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 Signalling Associated with Brain Dead Organ Donation: From Brain Injury to Brain Stem Death and Posttransplant Ischaemia Reperfusion Injury

DOI: 10.1155/2013/521369

Full-Text   Cite this paper   Add to My Lib

Abstract:

Brain death is associated with dramatic and serious pathophysiologic changes that adversely affect both the quantity and quality of organs available for transplant. To fully optimise the donor pool necessitates a more complete understanding of the underlying pathophysiology of organ dysfunction associated with transplantation. These injurious processes are initially triggered by catastrophic brain injury and are further enhanced during both brain death and graft transplantation. The activated inflammatory systems then contribute to graft dysfunction in the recipient. Inflammatory mediators drive this process in concert with the innate and adaptive immune systems. Activation of deleterious immunological pathways in organ grafts occurs, priming them for further inflammation after engraftment. Finally, posttransplantation ischaemia reperfusion injury leads to further generation of inflammatory mediators and consequent activation of the recipient’s immune system. Ongoing research has identified key mediators that contribute to the inflammatory milieu inherent in brain dead organ donation. This has seen the development of novel therapies that directly target the inflammatory cascade. 1. Introduction Organ transplantation is the gold standard treatment for patients with end stage solid organ failure. An ever increasing disparity between available organs and potential recipients is the cause of avoidable morbidity and mortality [1–4]. Ongoing efforts are being made to increase the quantity and quality of organs available for transplant. Although outcomes from non-heart-beating donors have become increasingly successful [5], the majority of organs are still donated from donors after brain death (BD). Significant brain injury of any aetiology will cause a systemic response [6], creating a proinflammatory environment prior to the occurrence of brain death itself. BD then also creates a variety of inflammatory, haemodynamic and endocrine effects, which induce adverse sequelae in distant organs [7–10]. Finally, ischaemia-reperfusion injury (IRI), inherent in transplantation, generates reactive oxygen species (ROS), activates complement, and independently drives cytokine release and inflammation [11, 12]. Every transplanted organ from a BD donor will face these stages of potential injury. Consequently, donor management must consider each step from donor to recipient in order to maximise recipient outcomes. The purpose of this paper is to explore the current understanding of the three main contributors to injury that an organ will travel through from donor to

References

[1]  Organ, “Procurement and Transplantation Network,” Organ by Status, 2011, http://optn.transplant.hrsa.gov/.
[2]  M. A. Fink, S. R. Berry, P. J. Gow et al., “Risk factors for liver transplantation waiting list mortality,” Journal of Gastroenterology and Hepatology, vol. 22, no. 1, pp. 119–124, 2007.
[3]  N. R. Banner, C. A. Rogers, R. S. Bonser, et al., “Effect of heart transplantation on survival in ambulatory and decompensated heart failure,” Transplantation, vol. 96, no. 11, article 8, 2008.
[4]  A. Reed, G. I. Snell, C. McLean, and T. J. Williams, “Outcomes of patients with interstitial lung disease referred for lung transplant assessment,” Internal Medicine Journal, vol. 36, no. 7, pp. 423–430, 2006.
[5]  S. I. De Vleeschauwer, S. Wauters, L. J. Dupont et al., “Medium-term outcome after lung transplantation is comparable between brain-dead and cardiac-dead donors,” Journal of Heart and Lung Transplantation, vol. 30, no. 9, pp. 975–981, 2011.
[6]  S.-T. Lee, K. Chu, K. H. Jung et al., “Cholinergic anti-inflammatory pathway in intracerebral hemorrhage,” Brain Research, vol. 1309, pp. 164–171, 2010.
[7]  W. N. Nijboer, T. A. Schuurs, J. A. B. Van Der Hoeven et al., “Effect of brain death on gene expression and tissue activation in human donor kidneys,” Transplantation, vol. 78, no. 7, pp. 978–986, 2004.
[8]  G. H. Naderi, D. Mehraban, S. M. Kazemeyni, M. Darvishi, and A. H. Latif, “Living or deceased donor kidney transplantation: a comparison of results and survival rates among Iranian patients,” Transplantation Proceedings, vol. 41, no. 7, pp. 2772–2774, 2009.
[9]  A. C. Anyanwu, N. R. Banner, R. Radley-Smith, A. Khaghani, and M. H. Yacoub, “Long-term results of cardiac transplantation from live donors: the domino heart transplant,” Journal of Heart and Lung Transplantation, vol. 21, no. 9, pp. 971–975, 2002.
[10]  A. Khaghani, E. J. Birks, A. C. Anyanwu, and N. R. Banner, “Heart transplantation from live donors: ‘Domino procedure’,” Journal of Heart and Lung Transplantation, vol. 23, no. 9, supplement 1, pp. S257–S259, 2004.
[11]  E. Manning, S. Pham, S. Li et al., “Interleukin-10 delivery via mesenchymal stem cells: a novel gene therapy approach to prevent lung ischemia-reperfusion injury,” Human Gene Therapy, vol. 21, no. 6, pp. 713–727, 2010.
[12]  J. Damman, M. R. Daha, W. J. Van Son, H. G. Leuvenink, R. J. Ploeg, and M. A. Seelen, “Crosstalk between complement and toll-like receptor activation in relation to donor brain death and renal ischemia-reperfusion injury,” American Journal of Transplantation, vol. 11, no. 4, pp. 660–669, 2011.
[13]  C. T. Weaver, L. E. Harrington, P. R. Mangan, M. Gavrieli, and K. M. Murphy, “Th17: an effector CD4 T cell lineage with regulatory T cell ties,” Immunity, vol. 24, no. 6, pp. 677–688, 2006.
[14]  F. Deknuydt, G. Bioley, D. Valmori, and M. Ayyoub, “IL-1β and IL-2 convert human Treg into TH17 cells,” Clinical Immunology, vol. 131, no. 2, pp. 298–307, 2009.
[15]  J.-Q. Li, H. Z. Qi, Z. J. He et al., “Cytoprotective effects of human interleukin-10 gene transfer against necrosis and apoptosis induced by hepatic cold ischemia/reperfusion injury,” Journal of Surgical Research, vol. 157, no. 1, pp. e71–e78, 2009.
[16]  C. H. Y. Wong, C. N. Jenne, W. Y. Lee, C. Léger, and P. Kubes, “Functional innervation of hepatic iNKT cells is immunosuppressive following stroke,” Science, vol. 334, no. 6052, pp. 101–105, 2011.
[17]  P. P. Wadia and A. R. Tambur, “Yin and yan of cytokine regulation in solid organ graft rejection and tolerance,” Clinics in Laboratory Medicine, vol. 28, no. 3, pp. 469–479, 2008.
[18]  T. Nakajima, V. Palchevsky, D. L. Perkins, J. A. Belperio, and P. W. Finn, “Lung transplantation: infection, inflammation, and the microbiome,” Seminars in Immunopathology, vol. 33, no. 2, pp. 135–156, 2011.
[19]  D. J. Huss, R. C. Winger, H. Peng, Y. Yang, M. K. Racke, and A. E. Lovett-Racke, “TGF-β enhances effector Th1 cell activation but promotes self-regulation via IL-10,” Journal of Immunology, vol. 184, no. 10, pp. 5628–5636, 2010.
[20]  S. Itoh, N. Kimura, R. C. Axtell, et al., “Interleukin-17 accelerates allograft rejection by suppressing regulatory T cell expansion,” Circulation, vol. 124, supplement 11, pp. S187–S196, 2011.
[21]  E. A. Kastelijn, G. T. Rijkers, C. H. Van Moorsel et al., “Systemic and exhaled cytokine and chemokine profiles are associated with the development of bronchiolitis obliterans syndrome,” The Journal of Heart and Lung Transplantation, vol. 29, no. 9, pp. 997–1008, 2010.
[22]  C. Atkinson, J. C. Varela, and S. Tomlinson, “Complement-dependent inflammation and injury in a murine model of brain dead donor hearts,” Circulation Research, vol. 105, no. 11, pp. 1094–1101, 2009.
[23]  N. Yoshida, T. Yoshikawa, Y. Nakamura et al., “Interactions of neutrophils and endothelial cells under low flow conditions in vitro,” Shock, vol. 8, no. 2, pp. 125–130, 1997.
[24]  T. Lattmann, M. Hein, S. Horber et al., “Activation of pro-inflammatory and anti-inflammatory cytokines in host organs during chronic allograft rejection: role of endothelin receptor signaling,” American Journal of Transplantation, vol. 5, no. 5, pp. 1042–1049, 2005.
[25]  H.-O. Pae, G. S. Oh, B. M. Choi et al., “Carbon monoxide produced by heme oxygenase-1 suppresses T cell proliferation via inhibition of IL-2 production,” Journal of Immunology, vol. 172, no. 8, pp. 4744–4751, 2004.
[26]  M. Ishizaki, T. Akimoto, R. Muromoto et al., “Involvement of tyrosine kinase-2 in both the IL-12/Th1 and IL-23/Th17 axes in vivo,” Journal of Immunology, vol. 187, no. 1, pp. 181–189, 2011.
[27]  J. L. Harden, T. Gu, M. O. Kilinc, R. B. Rowswell-Turner, L. P. Virtuoso, and N. K. Egilmez, “Dichotomous effects of IFN-γ on dendritic cell function determine the extent of IL-12 - driven antitumor T cell immunity,” Journal of Immunology, vol. 187, no. 1, pp. 126–132, 2011.
[28]  T. Ueno, A. Yamada, T. Ito, et al., “Role of nuclear factor of activated T cell (NFAT) transcription factors in skin and vascularized cardiac allograft rejection,” Transplantation, vol. 92, no. 5, pp. e26–e27, 2011.
[29]  X. Xiong, G. E. Barreto, L. Xu, Y. B. Ouyang, X. Xie, and R. G. Giffard, “Increased brain injury and worsened neurological outcome in interleukin-4 knockout mice after transient focal cerebral ischemia,” Stroke, vol. 42, no. 7, pp. 2026–2032, 2011.
[30]  G. L. Theodorou, S. Marousi, J. Ellul et al., “T helper 1 (Th1)/Th2 cytokine expression shift of peripheral blood CD4+ and CD8+ T cells in patients at the post-acute phase of stroke,” Clinical and Experimental Immunology, vol. 152, no. 3, pp. 456–463, 2008.
[31]  T. Hensler, S. Sauerland, P. Riess et al., “The effect of additional brain injury on systemic interleukin (IL)-10 and IL-13 levels in trauma patients,” Inflammation Research, vol. 49, no. 10, pp. 524–528, 2000.
[32]  F. D. Finkelman, I. M. Katona, J. F. Urban Jr., et al., “IL-4 is required to generate and sustain in vivo IgE responses,” Journal of Immunology, vol. 141, no. 7, pp. 2335–2341, 1988.
[33]  C. Wang, S. S. Tay, G. T. Tran et al., “Donor IL-4-treatment induces alternatively activated liver macrophages and IDO-expressing NK cells and promotes rat liver allograft acceptance,” Transplant Immunology, vol. 22, no. 3-4, pp. 172–178, 2010.
[34]  G. Bansal, C. M. Wong, L. Liu, Y. J. Suzuki, et al., “Oxidant signaling for interleukin-13 gene expression in lung smooth muscle cells,” Free Radical Biology and Medicine, vol. 52, no. 9, pp. 1552–1559, 2012.
[35]  M. Sandovici, L. E. Deelman, H. van Goor, W. Helfrich, D. de Zeeuw, and R. H. Henning, “Adenovirus-mediated interleukin-13 gene therapy attenuates acute kidney allograft injury,” The Journal of Gene Medicine, vol. 9, no. 12, pp. 1024–1032, 2007.
[36]  J. Corren, “Inhibition of interleukin-5 for the treatment of eosinophilic diseases,” Discovery Medicine, vol. 13, no. 71, pp. 305–312, 2012.
[37]  T. Fujisawa, R. Abu-Ghazaleh, H. Kita, C. J. Sanderson, and G. J. Gleich, “Regulatory effect of cytokines on eosinophil degranulation,” Journal of Immunology, vol. 144, no. 2, pp. 642–646, 1990.
[38]  L. Z. Grund, E. N. Komegae, M. Lopes-Ferreira, and C. Lima, “IL-5 and IL-17A are critical for the chronic IgE response and differentiation of long-lived antibody-secreting cells in inflamed tissues,” Cytokine, vol. 59, no. 2, pp. 335–351, 2012.
[39]  S. M. Knoblach and A. I. Faden, “Interleukin-10 improves outcome and alters proinflammatory cytokine expression after experimental traumatic brain injury,” Experimental Neurology, vol. 153, no. 1, pp. 143–151, 1998.
[40]  D. S. Nath, H. Ilias Basha, V. Tiriveedhi et al., “Characterization of immune responses to cardiac self-antigens myosin and vimentin in human cardiac allograft recipients with antibody-mediated rejection and cardiac allograft vasculopathy,” Journal of Heart and Lung Transplantation, vol. 29, no. 11, pp. 1277–1285, 2010.
[41]  X. C. Liu, A. Zhai, J. Q. Li, and H. Z. Qi, “Interleukin-23 promotes natural killer T-cell production of IL-17 during rat liver transplantation,” Transplantation Proceedings, vol. 43, no. 5, pp. 1962–1966, 2011.
[42]  M. Laan, Z. H. Cui, H. Hoshino, et al., “Neutrophil recruitment by human IL-17 via C-X-C chemokine release in the airways,” Journal of Immunology, vol. 162, no. 4, pp. 2347–2352, 1999.
[43]  T. Shichita, Y. Sugiyama, H. Ooboshi, et al., “Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury,” Nature Medicine, vol. 15, no. 8, pp. 946–950, 2009.
[44]  J. Parrish-Novak, S. R. Dillon, A. Nelson, et al., “Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function,” Nature, vol. 408, no. 6808, pp. 57–63, 2000.
[45]  M. Hagn, K. Sontheimer, K. Dahlke, et al., “Human B cells differentiate into granzyme B-secreting cytotoxic B lymphocytes upon incomplete T-cell help,” Immunology and Cell Biology, vol. 90, no. 4, pp. 457–467, 2012.
[46]  J. Scheller, A. Chalaris, D. Schmidt-Arras, and S. Rose-John, “The pro- and anti-inflammatory properties of the cytokine interleukin-6,” Biochimica et Biophysica Acta, vol. 1813, no. 5, pp. 878–888, 2011.
[47]  M. Akdis, “The cellular orchestra in skin allergy; Are differences to lung and nose relevant?” Current Opinion in Allergy and Clinical Immunology, vol. 10, no. 5, pp. 443–451, 2010.
[48]  Y. Chen, D. Li, J. Y. S. Tsang et al., “PPAR-γ signaling and IL-5 inhibition together prevent chronic rejection of MHC Class IImismatched cardiac grafts,” Journal of Heart and Lung Transplantation, vol. 30, no. 6, pp. 698–706, 2011.
[49]  R. Eulenfeld, et al., “Interleukin-6 signalling: more than Jaks and STATs,” European Journal of Cell Biology, vol. 91, no. 6-7, pp. 486–495, 2012.
[50]  G. Wang, A. Zhong, S. Wang, N. Dong, Z. Sun, and J. Xia, “Retinoic acid attenuates acute heart rejection by increasing regulatory t cell and repressing differentiation of th17 cell in the presence of TGF-β,” Transplant International, vol. 23, no. 10, pp. 986–997, 2010.
[51]  D. C. Neujahr and C. P. Larsen, “Regulatory T cells in lung transplantation-an emerging concept,” Seminars in Immunopathology, vol. 33, no. 2, pp. 117–127, 2011.
[52]  Organ Procurement and Transplantation Network, Deceased Donors Recovered in the U.S. by Cause of Death, 2011, http://optn.transplant.hrsa.gov/latestData/rptData.asp.
[53]  K. Inaba, B. C. Branco, L. Lam et al., “Organ donation and time to procurement: late is not too late,” Journal of Trauma, vol. 68, no. 6, pp. 1362–1366, 2010.
[54]  S. Lee, M. Shin, E. Kim et al., “Donor characteristics associated with reduced survival of transplanted kidney grafts in Korea,” Transplantation Proceedings, vol. 42, no. 3, pp. 778–781, 2010.
[55]  M. Zukowski, R. Bohatyrewicz, J. Biernawska et al., “Cause of death in multiorgan donors and its relation to the function of transplanted kidneys,” Transplantation Proceedings, vol. 41, no. 8, pp. 2972–2974, 2009.
[56]  S. Wauters, G. M. Verleden, A. Belmans et al., “Donor cause of brain death and related time intervals: does it affect outcome after lung transplantation?” European Journal of Cardio-thoracic Surgery, vol. 39, no. 4, pp. e68–e76, 2011.
[57]  J. S. Ganesh, C. A. Rogers, N. R. Banner, and R. S. Bonser, “Donor cause of death and medium-term survival after heart transplantation: a United Kingdom national study,” Journal of Thoracic and Cardiovascular Surgery, vol. 129, no. 5, pp. 1153–1159, 2005.
[58]  M. Godino, M. Lander, A. Cacciatore, S. Perez-Protto, and R. Mizraji, “Ventricular dysfunction associated with brain trauma is cause for exclusion of young heart donors,” Transplantation Proceedings, vol. 42, no. 5, pp. 1507–1509, 2010.
[59]  S. J. Campbell, V. H. Perry, F. J. Pitossi et al., “Central nervous system injury triggers Hepatic CC and CXC chemokine expression that is associated with leukocyte mobilization and recruitment to both the central nervous system and the liver,” American Journal of Pathology, vol. 166, no. 5, pp. 1487–1497, 2005.
[60]  S. J. Campbell, Y. Jiang, A. E. M. Davis et al., “Immunomodulatory effects of etanercept in a model of brain injury act through attenuation of the acute-phase response,” Journal of Neurochemistry, vol. 103, no. 6, pp. 2245–2255, 2007.
[61]  J. Lu, S. J. Goh, P. Y. L. Tng, Y. Y. Deng, E. A. Ling, and S. Moochhala, “Systemic inflammatory response following acute traumatic brain injury,” Frontiers in Bioscience, vol. 14, no. 10, pp. 3795–3813, 2009.
[62]  Y. Kitamura, M. Nomura, H. Shima et al., “Acute lung injury associated with systemic inflammatory response syndrome following subarachnoid hemorrhage: a survey by the shonan neurosurgical association,” Neurologia Medico-Chirurgica, vol. 50, no. 6, pp. 456–460, 2010.
[63]  A. K. H. Tam, D. Ilodigwe, J. Mocco et al., “Impact of systemic inflammatory response syndrome on vasospasm, cerebral infarction, and outcome after subarachnoid hemorrhage: exploratory analysis of CONSCIOUS-1 database,” Neurocritical Care, vol. 13, no. 2, pp. 182–189, 2010.
[64]  S. J. Campbell, P. M. Hughes, J. P. Iredale et al., “CINC-1 is an acute-phase protein induced by focal brain injury causing leukocyte mobilization and liver injury,” The FASEB Journal, vol. 17, no. 9, pp. 1168–1170, 2003.
[65]  S.-T. Lee, K. Chu, K. H. Jung et al., “Anti-inflammatory mechanism of intravascular neural stem cell transplantation in haemorrhagic stroke,” Brain, vol. 131, no. 3, pp. 616–629, 2008.
[66]  C. Adrie, M. Monchi, J. P. Fulgencio et al., “Immune status and apoptosis activation during brain death,” Shock, vol. 33, no. 4, pp. 353–362, 2010.
[67]  S. Hoeger, C. Bergstraesser, J. Selhorst et al., “Modulation of brain dead induced inflammation by vagus nerve stimulation,” American Journal of Transplantation, vol. 10, no. 3, pp. 477–489, 2010.
[68]  A. J. Rostron, V. S. Avlonitis, D. M. Cork, D. S. Grenade, J. A. Kirby, and J. H. Dark, “Hemodynamic resuscitation with arginine vasopressin reduces lung injury after brain death in the transplant donor,” Transplantation, vol. 85, no. 4, pp. 597–606, 2008.
[69]  Z.-D. Guo, X. C. Sun, and J. H. Zhang, “Mechanisms of early brain injury after SAH: matrix metalloproteinase 9,” Acta Neurochirurgica. Supplement, vol. 110, no. 1, pp. 63–65, 2011.
[70]  T. Frugier, M. C. Morganti-Kossmann, D. O'Reilly, and C. A. McLean, “In situ detection of inflammatory mediators in post mortem human brain tissue after traumatic injury,” Journal of Neurotrauma, vol. 27, no. 3, pp. 497–507, 2010.
[71]  E. Jeremitsky, L. Omert, C. M. Dunham, J. Protetch, and A. Rodriguez, “Harbingers of poor outcome the day after severe brain injury: hypothermia, hypoxia, and hypoperfusion,” Journal of Trauma, vol. 54, no. 2, pp. 312–319, 2003.
[72]  D. Graetz, A. Nagel, F. Schlenk, O. Sakowitz, P. Vajkoczy, and A. Sarrafzadeh, “High ICP as trigger of proinflammatory IL-6 cytokine activation in aneurysmal subarachnoid hemorrhage,” Neurological Research, vol. 32, no. 7, pp. 728–735, 2010.
[73]  J. Perez-Barcena, J. Ibá?ez, M. Brell et al., “Lack of correlation among intracerebral cytokines, intracranial pressure, and brain tissue oxygenation in patients with traumatic brain injury and diffuse lesions,” Critical Care Medicine, vol. 39, no. 3, pp. 533–540, 2011.
[74]  G. W. Hergenroeder, J. B. Redell, A. N. Moore, and P. K. Dash, “Biomarkers in the clinical diagnosis and management of traumatic brain injury,” Molecular Diagnosis and Therapy, vol. 12, no. 6, pp. 345–358, 2008.
[75]  S. G. Rhind, N. T. Crnko, A. J. Baker et al., “Prehospital resuscitation with hypertonic saline-dextran modulates inflammatory, coagulation and endothelial activation marker profiles in severe traumatic brain injured patients,” Journal of Neuroinflammation, vol. 7, no. 1, article 5, 2010.
[76]  D. Shlosberg, M. Benifla, D. Kaufer, and A. Friedman, “Blood-brain barrier breakdown as a therapeutic target in traumatic brain injury,” Nature Reviews Neurology, vol. 6, no. 7, pp. 393–403, 2010.
[77]  T. Higashida, C. W. Kreipke, J. A. Rafols et al., “The role of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury: laboratory investigation,” Journal of Neurosurgery, vol. 114, no. 1, pp. 92–101, 2011.
[78]  H. Wu, Z. Zhang, Y. Li et al., “Time course of upregulation of inflammatory mediators in the hemorrhagic brain in rats: correlation with brain edema,” Neurochemistry International, vol. 57, no. 3, pp. 248–253, 2010.
[79]  H. Wu, Z. Zhang, X. Hu et al., “Dynamic changes of inflammatory markers in brain after hemorrhagic stroke in humans: a postmortem study,” Brain Research, vol. 1342, pp. 111–117, 2010.
[80]  E. Mi?ambres, A. Cemborain, P. Sánchez-Velasco et al., “Correlation between transcranial interleukin-6 gradient and outcome in patients with acute brain injury,” Critical Care Medicine, vol. 31, no. 3, pp. 933–938, 2003.
[81]  C. A. Skrabal, L. O. Thompson, E. V. Potapov et al., “Organ-specific regulation of pro-inflammatory molecules in heart, lung, and kidney following brain death,” Journal of Surgical Research, vol. 123, no. 1, pp. 118–125, 2005.
[82]  J. A. Amado, F. Lopez-Espadas, A. Vazquez-Barquero et al., “Blood levels of cytokines in brain-dead patients: relationship with circulating hormones and acute-phase reactants,” Metabolism, vol. 44, no. 6, pp. 812–816, 1995.
[83]  H. Offner, S. Subramanian, S. M. Parker, M. E. Afentoulis, A. A. Vandenbark, and P. D. Hurn, “Experimental stroke induces massive, rapid activation of the peripheral immune system,” Journal of Cerebral Blood Flow and Metabolism, vol. 26, no. 5, pp. 654–665, 2006.
[84]  B. Maier, R. Lefering, M. Lehnert et al., “Early versus late onset of multiple organ failure is associated with differing patterns of plasma cytokine biomarker expression and outcome after severe trauma,” Shock, vol. 28, no. 6, pp. 668–674, 2007.
[85]  L. G. Koudstaal, N. A. 'T Hart, P. J. Ottens et al., “Brain death induces inflammation in the donor intestine,” Transplantation, vol. 86, no. 1, pp. 148–154, 2008.
[86]  H. Jedrzejowska-Szypulka, G. Straszak, M. Larysz-Brysz et al., “Interleukin-1β plays a role in the activation of peripheral leukocytes after blood-brain barrier rupture in the course of subarachnoid hemorrhage,” Current Neurovascular Research, vol. 7, no. 1, pp. 39–48, 2010.
[87]  K. A. Hanafy, B. Grobelny, L. Fernandez et al., “Brain interstitial fluid TNF-α after subarachnoid hemorrhage,” Journal of the Neurological Sciences, vol. 291, no. 1-2, pp. 69–73, 2010.
[88]  S. M. Lucas, N. J. Rothwell, and R. M. Gibson, “The role of inflammation in CNS injury and disease,” British Journal of Pharmacology, vol. 147, supplement 1, pp. S232–S240, 2006.
[89]  B. Maier, M. Lehnert, H. L. Laurer, A. E. Mautes, W. I. Steudel, and I. Marzi, “Delayed elevation of soluble tumor necrosis factor receptors p75 and p55 in cerebrospinal fluid and plasma after traumatic brain injury,” Shock, vol. 26, no. 2, pp. 122–127, 2006.
[90]  P. Mellerg?rd, O. ?neman, F. Sj?gren, C. S?berg, and J. Hillman, “Differences in cerebral extracellular response of interleukin-1β, interleukin-6, and interleukin-10 after subarachnoid hemorrhage or severe head trauma in humans,” Neurosurgery, vol. 68, no. 1, pp. 12–19, 2011.
[91]  S. Weiss, K. Kotsch, M. Francuski et al., “Brain death activates donor organs and is associated with a worse I/R injury after liver transplantation,” American Journal of Transplantation, vol. 7, no. 6, pp. 1584–1593, 2007.
[92]  S. J. Campbell, I. Zahid, P. Losey et al., “Liver Kupffer cells control the magnitude of the inflammatory response in the injured brain and spinal cord,” Neuropharmacology, vol. 55, no. 5, pp. 780–787, 2008.
[93]  K. M. McLean, J. Y. Duffy, P. K. Pandalai et al., “Glucocorticoids alter the balance between pro- and anti-inflammatory mediators in the myocardium in a porcine model of brain death,” Journal of Heart and Lung Transplantation, vol. 26, no. 1, pp. 78–84, 2007.
[94]  M. Katsuno, H. Yokota, Y. Yamamoto, and A. Teramoto, “Increased regional interleukin-4 during the acute stage of severe intracranial disorders,” Neurologia Medico-Chirurgica, vol. 46, no. 10, pp. 471–474, 2006.
[95]  M. S. Yang, E. J. Park, S. Sohn et al., “Interleukin-13 and -4 induce death of activated microglia,” GLIA, vol. 38, no. 4, pp. 273–280, 2002.
[96]  A. M. Planas, R. Gorina, and A. Chamorro, “Signalling pathways mediating inflammatory responses in brain ischaemia,” Biochemical Society Transactions, vol. 34, no. 6, pp. 1267–1270, 2006.
[97]  T. Hayakata, T. Shiozaki, O. Tasaki et al., “Changes in CSF S100B and cytokine concentrations in early-phase severe traumatic brain injury,” Shock, vol. 22, no. 2, pp. 102–107, 2004.
[98]  X.-Q. Wang, Y. P. Peng, J. H. Lu, B. B. Cao, and Y. H. Qiu, “Neuroprotection of interleukin-6 against NMDA attack and its signal transduction by JAK and MAPK,” Neuroscience Letters, vol. 450, no. 2, pp. 122–126, 2009.
[99]  I. Dimopoulou, S. Korfias, U. Dafni et al., “Protein S-100b serum levels in trauma-induced brain death,” Neurology, vol. 60, no. 6, pp. 947–951, 2003.
[100]  A. Quintana, M. Giralt, A. Molinero, I. L. Campbell, M. Penkowa, and J. Hidalgo, “Analysis of the cerebral transcriptome in mice subjected to traumatic brain injury: importance of IL-6,” NeuroImmunoModulation, vol. 14, no. 3-4, pp. 139–143, 2007.
[101]  J. Damman, W. N. Nijboer, T. A. Schuurs et al., “Local renal complement C3 induction by donor brain death is associated with reduced renal allograft function after transplantation,” Nephrology Dialysis Transplantation, vol. 26, no. 7, pp. 2345–2354, 2011.
[102]  M. Lv, Y. Liu, J. Zhang et al., “Roles of inflammation response in microglia cell through Toll-like receptors 2/interleukin-23/interleukin-17 pathway in cerebral ischemia/reperfusion injury,” Neuroscience, vol. 176, pp. 162–172, 2011.
[103]  F. Konoeda, T. Shichita, H. Yoshida et al., “Therapeutic effect of IL-12/23 and their signaling pathway blockade on brain ischemia model,” Biochemical and Biophysical Research Communications, vol. 402, no. 3, pp. 500–506, 2010.
[104]  D.-D. Wang, Y. F. Zhao, G. Y. Wang et al., “IL-17 potentiates neuronal injury induced by oxygen-glucose deprivation and affects neuronal IL-17 receptor expression,” Journal of Neuroimmunology, vol. 212, no. 1-2, pp. 17–25, 2009.
[105]  A. P. Comellas and A. Briva, “Role of endothelin-1 in acute lung injury,” Translational Research, vol. 153, no. 6, pp. 263–271, 2009.
[106]  B. P. Persson, P. Rossi, E. Weitzberg, and A. Oldner, “Inhaled tezosentan reduces pulmonary hypertension in endotoxin-induced lung injury,” Shock, vol. 32, no. 4, pp. 427–434, 2009.
[107]  D. Konrad, M. Haney, G. Johansson, M. Wanecek, E. Weitzberg, and A. Oldner, “Cardiac effects of endothelin receptor antagonism in endotoxemic pigs,” American Journal of Physiology, vol. 293, no. 2, pp. H988–H996, 2007.
[108]  H. H. Leuchte, T. Meis, M. El-Nounou, J. Michalek, and J. Behr, “Inhalation of endothelin receptor blockers in pulmonary hypertension,” American Journal of Physiology, vol. 294, no. 4, pp. L772–L777, 2008.
[109]  V. N. Kuklin, M. Y. Kirov, O. V. Evgenov et al., “Novel endothelin receptor antagonist attenuates endotoxin-induced lung injury in sheep,” Critical Care Medicine, vol. 32, no. 3, pp. 766–773, 2004.
[110]  S. Kallakuri, C. W. Kreipke, P. C. Schafer, S. M. Schafer, and J. A. Rafols, “Brain cellular localization of endothelin receptors A and B in a rodent model of diffuse traumatic brain injury,” Neuroscience, vol. 168, no. 3, pp. 820–830, 2010.
[111]  D. A. Chatfield, D. H. Brahmbhatt, T. Sharp, I. E. Perkes, J. G. Outrim, and D. K. Menon, “Juguloarterial endothelin-1 gradients after severe traumatic brain injury,” Neurocritical Care, vol. 14, no. 1, pp. 55–60, 2011.
[112]  H. Vatter, J. Konczalla, and V. Seifert, “Endothelin related pathophysiology in cerebral vasospasm: what happens to the cerebral vessels?” Acta Neurochirurgica. Supplement, vol. 110, no. 1, pp. 177–180, 2011.
[113]  R. Salonia, P. E. Empey, S. M. Poloyac et al., “Endothelin-1 is increased in cerebrospinal fluid and associated with unfavorable outcomes in children after severe traumatic brain injury,” Journal of Neurotrauma, vol. 27, no. 10, pp. 1819–1825, 2010.
[114]  M. Sabri, J. Ai, and R. L. MacDonald, “Dissociation of vasospasm and secondary effects of experimental subarachnoid hemorrhage by clazosentan,” Stroke, vol. 42, no. 5, pp. 1454–1460, 2011.
[115]  A. J. Sutherland, R. S. Ware, C. Winterford, and J. F. Fraser, “The endothelin axis and gelatinase activity in alveolar macrophages after brain-stem death injury: a pilot study,” Journal of Heart and Lung Transplantation, vol. 26, no. 10, pp. 1040–1047, 2007.
[116]  B. Reel, G. Oktay, S. Ozkal et al., “MMP-2 and MMP-9 alteration in response to collaring in rabbits: the effects of endothelin receptor antagonism,” Journal of Cardiovascular Pharmacology and Therapeutics, vol. 14, no. 4, pp. 292–301, 2009.
[117]  P. Rossi, B. Persson, P. J. M. Boels, A. Arner, E. Weitzberg, and A. Oldner, “Endotoxemic pulmonary hypertension is largely mediated by endothelin-induced venous constriction,” Intensive Care Medicine, vol. 34, no. 5, pp. 873–880, 2008.
[118]  H. Kimura, I. Gules, T. Meguro, and J. H. Zhang, “Cytotoxicity of cytokines in cerebral microvascular endothelial cell,” Brain Research, vol. 990, no. 1-2, pp. 148–156, 2003.
[119]  P. M. Cobelens, I. A. C. W. Tiebosch, R. M. Dijkhuizen et al., “Interferon-β attenuates lung inflammation following experimental subarachnoid hemorrhage,” Critical Care, vol. 14, no. 4, article R157, 2010.
[120]  V. I. Otto, U. E. Heinzel-Pleines, S. M. Gloor, O. Trentz, and T. Kossmann, “Morganti-Kossmann MCsICAM-1 and TNF-α induce MIP-2 with distinct kinetics in astrocytes and brain microvascular endothelial cells,” Journal of Neuroscience Research, vol. 60, no. 6, pp. 733–742, 2000.
[121]  E. J. Birks, P. B. J. Burton, V. Owen et al., “Elevated tumor necrosis factor-α and interleukin-6 in myocardium and serum of malfunctioning donor hearts,” Circulation, vol. 102, no. 19, pp. III352–III358, 2000.
[122]  H. H. Wei, X. C. M. Lu, D. A. Shear et al., “NNZ-2566 treatment inhibits neuroinflammation and pro-inflammatory cytokine expression induced by experimental penetrating ballistic-like brain injury in rats,” Journal of Neuroinflammation, vol. 6, no. 1, article 19, 2009.
[123]  J. A. Kellum, R. Venkataraman, D. Powner, M. Elder, G. Hergenroeder, and M. Carter, “Feasibility study of cytokine removal by hemoadsorption in brain-dead humans,” Critical Care Medicine, vol. 36, no. 1, pp. 268–272, 2008.
[124]  T. Bartfai, M. Sanchez-Alavez, S. Andell-Jonsson et al., “Interleukin-1 system in CNS stress: seizures, fever, and neurotrauma,” Annals of the New York Academy of Sciences, vol. 1113, no. 1, pp. 173–177, 2007.
[125]  A. Basu, J. K. Krady, J. R. Enterline, and S. W. Levison, “Transforming growth factor β1 prevents IL-1β-induced microglial activation, whereas TNFα- and IL-6-stimulated activation are not antagonized,” GLIA, vol. 40, no. 1, pp. 109–120, 2002.
[126]  R. Murugan, R. Venkataraman, A. S. Wahed et al., “Increased plasma interleukin-6 in donors is associated with lower recipient hospital-free survival after cadaveric organ transplantation,” Critical Care Medicine, vol. 36, no. 6, pp. 1810–1816, 2008.
[127]  G. Plenz, H. Eschert, M. Erren et al., “The interleukin-6/interleukin-6-receptor system is activated in donor hearts,” Journal of the American College of Cardiology, vol. 39, no. 9, pp. 1508–1512, 2002.
[128]  R. Murugan, R. Venkataraman, A. S. Wahed et al., “Preload responsiveness is associated with increased interleukin-6 and lower organ yield from brain-dead donors,” Critical Care Medicine, vol. 37, no. 8, pp. 2387–2393, 2009.
[129]  P. Gregoric, A. Sijacki, S. Stankovic, et al., “SIRS score on admission and initial concentration of IL-6 as severe acute pancreatitis outcome predictors,” Hepato-Gastroenterology, vol. 58, no. 105, p. 263, 2011.
[130]  L. Wang, J. Quan, W. E. Johnston et al., “Age-dependent differences of interleukin-6 activity in cardiac function after burn complicated by sepsis,” Burns, vol. 36, no. 2, pp. 232–238, 2010.
[131]  B. Maier, K. Schwerdtfeger, A. Mautes et al., “Differential release of interleukines 6, 8, and 10 in cerebrospinal fluid and plasma after traumatic brain injury,” Shock, vol. 15, no. 6, pp. 421–426, 2001.
[132]  A. J. Fisher, S. C. Donnelly, N. Hirani et al., “Elevated levels of interleukin-8 in donor lungs is associated with early graft failure after lung transplantation,” American Journal of Respiratory and Critical Care Medicine, vol. 163, no. 1, pp. 259–265, 2001.
[133]  S. C. Donnelly, R. M. Strieter, S. L. Kunkel et al., “Interleukin-8 and development of adult respiratory distress syndrome in at-risk patient groups,” The Lancet, vol. 341, no. 8846, pp. 643–647, 1993.
[134]  E. Csuka, M. C. Morganti-Kossmann, P. M. Lenzlinger, H. Joller, O. Trentz, and T. Kossmann, “IL-10 levels in cerebrospinal fluid and serum of patients with severe traumatic brain injury: relationship to IL-6, TNF-α, TGF-β1 and blood-brain barrier function,” Journal of Neuroimmunology, vol. 101, no. 2, pp. 211–221, 1999.
[135]  M. C. Morganti-Kossman, P. M. Lenzlinger, V. Hans et al., “Production of cytokines following brain injury: beneficial and deleterious for the damaged tissue,” Molecular Psychiatry, vol. 2, no. 2, pp. 133–136, 1997.
[136]  U. Malipiero, U. Koedel, W. Pfister, and A. Fontana, “Bacterial meningitis: the role of transforming growth factor-beta in innate immunity and secondary brain damage,” Neurodegenerative Diseases, vol. 4, no. 1, pp. 43–50, 2007.
[137]  M. R. Douglas, M. Daniel, C. Lagord et al., “High CSF transforming growth factor b levels after subarachnoid haemorrhage: association with chronic communicating hydrocephalus,” Journal of Neurology, Neurosurgery and Psychiatry, vol. 80, no. 5, pp. 545–550, 2009.
[138]  T. Okamoto, S. Takahashi, E. Nakamura, K. Nagaya, T. Hayashi, and K. Fujieda, “Transforming growth factor-β1 induces matrix metalloproteinase-9 expression in human meningeal cells via ERK and Smad pathways,” Biochemical and Biophysical Research Communications, vol. 383, no. 4, pp. 475–479, 2009.
[139]  Y. Oishi, Y. Nishimura, Y. Tanoue et al., “Endothelin-1 receptor antagonist prevents deterioration of left ventricular function and coronary flow reserve in brain-dead canine heart,” Journal of Heart and Lung Transplantation, vol. 24, no. 9, pp. 1354–1361, 2005.
[140]  R. Ferrera, G. Hadour, F. Tamion et al., “Brain death provokes very acute alteration in myocardial morphology detected by echocardiography: preventive effect of beta-blockers,” Transplant International, vol. 24, no. 3, pp. 300–306, 2011.
[141]  R. V. Venkateswaran, V. Dronavalli, P. A. Lambert et al., “The proinflammatory environment in potential heart and lung donors: prevalence and impact of donor management and hormonal therapy,” Transplantation, vol. 88, no. 4, pp. 582–588, 2009.
[142]  A. Catania, C. Lonati, A. Sordi, and S. Gatti, “Detrimental consequences of brain injury on peripheral cells,” Brain, Behavior, and Immunity, vol. 23, no. 7, pp. 877–884, 2009.
[143]  M. Valdivia, C. Chamorro, M. A. Romera, B. Balandín, and M. Pérez, “Effect of posttraumatic donor's disseminated intravascular coagulation in intrathoracic organ donation and transplantation,” Transplantation Proceedings, vol. 39, no. 7, pp. 2427–2428, 2007.
[144]  S. R. James, A. M. Ranasinghe, R. Venkateswaran, C. J. McCabe, J. A. Franklyn, and R. S. Bonser, “The effects of acute triiodothyronine therapy on myocardial gene expression in brain stem dead cardiac donors,” Journal of Clinical Endocrinology and Metabolism, vol. 95, no. 3, pp. 1338–1343, 2010.
[145]  R. V. Venkateswaran, R. P. Steeds, D. W. Quinn et al., “The haemodynamic effects of adjunctive hormone therapy in potential heart donors: a prospective randomized double-blind factorially designed controlled trial,” European Heart Journal, vol. 30, no. 14, pp. 1771–1780, 2009.
[146]  V. S. Avlonitis, C. H. Wigfield, J. A. Kirby, and J. H. Dark, “The hemodynamic mechanisms of lung injury and systemic inflammatory response following brain death in the transplant donor,” American Journal of Transplantation, vol. 5, no. 4 I, pp. 684–693, 2005.
[147]  A. Salim, M. Martin, C. Brown, H. Belzberg, P. Rhee, and D. Demetriades, “Complications of brain death: frequency and impact on organ retrieval,” American Surgeon, vol. 72, no. 5, pp. 377–381, 2006.
[148]  A. Salim, G. C. Velmahos, C. Brown, H. Belzberg, and D. Demetriades, “Aggressive organ donor management significantly increases the number of organs available for transplantation,” Journal of Trauma, vol. 58, no. 5, pp. 991–994, 2005.
[149]  H. Cushing, “Concerning a definite regulatory mechanism of the vaso-motor centre which controls blood pressure during cerebral compression,” The Johns Hopkins Hospital Bulletin, vol. 12, pp. 290–292, 1901.
[150]  H. Cushing, “Some experimental and clinical observations concerning states of increased intracranial tension. The Mütter Lecture for 1901,” American Journal of Medical Sciences, vol. 124, pp. 375–400, 1902.
[151]  C. Dictus, B. Vienenkoetter, M. Esmaeilzadeh, A. Unterberg, and R. Ahmadi, “Critical care management of potential organ donors: our current standard,” Clinical Transplantation, vol. 23, no. 21, pp. 2–9, 2009.
[152]  H. Schrader, C. Hall, and N. N. Zwetnow, “Effects of prolonged supratentorial mass expansion on regional blood flow and cardiovascular parameters during the Cushing response,” Acta Neurologica Scandinavica, vol. 72, no. 3, pp. 283–294, 1985.
[153]  C. Gao, X. Liu, H. Shi et al., “Relationship between sympathetic nervous activity and inflammatory response after subarachnoid hemorrhage in a perforating canine model,” Autonomic Neuroscience, vol. 147, no. 1-2, pp. 70–74, 2009.
[154]  R. Ferrera, M. Ovize, B. Claustrat, and G. Hadour, “Stable myocardial function and endocrine dysfunction during experimental brain death,” Journal of Heart and Lung Transplantation, vol. 24, no. 7, pp. 921–927, 2005.
[155]  H. Marthol, T. Intravooth, J. Bardutzky, P. De Fina, S. Schwab, and M. J. Hilz, “Sympathetic cardiovascular hyperactivity precedes brain death,” Clinical Autonomic Research, vol. 20, no. 6, pp. 363–369, 2010.
[156]  C. Chamorro, J. A. Falcón, and J. C. Michelena, “Controversial points in organ donor management,” Transplantation Proceedings, vol. 41, no. 8, pp. 3473–3475, 2009.
[157]  P. Herijgers, M. Borgers, and W. Flameng, “The effect of brain death on cardiovascular function in rats. Part I. Is the heart damaged?” Cardiovascular Research, vol. 38, no. 1, pp. 98–106, 1998.
[158]  L. V. Borovikova, S. Ivanova, M. Zhang et al., “Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin,” Nature, vol. 405, no. 6785, pp. 458–462, 2000.
[159]  M. A. Flierl, D. Rittirsch, B. A. Nadeau et al., “Phagocyte-derived catecholamines enhance acute inflammatory injury,” Nature, vol. 449, no. 7163, pp. 721–725, 2007.
[160]  M. Takada, K. C. Nadeau, W. W. Hancock et al., “Effects of explosive brain death on cytokine activation of peripheral organs in the rat,” Transplantation, vol. 65, no. 12, pp. 1533–1542, 1998.
[161]  V. S. Avlonitis, C. H. Wigfield, J. A. Kirby, and J. H. Dark, “Treatment of the brain-dead lung donor with aprotinin and nitric oxide,” Journal of Heart and Lung Transplantation, vol. 29, no. 10, pp. 1177–1184, 2010.
[162]  C. Zhu, J. Li, G. Zhang et al., “Brain death disrupts structure and function of pig liver,” Transplantation Proceedings, vol. 42, no. 3, pp. 733–736, 2010.
[163]  M. Cypel, H. Kaneda, J. C. Yeung et al., “Increased levels of interleukin-1β and tumor necrosis factor-α in donor lungs rejected for transplantation,” Journal of Heart and Lung Transplantation, vol. 30, no. 4, pp. 452–459, 2011.
[164]  D. Kaminska, B. Tyran, O. Mazanowska et al., “Cytokine gene expression in kidney allograft biopsies after donor brain death and ischemia-reperfusion injury using in situ reverse-transcription polymerase chain reaction analysis,” Transplantation, vol. 84, no. 9, pp. 1118–1124, 2007.
[165]  J. Q. Li, H. Z. Qi, Z. J. He, W. Hu, Z. Z. Si, and Y. N. Li, “Induction of lymphocyte apoptosis in rat liver allograft by adenoviral gene transfection of human interleukin-10,” European Surgical Research, vol. 44, no. 3-4, pp. 133–141, 2010.
[166]  W. N. Nijboer, P. J. Ottens, A. Van Dijk, H. Van Goor, R. J. Ploeg, and H. G. D. Leuvenink, “Donor pretreatment with carbamylated erythropoietin in a brain death model reduces inflammation more effectively than erythropoietin while preserving renal function,” Critical Care Medicine, vol. 38, no. 4, pp. 1155–1161, 2010.
[167]  C. F. Bulcao, K. M. D'Souza, R. Malhotra et al., “Activation of JAK-STAT and nitric oxide signaling as a mechanism for donor heart dysfunction,” Journal of Heart and Lung Transplantation, vol. 29, no. 3, pp. 346–351, 2010.
[168]  K. P. Kotsch, F. Ulrich, A. Reutzel-Selke et al., “Methylprednisolone therapy in deceased donors reduces inflammation in the donor liver and improves outcome after liver transplantation a prospective randomized controlled trial,” Annals of Surgery, vol. 248, no. 6, pp. 1042–1049, 2008.
[169]  H. Kaneda, T. K. Waddell, M. De Perrot et al., “Pre-implantation multiple cytokine mRNA expression analysis of donor lung grafts predicts survival after lung transplantation in humans,” American Journal of Transplantation, vol. 6, no. 3, pp. 544–551, 2006.
[170]  A. Loverre, C. Divella, G. Castellano et al., “T helper 1, 2 and 17 cell subsets in renal transplant patients with delayed graft function,” Transplant International, vol. 24, no. 3, pp. 233–242, 2011.
[171]  I. Inci, B. Erne, S. Arni et al., “Prevention of primary graft dysfunction in lung transplantation by N-acetylcysteine after prolonged cold ischemia,” Journal of Heart and Lung Transplantation, vol. 29, no. 11, pp. 1293–1301, 2010.
[172]  E. C. Lascano, A. Bertolotti, C. B. Gómez et al., “Failure of IL-8 to assess early reperfusion injury following lung transplantation of cardiac death donor pigs,” Transplant International, vol. 22, no. 5, pp. 574–582, 2009.
[173]  S. J. Zhang and S. Chen, “The influence of brain death on liver in rats,” Transplantation Proceedings, vol. 36, no. 7, pp. 1925–1927, 2004.
[174]  M. Salama, O. Andrukhova, M. A. Hoda et al., “Concomitant endothelin-1 overexpression in lung transplant donors and recipients predicts primary graft dysfunction,” American Journal of Transplantation, vol. 10, no. 3, pp. 628–636, 2010.
[175]  M. Kusaka, Y. Kuroyanagi, H. Kowa et al., “Genomewide expression profiles of rat model renal isografts from brain dead donors,” Transplantation, vol. 83, no. 1, pp. 62–70, 2007.
[176]  S. Shiotani, M. Shimada, T. Suehiro et al., “Involvement of Rho-kinase in cold ischemia-reperfusion injury after liver transplantation in rats,” Transplantation, vol. 78, no. 3, pp. 375–382, 2004.
[177]  R. Tuuminen, S. Syrj?l?, R. Krebs, et al., “Donor simvastatin treatment abolishes rat cardiac allograft ischemia/reperfusion injury and chronic rejection through microvascular protection,” Circulation, vol. 124, no. 10, pp. 1138–1150, 2011.
[178]  S. Ulukaya, E. Ulukaya, I. Alper, A. Yilmaztepe-Oral, and M. Kilic, “Soluble cytokeratin 18 biomarkers may provide information on the type of cell death during early ischemia and reperfusion periods of liver transplantation,” Clinical Transplantation, vol. 24, no. 6, pp. 848–854, 2010.
[179]  Y. F. Xu, M. Liu, B. Peng et al., “Protective effects of SP600125 on renal ischemia-reperfusion injury in rats,” Journal of Surgical Research, vol. 169, no. 1, pp. e77–e84, 2011.
[180]  M. L. Blagonravov, M. M. Azova, M. V. Onufriev, and V. A. Frolov, “Activities of some caspase cascade enzymes and myocardial contractility in experimental left ventricular focal ischemia,” Bulletin of Experimental Biology and Medicine, vol. 150, no. 6, pp. 672–675, 2011.
[181]  C. Ballet, K. Renaudin, N. Degauque et al., “Indirect CD4+ TH1 response, antidonor antibodies and diffuse C4d graft deposits in long-term recipients conditioned by donor antigens priming,” American Journal of Transplantation, vol. 9, no. 4, pp. 697–708, 2009.
[182]  M. Zhang, L. H. Michael, S. A. Grosjean, R. A. Kelly, M. C. Carroll, and M. L. Entman, “The role of natural IgM in myocardial ischemia-reperfusion injury,” Journal of Molecular and Cellular Cardiology, vol. 41, no. 1, pp. 62–67, 2006.
[183]  A. E. Gelman, M. Okazaki, S. Sugimoto et al., “CCR2 regulates monocyte recruitment as well as CD4+ T h1 allorecognition after lung transplantation,” American Journal of Transplantation, vol. 10, no. 5, pp. 1189–1199, 2010.
[184]  D. K. de Vries, J. H. N. Lindeman, J. Ringers, M. E. J. Reinders, T. J. Rabelink, and A. F. M. Schaapherder, “Donor brain death predisposes human kidney grafts to a proinflammatory reaction after transplantation,” American Journal of Transplantation, vol. 11, no. 5, pp. 1064–1070, 2011.
[185]  S. A. Hosgood, I. H. Mohamed, A. Bagul, and M. L. Nicholson, “Hypothermic machine perfusion after static cold storage does not improve the preservation condition in an experimental porcine kidney model,” British Journal of Surgery, vol. 98, no. 7, pp. 943–950, 2011.
[186]  J. A. Duran, A. A. González, D. D. García et al., “Variation in the levels of inflammatory cytokines depending on ischemic time: effects on respiratory variables,” Transplantation Proceedings, vol. 41, no. 3, pp. 980–982, 2009.
[187]  M. Ilmakunnas, K. H?ckerstedt, H. M?kisalo, S. Siitonen, H. Repo, and E. J. Pesonen, “Hepatic IL-8 release during graft procurement is associated with impaired graft function after human liver transplantation,” Clinical Transplantation, vol. 24, no. 1, pp. 29–35, 2010.
[188]  H. E. Merry, P. S. Wolf, E. Fitzsullivan, J. C. Keech, and M. S. Mulligan, “Lipopolysaccharide pre-conditioning is protective in lung ischemia-reperfusion injury,” Journal of Heart and Lung Transplantation, vol. 29, no. 4, pp. 471–478, 2010.
[189]  A. Benson, S. Murray, P. Divakar, et al., “Microbial infection-induced expansion of effector T cells overcomes the suppressive effects of regulatory T cells via an IL-2 deprivation mechanism,” The Journal of Immunology, vol. 188, no. 2, pp. 800–810, 2012.
[190]  D. Jonigk, M. Merk, K. Hussein et al., “Obliterative airway remodeling: molecular evidence for shared pathways in transplanted and native lungs,” American Journal of Pathology, vol. 178, no. 2, pp. 599–608, 2011.
[191]  B. Ke, X. D. Shen, C. R. Lassman, F. Gao, R. W. Busuttil, and J. W. Kupiec-Weglinski, “Cytoprotective and antiapoptotic effects of IL-13 in hepatic cold ischemia/reperfusion injury are heme oxygenase-1 dependent,” American Journal of Transplantation, vol. 3, no. 9, pp. 1076–1082, 2003.
[192]  L. Sun, T. Shi, H. Qiao et al., “Hepatic overexpression of heme oxygenase-1 improves liver allograft survival by expanding t regulatory cells,” Journal of Surgical Research, vol. 166, no. 2, pp. e187–e194, 2011.
[193]  M. Schaub, C. J. Ploetz, D. Gerbaulet et al., “Effect of dopamine on inflammatory status in kidneys of brain-dead rats,” Transplantation, vol. 77, no. 9, pp. 1333–1340, 2004.
[194]  H. Zhou, J. Liu, P. Pan, D. Jin, W. Ding, and W. Li, “Carbon monoxide inhalation decreased lung injury via anti-inflammatory and anti-apoptotic effects in brain death rats,” Experimental Biology and Medicine, vol. 235, no. 10, pp. 1236–1243, 2010.
[195]  H. Zhou, H. Qian, J. Liu et al., “Protection against lung graft injury from brain-dead donors with carbon monoxide, biliverdin, or both,” Journal of Heart and Lung Transplantation, vol. 30, no. 4, pp. 460–466, 2011.
[196]  E. Sierra-Filardi, M. A. Vega, P. Sánchez-Mateos, A. L. Corbí, and A. Puig-Kr?ger, “Heme Oxygenase-1 expression in M-CSF-polarized M2 macrophages contributes to LPS-induced IL-10 release,” Immunobiology, vol. 215, no. 9-10, pp. 788–795, 2010.
[197]  B. Ke, X. D. Shen, Y. Zhai et al., “Heme oxygenase 1 mediates the immunomodulatory and antiapoptotic effects of interleukin 13 gene therapy in vivo and in vitro,” Human Gene Therapy, vol. 13, no. 15, pp. 1845–1857, 2002.
[198]  F. W. Selck, P. Deb, and E. B. Grossman, “Deceased organ donor characteristics and clinical interventions associated with organ yield,” American Journal of Transplantation, vol. 8, no. 5, pp. 965–974, 2008.
[199]  A. Salim, M. Martin, C. Brown, P. Rhee, D. Demetriades, and H. Belzberg, “The effect of a protocol of aggressive donor management: implications for the national organ donor shortage,” Journal of Trauma, vol. 61, no. 2, pp. 429–432, 2006.
[200]  J. D. Rosendale, H. M. Kauffman, M. A. McBride et al., “Aggressive pharmacologic donor management results in more transplanted organs,” Transplantation, vol. 75, no. 4, pp. 482–487, 2003.
[201]  Critical Pathway for the Organ Donor, United Network for Organ Sharing, 2002, http://www.unos.org/docs/Critical_Pathway.pdf.
[202]  A. Dare, A. Bartlett, and J. Fraser, “Critical care of the potential organ donor,” Current Neurology and Neuroscience Reports, vol. 12, no. 4, pp. 456–465, 2012.
[203]  A. J. M. Lewis, A. J. Rostron, D. M. W. Cork, J. A. Kirby, and J. H. Dark, “Norepinephrine and arginine vasopressin increase hepatic but not renal inflammatory activation during hemodynamic resuscitation in a rodent model of brain-dead donors,” Experimental and Clinical Transplantation, vol. 7, no. 2, pp. 119–123, 2009.
[204]  M. Cypel, M. Liu, M. Rubacha et al., “Functional repair of human donor lungs by IL-10 gene therapy,” Science Translational Medicine, vol. 1, no. 4, article 4ra9, 2009.
[205]  M. R. Sadaria, P. D. Smith, D. A. Fullerton et al., “Cytokine expression profile in human lungs undergoing normothermic ex-vivo lung perfusion,” Annals of Thoracic Surgery, vol. 92, no. 2, pp. 478–484, 2011.
[206]  T. Kakishita, T. Oto, S. Hori et al., “Suppression of inflammatory cytokines during ex vivo lung perfusion with an adsorbent membrane,” Annals of Thoracic Surgery, vol. 89, no. 6, pp. 1773–1779, 2010.
[207]  F. M. Santiago, P. Bueno, C. Olmedo et al., “Effect of N-acetylcysteine administration on intraoperative plasma levels of interleukin-4 and interleukin-10 in liver transplant recipients,” Transplantation Proceedings, vol. 40, no. 9, pp. 2978–2980, 2008.
[208]  O. Brissaud, F. Villega, J. P. Konsman et al., “Short-term effect of erythropoietin on brain lesions and aquaporin-4 expression in a hypoxic-ischemic neonatal rat model assessed by magnetic resonance diffusion weighted imaging and immunohistochemistry,” Pediatric Research, vol. 68, no. 2, pp. 123–127, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133