B, Smith PK, Newman MF, Neurologic Outcome Research Group (NORG), Cardiothoracic Anesthesiology Research Endeavors (CARE) Investigators of the Duke Heart Center: Postoperative hyperthermia is associated with cognitive dysfunction after coronary artery bypass graft surgery. Stroke 2002, 33:537-541. 14. Mathew JP, Podgorenau MV, Grocott HP, White WD, Morris RW, Stafford-Smith M, MacKensen GB, Rinder CS, Blumenthal JA, Schwinn DA et al.: Genetic variants in P-selectine and Creactive protein inuence susceptibility to cognitive decline after cardiac surgery. J Am Coll Cardiol 2007, 49:1934-1942. 15. Hogue CW, Murphy SF, Schechtman KB, Davila-Roman VG: Risk factors for early or delayed stroke after cardiac surgery. Circulation 1999, 100:642-647. 16. Ramlawi B, Rudolph JL, Mieno S, Feng J, Boodhwani M, Khabbaz K, Levkoff SE, Marcantonio ER, Bianchi C, Sellke FW: Creactive protein and inammatory response associated to neurocognitive decline following cardiac surgery. Surgery 2006, 140:221-226. 17. Hogue CW, Creswell LL, Gutterman DD, Fleisher LA, American College of Chest Physicians: Epidemiology, mechanism and www.sciencedirect.com

References and recommended reading


Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest 1. Hogue CW, Palin CA, Arrowsmith JE: Cardiopulmonary bypass management and Neurologic outcomes: an evidence-based appraisal of current practices. Anesth Analg 2006, 103:21-37. Newman MF, Grocott HP, Mathew JP, White WD, Landolfo K, Reves JG, Laskowitz DT, Mark DB, Blumenthal JA, Neurologic Outcome research Group and Cardiothoracic Anesthesia Research Endeavors (CARE) Investigators of the Duke Heart Center: Report of the substudy assessing impact of neurocognitive function on quality of life 5 years after cardiac surgery. Stroke 2001, 32:2874-2881. Gottseman RF, Grega MA, Bailey MM, Pham LD, Zeger SL, Baumgartner WA, Selnes OA, McKhann GM: Delirium after coronary artery bypass graft surgery and late mortality. Ann Neurol 2010, 67:338-344.

2.

3.

Current Opinion in Pharmacology 2012, 12:189194

Brain injury and heart surgery Dabrowski et al. 193

risk: American College of Chest Physicians guidelines for the prevention and management of postoperative atrial brillation after cardiac surgery. Chest 2005, 128:9S-16S. 18. Litosky DS, Marrin CA, Caplan LR, Baribeau YR, Morton JR, Weintraub RM, Hartmen GS, Hernandez F Jr, Braff SP, Charlesworth DC et al.: Determination of etiologic mechanisms of strokes secondary to coronary artery bypass graft surgery. Stroke 2003, 34:2830-2834. 19. Caplan LR, Hennerici M: Impaired clearance of emboli (washout) is an important link between hypoperfusion, embolism and ischemic stroke. Arch Neurol 1998, 55:1475-1482. 20. Litosky DS, Caplan LR, Weintraub RM, Hartmen GS, Malenka DJ, Ross CS, Landis ES, Applebaum B, Braff SP, OConor GT, Northern New England Cardiovascular Disease Study Group, Lebanon, New Hampshire: Intraoperative and postoperative variables associated with strokes following cardiac surgery. Heart Surg Forum 2004, 7:E271-E276. 21. Roach GW, Kanchuger M, Mangano C, Newman M, Nussmeier N, Wolman R, Aggarwal A, Marschall K, Graham SH, Ley C: Adverse cerebral outcomes after coronary bypass surgery. Multicenter Study of Perioperative Ischemia Research Group and the Ischemia Research and Education Foundation Investigators. NEJM 1996, 335:1857-1863. 22. Kamenar E, Burger P: Cerebral fat embolism: a neuropathological study of microembolic stage. Stroke 1980, 11:477-484. 23. Brooker RF, Brown WR, Moody DM, Hammon JW Jr, Reboussin DM, Deal DD, Ghazi-Birry HS, Stump DA: Cardiotomy suction: a major source of brain lipid emboli during cardiopulmonary bypass. Ann Thorac Surg 1998, 65:1651-1655. 24. Carrascal Y, Guerro AL: Neurological damage related to cardiac surgery: pathophysiology, diagnostic tools and prevention strategies. Using actual knowledge for planning future. Neurologist 2010, 16:152-164. 25. Gopaldas RR, Chu D, Dao TK, Huh J, LeMaire SA, Lin P, Coselli JS, Bakaeen FG: Staged versus synchronous carotid endarterectomy and coronary artery bypass grafting: analysis of 10-year nationwide outcomes. Ann Thorac Surg 2011, 91:1323-1329. 26. Berens ES, Kouchoukos NT, Murphy SF, Wareing TH: Preoperative carotid screening in eldery patients undergoing cardiac surgery. J Vasc Surg 1992, 15:313-321. 27. Timaran CH, Rosero EB, Smith ST, Valentine RJ, Modrall JG, Claqett GP: Trends and outcome of concurrent carotid revascularization and coronary bypass. J Vasc Surg 2008, 42:355-360. 28. Perttila J, Salo M, Petola O: Comparison of the effect of centrifugal versus roller pump on the immune response in open-heart surgery. Perfusion 1995, 10:249-256. 29. Scott DA, Silbert BS, Doyle TJ, Blyth C, Borton MC, O/Brien JL, de L, Horne DJ: Centrifugal versus roller head pumps for cardiopulmonary bypass: effect on early neuropsychologic outcomes after coronary artery surgery. J Cardiothorac Vasc Anesth 2002, 16:715-722. 30. Usman U, Wasay M: Mechanism of neuronal injury in cerebral venous thrombosis. J Pak Med Assoc 2006, 56:509-512. 31. Zeitlhofer J, Asenbaum S, Spiss C, Wimmer A, Mayr N, Wolner E, Deecke L: Central nervous system function after cardiopulmonary bypass. Eur Heart J 1993, 14:885-890. 32. Ramlawi B, Rudolph JL, Mieno S, Khabbaz K, Sodha NR, Boodhwani M, Levkoff SE, Marcantonio ER, Sellke FW: Serologic markers of brain injury and cognitive function after cardiopulmonary bypass. Ann Surg 2006, 244:593-601. 33. Caputo M, Mokhtari A, Rogers CA, Panayitou N, Chen Q, Ghorbel MT, Angelini GD, Parry AJ: The effects of normoxic versus hyperoxic cardiopulmonary bypass on oxidative stress and inammatory response in cyanotic pediatric patients undergoing open cardiac surgery; a randomized controlled trial. J Thorac Cardiovasc Surg 2009, 138:206-214. www.sciencedirect.com

34. Gessler P, Schmitt B, Pretre R, Latal B: Inammatory response and neurodevelopmental outcome after open-heart surgery in children. Pediatr Cardiol 2009, 30:301-305. 35. Gao L, Taha R, Gauvin D, Othmen LB, Wang Y, Blaise G: Postoperative cognitive dysfunction after cardiac surgery. Chest 2005, 128:3664-3670. 36. Ben-Abraham R, Weinbroum AA, Dekel B, Paret G: Chemokines and the inammatory response following cardiopulmonary bypass: a new target for therapeutic intervention? A review. Paediatr Anaesth 2003, 13:655-661. 37. Weber R, Diener HC, Weimar C: Prevention of cardioembolic stroke in patients with atrial brillation. Expert Rev Cardiovasc Ther 2010, 8:1405-1415. 38. Funk M, Richards SB, Desjardns J, Bebon C, Wilcox H: Incidence, timing, symptoms and risk factors for atrial brillation after cardiac surgery. Am J Crit Care 2003, 12:424-433. 39. Lahtinen J, Biancari F, Salmela E, Mosorin M, Satta J, Rainio P, inen J, Lepoja rvi M, Juvonen T: Postoperative atrial Rimpila brillation is a major cause of stroke after off-pump coronary artery bypass surgery. Ann Thorac Surg 2004, 77:1241-1244. 40. Fuller S, Nord AS, Gerdes M, Wernowsky G, Jarvik GP, Bernbaum J, Zackai E, Gaynor JW: Predictors of impaired neurodevelopmental outcomes at one year of age after infant cardiac surgery. Eur J Cardiothorac Surg 2009, 36:40-47. 41. Grocott HP, White WD, Morris RW, Podgoreanu MV, Mathew JP, Nielsen DM, Schwinn DA, Newman MF, Perioperative Genetics and Safety Outcomes Study (PEGASUS) Investigate Team: Genetic polymorphisms and the risk of stroke after cardiac surgery. Stroke 2005, 36:1854-1858. 42. Mathew JP, Podgoreanu MV, Grocott HP, White WD, Morris RW, Stafford-Smith M, Mackensen GB, Rinder CS, Blumenthal JA, Schwinn DA et al.: Genetic variants in P-selectin and C-reactive protein inuebce susceptibility to cognitive decline after cardiac surgery. J Am Coll Cardiol 2007, 49:1934-1942. 43. Grigore AM, Mathew J, Grocott HP, Reves JG, Blumenthal JA, White WD, Smith PK, Jones RH, Kirchner JL, Mark DB et al.: Prospective randomized trial of normothermic versus hypothermic cardiopulmonary bypass on cognitive function after coronary artery bypass graft surgery. Anesthesiology 2001, 95:1110-1119. 44. Campos JM, Paniagua P: Hypothermia during cardiac surgery. Best Pract Res Clin Anaesthesiol 2008, 22:695-709. 45. Cavus E, Hoffman G, Bein B, Scheewe J, Meybohm P, Renner J, Scholz J, Boening A: Cerebral metabolism during deep hypothermic circulatory arrest vs moderate hypothermic selective cerebral perfusion in piglet model: a microdialysis study. Paerdiatr Anaesth 2009, 19:770-778. 46. Nordmark J, Enblad P, Rubertson S: Cerebral energy failure following experimental cardiac arrest. Hypothermia treatment reduces secondary lactate/puryvate-ratio increase. Resuscitation 2009, 80:573-579. 47. Tang XN, Yenari MA: Hypothermia as a cytoprotective strategy in ischemic tissue injury. Ageing Res Rev 2010, 9:61-68. 48. Paolin A, Michielon P, Betetto M, Satori G, Velfre C, Rodriquez G, Murkin M: Lower perfusion pressure during hypothermic cardiopulmonary bypass is associated with decreased cerebral blood ow and impaired memory performance 6 months postoperatively. Heart Surg Forum 2010, 13:E7-E12. 49. Grogan K, Stearns J, Hogue W: Brain protection in cardiac surgery. Anesthesiol Clin 2008, 26:521-538.  In this up-to-date review, the authors presented pathophysiology of brain injury in patients undergoing cardiac surgery and provided an analysis of methods reducing the occurrence of postoperative neuropsychological dysfunctions. 50. Nathan HJ, Rodriguez R, Wozny D, Dupuis JY, Rubens FD, Bryson GL, Wells GA: Neuroprotective effect of mild hypothermia in patients undergoing coronary artery surgery with cardiopulmonary bypass: ve-year follow-up of a randomized trial. J Thorac Cardiovasc Surg 2007, 133:1206-1211. Current Opinion in Pharmacology 2012, 12:189194

194 Cardiovascular and renal

51. Abdul Aziz KA, Meduoye A: Is ph-stat or alpha stat the best technique to follow in patients undergoing deep hypothermic circulatory arrest? Interact Cardiovasc Thorac Surg 2010, 10:271-282. 52. Brady KM, Mytar JO, Lee JK, Cameron DE, Vricella LA, Thompson WR, Hogue CW, Easley RB: Monitoring cerebral blood ow pressure autoregulation in pediatric patients during cardiac surgery. Stroke 2010, 41:1957-1962. 53. Wang S, Dai ZG, Dong XW, Guo SX, Liu Y, Wang ZP, Zeng YM: Duplicate preconditioning with sevourane in vitro improves neuroprotection in rat brain via activating the extracellular signal-regulated protein kinase. Neurosci Bull 2010, 26:437-444. 54. Murkin JM, Arango M: Near-infrared spectroscopy as an index of brain and tissue oxygenation. Br J Anaesth 2009, 103:i3-i13. 55. Schoen J, Heringlake M, Tiemeyer C, Husemann L, Paarmann H: The association between gender, cognitive function, markers of inammation and preoperative cerebral oxygen saturation in cohort of preoperative cardiac surgery patients. Appl Cardiopulm Pathophysiol 2011, 15:62-70. 56. Zhang J, Zhou W, Qiao H: Bioenergetic homeostasis decides neuroprotection or neurotoxicity induced by volatile anesthetics: a uniform mechanism of dual effects. Med Hypotheses 2011, 77:223-229. 57. Bains R, Moe MC, Larsen GA, Berg-Johnsen J, Vinje ML: Volatile anaesthetics depolarize neural mitochondria by inhibition of the electron transport chain. Acta Anaesth Scand 2006, 50:572-579. 58. Bickler PE, Fahlman CS: The inhalated anesthetic, isourane, enhances Ca+-dependent survival signaling in cortical neurons and modulates MAP kinases, apoptosis protein and transcription factors during hypoxia. Anesth Analg 2006, 103:419-429. 59. Kitamura T, Sato K, Kawamura G, Yamada Y: The involvement of adenosine triphosphate-sensitive potassium channels in the different effects of sevourane and propofol on glucose metabolism in fed rats. Anesth Analg 2012, 114:110-116. 60. Codaccioni JL, Velly LJ, Moubarik C, Bruder NJ, Pisano PS, Guillet BA: Sevourane preconditioning against focal cerebral ischemia. Anesthesiology 2009, 110:1271-1278. 61. Dabrowski W, Rzecki Z, Czajkowski M, Pilat J, Wacinski P,  Kotlinska E, Sztanke M, Sztanke K, Stazka K, Pasternak K: Volatile anesthetics reduce biochemical markers of brain injury and brain magnesium disorders in patients undergoing coronary artery bypass graft surgery. J Cardiothorac Vasc Anesth 2011. doi:org/10.1053/j.jvca.2011.10.014. The authors presented that isourane and sevourane signicantly decreased plasma glial brillary acidic protein (GFAP) and matrix metalloproteinase 9 (MMP-9) concentrations. Reduced brain injury markers concentration conrmed lower brain damage following cardiopulmonary bypass documenting the neuroprotective action of volatile anaesthetics. 62. Vink R, Cernak I: Regulation of intracellular free magnesium in central nervous system injury. Front Biosci 2000, 5:d656-d665. 63. Dabrowski W, Rzecki Z, Sztanke M, Visconti J, Wacinski P,  Pasternak K: The efciency of magnesium supplementation in patients undergoing cardiopulmonary bypass: changes in serum magnesium concentrations and atrial brillation episodes. Magnes Res 2008, 21:205-221. This study demonstrated the changes in plasma magnesium concentration in patients undergoing coronary artery bypass graft surgery and the most protable dose of MgSO4 (10 mg MgSO4 per min intravenously) correcting plasma magnesium disorders and reducing frequency of perioperative atrial brillation episodes. 64. Dabrowski W, Rzecki Z, Czajkowski M, Pilat J, Biernacka J, ka K, Sztanke M, Sztanke K: Plasma z Kotlinska E, Pasternak K, Sta Martix metalloproteinases 9 correlates with disorders of brain

magnesium homeostasis in patients undergoing coronary artery bypass surgery. Magnes Res 2010, 23:169-179. 65. Dabrowski W: Magnesium supplementation signicantly reduces serum S100beta concentrations in patients who have undergone coronary artery bypass surgery. Magnes Res 2009, 22:21-31. 66. Bhudia SK, Cosgrove DM, Nagle RI, Rajeswaran J, Lam B-K,  Walter E, Petrich J, Palumbo RC, Gillinov AM, Apperson-Hansen C et al.: Magnesium as a neuroprotectant in cardiac surgery: a randomized clinical trial. J Thorac Cardiovasc Surg 2006, 131:853-861. This is important study documented the protable effect of magnesium supplementation on postoperative neuropsychological disorders. The authors presented a signicant reduction of short and long-term neuropsychological dysfunction in patients receiving extra dose of magnesium during cardiopulmonary bypass. 67. Popp SS, Lei B, Kelemen E, Fenton AA, Cottrell JE, Kass IS: Intravenous antiarrhythmic doses of lidocaine increase the survival rate of CA1 neurons and improve cognitive outcome after transient global cerebral ischemia in rats. Neuroscience 2011, 192:537-549. 68. Wang D, Wu X, Li J, Xiao F, Liu X, Meng M: The effect of lidocaine on early postoperative cognitive dysfunction after coronary artery bypass surgery. Anesth Analg 2002, 95:1134-1141. 69. Mitchell SJ, Merry AF, Frampton C, Davies E, Grieve D, Mills BP, Webster CS, Milsom FP, Willcox TW, Gorman DF: Cerebral protection by lidocaine during cardiac operations: a follow-up study. Ann Thorac Surg 2009, 87:820-825. 70. Wang H, Luo M, Li C, Wang G: Propofol post-conditioning induced long-term neuroprotection and reduced internalization of AMPAR GluR2 subunit in a rat model of focal cerebral ischemia/reperfusion. J Neurochem 2011, 119:210-219. 71. Kotani Y, Shimazawa M, Yoshimura S, Iwama T, Hara H: The experimental and clinical pharmacology of propofol, an anesthetic agent with neuroprotective properties. CNS Neurosci Ther 2008, 14:95-106. V, Milanovic D, Tanic N, Popic J, Kanazir S, Jevtovic 72. Pesic S: Potential mechanism of cell death in the V, Ruzdijic Todorovic developing rat brain induced by propofol anesthesia. Int J Dev Neurosci 2009, 27:279-287. 73. Schilliti D, Grasso G, Conti A, Fodale V: Anaesthetic-related  neuroprotection. Intravenous or inhalational agents? CNS Drugs 2010, 24:893-907. In this up-to-date review, the authors performed a systematic review of articles published between January 1980 and April 2010, including studies in animals and humans describing pharmacological neuroprotection. They analysed advantages and disadvantages effect of volatile and intravenous anaesthetics in patients with traumatic brain injury. 74. Tachibana K, Hashimoto T, Kato R, Tsuruga K, Ito R, Morimoto Y: Long-lasting effects of neonatal pentobarbital administration on spatial learning and hippocampal synaptic plasticity. Brain Res 2011, 1388:69-76. 75. Bouchard D, Carrier M, Demers P, Cartier R, Pellerin M, Perrault LP, Lambert J: Statin in combination with b-blocker therapy reduces postoperative stroke after coronary artery bypass graft surgery. Ann Thorac Surg 2011, 91:654-659. 76. Amory DW, Grigore A, Amory JK, Gerhardt MA, White WD, Smith PK, Schwinn DA, Reves JG, Newman MF: Neuroprotection is associated with beta-adrenergic receptor antagonists during cardiac surgery: evidence from 2,575 patients. J Cardiothorac Vasc Anesth 2002, 16:270-277. 77. Hanney M, Prasher V, Williams N, Jones EL, Aarsland D, Corbett A, Lawrence D, Yu LM, Tyrer S, Francis PT et al.: Memantine for dementia in adults older than 40 years with Downs syndrome (MEADOWS): a randomised, double-blind, placebo-controlled trial. Lancet 2012 doi: 10.1016/S0140-6736(11)61676-0.

Current Opinion in Pharmacology 2012, 12:189194

www.sciencedirect.com

Das könnte Ihnen auch gefallen