289
Views
10
CrossRef citations to date
0
Altmetric
INVITED REVIEW

Pharmacology of Ischemia–Reperfusion. Translational Research Considerations

, , , , , , & show all
Pages 234-249 | Received 12 May 2015, Accepted 28 Oct 2015, Published online: 23 May 2016

REFERENCES

  • Castaneda MP, Swiatecka-Urban A, Mitsnefes MM, et al. Activation of mitochondrial apoptotic pathways in human renal allografts after ischemia reperfusion injury. Transplantation. 2003;76(1):50–54.
  • Devarajan P. Update on mechanisms of ischemic acute kidney injury. J Am Soc Nephrol. 2006;17(6):1503–1520.
  • Kennedy SE, Erlich JH. Murine renal ischaemia-reperfusion injury. Nephrology (Carlton). 2008;13(5):390–396.
  • Schoenberg MH, Beger HG. Reperfusion injury after intestinal ischemia. Crit Care Med. 1993;21(9):1376–1386.
  • Yun Y, Duan WG, Chen P, et al. Ischemic postconditioning modified renal oxidative stress and lipid peroxidation caused by ischemic reperfusion injury in rats. Transplant Proc. 2009;41(9):3597–3602.
  • Hertle L, Garthoff B. Calcium channel blocker nisoldipine limits ischemic damage in rat kidney. J Urol. 1985;134(6):1251–1254.
  • Shapiro JI, Cheung C, Itabashi A, et al. The effect of verapamil on renal function after warm and cold ischemia in the isolated perfused rat kidney. Transplantation. 1985;40(6):596–600.
  • Niños JA, Toledo AH, López-Neblina F, et al. “Effects of dantrolene on ischemia–reperfusion injury in animals models: a review of outcomes in Heart, Brain, Liver, and Kidney”. Universidad estatal de Michigan. J Surg Res. 2010.
  • Schreinemachers MC, Doorschodt BM, Florquin S, et al. Comparison of preservation solutions for washout of kidney grafts: an experimental study. Transplant Proc. 2009;41(10):4072–4079.
  • Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74(5):1124–1136.
  • Przyklenk K, Bauer B, Ovize M, et al. Regional ischemic “preconditioning” protects remote virgin myocardium from subsequent sustained coronary occlusion. Circulation. 1993;87(3):893–899.
  • Cohen M, Downey JM. Precondicionamiento isquémico: de los mecanismos básicos a las aplicaciones clínicas. Revista Argentina de Cardiología. 2000;62(2):285–299.
  • Linden J. Molecular approach to adenosine receptors: receptor-mediated mechanisms of tissue protection. Annu Rev Pharmacol Toxicol. 2001;41:775–787.
  • Zaugg M, Lucchinetti E, Uecker M, et al.. Anaesthetics and cardiac preconditioning. Part I. Signalling and cytoprotective mechanism. Br J Anaesth. 2003;91:551–565.
  • Minamino T, Kitakaze M, Morioka T, et al. Cardioprotection due to preconditioning correlates with increased ecto-5′-nucleotidase activity. Am J Physiol. 1996;270(1 Pt 2):H238–H244.
  • Kharbanda RK, Mortensen UM, White PA, et al. Transient limb ischemia induces remote ischemic preconditioning in vivo. Circulation. 2002;106(23):2881–2883.
  • Dong JH, Liu YX, Ji ES, et al. Limb ischemic preconditioning reduces infarct size following myocardial ischemia–reperfusion in rats. Sheng Li Xue Bao. 2004;56(1):41–46.
  • Gho BC, Schoemaker RG, Van den Doel MA, et al. Myocardial protection by brief ischemia in noncardiac tissue. Circulation. 1996;94(9):2193–2200.
  • Lang SC, Elsasser A, Scheler C, et al. Myocardial preconditioning and remote renal preconditioning-identifying a protective factor using proteomic methods?. Basic Res Cardiol. 2006;101(2):149–158.
  • Dong HL, Zhang Y, Su BX, et al. Limb remote ischemic preconditioning protects the spinal cord from ischemia–reperfusion injury: a newly identified nonneuronal but reactive oxygen species-dependent pathway. Anesthesiology 2010;112(4):881–891.
  • Lazaris AM, Maheras AN, Vasdekis SN, et al. Protective effect of remote ischemic preconditioning in renal ischemia/ reperfusion injury, in a model of thoracoabdominal aorta approach. J Surg Res. 2009;154(2):267–273.
  • Oehmann C, Benz S, Drognitz O, et al. Remote preconditioning reduces microcirculatory disorders in pancreatic ischemia/reperfusion injury. Pancreas 2007;35(4): e45–e50.
  • Takagi H, Manabe H, Kawai N, et al. Review and meta-analysis of randomized controlled clinical trials of remote ischemic preconditioning in cardiovascular surgery. Am J Cardiol. 2008;102(11):1487–1488.
  • Zhou W, Zeng D, Chen R, et al. Limb ischemic preconditioning reduces heart and lung injury after an open heart operation in infants. Pediatr Cardiol. 2010;31(1):22–29.
  • Hu S, Dong HL, Li YZ, et al. Effects of remote ischemic preconditioning on biochemical markers and neurologic outcomes in patients undergoing elective cervical decompression surgery: a prospective randomized controlled trial. J Neurosurg Anesthesiol. 2010;22(1):46–52.
  • Ali ZA, Callaghan CJ, Lim E, et al. Remote ischemic preconditioning reduces myocardial and renal injury after elective abdominal aortic aneurysm repair: a randomized controlled trial. Circulation. 2007;116(11-supl):I98–I105.
  • Kloner RA, Jennings RB. Consequences of brief ischemia: stunning, preconditioning, and their clinical implications: Part 1. Circulation. 2001;104(24):2981–2989.
  • Kis A, Vegh A, Papp J y col. Pacing-induced delayed protection against arrhythmias is attenuated by aminoguanidine, an inhibitor of nitric oxide synthase. Br J Pharmacol. 1999;127:1545–1550.
  • Parra tt JR. Protection of the hearth by ischemic preconditioning: Mechanisms and possibilities for pharmacological exploitation. Trenda Pharmacol Sci. 1994;15: 19–25.
  • Bolli R, Dawn B, Tang X-L y col. The nitric oxide hypothesis of late preconditioning. Basic Res Cardiol. 1998;93:325–338.
  • Wu Z-K, Livainen T, Pehkonen E, et al. Ischemic preconditioning suppresses ventricular tachyarrhythmias after myocardial revascularization. Circulation. 2002;106(24):3091–3096.
  • Clavien PA, Yadav S, Sindram D, et al. Protective effects of ischemic preconditioning for liver resection performed under inflow occlusion in humans. Ann Surg. 2000;232(2):155–162.
  • Atalla SL, Toledo-Pereyra LH, MacKenzie GH, et al. Influence of oxygen-derived free radical scavengers on ischemic livers. Transplantation. 1985;40(6):584–590.
  • Suzuki M, Takeuchi H, Kakita T, et al. The involvement of the intracellular superoxide production system in hepatic ischemia–reperfusion injury. In vivo and in vitro experiments using transgenic mice manifesting excessive CuZn–SOD activity. Free Radic Biol Med. 2000;29(8):756–763.
  • Bulger E, Maier R. Antioxidants in critical illness. Arch Surg. 2001;136(10):1201–1207.
  • Origassa CS, Camara NO. Cytoprotective role of heme oxygenase-1 and heme degradation derived end products in liver injury. World J Hepatol. 2013;5(10):541–549.
  • Amersi F, Buelow R, Kato H, et al. Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. J Clin Invest. 1999;104(11):1631–1639.
  • Takeda A, Onodera H, Sugimoto A, et al. Increased expression of heme oxygenase mRNA in rat brain following transient forebrain ischemia. Brain Res. 1994;666(1):120–124.
  • Yamaguchi T, Terakado M, Horio F, et al. Role of bilirubin as an antioxidant in an ischemia–reperfusion of rat liver and induction of heme oxygenase. Biochem Biophys Res Commun. 1996;223(1):129–135.
  • Tsuchihashi S, Fondevila C, Kupiec-Weglinski JW. Heme oxygenase system in ischemia and reperfusion injury. Ann Transplant. 2004;9(1):84–87.
  • Ohno K, Ito M, Ichihara M et al. Molecular hydrogen as an emerging therapeutic medical gas for neurodegenerative and other diseases. Oxid Med Cell Longev. 2012;353152, 11 pages.
  • Roberts BW, Mitchell J, Kilgannon JH, et al. Nitric oxide donor agents for the treatment of ischemia/ reperfusion injury in human subjects: a systematic review. Shock. 2013;39(3):229–239.
  • Siriussawakul A, Ahmed Zaky, Lang J D. Role of nitric oxide in hepatic ischemia–reperfusion injury. World J Gastroenterol. 2010;16(48):6079–6086.
  • Yamashita M, Schmid RA, Ando K, et al. Nitroprusside ameliorates lung allograft reperfusion injury. Ann Thorac Surg. 1996;62(3):791–796.
  • Albrecht EW, Stegeman CA, Heeringa P, et al. Protective role of endothelial nitric oxide synthase. J Pathol. 2003;199(1):8–17.
  • Moens AL, Takimoto E, Tocchetti CG, et al. Reversal of cardiac hypertrophy and fibrosis from pressure overload by tetrahydrobiopterin: efficacy of recoupling nitric oxide synthase as a therapeutic strategy. Circulation. 2008;117(20):2626–2636.
  • Yuan L, Kaplowitz N. Glutathione in liver diseases and hepatotoxicity. Mol Aspects Med. 2009;30:29–41.
  • Lu SC. Regulation of glutathione synthesis. Mol Aspects Med. 2009;30(1–2):42–59.
  • Knight TR, Ho YS, Farhood A, et al. Peroxynitrite is a critical mediator of acetaminophen hepatotoxicity in murine livers: protection by glutathione. J Pharmacol Exp Ther. 2002 303(2):468–475.
  • Liu P, Fisher MA, Farhood A, et al. Beneficial effects of extracellular glutathione against endotoxin-induced liver injury during ischemia and reperfusion. Circ Shock. 1994;43(2):64–70.
  • Jaeschke H, Farhood A. Neutrophil and Kupffer cell-induced oxidant stress and ischemia–reperfusion injury in rat liver. Am J Physiol. 1991;260(3 Pt 1):G355–G362.
  • Bilzer M, Baron A, Schauer R, et al. Glutathione treatment protects the rat liver against injury after warm ischemia and Kupffer cell activation. Digestion. 2002;66(1): 49–57.
  • Schauer RJ, Kalmuk S, Gerbes AL, et al. Intravenous administration of glutathione protects parenchymal and non-parenchymal liver cells against reperfusion injury following rat liver transplantation. World J Gastroenterol. 2004;10(6):864–870.
  • Prieto-Moure B, Carabén-Redaño A, Aliena-Valero A, et al. Allopurinol in renal ischemia. J Invest Surg. 2014;27:304–316.
  • Perry BC, Soltys D, Toledo AH, et al. “Tumor necrosis factor-α in liver ischemia/reperfusion injury”. J Invest Surg. 2011;24:178–188.
  • Adkins WK, Taylor AE. Role of xanthine oxidase and neutrophils in ischemia–reperfusion injury in rabbit lung. J Appl Physiol. 1990;69(6):2012–2018.
  • Al-Mehdi AB, Shuman H, Fisher AB. Intracellular generation of reactive oxygen species during nonhypoxic lung ischemia. Am J Physiol. 1997;272(2 Pt 1):L294–L300.
  • Kennedy TP, Rao NV, Hopkins C, et al. Role of reactive oxygen species in reperfusion injury of the rabbit lung. J Clin Invest. 1989;83(4):1326–1335.
  • Zhao G, al-Mehdi AB, Fisher AB. Anoxia-reoxygenation versus ischemia in isolated rat lungs. Am J Physiol. 1997;273(6 Pt 1):L1112–L1117.
  • Mashiach E, Sela S, Weinstein T, et al. Mesna: a novel renoprotective antioxidant in ischemia acute renal failure. Nephrol Dial Transplant. 2001;16(3):273–276.
  • Jaeschke H, Woolbright BL. Current strategies to minimize hepatic ischemia–reperfusion injury by targeting reactive oxygen species. Transplant Rev (Orlando). 2012;26(2):103–114.
  • Zhu C, Bilali A, Georgieva GS, et al. Salvage of nonischemic control lung from injury by unilateral ischemic lung with apocynin, a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, in isolated perfused rat lung. Transl Res. 2008;152(6):273–282.
  • Dodd O, Pearse DB. Effect of the NADPH oxidase inhibitor apocynin on ischemia–reperfusion lung injury. Am J Physiol Heart Circ Physiol. 2000;279(1):H303–H312.
  • Dinauer MC. The respiratory burst oxidase and the molecular genetics of chronic granulomatous disease. Crit Rev Clin Lab Sci. 1993;30(4):329–369.
  • Heumuller S, Wind S, Barbosa-Sicard E, et al. Apocynin is not an inhibitor of vascular NADPH oxidases but an antioxidant. Hypertension. 2008;51(2):211–217.
  • Chou TC, Lin YF, Wu WC, et al. Enhanced nitric oxide and cyclic GMP formation plays a role in the anti-platelet activity of simvastatin. Br J Pharmacol. 2008;153(6):1281–1287.
  • Gaddam V, Li DH, Mehte JL. Anti-thrombotic effect of atorvastatin-an effect unrelated to lipid lowering. J Cardiovasc Pharmacol Ther. 2002;7(4):247–253.
  • Lee TS, Chang CC, Zhu Yet al. Simvastatin induces heme oxygenase-1: a novel mechanism of vessel protection. Circulation. 2004;110(10):1296–1302.
  • Yet SF, Tian R, Layne MD, et al. Cardiac-specific expression of heme oxygenase-1 protects against ischemia and reperfusion injury in transgenic mice. Circ Res. 2001;89(2):168–173.
  • Taha MO, Caricati-Neto A, Ferreira RM, et al. L-arginine in the ischemic phase protects against liver ischemia–reperfusion injury. Acta Cir Bras. 2012;27(9):616–623.
  • Harward TR, Coe D, Souba WW, et al. Glutamine preserves gut glutathione levels during intestinal ischemia/reperfusion. J Surg Res. 1994;56(4):351–355.
  • Sozen S, Kisakürek M, Yildiz F, et al. The effect of glutamine on hepatic ischemia reperfusion injury in rats. Hippokratia. 2011;15(2):161–166.
  • Chinda K, Chattipakorn & Chattipakon N. Cardioprotective effects of incretin during ischaemia-reperfusion. Diab. Vasc. Dis Res. 2012;9(4):256–269.
  • Mason J. Vitamins and trace elements in the critically ill patient. In: Nutritional Considerations in the Intensive Care Unit: Science, Rationale and Practice. Shikora S, Martindale R, Schwaitzberg, S, Eds. Iowa (USA): Kendall/Hunt Publishing Company. 2002;61–79.
  • Mallick IH, Winslet MC, Seifalian AM. Pyrrolidine dithiocarbamate protects the small bowel from warm ischaemia/reperfusion injury of the intestine: the role of haem oxygenase. Clin Sci (Lond). 2006;111(6): 373–380.
  • Tian XF, Yao JH, Li YH, et al. protective effect of pyrrolidine dithiocarbamate on liver injury induced by intestinal ischemia–reperfusion in rats. Hepatobiliary Pancreat Dis Int. 2006;5(1):90–95.
  • Isselhard W, Eitenmúller J, Mäurer W, et al. Increase in myocardial adenine nucleotides induced by adenosine: dosage, mode of application and duration, species differences. J Mol Cell Cardiol. 1980;12(6):619–634.
  • Ely SW, Berne RM. Protective effects of adenosine in myocardial ischemia. Circulation. 1992;85(3):893–904.
  • Dominguez-Rodriguez A, Abreu-Gonzalez P. Myocardial ischemia–reperfusion injury: Possible role of melatonin. World Cardiol. 2010;2(8):233–236.
  • Petrosillo G, Moro N, Ruggiero FM, et al. Melatonin inhibits cardiolipin peroxidation in mitochondria and prevents the mitochondrial permeability transition an cytochrome c release. Free Radic Biol Med. 2009;47: 969–974.
  • Lochner A, Huisamen B, Nduhirabandi F. Cardioprotective effect of melatonin against ischemia/reperfusion damage. Front Biosci (Elite Ed). 2013;5:305–315.
  • Verma R, Mishra V, Sasmal D, et al. Pharmacological evaluation of glutamate transporter 1 (GLT-1) mediated neuroprotection following cerebral ischemia/reperfusion injury. Eur J Pharmacol. 2010;638(1–3):65–71.
  • Altas M, Meydan S, Aras M, et al. Effects of ceftriaxone on ischemia/reperfusion injury in rat brain. J Clin Neurosci. 2013;20(3):457–461.
  • High WA. Medicolegal aspects of dermatology. Semin Cutan Med Surg. 2013;32(4):183–184.
  • Ota Y, Kuyotaka K, Sugiyama S, et al. Impairment of endothelium-dependent relaxation of rabbit aortas by cigarette smoke extract-role of free radicals and attenuation by captopril. Atherosclerosis. 1997;131(2):195–202.
  • De-Cavanagh EM, Inserra F, Fraga CG. Enalapril and captopril enhance glutathione-dependent antioxidant defenses in mouse tissues. Am J Physiol Integr Comp Physiol. 2000;278(3):R572–R577.
  • Djordjevic VB, Pavlovic D, Pejovic M, et al. Changes of lipid peroxides and antioxidative factors levels in blood of patients treated with ACE inhibitors. Clin Nephrol. 1997;47(4):243–247.
  • Khaper N, Singal PK. Effects of after load-reducing drugs on pathogenesis of antioxidant changes and congestive heart failure in rats. J Am Coll Cardiol. 1997;29(4): 856–861.
  • Noda Y, Mori A, Parker L. Free radical scavenging properties of alacepril metabolites and lisinopril. Res Commun Mol Pathol Pharmacol. 1997;96(2):125–136.
  • Anderson B, Khaper N, Dhalla AK, et al. Anti-free radical mechanisms in captopril protection against reperfusion injury in isolated rat hearts. Can J Cardiol. 1996;12(10):1099–1104.
  • Bagchi D, Prasad R, Das DK. Direct scavenging of free radicals by captopril, an angiotensin converting enzyme inhibitor. Biochem Biophys Res Commun. 1989;158(1): 52–57.
  • Chopra M, Scott N, McMurray J, et al. Captopril: a free radical scavenger. Br J Pharmacol 1989;27:396–397.
  • De-Cavanagh EM, Inserra F, Ferder L, et al. Superoxide dismutase and glutathione peroxidase activities are increased by enalapril and captopril in mouse liver. FEBS Lett. 1995;361(1):22–24.
  • Güllüoglu BM, Aktan AO, Yegen C, et al. Endothelin release is augmented with captopril in rat ischemia–reperfusion of the liver. Res Exp Med (Berl). 1996;196(4):227–233.
  • Ross SD, Kron IL, Gangemi JJ, et al. Attenuation of lung reperfusion injury after transplantation using an inhibitor of nuclear factor-κB. Am J Physiol Lung Cell Mol Physiol. 2000;279(3):L528–L536.
  • Molinero LL, Alegre ML. Role of T cell-NF-κB in transplantation. Transplant Rev (Orlando). 2012:26(3):189–200.
  • Chiang CH, Pai HI, Liu SL. Ventilator-induced lung injury (VILI) promotes ischemia/reperfusion lung injury (I/R) and NF-kappaB antibody attenuates both injuries. Resuscitation. 2008;79(1):147–154.
  • Krishnadasan B, Naidu B, Rosengart M, et al. Decreased lung ischemia–reperfusion injury in rats after preoperative administration of cyclosporine and tacrolimus. J Thorac Cardiovasc Surg. 2002;123(4):756–767.
  • Ishii M, Suzuki Y, Takeshita K, et al. Inhibition of c-Jun NH2-terminal kinase activity improves ischemia/reperfusion injury in rat lungs. J Immunol. 2004;172(4):2569–2577.
  • Bennett BL, Sasaki DT, Murray BW, et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc Natl Acad Sci U S A. 2001;98(24):13681–13686.
  • Ozaki KS, Kimura S, Murase N. Use of carbon monoxide in minimizing ischemia reperfusion injury in transplantation. Transplant Rev. 2012;26(2):125–139.
  • Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998;391(6662):82–86.
  • Ricote M, Li AC, Willson TM, et al.. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature. 1998;391(6662):79–82.
  • Ito K, Shimada J, Kato D, et al. Protective effects of preischemic treatment with pioglitazone, a peroxisome proliferator-activated receptor-gamma ligand, on lung ischemia–reperfusion injury in rats. Eur J Cardiothorac Surg. 2004;25(4):530–536.
  • Wang H, Zhu QW, Ye P, et al. Pioglitazone attenuates myocardial ischemia–reperfusion injury via up-regulation of ERK and COX-2. Biosci Trends. 2012;6(6):325–332.
  • Petrat F, Drowatzky J, Boengler K, et al. Protection from glycine at low doses in ischemia–reperfusion injury of the rat small intestine. Eur Surg Res. 2011;46: 180–187.
  • Aldemir M, Boşnak M, Al B, et al.. Effects of molsidomine and lexipafant in hepatic ischemia–reperfusion injury. Injury. 2004;35(3):232–237.
  • Naidu BV, Krishnadasan B, Farivar AS, et al. Early activation of the alveolar macrophage is critical to the development of lung ischemia–reperfusion injury. J Thorac Cardiovasc Surg. 2003;126(1):200–207.
  • Zhao M, Fernandez LG, Doctor A, et al. Alveolar macrophage activation is a key initiation signal for acute lung ischemia–reperfusion injury. Am J Physiol Lung Cell Mol Physiol. 2006;291(5):L1018–L1026.
  • Naidu BV, Krishnadasan B, Byrne K, et al. Regulation of chemekine expression by cyclosporine A in alveolar macrophages exposed to hypoxia and reoxygenation. Ann Thorac Surg. 2002;74(3):899–905.
  • Mizutani A, Okajima K, Uchiba M, et al. Antithrombin reduces ischemia/reperfusion-induced renal injury in rats by inhibiting leukocyte antivation through promotion of prostacyclin production. Blood. 2003;101(8):3029–3036.
  • Zambas NA, Karkos CD, Kambaroudis AG, et al. Protective effect of antithrombin III against lung and myocardial injury in lower-limb ischemia–reperfusion syndrome. Ann Vasc Surg. 2012;26(4):566–570.
  • Goto T, Ishizaka A, Kobayashi F, et al. Importance of tumor necrosis factor-alpha cleavage process in post-transplantation lung injury in rats. Am J Respir Crit Care Med. 2004;170(11):1239–1246.
  • Khimenko PL, Bagby GJ, Fuseler J, et al. Tumor necrosis factor-alpha in ischemia and reperfusion injury in rat lungs. J Appl Physiol (1985). 1998;85(6):2005–2011.
  • Genovés P, García D, Cejalvo D, et al. Pentoxyfilline in liver ischemia and reperfusion. J Invest Surg. 2013;27:114–124.
  • Eun BL, Liu XH, Barks JD. Pentoxifylline attenuates hypoxic-ischemic brain injury in immature rats. Pediatr Res. 2000;47(1):73–78.
  • Clark SC. Lung injury after cardiopulmonary bypass. Perfusion. 2006;21:225–228.
  • Ji Q, Zhang L, Jia H, et al. Pentoxifylline inhibits endotoxin-induced NF-kappa B activation and associated production of proinflammatory cytokines. Ann Clin Lab Sci. 2004;34(4):427–436.
  • Lloris Carsi JM, Cejalvo Lapeña D, et al. Pentoxifylline protects the small intestine after severe ischemia and reperfusion. Exp Clin Transplant. 2013;11(3): 250–258.
  • Davila-Esqueda ME, Martinez-Morales F. Pentoxifylline diminishes the oxidative damage to renal tissue induced by streptozotocin in the rat. Exp Diabesity Res. 2004;5(4):245–251.
  • Guggilam A, Haque M, Kerut EK, et al. TNF-alpha blockade decreases oxidative stress in the paraventricular nucleus and attenuates sympatho-excitation in heart failure rats. Am J Physiol Heart Circ Physiol. 2007;293(1):H599–H609.
  • Sirin BH, Yilik L, Coskun E, et al. Pentoxifylline reduces injury of the brain in transient ischemia. Acta Cardiol. 1998;53(2):89–95.
  • Thabut G, Brugiere O, Leseche G, et al. Preventive effect of inhaled nitric oxide and pentoxifylline on ischemia/reperfusion injury after lung transplantation. Transplantation. 2001;71(9):1295–1300.
  • Zhang M, Xu YJ, Mengi SA, et al. Therapeutic potentials of pentoxifylline for treatment of cardiovascular diseases. Exp Clin Cardiol. 2004;9(2):103–111.
  • Ciuffetti G, Mercuri M, Ott C, et al. Use of pentoxifylline as a inhibitor of free radical generation in peripheral vascular disease. Eur J Clin Pharmacol. 1991;41(6):511–515.
  • Chapelier A, Reignier J, Mazmanian M, et al. Pentoxifylline and lung ischemia–reperfusion injury: application to lung transplantation. J. Cardiovasc. Pharmacol. 1995;25(Suppl 2):S130–S133.
  • Adams JG Jr, Dhar A, Shukla SD, et al. Effect of pentoxifylline on tissue injury and platelet-activating factor production during ischemia–reperfusion injury. J. Vasc Surg. 1995;21(5):742–748.
  • Berens KL, Luke DR. Pentoxifylline in the isolated perfused rat kidney. Transplantation. 1990;49(5):876–879.
  • Myers SI, Horton JW, Hernandez R, et al. Pentoxifylline protects splanchnic prostacyclin synthesis during mesenteric ischemia/reperfusion. Prostaglandins. 1994;47(2):137–150.
  • Ege T, Arar C, Canbaz S, et al. The importance of aprotinin and pentoxifylline in preventing leukocyte sequestration and lung injury caused by protamine at the end of cardiopulmonary bypass surgery. Thorac Cardiovasc Surg. 2004;52(1):10–15.
  • Hishikari K, Suzuki J, Ogawa M, et al. Pharmacological activation of the prostaglandin E2 receptor EP4 improves cardiac function after myocardial ischaemia/reperfusion injury. Cardiovasc Res. 2009;81(1):123–132.
  • Kazar R, MacQuiggan M. Nutritional support in trauma patients. In: Nutritional Considerations in the Intensive Care Unit: Science, Rationale and Practice. Shikora S, Martindale R, Schwaitzberg, S, eds. Iowa (USA): Kendall/Hunt Publishing Company; 2002;229–244.
  • Mora, R. Inmunonutrición. In: Soporte Nutricional Especial. Bogotá: Panamericana Ed; 2000;338–341.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.