全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Inflammatory Response in Patients under Coronary Artery Bypass Grafting Surgery and Clinical Implications: A Review of the Relevance of Dexmedetomidine Use

DOI: 10.1155/2014/905238

Full-Text   Cite this paper   Add to My Lib

Abstract:

Despite the fact that coronary artery bypass grafting surgery (CABG) with cardiopulmonary bypass (CPB) prolongs life and reduces symptoms in patients with severe coronary artery diseases, these benefits are accompanied by increased risks. Morbidity associated with cardiopulmonary bypass can be attributed to the generalized inflammatory response induced by blood-xenosurfaces interactions during extracorporeal circulation and the ischemia/reperfusion implications, including exacerbated inflammatory response resembling the systemic inflammatory response syndrome (SIRS). The use of specific anesthetic agents with anti-inflammatory activity can modulate the deleterious inflammatory response. Consequently, anti-inflammatory anesthetics may accelerate postoperative recovery and better outcomes than classical anesthetics. It is known that the stress response to surgery can be attenuated by sympatholytic effects caused by activation of central (α-)2-adrenergic receptor, leading to reductions in blood pressure and heart rate, and more recently, that they can have anti-inflammatory properties. This paper discusses the clinical significance of the dexmedetomidine use, a selective (α-)2-adrenergic agonist, as a coadjuvant in general anesthesia. Actually, dexmedetomidine use is not in anesthetic routine, but this drug can be considered a particularly promising agent in perioperative multiple organ protection. 1. Introduction 1.1. Inflammatory Response and Ischemia/Reperfusion in CABG Surgery Surgery induces a variety of metabolic, endocrine, and immune changes known as the “stress response," which may lead to prolonged in-hospital stay. The clinical manifestation of this reaction includes postoperative complications such as respiratory failure, wound infections [1], myocardial damage with contractile dysfunction, renal impairment, coagulopathy, neurologic dysfunction [2], and altered liver function with an increased mortality [3]. Inflammatory response in cardiac surgical patients is produced by complex interactions with numerous pathways including generation or activation of complement, cytokines, neutrophils, thrombin, mast cells, and other multiple inflammatory mediators. Cardiopulmonary bypass responses have often been compared with the pathophysiologic changes occurring in systemic inflammatory response syndrome (SIRS) [4] and remain not fully understood. Several interlinked mechanisms could play a role in the pathological effects associated with cardiopulmonary bypass, for instance, the exposure of blood to nonphysiologic surfaces, surgical trauma, anesthesia,

References

[1]  M. Sander, C. von Heymann, V. von Dossow et al., “Increased interleukin-6 after cardiac surgery predicts infection,” Anesthesia and Analgesia, vol. 102, no. 6, pp. 1623–1629, 2006.
[2]  J. M. Murkin, “Panvascular inflammation and mechanisms of injury in perioperative CNS outcomes,” Seminars in Cardiothoracic and Vascular Anesthesia, vol. 14, no. 3, pp. 190–195, 2010.
[3]  M. E. Plomondon, J. C. Cleveland Jr., S. T. Ludwig et al., “Off-pump coronary artery bypass is associated with improved risk-adjusted outcomes,” Annals of Thoracic Surgery, vol. 72, no. 1, pp. 114–119, 2001.
[4]  S. Wan, J.-L. Leclerc, and J.-L. Vincent, “Inflammatory response to cardiopulmonary bypass: mechanisms involved and possible therapeutic strategies,” Chest, vol. 112, no. 3, pp. 676–692, 1997.
[5]  M. M. Elahi, J. S. Khan, and B. M. Matata, “Deleterious effects of cardiopulmonary bypass in coronary artery surgery and scientific interpretation of off-pump's logic,” Acute Cardiac Care, vol. 8, no. 4, pp. 196–209, 2006.
[6]  B. M. Matata, A. W. Sosnowski, and M. Gali?anes, “Off-pump bypass graft operation significantly reduces oxidative stress and inflammation,” Annals of Thoracic Surgery, vol. 69, no. 3, pp. 785–791, 2000.
[7]  B. M. Matata and M. Gali?anes, “Cardiopulmonary bypass exacerbates oxidative stress but does not increase proinflammatory cytokine release in patients with diabetes compared with patients without diabetes: regulatory effects of exogenous nitric oxide,” Journal of Thoracic and Cardiovascular Surgery, vol. 120, no. 1, pp. 1–11, 2000.
[8]  E. P. Stover, L. C. Siegel, R. Parks et al., “Variability in transfusion practice for coronary artery bypass surgery persists despite national consensus guidelines: a 24-institution study,” Anesthesiology, vol. 88, no. 2, pp. 327–333, 1998.
[9]  S. C. Stamou, P. C. Hill, G. Dangas et al., “Stroke after coronary artery bypass: incidince, predictors, and clinical outcome,” Stroke, vol. 32, no. 7, pp. 1508–1512, 2001.
[10]  J. P. Mathew, R. Parks, J. S. Savino et al., “Atrial fibrillation following coronary artery bypass graft surgery: predictors, outcomes, and resource utilization. MultiCenter Study of Perioperative Ischemia Research Group,” Journal of the American Medical Association, vol. 276, no. 4, pp. 300–306, 1996.
[11]  E. A. Rose, “Off-pump coronary-artery bypass surgery,” The New England Journal of Medicine, vol. 348, no. 5, pp. 379–380, 2003.
[12]  R. Ascione, M. Caputo, and G. D. Angelini, “Off-pump coronary artery bypass grafting: not a flash in the pan,” Annals of Thoracic Surgery, vol. 75, no. 1, pp. 306–313, 2003.
[13]  J. E. Hall, T. D. Uhrich, J. A. Barney, S. R. Arain, and T. J. Ebert, “Sedative, amnestic, and analgesic properties of small-dose dexmedetomidine infusions,” Anesthesia and Analgesia, vol. 90, no. 3, pp. 699–705, 2000.
[14]  J. H. Gibbon Jr., “Application of a mechanical heart and lung apparatus to cardiac surgery,” Minnesota medicine, vol. 37, no. 3, pp. 171–185, 1954.
[15]  Y.-F. Chen, W.-C. Tsai, C.-C. Lin et al., “Effect of leukocyte depletion on endothelial cell activation and transendothelial migration of leukocytes during cardiopulmonary bypass,” Annals of Thoracic Surgery, vol. 78, no. 2, pp. 634–642, 2004.
[16]  T. Okamura, N. Ishibashi, D. Zurakowski, and R. A. Jonas, “Cardiopulmonary Bypass Increases Permeability of the Blood-Cerebrospinal Fluid Barrier,” Annals of Thoracic Surgery, vol. 89, no. 1, pp. 187–194, 2010.
[17]  O. J. Warren, A. J. Smith, C. Alexiou et al., “The inflammatory response to cardiopulmonary bypass: part 1—mechanisms of pathogenesis,” Journal of Cardiothoracic and Vascular Anesthesia, vol. 23, no. 2, pp. 223–231, 2009.
[18]  K. C. Wood, L. L. Hsu, and M. T. Gladwin, “Sickle cell disease vasculopathy: a state of nitric oxide resistance,” Free Radical Biology and Medicine, vol. 44, no. 8, pp. 1506–1528, 2008.
[19]  S. Kumar and U. Bandyopadhyay, “Free heme toxicity and its detoxification systems in human,” Toxicology Letters, vol. 157, no. 3, pp. 175–188, 2005.
[20]  D. J. Schaer, P. W. Buehler, A. I. Alayash, J. D. Belcher, and G. M. Vercellotti, “Hemolysis and free hemoglobin revisited: exploring hemoglobin and hemin scavengers as a novel class of therapeutic proteins,” Blood, vol. 121, pp. 1276–1284, 2013.
[21]  I. C. Vermeulen Windsant, S. J. Hanssen, W. A. Buurman, and M. J. Jacobs, “Cardiovascular surgery and organ damage: time to reconsider the role of hemolysis,” Journal of Thoracic and Cardiovascular Surgery, vol. 142, no. 1, pp. 1–11, 2011.
[22]  M. Haase, R. Bellomo, and A. Haase-Fielitz, “Novel biomarkers, oxidative stress, and the role of labile iron toxicity in cardiopulmonary bypass-associated acute kidney injury,” Journal of the American College of Cardiology, vol. 55, no. 19, pp. 2024–2033, 2010.
[23]  F. Farina, D. S. Davila, J. B. T. Rocha et al., “Metals, oxidative stress and neurodegeneration: a focus on iron, manganese and mercury,” Neurochemistry International, vol. 62, no. 5, pp. 575–594, 2012.
[24]  M. Haase, A. Haase-Fielitz, and R. Bellomo, “Cardiopulmonary bypass, hemolysis, free iron, acute kidney injury and the impact of bicarbonate,” Contributions to Nephrology, vol. 165, pp. 28–32, 2010.
[25]  C. Ritter, M. E. Andrades, A. Reinke, S. Menna-Barreto, J. C. F. Moreira, and F. Dal-Pizzol, “Treatment with N-acetylcysteine plus deferoxamine protects rats against oxidative stress and improves survival in sepsis,” Critical Care Medicine, vol. 32, no. 2, pp. 342–349, 2004.
[26]  M. Vulcano, R. P. Meiss, and M. A. Isturiz, “Deferoxamine reduces tissue injury and lethality in LPS-treated mice,” International Journal of Immunopharmacology, vol. 22, no. 8, pp. 635–644, 2000.
[27]  D. Vlahakos, N. Arkadopoulos, G. Kostopanagiotou et al., “Deferoxamine attenuates lipid peroxidation, blocks interleukin-6 production, ameliorates sepsis inflammatory response syndrome, and confers renoprotection after acute hepatic ischemia in pigs,” Artificial Organs, vol. 36, no. 4, pp. 400–408, 2012.
[28]  L. H. Edmunds, “Extracorporeal perfusion,” in Cardiac Surgery in the Adult, L. H. Edmunds, Ed., pp. 255–294, McGraw-Hill, New York, NY, USA, 1997.
[29]  M. Rossi, G. Sganga, M. Mazzone et al., “Cardiopulmonary bypass in man: role of the intestine in a self-limiting inflammatory response with demonstrable bacterial translocation,” Annals of Thoracic Surgery, vol. 77, no. 2, pp. 612–618, 2004.
[30]  B. Krishnadasam, J. Griscavage-Ennis, and G. S. Aldea, “Reperfusion injury during cardiopulmonary bypass,” in Leukocyte Depletion in Cardiac Surgery and Cardiology, G. Matheis, A. Moritz, and M. Scholz, Eds., p. 54, Karger, Basel, Switzerland, 2002.
[31]  N. B. Aydin, H. Gercekoglu, B. Aksu et al., “Endotoxemia in coronary artery bypass surgery: a comparison of the off-pump technique and conventional cardiopulmonary bypass,” Journal of Thoracic and Cardiovascular Surgery, vol. 125, no. 4, pp. 843–848, 2003.
[32]  H. M. Oudemans-van Straaten, P. G. M. Jansen, F. J. Hoek et al., “Intestinal permeability, circulating endotoxin, and postoperative systemic responses in cardiac surgery patients,” Journal of Cardiothoracic and Vascular Anesthesia, vol. 10, no. 2, pp. 187–194, 1996.
[33]  U. Koca, ?. G. Olguner, B. U. Ergür et al., “The effects of dexmedetomidine on secondary acute lung and kidney injuries in the rat model of intra-abdominal sepsis,” The Scientific World Journal, vol. 2013, Article ID 292687, 11 pages, 2013.
[34]  H. A. Vohra, R. Whistance, A. Modi, and S. K. Ohri, “The inflammatory response to miniaturised extracorporeal circulation: a review of the literature,” Mediators of Inflammation, vol. 2009, Article ID 707042, 7 pages, 2009.
[35]  P. Menasché, “The inflammatory response to cardiopulmonary bypass and its impact on post-operative myocardial function,” Current Opinion in Cardiology, vol. 10, pp. 597–604, 1995.
[36]  D. Journois, “Hemofiltration during cardiopulmonary bypass,” Kidney International, vol. 53, no. 66, pp. S-174–S-177, 1998.
[37]  G. Clermont, C. Vergely, S. Jazayeri et al., “Systemic free radical activation is a major event involved in myocardial oxidative stress related to cardiopulmonary bypass,” Anesthesiology, vol. 96, no. 1, pp. 80–87, 2002.
[38]  S. W. Davies, J. P. Duffy, D. G. Wickens et al., “Time-course of free radical activity during coronary artery operations with cardiopulmonary bypass,” Journal of Thoracic and Cardiovascular Surgery, vol. 105, no. 6, pp. 979–987, 1993.
[39]  P. E. Ballmer, W. H. Reinhart, P. Jordan, E. Buhler, U. K. Moser, and K. F. Gey, “Depletion of plasma vitamin C but not of vitamin E in response to cardiac operations,” Journal of Thoracic and Cardiovascular Surgery, vol. 108, no. 2, pp. 311–320, 1994.
[40]  H. J. Toivonen and M. Ahotupa, “Free radical reaction products and antioxidant capacity in arterial plasma during coronary artery bypass grafting,” Journal of Thoracic and Cardiovascular Surgery, vol. 108, no. 1, pp. 140–147, 1994.
[41]  A. Akila, B. D'souza, P. Vishwanath, and V. D'souza, “Oxidative injury and antioxidants in coronary artery bypass graft surgery: off-pump CABG significantly reduces oxidative stress,” Clinica Chimica Acta, vol. 375, no. 1-2, pp. 147–152, 2007.
[42]  J. Milei, P. Forcada, C. G. Fraga et al., “Relationship between oxidative stress, lipid peroxidation, and ultrastructural damage in patients with coronary artery disease undergoing cardioplegic arrest/reperfusion,” Cardiovascular Research, vol. 73, no. 4, pp. 710–719, 2007.
[43]  C. W. Hogue Jr., C. A. Palin, and J. E. Arrowsmith, “Cardiopulmonary bypass management and neurologic outcomes: an evidence-based appraisal of current practices,” Anesthesia and Analgesia, vol. 103, no. 1, pp. 21–37, 2006.
[44]  P. L. C. Smith, “The systemic inflammatory response to cardiopulmonary bypass and the brain,” Perfusion, vol. 11, no. 3, pp. 196–199, 1996.
[45]  J. M. Murkin, “Etiology and incidence of brain dysfunction after cardiac surgery,” Journal of Cardiothoracic and Vascular Anesthesia, vol. 13, no. 4, pp. 12–17, 1999.
[46]  F. de Lange, J. M. Dieleman, B. Jungwirth, and C. J. Kalkman, “Effects of cardiopulmonary bypass on neurocognitive performance and cytokine release in old and diabetic rats,” British Journal of Anaesthesia, vol. 99, no. 2, pp. 177–183, 2007.
[47]  S. Westaby, K. Saatvedt, S. White, T. Katsumata, W. van Oeveren, and P. W. Halligan, “Is there a relationship between cognitive dysfunction and systemic inflammatory response after cardiopulmonary bypass?” Annals of Thoracic Surgery, vol. 71, no. 2, pp. 667–672, 2001.
[48]  A. Parolari, M. Camera, F. Alamanni et al., “Systemic inflammation after on-pump and off-pump coronary bypass surgery: a one-month follow-up,” Annals of Thoracic Surgery, vol. 84, no. 3, pp. 823–828, 2007.
[49]  C. Coimba, M. Drake, F. Boris-Moller, et al., “Long-lasting neuroprotective effect of postischemic hypothermia and treatment with an antiimflammatory/antipyretic drug,” Stroke, vol. 27, pp. 1578–1585, 1996.
[50]  D. S. Warner, H. Sheng, and I. Batini?-Haberle, “Oxidants, antioxidants and the ischemic brain,” Journal of Experimental Biology, vol. 207, no. 18, pp. 3221–3231, 2004.
[51]  A. E. van Harten, T. W. L. Scheeren, and A. R. Absalom, “A review of postoperative cognitive dysfunction and neuroinflammation associated with cardiac surgery and anaesthesia,” Anaesthesia, vol. 67, no. 3, pp. 280–293, 2012.
[52]  P. W?hrborg, J. E. Booth, T. Clayton et al., “Neuropsychological outcome after percutaneous coronary intervention or coronary artery bypass grafting: results from the Stent or Surgery (SoS) trial,” Circulation, vol. 110, no. 22, pp. 3411–3417, 2004.
[53]  B. ?. Jensen, L. S. Rasmussen, and D. A. Steinbrüchel, “Cognitive outcomes in elderly high-risk patients 1 year after off-pump versus on-pump coronary artery bypass grafting. A randomized trial,” European Journal of Cardio-thoracic Surgery, vol. 34, no. 5, pp. 1016–1021, 2008.
[54]  D. van Dijk, M. Spoor, R. Hijman et al., “Cognitive and cardiac outcomes 5 years after off-pump vs on-pump coronary artery bypass graft surgery,” Journal of the American Medical Association, vol. 297, no. 7, pp. 701–708, 2007.
[55]  M. F. Newman, J. P. Mathew, H. P. Grocott et al., “Central nervous system injury associated with cardiac surgery,” The Lancet, vol. 368, no. 9536, pp. 694–703, 2006.
[56]  C. S. Ernest, B. M. Murphy, M. U. C. Worcester et al., “Cognitive function in candidates for coronary artery bypass graft surgery,” Annals of Thoracic Surgery, vol. 82, no. 3, pp. 812–818, 2006.
[57]  O. A. Selnes, M. A. Grega, M. M. Bailey et al., “Cognition 6 years after surgical or medical therapy for coronary artery disease,” Annals of Neurology, vol. 63, no. 5, pp. 581–590, 2008.
[58]  D. van Dijk, K. G. M. Moons, H. M. Nathoe et al., “Cognitive outcomes five years after not undergoing coronary artery bypass graft surgery,” Annals of Thoracic Surgery, vol. 85, no. 1, pp. 60–64, 2008.
[59]  O. A. Selnes, L. Pham, S. Zeger, and G. M. McKhann, “Defining cognitive change after CABG: decline versus normal variability,” Annals of Thoracic Surgery, vol. 82, no. 2, pp. 388–390, 2006.
[60]  D. van Dijk and C. J. Kalkman, “Why are cerebral microemboli not associated with cognitive decline?” Anesthesia and Analgesia, vol. 109, no. 4, pp. 1006–1008, 2009.
[61]  B. Jungwirth, B. Eckel, M. Blobner, K. Kellermann, E. F. Kochs, and G. B. Mackensen, “The impact of cardiopulmonary bypass on systemic interleukin-6 release, cerebral nuclear factor-kappa B expression, and neurocognitive outcome in rats,” Anesthesia and Analgesia, vol. 110, no. 2, pp. 312–320, 2010.
[62]  B. S. Silbert, D. A. Scott, L. A. Evered, M. S. Lewis, and P. T. Maruff, “Preexisting cognitive impairment in patients scheduled for elective coronary artery bypass graft surgery,” Anesthesia and Analgesia, vol. 104, no. 5, pp. 1023–1028, 2007.
[63]  F. Winningham-Major, J. L. Staecker, S. W. Barger, S. Coats, and L. J. van Eldik, “Neurite extension and neuronal survival activities of recombinant S100β proteins that differ in the content and position of cysteine residues,” Journal of Cell Biology, vol. 109, no. 6, pp. 3063–3071, 1989.
[64]  F. Tramontina, M. C. Leite, D. Gon?alves et al., “High glutamate decreases S100B secretion by a mechanism dependent on the glutamate transporter,” Neurochemical Research, vol. 31, no. 6, pp. 815–820, 2006.
[65]  R. Donato, G. Sorci, F. Riuzzi et al., “S100B's double life: intracellular regulator and extracellular signal,” Biochimica et Biophysica Acta, vol. 1793, no. 6, pp. 1008–1022, 2009.
[66]  D. F. de Souza, K. Wartchow, F. Hansen et al., “Interleukin-6-induced S100B secretion is inhibited by haloperidol and risperidone,” Progress in Neuro-Psychopharmacology & Biological Psychiatry, vol. 43, pp. 14–22, 2013.
[67]  D. F. de Souza, M. C. Leite, A. Quincozes-Santos et al., “S100B secretion is stimulated by IL-1β in glial cultures and hippocampal slices of rats: likely involvement of MAPK pathway,” Journal of Neuroimmunology, vol. 206, no. 1-2, pp. 52–57, 2009.
[68]  C. Beer, D. Blacker, M. Bynevelt, G. J. Hankey, and I. B. Puddey, “Systemic markers of inflammation are independently associated with S100B concentration: results of an observational study in subjects with acute ischaemic stroke,” Journal of Neuroinflammation, vol. 7, article 71, 2010.
[69]  A. Reichenberg, R. Yirmiya, A. Schuld et al., “Cytokine-associated emotional and cognitive disturbances in humans,” Archives of General Psychiatry, vol. 58, no. 5, pp. 445–452, 2001.
[70]  H. J. Huttunen, J. Kuja-Panula, G. Sorci, A. L. Agneletti, R. Donato, and H. Rauvala, “Coregulation of neurite outgrowth and cell survival by amphoterin and S100 proteins through receptor for advanced glycation end products (RAGE) activation,” Journal of Biological Chemistry, vol. 275, no. 51, pp. 40096–40105, 2000.
[71]  A. Bierhaus, P. M. Humpert, M. Morcos et al., “Understanding RAGE, the receptor for advanced glycation end products,” Journal of Molecular Medicine, vol. 83, no. 11, pp. 876–886, 2005.
[72]  C. Adami, G. Sorci, E. Blasi, A. L. Agneletti, F. Bistoni, and R. Donato, “S100B expression in and effects on microglia,” Glia, vol. 33, pp. 131–142, 2001.
[73]  J. Hu, F. Castets, J. L. Guevara, and L. J. van Eldiki, “S100β stimulates inducible nitric oxide synthase activity and mRNA levels in rat cortical astrocytes,” Journal of Biological Chemistry, vol. 271, no. 5, pp. 2543–2547, 1996.
[74]  T. V. Petrova, J. Hu, and L. J. van Eldik, “Modulation of glial activation by astrocyte-derived protein S100B: differential responses of astrocyte and microglial cultures,” Brain Research, vol. 853, no. 1, pp. 74–80, 2000.
[75]  J. Hu, A. Ferreira, and L. J. van Eldik, “S100β induces neuronal cell death through nitric oxide release from astrocytes,” Journal of Neurochemistry, vol. 69, no. 6, pp. 2294–2301, 1997.
[76]  S. H. Kim, C. J. Smith, and L. J. van Eldik, “Importance of MAPK pathways for microglial pro-inflammatory cytokine IL-1β production,” Neurobiology of Aging, vol. 25, no. 4, pp. 431–439, 2004.
[77]  S. Westaby, P. Johnsson, A. J. Parry et al., “Serum S100 protein: a potential marker for cerebral events during cardiopulmonary bypass,” Annals of Thoracic Surgery, vol. 61, no. 1, pp. 88–92, 1996.
[78]  S. Blomquist, P. Johnsson, C. Lührs et al., “The appearance of S-100 protein in serum during and immediately after cardiopulmonary bypass surgery: a possible marker for cerebral injury,” Journal of Cardiothoracic and Vascular Anesthesia, vol. 11, no. 6, pp. 699–703, 1997.
[79]  D. Barbut, F. S. Yao, D. N. Hager, P. Kavanaugh, R. R. Trifiletti, and J. P. Gold, “Comparison of transcranial Doppler ultrasonography and transesophageal echocardiography to monitor emboli during coronary artery bypass surgery,” Stroke, vol. 27, no. 1, pp. 87–90, 1996.
[80]  H. J?nsson, P. Johnsson, C. Alling, S. Westaby, and S. Blomquist, “Significance of serum S100 release after coronary artery bypass grafting,” Annals of Thoracic Surgery, vol. 65, no. 6, pp. 1639–1644, 1998.
[81]  H. P. Grocott, N. D. Croughwell, D. W. Amory et al., “Cerebral emboli and serum S100β during cardiac operations,” Annals of Thoracic Surgery, vol. 65, no. 6, pp. 1645–1650, 1998.
[82]  A. Jonsson, “As compared to neuron-specific enolase, S100B protein correlate more specific to the stroke volume and clinical outcome in ischemic stroke,” Kaka-Orinska, 2010.
[83]  B. C. van Munster, J. C. Korevaar, A. H. Zwinderman, M. Levi, W. J. Wiersinga, and S. E. De Rooij, “Time-course of cytokines during delirium in elderly patients with hip fractures,” Journal of the American Geriatrics Society, vol. 56, no. 9, pp. 1704–1709, 2008.
[84]  H. Worthmann, A. B. Tryc, A. Goldbecker et al., “The temporal profile of inflammatory markers and mediators in blood after acute ischemic stroke differs depending on stroke outcome,” Cerebrovascular Diseases, vol. 30, no. 1, pp. 85–92, 2010.
[85]  J. W. W. Thomason, A. Shintani, J. F. Peterson, B. T. Pun, J. C. Jackson, and E. W. Ely, “Intensive care unit delirium is an independent predictor of longer hospital stay: a prospective analysis of 261 non-ventilated patients,” Critical Care, vol. 9, no. 4, pp. R375–381, 2005.
[86]  P. Pandharipande, B. A. Cotton, A. Shintani et al., “Prevalence and risk factors for development of delirium in surgical and trauma intensive care unit patients,” The Journal of trauma, vol. 65, no. 1, pp. 34–41, 2008.
[87]  M. Lundstr?m, A. Edlund, G. Bucht, S. Karlsson, and Y. Gustafson, “Dementia after delirium in patients with femoral neck fractures,” Journal of the American Geriatrics Society, vol. 51, no. 7, pp. 1002–1006, 2003.
[88]  V. H. Perry, “The influence of systemic inflammation on inflammation in the brain: implications for chronic neurodegenerative disease,” Brain, Behavior, and Immunity, vol. 18, no. 5, pp. 407–413, 2004.
[89]  M. Herrmann, A. D. Ebert, I. Galazky, M. T. Wunderlich, W. S. Kunz, and C. Huth, “Neurobehavioral outcome prediction after cardiac surgery: role of neurobiochemical markers of damage to neuronal and glial brain tissue,” Stroke, vol. 31, no. 3, pp. 645–650, 2000.
[90]  Z. P. Khan, C. N. Ferguson, and R. M. Jones, “Alpha-2 and imidazoline receptor agonists. Their pharmacology and therapeutic role,” Anaesthesia, vol. 54, no. 2, pp. 146–165, 1999.
[91]  D. S. Carollo, B. D. Nossaman, and U. Ramadhyani, “Dexmedetomidine: a review of clinical applications,” Current Opinion in Anaesthesiology, vol. 21, no. 4, pp. 457–461, 2008.
[92]  J. C. Eisenach, M. de Kock, and W. Klimscha, “α2-Adrenergic agonists for regional anesthesia: a clinical review of clonidine (1984–1995),” Anesthesiology, vol. 85, no. 3, pp. 655–674, 1996.
[93]  M. Maze and W. Tranquilli, “Alpha-2 adrenoceptor agonists: defining the role in clinical anesthesia,” Anesthesiology, vol. 74, no. 3, pp. 581–605, 1991.
[94]  S. Fürst, “Transmitters involved in antinociception in the spinal cord,” Brain Research Bulletin, vol. 48, no. 2, pp. 129–141, 1999.
[95]  H. M. Loick, C. Schmidt, H. van Aken et al., “High thoracic epidural anesthesia, but not clonidine, attenuates the perioperative stress response via sympatholysis and reduces the release of troponin T in patients undergoing coronary artery bypass grafting,” Anesthesia and Analgesia, vol. 88, no. 4, pp. 701–709, 1999.
[96]  A. W. Wallace, D. Galindez, A. Salahieh et al., “Effect of clonidine on cardiovascular morbidity and mortality after noncardiac surgery,” Anesthesiology, vol. 101, no. 2, pp. 284–293, 2004.
[97]  R. D. Stevens, H. Burri, and M. R. Tramèr, “Efficacy of clonidine for prevention of perioperative myocardial ischemia: a critical appraisal and meta-analysis of the literature,” Anesthesia & Analgesia, vol. 97, pp. 623–633, 2003.
[98]  K. Nishina, K. Mikawa, T. Uesugi et al., “Efficacy of clonidine for prevention of perioperative myocardial ischemia: a critical appraisal and meta-analysis of the literature,” Anesthesiology, vol. 96, no. 2, pp. 323–329, 2002.
[99]  V. von Dossow, N. Baehr, M. Moshirzadeh et al., “Clonidine attenuated early proinflammatory response in T-cell subsets after cardiac surgery,” Anesthesia and Analgesia, vol. 103, no. 4, pp. 809–814, 2006.
[100]  J. E. Ellis, S. Pedlow, and J. Bains, “Premedication with clonidine does not attenuate suppression of certain lymphocyte subsets after surgery,” Anesthesia and Analgesia, vol. 87, no. 6, pp. 1426–1430, 1998.
[101]  T. Dorman, K. Clarkson, B. A. Rosenfeld, C. Shanholtz, P. A. Lipsett, and M. J. Breslow, “Effects of clonidine on prolonged postoperative sympathetic response,” Critical Care Medicine, vol. 25, no. 7, pp. 1147–1152, 1997.
[102]  P. J. Kulka, M. Tryba, and M. Zenz, “Dose-response effects of intravenous clonidine on stress response during induction of anesthesia in coronary artery bypass graft patients,” Anesthesia and Analgesia, vol. 80, no. 2, pp. 263–268, 1995.
[103]  N. D. Nader, T. A. Ignatowski, C. J. Kurek, P. R. Knight, and R. N. Spengler, “Clonidine suppresses plasma and cerebrospinal fluid concentrations of TNF-α during the perioperative period,” Anesthesia and Analgesia, vol. 93, no. 2, pp. 363–369, 2001.
[104]  J. Persec, Z. Persec, and I. Husedzinovic, “Postoperative pain and systemic inflammatory stress response after preoperative analgesia with clonidine or levobupivacaine: a randomized controlled trial,” Wiener Klinische Wochenschrift, vol. 121, no. 17-18, pp. 558–563, 2009.
[105]  A. T. Guerlach and J. F. Dasta, “Dexmedetomidine: an updated review,” Annals of Pharmacotherapy, vol. 41, pp. 245–252, 2007.
[106]  D. Ma, N. Rajakumaraswamy, and M. Maze, “α2-adrenoceptor agonists: shedding light on neuroprotection?” British Medical Bulletin, vol. 71, pp. 77–92, 2005.
[107]  V. Fagerholm, M. Scheinin, and M. Haaparanta, “α2A-Adrenoceptor antagonism increases insulin secretion and synergistically augments the insulinotropic effect of glibenclamide in mice,” British Journal of Pharmacology, vol. 154, no. 6, pp. 1287–1296, 2008.
[108]  K. Takada, D. J. Clark, M. F. Davies et al., “Meperidine exerts agonist activity at the α2B-adrenoceptor subtype,” Anesthesiology, vol. 96, no. 6, pp. 1420–1426, 2002.
[109]  V. Fagerholm, J. Rokka, L. Nyman et al., “Autoradiographic characterization of α2C-adrenoceptors in the human striatum,” Synapse, vol. 62, no. 7, pp. 508–515, 2008.
[110]  E. Moura, J. Afonso, L. Hein, and M. A. Vieira-Coelho, “α2-adrenoceptor subtypes involved in the regulation of catecholamine release from the adrenal medulla of mice,” British Journal of Pharmacology, vol. 149, no. 8, pp. 1049–1058, 2006.
[111]  I. Takamatsu, A. Iwase, M. Ozaki, T. Kazama, K. Wada, and M. Sekiguchi, “Dexmedetomidine reduces long-term potentiation in mouse hippocampus,” Anesthesiology, vol. 108, no. 1, pp. 94–102, 2008.
[112]  R. M. Venn, M. D. Karol, and R. M. Grounds, “Pharmacokinetics of dexmedetomidine infusions for sedation of postoperative patients requiring intensive care,” British Journal of Anaesthesia, vol. 88, no. 5, pp. 669–675, 2002.
[113]  A. M. de Wolf, R. J. Fragen, M. J. Avram, P. C. Fitzgerald, and F. Rahimi-Danesh, “The pharmacokinetics of dexmedetomidine in volunteers with severe renal impairment,” Anesthesia and Analgesia, vol. 93, no. 5, pp. 1205–1209, 2001.
[114]  P. Talke, C. A. Richardson, M. Scheinin, and D. M. Fisher, “Postoperative pharmacokinetics and sympatholytic effects of dexmedetomidine,” Anesthesia and Analgesia, vol. 85, no. 5, pp. 1136–1142, 1997.
[115]  A. Kallio, M. Scheinin, M. Koulu et al., “Effects of dexmedetomidine, a selective α2-adrenoceptor agonist, on hemodynamic control mechanisms,” Clinical Pharmacology and Therapeutics, vol. 46, no. 1, pp. 33–42, 1989.
[116]  T. J. Ebert, J. E. Hall, J. A. Barney, T. D. Uhrich, and M. D. Colinco, “The effects of increasing plasma concentrations of dexmedetomidine in humans,” Anesthesiology, vol. 93, no. 2, pp. 382–394, 2000.
[117]  V. S. B. Jorden, R. M. Pousman, M. M. Sanford, P. A. J. Thorborg, and M. P. Hutchens, “Dexmedetomidine overdose in the perioperative setting,” Annals of Pharmacotherapy, vol. 38, no. 5, pp. 803–807, 2004.
[118]  M. A. E. Ramsay and D. L. Luterman, “Dexmedetomidine as a total intravenous anesthetic agent,” Anesthesiology, vol. 101, no. 3, pp. 787–790, 2004.
[119]  J. Sleigh, “All hands on dex,” Anaesthesia, vol. 67, pp. 1193–1197, 2012.
[120]  M. Aho, O. Erkola, A. Kallio, H. Scheinin, and K. Korttila, “Dexmedetomidine infusion for maintenance of anesthesia in patients undergoing abdominal hysterectomy,” Anesthesia and Analgesia, vol. 75, no. 6, pp. 940–946, 1992.
[121]  H. Ishii, T. Kohno, T. Yamakura, M. Ikoma, and H. Baba, “Action of dexmedetomidine on the substantia gelatinosa neurons of the rat spinal cord,” European Journal of Neuroscience, vol. 27, no. 12, pp. 3182–3190, 2008.
[122]  L. E. Nelson, T. You, M. Maze, and N. P. Franks, “Evidence that the mechanism of hypnotic action in dexmedetomidine and muscimol-induced anesthesia converges on the endogenous sleep pathway,” Anesthesiology, vol. 95, p. A1368, 2001.
[123]  R. R. Al-Metwalli, H. A. Mowafi, S. A. Ismail et al., “Effect of intra-articular dexmedetomidine on postoperative analgesia after arthroscopic knee surgery,” British Journal of Anaesthesia, vol. 101, no. 3, pp. 395–399, 2008.
[124]  T. Yoshitomi, A. Kohjitani, S. Maeda, H. Higuchi, M. Shimada, and T. Miyawaki, “Dexmedetomidine enhances the local anesthetic action of lidocaine via an α-2a adrenoceptor,” Anesthesia and Analgesia, vol. 107, no. 1, pp. 96–101, 2008.
[125]  A. Turkmen, A. Altan, N. Turgut, S. Vatansever, and S. Gokkaya, “The correlation between the richmond agitation-sedation scale and bispectral index during dexmedetomidine sedation,” European Journal of Anaesthesiology, vol. 23, no. 4, pp. 300–304, 2006.
[126]  R. Aantaa, “Assessment of the sedative effects of dexmedetomidine, an α2-adrenoceptor agonist, with analysis of saccadic eye movements,” Pharmacology and Toxicology, vol. 68, no. 5, pp. 394–398, 1991.
[127]  W. J. Elias, M. E. Durieux, D. Huss, and R. C. Frysinger, “Dexmedetomidine and arousal affect subthalamic neurons,” Movement Disorders, vol. 23, no. 9, pp. 1317–1320, 2008.
[128]  Y.-W. Hsu, L. I. Cortinez, K. M. Robertson et al., “Dexmedetomidine pharmacodynamics: part I—crossover comparison of the respiratory effects of dexmedetomidine and remifentanil in healthy volunteers,” Anesthesiology, vol. 101, no. 5, pp. 1066–1076, 2004.
[129]  J. T. Coull, M. E. P. Jones, T. D. Egan, C. D. Frith, and M. Maze, “Attentional effects of noradrenaline vary with arousal level: selective activation of thalamic pulvinar in humans,” NeuroImage, vol. 22, no. 1, pp. 315–322, 2004.
[130]  E. Deutsch and J. D. Tobias, “Hemodynamic and respiratory changes following dexmedetomidine administration during general anesthesia: sevoflurane vs desflurane,” Paediatric Anaesthesia, vol. 17, no. 5, pp. 438–444, 2007.
[131]  A. Koroglu, H. Teksan, O. Sagir, A. Yucel, H. I. Toprak, and O. M. Ersoy, “A comparison of the sedative, hemodynamic, and respiratory effects of dexmedetomidine and propofol in children undergoing magnetic resonance imaging,” Anesthesia and Analgesia, vol. 103, no. 1, pp. 63–67, 2006.
[132]  P. A. Arpino, K. Kalafatas, and B. T. Thompson, “Feasibility of dexmedetomidine in facilitating extubation in the intensive care unit,” Journal of Clinical Pharmacy and Therapeutics, vol. 33, no. 1, pp. 25–30, 2008.
[133]  T. B. Corcoran, A. Engel, H. Sakamoto, A. O'Shea, S. O'Callaghan-Enright, and G. D. Shorten, “The effects of propofol on neutrophil function, lipid peroxidation and inflammatory response during elective coronary artery bypass grafting in patients with impaired ventricular function,” British Journal of Anaesthesia, vol. 97, no. 6, pp. 825–831, 2006.
[134]  M. Arslan, F. M. ?omu, A. Ku?uk, L. Ozturk, and F. Yaylak, “Dexmedetomidine protects against lipid peroxidation and erythrocyte deformability alterations in experimental hepatic ischemia reperfusion injury,” Libyan Journal of Medicine, vol. 7, article 18185, 2012.
[135]  J. B. T. Rocha, N. M. Heinzmann Bulow, E. F. M. Correa, C. Scholze, C. W. Nogueira, and N. B. Barbosa, “Dexmedetomidine protects blood γ-aminolevulinate dehydratase from inactivation caused by hyperoxygenation in total intravenous anesthesia,” Human and Experimental Toxicology, vol. 30, no. 4, pp. 289–295, 2011.
[136]  R. H. Straub, M. Herrmann, G. Berkmiller et al., “Neuronal regulation of interleukin 6 secretion in murine spleen: adrenergic and opioidergic control,” Journal of Neurochemistry, vol. 68, no. 4, pp. 1633–1639, 1997.
[137]  J. Szelényi, J. P. Kiss, and E. S. Vizi, “Differential involvement of sympathetic nervous system and immune system in the modulation of TNF-α production by α2- and β-adrenoceptors in mice,” Journal of Neuroimmunology, vol. 103, no. 1, pp. 34–40, 2000.
[138]  T. Taniguchi, Y. Kidani, H. Kanakura, Y. Takemoto, and K. Yamamoto, “Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats,” Critical Care Medicine, vol. 32, no. 6, pp. 1322–1326, 2004.
[139]  S. Hofer, J. Steppan, T. Wagner et al., “Central sympatholytics prolong survival in experimental sepsis,” Critical Care, vol. 13, no. 1, article R11, 2009.
[140]  M. Can, S. Gul, S. Bektas, V. Hanci, and S. Acikgoz, “Effects of dexmedetomidine or methylprednisolone on inflammatory responses in spinal cord injury,” Acta Anaesthesiologica Scandinavica, vol. 53, no. 8, pp. 1068–1072, 2009.
[141]  M. Maze, R. Virtanen, D. Daunt, S. J. M. Banks, E. P. Stover, and D. Feldman, “Effects of dexmedetomidine, a novel imidazole sedative-anesthetic agent, on adrenal steroidogenesis: in vivo and in vitro studies,” Anesthesia and Analgesia, vol. 73, no. 2, pp. 204–208, 1991.
[142]  R. M. Venn, A. Bryant, G. M. Hall, and R. M. Grounds, “Effects of dexmedetomidine on adrenocortical function, and the cardiovascular, endocrine and inflammatory responses in post-operative patients needing sedation in the intensive care unit,” British Journal of Anaesthesia, vol. 86, no. 5, pp. 650–656, 2001.
[143]  N. M. H. Bulow, N. B. V. Barbosa, and J. B. T. Rocha, “Opioid consumption in total intravenous anesthesia is reduced with dexmedetomidine: a comparative study with remifentanil in gynecologic videolaparoscopic surgery,” Journal of Clinical Anesthesia, vol. 19, no. 4, pp. 280–285, 2007.
[144]  P. P. Pandharipande, B. T. Pun, D. L. Herr et al., “Effect of sedation with dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial,” Journal of the American Medical Association, vol. 298, no. 22, pp. 2644–2653, 2007.
[145]  A. T. Gerlach, C. V. Murphy, and J. F. Dasta, “An updated focused review of dexmedetomidine in adults,” Annals of Pharmacotherapy, vol. 43, no. 12, pp. 2064–2074, 2009.
[146]  S. Sulaiman, R. B. Karthekeyan, M. Vakamudi, A. S. Sundar, H. Ravullapalli, and R. Gandham, “The effects of dexmedetomidine on attenuation of stress response to endotracheal intubation in patients undergoing elective off-pump coronary artery bypass grafting,” Annals of Cardiac Anaesthesia, vol. 15, no. 1, pp. 39–43, 2012.
[147]  B. Scheinin, L. Lindgren, T. Randell, H. Scheinin, and M. Scheinin, “Dexmedetomidine attenuates sympathoadrenal responses to tracheal intubation and reduces the need for thiopentone and peroperative fentanyl,” British Journal of Anaesthesia, vol. 68, no. 2, pp. 126–131, 1992.
[148]  S. Sukegawa, M. Inoue, H. Higuchi, Y. Tomoyasu, S. Maeda, and T. Miyawaki, “Locally injected dexmedetomidine inhibits carrageenin-induced inflammatory reactions in injected region,” in Proceedings of the American Society of Anesthesiologists Annual Meeting, 2011, A1590.
[149]  Y.-C. Lai, P.-S. Tsai, and C.-J. Huang, “Effects of dexmedetomidine on regulating endotoxin-induced up-regulation of inflammatory molecules in murine macrophages,” Journal of Surgical Research, vol. 154, no. 2, pp. 212–219, 2009.
[150]  H. Yagmurdur, N. Ozcan, F. Dokumaci, K. Kilinc, F. Yilmaz, and H. Basar, “Dexmedetomidine reduces the ischemia-reperfusion injury markers during upper extremity surgery with tourniquet,” Journal of Hand Surgery, vol. 33, no. 6, pp. 941–947, 2008.
[151]  A. Bekker, M. Haile, R. Kline et al., “The effect of intraoperative infusion of dexmedetomidine on the quality of recovery after major spinal surgery,” Journal of Neurosurgical Anesthesiology, vol. 25, no. 1, pp. 16–24, 2012.
[152]  J. Gu, J. Chen, P. Xia, G. Tao, H. Zhao, and D. Ma, “Dexmedetomidine attenuates remote lung injury induced by renal ischemia-reperfusion in mice,” Acta Anaesthesiologica Scandinavica, vol. 55, no. 10, pp. 1272–1278, 2011.
[153]  X. Wu, X. Song, N. Li, L. Zhan, Q. Meng, and Z. Xia, “Protective effects of dexmedetomidine on blunt chest trauma-induced pulmonary contusion in rats,” Journal of Trauma and Acute Care Surgery, vol. 74, pp. 524–530, 2013.
[154]  S.-H. Kang, Y.-S. Kim, T.-H. Hong et al., “Effects of dexmedetomidine on inflammatory responses in patients undergoing laparoscopic cholecystectomy,” Acta Anaesthesiologica Scandinavica, vol. 57, pp. 480–487, 2013.
[155]  M. Tasdogan, D. Memis, N. Sut, and M. Yuksel, “Results of a pilot study on the effects of propofol and dexmedetomidine on inflammatory responses and intraabdominal pressure in severe sepsis,” Journal of Clinical Anesthesia, vol. 21, no. 6, pp. 394–400, 2009.
[156]  R. D. Sanders, D. Ma, and M. Maze, “Anaesthesia induced neuroprotection,” Best Practice and Research, vol. 19, no. 3, pp. 461–474, 2005.
[157]  E. L. Janke and S. Samra, “Dexmedetomidine and neuroprotection,” Seminars in Anesthesia, Perioperative Medicine and Pain, vol. 25, no. 2, pp. 71–76, 2006.
[158]  M. F. Newman, “Open heart surgery and cognitive decline,” Cleveland Clinic journal of medicine, vol. 74, pp. S52–S55, 2007.
[159]  N. Stroobant, G. van Nooten, D. de Bacquer, Y. van Belleghem, and G. Vingerhoets, “Neuropsychological functioning 3-5 years after coronary artery bypass grafting: does the pump make a difference?” European Journal of Cardio-thoracic Surgery, vol. 34, no. 2, pp. 396–401, 2008.
[160]  H. P. Grocott, “Genetic influences on cerebral outcome after cardiac surgery,” Seminars in Cardiothoracic and Vascular Anesthesia, vol. 10, no. 4, pp. 291–296, 2006.
[161]  K. M. Taylor, “Central nervous system effects of cardiopulmonary bypass,” Annals of Thoracic Surgery, vol. 66, no. 5, pp. S20–S24, 1998.
[162]  C. W. Hogue Jr., T. Hershey, D. Dixon et al., “Preexisting cognitive impairment in women before cardiac surgery and its relationship with C-reactive protein concentrations,” Anesthesia and Analgesia, vol. 102, no. 6, pp. 1602–1608, 2006.
[163]  P. M. Ho, D. B. Arciniegas, J. Grigsby et al., “Predictors of cognitive decline following coronary artery bypass graft surgery,” Annals of Thoracic Surgery, vol. 77, no. 2, pp. 597–603, 2004.
[164]  C. G. Koch, L. Li, M. Shishehbor et al., “Socioeconomic status and comorbidity as predictors of preoperative quality of life in cardiac surgery,” Journal of Thoracic and Cardiovascular Surgery, vol. 136, no. 3, pp. 665–672, 2008.
[165]  N. Stroobant and G. Vingerhoets, “Depression, anxiety, and neuropsychological performance in coronary artery bypass graft patients: a follow-up study,” Psychosomatics, vol. 49, no. 4, pp. 326–331, 2008.
[166]  R. Ille, T. Lahousen, S. Schweiger, P. Hofmann, and H.-P. Kapfhammer, “Influence of patient-related and surgery-related risk factors on cognitive performance, emotional state, and convalescence after cardiac surgery,” Cardiovascular Revascularization Medicine, vol. 8, no. 3, pp. 166–169, 2007.
[167]  N. A. Nussmeier and B. E. Searles, “Inflammatory brain injury after cardiopulmonary bypass: is it real?” Anesthesia and Analgesia, vol. 110, no. 2, pp. 288–290, 2010.
[168]  S. Singh, P. Kapoor, U. Chowdhury, and U. Kiran, “Comparison of S100 levels, and their correlation with hemodynamic indices in patients undergoing coronary artery bypass grafting with three different anesthetic techniques,” Annals of Cardiac Anaesthesia, vol. 14, no. 3, pp. 197–202, 2011.
[169]  A. Kleindienst, F. Hesse, M. R. Bullock, and M. Buchfelder, “The neurotrophic protein S100B: value as a marker of brain damage and possible therapeutic implications,” Progress in Brain Research, vol. 161, pp. 317–325, 2007.
[170]  R. R. Riker, Y. Shehabi, P. M. Bokesch et al., “Dexmedetomidine vs midazolam for sedation of critically Ill patients A randomized trial,” Journal of the American Medical Association, vol. 301, no. 5, pp. 489–499, 2009.
[171]  J. R. Maldonado, A. Wysong, P. J. A. van der Starre, T. Block, C. Miller, and B. A. Reitz, “Dexmedetomidine and the reduction of postoperative delirium after cardiac surgery,” Psychosomatics, vol. 50, no. 3, pp. 206–217, 2009.
[172]  E. Ruokonen, I. Parviainen, S. M. Jakob et al., “Dexmedetomidine versus propofol/midazolam for long-term sedation during mechanical ventilation,” Intensive Care Medicine, vol. 35, no. 2, pp. 282–290, 2009.
[173]  W. Mayo, V. Lemaire, J. Malaterre et al., “Pregnenolone sulfate enhances neurogenesis and PSA-NCAM in young and aged hippocampus,” Neurobiology of Aging, vol. 26, no. 1, pp. 103–114, 2005.
[174]  E. A. Keller, A. Zamparini, L. N. Borodinsky, M. C. Gravielle, and M. L. Fiszman, “Role of allopregnanolone on cerebellar granule cells neurogenesis,” Developmental Brain Research, vol. 153, no. 1, pp. 13–17, 2004.
[175]  C. Correa-Sales, B. C. Rabin, and M. Maze, “A hypnotic response to dexmedetomidine, an α2 agonist, is mediated in the locus coeruleus in rats,” Anesthesiology, vol. 76, no. 6, pp. 948–952, 1992.
[176]  V. A. Kulkarni, S. Jha, and V. A. Vaidya, “Depletion of norepinephrine decreases the proliferation, but does not influence the survival and differentiation, of granule cell progenitors in the adult rat hippocampus,” European Journal of Neuroscience, vol. 16, no. 10, pp. 2008–2012, 2002.
[177]  P. Mohapel, G. Leanza, M. Kokaia, and O. Lindvall, “Forebrain acetylcholine regulates adult hippocampal neurogenesis and learning,” Neurobiology of Aging, vol. 26, no. 6, pp. 939–946, 2005.
[178]  A. Tung, S. Herrera, C. A. Fornal, and B. L. Jacobs, “The effect of prolonged anesthesia with isoflurane, propofol, dexmedetomidine, or ketamine on neural cell proliferation in the adult rat,” Anesthesia and Analgesia, vol. 106, no. 6, pp. 1772–1777, 2008.
[179]  W. E. Hoffman, E. Kochs, C. Werner, C. Thomas, and R. F. Albrecht, “Dexmedetomidine improves neurologic outcome from incomplete ischemia in the rat. Reversal by the α2-adrenergic antagonist atipamezole,” Anesthesiology, vol. 75, no. 2, pp. 328–332, 1991.
[180]  W. E. Hoffman, V. L. Baughman, and R. F. Albrecht, “Interaction of catecholamines and nitrous oxide ventilation during incomplete brain ischemia in rats,” Anesthesia and Analgesia, vol. 77, no. 5, pp. 908–912, 1993.
[181]  C. Maier, G. K. Steinberg, G. H. Sun, G. T. Zhi, and M. Maze, “Neuroprotection by the α2-adrenoreceptor agonist dexmedetomidine in a focal model of cerebral ischemia,” Anesthesiology, vol. 79, no. 2, pp. 306–312, 1993.
[182]  J. Kuhmonen, J. Pokorny, R. Miettinen et al., “Neuroprotective effects of dexmedetomidine in the gerbil hippocampus after transient global ischemia,” Anesthesiology, vol. 87, no. 2, pp. 371–377, 1997.
[183]  V. Laudenbach, J. Mantz, H. Lagercrantz, J.-M. Desmonts, P. Evrard, and P. Gressens, “Effects of α2-adrenoceptor agonists on perinatal excitotoxic brain injury: comparison of clonidine and dexmedetomidine,” Anesthesiology, vol. 96, no. 1, pp. 134–141, 2002.
[184]  L. Peng, A. C. H. Yu, K. Y. Fung, V. Prévot, and L. Hertz, “α-adrenergic stimulation of ERK phosphorylation in astrocytes is α2-specific and may be mediated by transactivation,” Brain Research, vol. 978, no. 1-2, pp. 65–71, 2003.
[185]  B. Li, T. Du, H. Li et al., “Signalling pathways for transactivation by dexmedetomidine of epidermal growth factor receptors in astrocytes and its paracrine effect on neurons,” British Journal of Pharmacology, vol. 154, no. 1, pp. 191–203, 2008.
[186]  L. Hertz, D. Lovatt, S. A. Goldman, and M. Nedergaard, “Adrenoceptors in brain: cellular gene expression and effects on astrocytic metabolism and [Ca2+]i,” Neurochemistry International, vol. 57, no. 4, pp. 411–420, 2010.
[187]  L. Peng, T. Du, J. Xu, et al., “Adrenergic and V1-ergic agonists/antagonists affecting recovery from Brain Trauma in the Lund project Act on astrocytes,” Current Signal Transduction Therapy, vol. 7, pp. 43–55, 2012.
[188]  L. Liu, F. Ji, J. Liang, H. He, Y. Fu, and M. Cao, “Inhibition by dexmedetomidine of the activation of spinal dorsal horn glias and the intracellular ERK signaling pathway induced by nerve injury,” Brain Research, vol. 1427, pp. 1–9, 2012.
[189]  B. Xu, W. S. Zhang, J. L. Yang et al., “Evidence for suppression of spinal glial activation by dexmedetomidine in a rat model of monoarthritis.,” Clinical and Experimental Pharmacology and Physiology, vol. 37, pp. 158–166, 2010.
[190]  C. Chrysostomou and C. G. Schmitt, “Dexmedetomidine: sedation, analgesia and beyond,” Expert Opinion on Drug Metabolism and Toxicology, vol. 4, no. 5, pp. 619–627, 2008.
[191]  M. Schoeler, P. D. Loetscher, R. Rossaint et al., “Dexmedetomidine is neuroprotective in an in vitro model for traumatic brain injury,” BMC Neurology, vol. 12, p. 20, 2012.
[192]  M. Zhang, X. Shan, L. Gu, L. Hertz, and L. Peng, “Dexmedetomidine causes neuroprotection via astrocytic α2-adrenergic receptor stimulation and HB-EGF release,” Journal of Anesthesiology & Clinical Science, 2013.
[193]  E. D. Hall, R. A. Vaishnav, and A. G. Mustafa, “Antioxidant Therapies for Traumatic Brain Injury,” Neurotherapeutics, vol. 7, no. 1, pp. 51–61, 2010.
[194]  P. S. Eriksson, E. Perfilieva, T. Bj?rk-Eriksson et al., “Neurogenesis in the adult human hippocampus,” Nature Medicine, vol. 4, no. 11, pp. 1313–1317, 1998.
[195]  T. J. Shors, G. Miesegaes, A. Beylin, M. Zhao, T. Rydel, and E. Gould, “Neurogenesis in the adult is involved in the formation of trace memories,” Nature, vol. 410, pp. 372–376, 2001.
[196]  B. Leuner, E. Gould, and T. J. Shors, “Is there a link between adult neurogenesis and learning?” Hippocampus, vol. 16, no. 3, pp. 216–224, 2006.
[197]  L. Henriksen, E. Hjelms, and T. Lindeburgh, “Brain hyperperfusion during cardiac operations. Cerebral blood flow measured in man by intra-arterial injection of xenon 133: evidence suggestive of intraoperative microembolism,” Journal of Thoracic and Cardiovascular Surgery, vol. 86, no. 2, pp. 202–208, 1983.
[198]  A. V. Govier, J. G. Reves, and R. D. McKay, “Factors and their influence on regional cerebral blood flow during nonpulsatile cardiopulmonary bypass,” Annals of Thoracic Surgery, vol. 38, no. 6, pp. 592–600, 1984.
[199]  J. M. Murkin, J. K. Farrar, W. A. Tweed, F. N. McKenzie, and G. Guiraudon, “Cerebral autoregulation and flow/metabolism coupling during cardiopulmonary bypass: the influence of PaCO2,” Anesthesia and Analgesia, vol. 66, no. 9, pp. 825–832, 1987.
[200]  Y. M. Ganushchak, E. J. Fransen, C. Visser, D. S. de Jong, and J. G. Maessen, “Neurological complications after coronary artery bypass grafting related to the performance of cardiopulmonary bypass,” Chest, vol. 125, no. 6, pp. 2196–2205, 2004.
[201]  W. E. Hoffman, E. Kochs, C. Werner, C. Thomas, and R. F. Albrecht, “Dexmedetomidine improves neurologic outcome from incomplete ischemia in the rat. Reversal by the α2-adrenergic antagonist atipamezole,” Anesthesiology, vol. 75, no. 2, pp. 328–332, 1991.
[202]  S. Dahmani, D. Rouelle, P. Gressens, and J. Mantz, “Characterization of the postconditioning effect of dexmedetomidine in mouse organotypic hippocampal slice cultures exposed to oxygen and glucose deprivation,” Anesthesiology, vol. 112, no. 2, pp. 373–383, 2010.
[203]  K. Engelhard, C. Werner, E. Ebersp?cher et al., “The effect of the α2-agonist dexmedetomidine and the N-methyl-D-aspartate antagonist S(+)-ketamine on the expression of apoptosis-regulating proteins after incomplete cerebral ischemia and reperfusion in rats,” Anesthesia and Analgesia, vol. 96, no. 2, pp. 524–531, 2003.
[204]  K. Sato, T. Kimura, T. Nishikawa, Y. Tobe, and Y. Masaki, “Neuroprotective effects of a combination of dexmedetomidine and hypothermia after incomplete cerebral ischemia in rats,” Acta Anaesthesiologica Scandinavica, vol. 54, no. 3, pp. 377–382, 2010.
[205]  O. Eser, H. Fidan, O. Sahin et al., “The influence of dexmedetomidine on ischemic rat hippocampus,” Brain Research, vol. 1218, pp. 250–256, 2008.
[206]  J. Afonso and F. Reis, “Dexmedetomidine: current role in anesthesia and intensive care,” Revista Brasileira de Anestesiologia, vol. 62, no. 1, pp. 118–133, 2012.
[207]  S. Dahmani, A. Paris, V. Jannier et al., “Dexmedetomidine increases hippocampal phosphorylated extracellular signal-regulated protein kinase 1 and 2 content by an α2-adrenoceptor- independent mechanism: evidence for the involvement of imidazoline I1 receptors,” Anesthesiology, vol. 108, no. 3, pp. 457–466, 2008.
[208]  J. Shen, Y. Wu, J.-Y. Xu et al., “ERK- and Akt-dependent neuroprotection by erythropoietin (EPO) against glyoxal-AGEs via modulation of Bcl-xL, Bax, and BAD,” Investigative Ophthalmology and Visual Science, vol. 51, no. 1, pp. 35–46, 2010.
[209]  H. Kocoglu, K. Karaaslan, E. Gonca, O. Bozdogan, and N. Gulcu, “Preconditionin effects of dexmedetomidine on myocardial ischemia/reperfusion injury in rats,” Current Therapeutic Research, vol. 69, no. 2, pp. 150–158, 2008.
[210]  H. Kocoglu, H. Ozturk, H. Ozturk, F. Yilmaz, and N. Gulcu, “Effect of dexmedetomidine on ischemia-reperfusion injury in rat kidney: a histopathologic study,” Renal Failure, vol. 31, no. 1, pp. 70–74, 2009.
[211]  S. Dahmani, D. Rouelle, P. Gressens, and J. Mantz, “Effects of dexmedetomidine on hippocampal focal adhesion kinase tyrosine phosphorylation in physiologic and ischemic conditions,” Anesthesiology, vol. 103, no. 5, pp. 969–977, 2005.
[212]  R. C. Prielipp, M. H. Wall, J. R. Tobin et al., “Dexmedetomidine-induced sedation in volunteers decreases regional and global cerebral blood flow,” Anesthesia and Analgesia, vol. 95, no. 4, pp. 1052–1059, 2002.
[213]  M. H. Zornow, M. Maze, J. B. Dyck, and S. L. Shafer, “Dexmedetomidine decreases cerebral blood flow velocity in humans,” Journal of Cerebral Blood Flow and Metabolism, vol. 13, no. 2, pp. 350–353, 1993.
[214]  J. C. Drummond, A. V. Dao, D. M. Roth et al., “Effect of dexmedetomidine on cerebral blood flow velocity, cerebral metabolic rate, and carbon dioxide response in normal humans,” Anesthesiology, vol. 108, no. 2, pp. 225–232, 2008.
[215]  M. Schoeler, P. D. Loetscher, R. Rossaint et al., “Dexmedetomidine is neuroprotective in an in vitro model for traumatic brain injury,” BMC Neurology, vol. 12, p. 20, 2012.
[216]  D. S. Sulemanji, A. D?nmez, D. Aldemir, A. Sezgin, and S. Türkoglu, “Dexmedetomidine during coronary artery bypass grafting surgery: is it neuroprotective?—a preliminary study,” Acta Anaesthesiologica Scandinavica, vol. 51, no. 8, pp. 1093–1098, 2007.
[217]  M. T. Bell, F. Ferenc Puskas, P. D. Smith et al., “Attenuation of spinal cord ischemia-reperfusion injury by specific α-2a receptor activation with dexmedetomidine,” Journal of Vascular Surgery, vol. 56, pp. 1398–1402, 2012.
[218]  J. Gu, P. Sun, H. Zhao et al., “Dexmedetomidine provides renoprotection against ischemia-reperfusion injury in mice,” Critical Care, vol. 15, no. 3, article R153, 2011.
[219]  B. Tufanogullari, P. F. White, M. P. Peixoto et al., “Dexmedetomidine infusion during laparoscopic bariatric surgery: the effect on recovery outcome variables,” Anesthesia and Analgesia, vol. 106, no. 6, pp. 1741–1748, 2008.
[220]  P. Jalowiecki, R. Rudner, M. Gonciarz, P. Kawecki, M. Petelenz, and P. Dziurdzik, “Sole use of dexmedetomidine has limited utility for conscious sedation during outpatient colonoscopy,” Anesthesiology, vol. 103, no. 2, pp. 269–273, 2005.
[221]  M. Muntazar and F. C. Kumar, “Cardiac arrest, a preventable yet a possible risk of dexmedetomidine: fact or fiction?” Anesthesiology, vol. 101, no. 6, pp. 1478–1480, 2004.
[222]  A. N. Shah, J. Koneru, A. Nicoara, L. B. Goldfeder, K. Thomas, and F. A. Ehlert, “Dexmedetomidine related cardiac arrest in a patient with permanent pacemaker; a cautionary tale,” Pacing and Clinical Electrophysiology, vol. 30, no. 9, pp. 1158–1160, 2007.
[223]  D. L. Herr, S. T. J. Sum-Ping, and M. England, “ICU sedation after coronary artery bypass graft surgery: dexmedetomidine-based versus propofol-based sedation regimens,” Journal of Cardiothoracic and Vascular Anesthesia, vol. 17, no. 5, pp. 576–584, 2003.
[224]  J. Jalonen, M. Hynynen, A. Kuitunen et al., “Dexmedetomidine as an anesthetic adjunct in coronary artery bypass grafting,” Anesthesiology, vol. 86, no. 2, pp. 331–345, 1997.
[225]  H. Karakaya Kabuk?u, N. Sahin, Y. Temel, and T. Aydogdu Titiz, “Hemodynamics in coronary artery bypass surgery: effects of intraoperative dexmedetomidine administration,” Anaesthesist, vol. 60, no. 5, pp. 427–431, 2011.
[226]  H. Guo, S. Takahashi, S. Cho, T. Hara, S. Tomiyasu, and K. Sumikawa, “The effects of dexmedetomidine on left ventricular function during hypoxia and reoxygenation in isolated rat hearts,” Anesthesia and Analgesia, vol. 100, no. 3, pp. 629–635, 2005.
[227]  H. Chen, H. Higashino, K. Maeda et al., “Reduction of cardiac norepinephrine improves postischemic heart function in stroke-prone spontaneously hypertensive rats,” Journal of Cardiovascular Pharmacology, vol. 38, no. 6, pp. 821–832, 2001.
[228]  P. M. Mertes, J. P. Carteaux, Y. Jaboin et al., “Estimation of myocardial interstitial norepinephrine release after brain death using cardiac microdialysis,” Transplantation, vol. 57, no. 3, pp. 371–377, 1994.
[229]  K. A. Bybee and A. Prasad, “Stress-related cardiomyopathy syndromes,” Circulation, vol. 118, no. 4, pp. 397–409, 2008.
[230]  V. C. Hachinski, K. E. Smith, and M. D. Silver, “Acute myocardial and plasma catecholamine changes in experimental stroke,” Stroke, vol. 17, no. 3, pp. 387–390, 1986.
[231]  V. H. Lee, J. K. Oh, S. L. Mulvagh, and E. F. M. Wijdicks, “Mechanisms in neurogenic stress cardiomyopathy after aneurysmal subarachnoid hemorrhage,” Neurocritical Care, vol. 5, no. 3, pp. 243–249, 2006.
[232]  H. M. Willigers, F. W. Prinzen, P. M. Roekaerts, S. de Lange, and M. E. Durieux, “Dexmedetomidine decreases perioperative myocardial lactate release in dogs,” Anesthesia and Analgesia, vol. 96, no. 3, pp. 657–664, 2003.
[233]  M. Karamazyn, R. E. Beamish, and N. S. Dhalla, “Involvement of calcium in coronary vasoconstriction due to prolonged hypoxia,” American Heart Journal, vol. 107, no. 2, pp. 293–297, 1984.
[234]  D. Pinsky, M. Oz, H. Liao et al., “Restoration of the cAMP second messenger pathway enhances cardiac preservation for transplantation in a heterotopic rat model,” Journal of Clinical Investigation, vol. 92, no. 6, pp. 2994–3002, 1993.
[235]  M. Kitakaze, M. Hori, K. Gotoh et al., “Beneficial effects of α2-adrenoceptor activity on ischemic myocardium during coronary hypoperfusion in dogs,” Circulation Research, vol. 65, no. 6, pp. 1623–1645, 1989.
[236]  M. F. Oliver, L. Goldman, D. G. Julian, and I. Holme, “Effect of mivazerol on perioperative cardiac complications during non-cardiac surgery in patients with coronary heart disease: the European Mivazerol Trial (EMIT),” Anesthesiology, vol. 91, no. 4, pp. 951–961, 1999.
[237]  D. N. Wijeysundera, J. S. Naik, and W. S. Beattie, “Alpha-2 adrenergic agonists to prevent perioperative cardiovascular complications: a meta-analysis,” American Journal of Medicine, vol. 114, no. 9, pp. 742–752, 2003.
[238]  A. J. Rough, L. H. Kudo, and C. Hébert, “Dexmedetomidine inhibits osmotic water permeability in the rat cortical collecting duct,” Journal of Pharmacology and Experimental Therapeutics, vol. 281, no. 1, pp. 62–69, 1997.
[239]  F. T. Billings IV, S. W. C. Chen, M. Kim et al., “α2-Adrenergic agonists protect against radiocontrast-induced nephropathy in mice,” American Journal of Physiology—Renal Physiology, vol. 295, no. 3, pp. F741–F748, 2008.
[240]  M. Taoda, Y. U. Adachi, Y. Uchihashi, K. Watanabe, T. Satoh, and E. S. Vizi, “Effect of dexmedetomidine on the release of [3H]-noradrenaline from rat kidney cortex slices: characterization of α2-adrenoceptor,” Neurochemistry International, vol. 38, no. 4, pp. 317–322, 2001.
[241]  R. M. Venn, P. J. Newman, and R. M. Grounds, “A phase II study to evaluate the efficacy of dexmedetomidine for sedation in the medical intensive care unit,” Intensive Care Medicine, vol. 29, no. 2, pp. 201–207, 2003.
[242]  K. Reid, Y. Hayashi, T.-Z. Guo, C. Correa-Sales, C. Nacif-Coelho, and M. Maze, “Chronic administration of an α2 adrenergic agonist desensitizes rats to the anesthetic effects of dexmedetomidine,” Pharmacology Biochemistry and Behavior, vol. 47, no. 1, pp. 171–175, 1994.
[243]  A. G. Doufas, C.-M. Lin, M.-I. Suleman et al., “Dexmedetomidine and meperidine additively reduce the shivering threshold in humans,” Stroke, vol. 34, no. 5, pp. 1218–1223, 2003.
[244]  E. G. Elvan, B. ??, ?. Uzun, E. Karabulut, F. Co?kun, and ü. Aypar, “Dexmedetomidine and postoperative shivering in patients undergoing elective abdominal hysterectomy,” European Journal of Anaesthesiology, vol. 25, no. 5, pp. 357–364, 2008.
[245]  G. A. Maccioli, “Dexmedetomidine to facilitate drug withdrawal,” Anesthesiology, vol. 98, no. 2, pp. 575–577, 2003.
[246]  A. S. Multz, “Prolonged dexmedetomidine infusion as an adjunct in treating sedation-induced withdrawal,” Anesthesia and Analgesia, vol. 96, no. 4, pp. 1054–1055, 2003.
[247]  K. Baddigam, P. Russo, J. Russo, and J. D. Tobias, “Dexmedetomidine in the treatment of withdrawal syndromes in cardiothoracic surgery patients,” Journal of Intensive Care Medicine, vol. 20, no. 2, pp. 118–123, 2005.
[248]  A. Rovasalo, H. Tohmo, R. Aantaa, E. Kettunen, and R. Palojoki, “Dexmedetomidine as an adjuvant in the treatment of alcohol withdrawal delirium: a case report,” General Hospital Psychiatry, vol. 28, no. 4, pp. 362–363, 2006.
[249]  J. Darrouj, N. Puri, E. Prince, A. Lomonaco, A. Spevetz, and D. R. Gerber, “Dexmedetomidine infusion as adjunctive therapy to benzodiazepines for acute alcohol withdrawal,” Annals of Pharmacotherapy, vol. 42, no. 11, pp. 1703–1705, 2008.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413