Acessibilidade / Reportar erro

Effect of the systemic administration of methylprednisolone on the lungs of brain-dead donor rats undergoing pulmonary transplantation

Abstract

OBJECTIVE:

Most lung transplants are obtained from brain-dead donors. The physiopathology of brain death involves hemodynamics, the sympathetic nervous system, and inflammatory mechanisms. Administering methylprednisolone 60 min after inducing brain death in rats has been shown to modulate pulmonary inflammatory activity. Our objective was to evaluate the effects of methylprednisolone on transplanted rat lungs from donors treated 60 min after brain death.

METHODS:

Twelve Wistar rats were anesthetized, and brain death was induced. They were randomly divided into two groups (n = 6), namely a control group, which was administered saline solution, and a methylprednisolone group, which received the drug 60 min after the induction of brain death. All of the animals were observed and ventilated for 2 h prior to being submitted to lung transplantation. We evaluated the hemodynamic and blood gas parameters, histological score, lung tissue levels of thiobarbituric acid-reactive substances, level of superoxide dismutase, level of tumor necrosis factor-alpha, and level of interleukin-1 beta.

RESULTS:

After transplantation, a significant reduction in the levels of tumor necrosis factor-alpha and IL-1β was observed in the group that received methylprednisolone (p = 0.0084 and p = 0.0155, respectively). There were no significant differences in tumor necrosis factor-alpha and superoxide dismutase levels between the control and methylprednisolone groups (p = 0.2644 and p = 0.7461, respectively). There were no significant differences in the blood gas parameters, hemodynamics, and histological alterations between the groups.

CONCLUSION:

The administration of methylprednisolone after brain death in donor rats reduces inflammatory activity in transplanted lungs but has no influence on parameters related to oxidative stress.

Lung Transplantation; Brain Death; Experimental Model; Methylprednisolone; Oxidative Stress


INTRODUCTION

The availability of donor organs remains a challenge for transplantation programs worldwide. This issue is a particular problem for lung transplantation, which is the ultimate option for patients with end-stage pulmonary disease. Approximately 5%-20% of donors who elect for organ retrieval have lungs that are considered viable for lung transplantation. However, the number of patients enrolled on waiting lists has been increasing (11. Trulock EP, Christie JD, Edwards LB, Boucek MM, Aurora P, et al. Registry of the International Society for Heart and Lung Transplantation: twenty-fourth official adult lung and heart-lung transplantation report-2007. J Heart Lung Transplant. 2007;26(8):782-95, http://dx.doi.org/10.1016/j.healun.2007.06.003.
http://dx.doi.org/10.1016/j.healun.2007....

2. Fernandes PM, Samano MN, Junqueira JJ, Waisberg DR, Jatene FB, et al. Lung donor profile in the State of Sao Paulo, Brazil, in 2006. J Bras Pneumol. 2008;34(7):497-505, http://dx.doi.org/10.1590/S1806-37132008000700010.
http://dx.doi.org/10.1590/S1806-37132008...
-33. Organ and tissue donation and transplantation (update 2000). Canadian Medical Association. Cmaj. 2000;163(2):206-11.).

Brain-dead donors still represent the main source of organs for transplantation. It is well known that brain death compromises the viability of transplanted solid organs, and it can affect lung donor viability through sympathetic, hemodynamic, and inflammatory mechanisms that lead to adrenergic storm and neurogenic pulmonary edema (44. Avlonitis VS, Fisher AJ, Kirby JA, Dark JH. Pulmonary transplantation: the role of brain death in donor lung injury. Transplantation. 2003;75(12):1928-33, http://dx.doi.org/10.1097/01.TP.0000066351.87480.9E.
http://dx.doi.org/10.1097/01.TP.00000663...
). In addition, these lungs are at risk for acute lung injury secondary to trauma, prolonged mechanical ventilation, transfusion, ischemia, aspiration, and infection (44. Avlonitis VS, Fisher AJ, Kirby JA, Dark JH. Pulmonary transplantation: the role of brain death in donor lung injury. Transplantation. 2003;75(12):1928-33, http://dx.doi.org/10.1097/01.TP.0000066351.87480.9E.
http://dx.doi.org/10.1097/01.TP.00000663...
,55. de Perrot M, Liu M, Waddell TK, Keshavjee S. Ischemia-reperfusion-induced lung injury. Am J Respir Crit Care Med. 2003;167(4):490-511, http://dx.doi.org/10.1164/rccm.200207-670SO.
http://dx.doi.org/10.1164/rccm.200207-67...
).

Currently, there is mounting evidence to suggest that organ grafts are not immunologically inert. Donor risk factors, such as previous disease, age, cause of death, donor management, and, most importantly, brain death, reprogram the graft into an immunologically active organ (55. de Perrot M, Liu M, Waddell TK, Keshavjee S. Ischemia-reperfusion-induced lung injury. Am J Respir Crit Care Med. 2003;167(4):490-511, http://dx.doi.org/10.1164/rccm.200207-670SO.
http://dx.doi.org/10.1164/rccm.200207-67...
). Accordingly, treating potential brain-dead donors may reduce immune activation and improve the condition of the lung that is to be transplanted (66. Pratschke J, Neuhaus P, Tullius SG. What can be learned from brain-death models? Transpl Int. 2005;18(1):15-21, http://dx.doi.org/10.1111/j.1432-2277.2004.00018.x.
http://dx.doi.org/10.1111/j.1432-2277.20...
).

The administration of systemic corticosteroids to brain-dead donors is known to be beneficial, mostly because it modulates the systemic inflammatory response caused by brain death (77. Follette DM, Rudich SM, Babcock WD. Improved oxygenation and increased lung donor recovery with high-dose steroid administration after brain death. J Heart Lung Transplant. 1998;17(4):423-9.). This modulation improves graft viability by reducing both the release of pro-inflammatory molecules and the production of leukocyte adhesion molecules, thereby increasing the clearance of alveolar fluids (77. Follette DM, Rudich SM, Babcock WD. Improved oxygenation and increased lung donor recovery with high-dose steroid administration after brain death. J Heart Lung Transplant. 1998;17(4):423-9.,88. Wigfield C, Golledge H, Shenton B, Kirby J, Dark J. Ameliorated reperfusion injury in lung transplantation after reduction of brain death induced inflammatory graft damage in the donor. J Heart Lung Transplant. 2002;21(1):57, http://dx.doi.org/10.1016/S1053-2498(01)00440-5.
http://dx.doi.org/10.1016/S1053-2498(01)...
). However, the optimal timing of the administration of corticosteroids during brain death remains unknown. Previously, we showed that in a model of brain death, the administration of methylprednisolone at 5 and 60 min reduced the levels of TNF-α to a similar extent (99. Pilla E, Pereira RB, Forgiarini Jr LA, Forgiarini LF, Paludo AO, Kulczynski JMU, Cardoso PFG, Andrade CF. Effects of methylprednisolone on inflammatory activity and oxidative stress in the lungs of brain-dead rats. J Bras Pneumol. 2013;39(2):173-180, http://dx.doi.org/10.1590/S1806-37132013000200008.
http://dx.doi.org/10.1590/S1806-37132013...
). In the present study, we hypothesized that the transplanted lungs from donors treated with methylprednisolone after 60 min of brain death would exhibit less inflammatory activity than those of untreated donors.

MATERIALS AND METHODS

The Ethical and Research Committee of Hospital de Clínicas de Porto Alegre approved the protocols used in this study. All of the animals received humane care that was in compliance with the Principles of Laboratory Animal Care formulated by the National Society for Medical Research and the Guide for the Care and Use of Laboratory Animals (Institute of Laboratory Animal Resources, published by the National Institutes of Health, Publication No. 86-23, revised 1996).

Surgical procedure

Twelve male rats, weighing 300-400 g each, underwent general anesthesia induced by an intraperitoneal injection of ketamine (100 mg/kg) and xylazine (15 mg/kg). Anesthesia was followed by a tracheostomy using an indwelling 14-gauge cannula (Abbocath® #14; Abbott Laboratories, Abbott Park, IL, EUA) and ventilation at a rate of 70-80 breaths/min with a tidal volume of 10 ml/kg of inspired room air (Harvard Rodent Ventilator, model 683; Harvard Apparatus Co., Millis, MA). The right carotid artery was dissected and cannulated using a 24-gauge cannula (Becton Dickinson, Franklin Lakes, NJ, USA) to record the mean arterial pressure (MAP) and heart rate (HR) (Sirecust 730 Siemens, Solna, Sweden). The right jugular vein was also dissected and cannulated in the same manner. Normal saline was used to flush the lines, with a total volume of 5 ml/kg/h used in all of the animals, and a warmed surgical table was used to maintain the body temperature at 37°C during the procedure.

Two hours following brain death, the donor rat lungs were flushed in an antegrade manner with 20 ml of cold (4°C), low-potassium dextran solution (LPD-Perfadex®; Vitrolife, Göteborg, Sweden) at a pressure of 20 cmH2O via the pulmonary artery, as described elsewhere (1010. Sanchez PG, Martins LK, Martins FK, Cardoso PF, Andrade CF, et al. Technical modification of unilateral lung transplantation in rats. J Bras Pneumol. 2007;33(4):448-53, http://dx.doi.org/10.1590/S1806-37132007000400015.
http://dx.doi.org/10.1590/S1806-37132007...
). The heart-lung block was extracted with the lungs inflated at the end-tidal volume. The left lung graft was isolated, prepared, and stored in LPD at 4°C for 90 min.

Twelve recipients were anaesthetized, intubated (14-gauge catheter), and ventilated. Then, they underwent a left thoracotomy, and the pulmonary vessels and left bronchus were anastomosed using a standard cuff technique (1010. Sanchez PG, Martins LK, Martins FK, Cardoso PF, Andrade CF, et al. Technical modification of unilateral lung transplantation in rats. J Bras Pneumol. 2007;33(4):448-53, http://dx.doi.org/10.1590/S1806-37132007000400015.
http://dx.doi.org/10.1590/S1806-37132007...
). The arterial clamps were released approximately 3 h after harvest. The recipient animals were then observed for 2 h. At the end of the observation period, the pulmonary hilum was clamped, and the lower half of the lung was immediately snap-frozen in liquid nitrogen and stored at −80°C. The upper half of the lung was embedded in 10% formaldehyde for histopathology.

Induction of brain death

The brain death model used here has previously been described in detail (1111. Bittner HB, Kendall SW, Campbell KA, Montine TJ, Van Trigt P. A valid experimental brain death organ donor model. J Heart Lung Transplant. 1995;14(2):308-17.,1212. Herijgers P, Leunens V, Tjandra-Maga TB, Mubagwa K, Flameng W. Changes in organ perfusion after brain death in the rat and its relation to circulating catecholamines. Transplantation. 1996;62(3):330-5, http://dx.doi.org/10.1097/00007890-199608150-00005.
http://dx.doi.org/10.1097/00007890-19960...
). Briefly, a frontolateral trepanation (1×1 mm with a dental drill) was performed, and a 14-gauge Fogarty balloon catheter (Baxter Health Care Corp., Irvine, CA, USA) was introduced into the extradural space with the tip pointed caudally. The balloon was inflated with 0.75 ml of water for 1 min, producing a sudden increase in intracranial pressure, which resulted in rapid, progressive brain injury leading to immediate brain death. A sharp rise, followed by a subsequent drop, of blood pressure and heart rate defined the initiation of brain death. The presence of brain death was confirmed by the absence of corneal reflexes and by the apnea test.

Study groups

After the initial procedures, the animals were randomly assigned to two brain-dead donor groups, namely the control (n = 6) and corticosteroid (n = 6) groups. In the control group (CONTROL), an IV bolus of 0.9% saline solution (0.3 ml) was administered 60 min after the induction of brain death. In the corticosteroid group (MET), an IV bolus of methylprednisolone (30 mg/kg) diluted in 0.2 ml of normal saline was administered 60 min after the induction of brain death. The timing and dose of methylprednisolone were based on a previous study (99. Pilla E, Pereira RB, Forgiarini Jr LA, Forgiarini LF, Paludo AO, Kulczynski JMU, Cardoso PFG, Andrade CF. Effects of methylprednisolone on inflammatory activity and oxidative stress in the lungs of brain-dead rats. J Bras Pneumol. 2013;39(2):173-180, http://dx.doi.org/10.1590/S1806-37132013000200008.
http://dx.doi.org/10.1590/S1806-37132013...
).

Sampling

All of the donor and recipient animals were monitored for 120 min and were submitted to the same ventilation procedures. Arterial blood samples were obtained at the time of insertion of the arterial line (basal) and at 60 and 120 min in the donor groups. In the recipient groups, the samples were drawn at 5 and 120 min after transplantation for blood gas analysis.

The lower half of the left lung was snap-frozen in liquid nitrogen and stored at −80°C for analyses of lipid peroxidation and superoxide dismutase (SOD) activity and for the protein quantification of TNF-α and IL-1β (1313. Pearce ML, Yamashita J, Beazell J. Measurement of Pulmonary Edema. Circulation research. 1965;16:482-8, http://dx.doi.org/10.1161/01.RES.16.5.482.
http://dx.doi.org/10.1161/01.RES.16.5.48...
).

Pulmonary TNF-α and IL-1β assay

After the samples were thawed, a 96-well plate was coated with monoclonal antibodies against TNF-α and IL-1β. The wells were filled with either 100 µl of homogenized lung (diluted 1∶2), 100 µl of positive or negative controls, or 100 µl of recombinant TNF-α or IL-1β at concentrations established by the manufacturer (Creative Biomart, NY, USA). Then, 100 µl of polyclonal anti-TNF-α and anti-IL-1β conjugated to peroxidase was added to the wells, and the samples were incubated for 3 h at room temperature. Following the incubation, the plate was washed four times with a detergent solution. The color change was then induced by adding hydrogen peroxide (0.02%) and tetramethylbenzene (2%). The reaction was interrupted 30 min later using sulfuric acid (1 M). The color intensity was assessed by obtaining optical density measurements using an ELISA automatic reader (Titertek Multiskan®) at a wavelength of 450 nm. The TNF-α and IL-1β concentrations in the homogenized lung samples were calculated based on the results of a standard curve.

Determination of thiobarbituric acid reactive substances (TBARS)

The products generated by lipid peroxidation were quantified by the TBARS reaction using 3 mg of protein per sample. The samples were first incubated at 90°C for 30 min. Then, 500 µl of 0.37% thiobarbituric acid and 15% trichloroacetic acid was added to the samples, and they were centrifuged at 4°C at 2000xg for 15 min. The absorbance was then determined by spectrophotometry at 535 nm (1414. Buege JA, Aust SD. Microsomal lipid peroxidation. Methods Enzymol. 1978;52:302-10.).

Superoxide dismutase (SOD)

The activity of SOD was determined using the pulse radiolytic method based on the auto-oxidation of epinephrine, as described by Misra and Fridovich (1515. Misra HP, Fridovich I. The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J Biol Chem. 1972;247(10):3170-5.).

Histology

The portion of lung fixed in formalin was embedded in paraffin, cut into 3-mm sections, and stained with hematoxylin and eosin. A pathologist blinded to the experimental protocol performed a quantitative examination under light microscopy. Each lung sample was examined under both low- and high-power fields, and 20 fields were randomly selected and analyzed. The severity of the histological lesions was assigned a score based on five parameters, namely intra-alveolar edema, hyaline membrane formation, hemorrhage, focal alveolar collapse or consolidation, and epithelial desquamation or the necrosis of airways or alveoli. Each parameter was evaluated semi-quantitatively according to the following scale: 0 =  absent, 1 =  mild, 2 =  moderate, and 3 =  prominent. For each animal, the scored value of each parameter was added to produce a final score (1616. Fujino Y, Goddon S, Chiche JD, Hromi J, Kacmarek RM. Partial liquid ventilation ventilates better than gas ventilation. Am J Respir Crit Care Med. 2000;162(2 Pt 1):650-7.).

Statistical analysis

All of the data collected from the experiments were coded, recorded, and analyzed using SPSS 16.0 for Windows (Chicago, IL, USA). To analyze the differences in the demographic variables over time in each group, the Wilcoxon nonparametric test was used. To analyze the differences in the demographic variables between groups, the Mann-Whitney U nonparametric test was used. For each test, the data are expressed as the median ± the interquartile range (IR), and p<0.05 was accepted as statistically significant. For the cytokine, TBARS, SOD, and histology analyses, the Student's t-test was used; the data are expressed as the mean value ± standard error (SE), and p<0.05 was accepted as statistically significant.

RESULTS

There were no differences between the two groups with respect to procedure time, blood gas measurements, and mean arterial pressure. In the donor rats, the mean arterial pressure decreased significantly in both groups after the induction of brain death (p<0.05) (Table 1). There were no differences between the two groups with respect to MAP and blood gas analyses following transplantation (Table 2).

Table 1
The level of arterial blood gases and the mean arterial pressure in donor rats before and 2 h following brain death.
Table 2
The levels of arterial blood gases and the mean arterial pressure in recipients at baseline and 2 h following lung transplantation.

There were no significant differences in the results of the TBARS and SOD assays between the MET and CONTROL groups (p = 0.2644 and p = 0.7461, respectively). The TNF-α and IL-1β tecidual concentrations were significantly lower in the MET group (p = 0.0084 and p = 0.0155, respectively) (Figures 1 and 2).

Figure 1
Pulmonary tumor necrosis factor-alpha assay. After transplantation, there was a significant decrease in the levels of tumor necrosis factor-alpha in the methylprednisolone group compared with the control group (*p = 0.0084). The values are expressed as the mean ± standard error of the mean.
Figure 2
Pulmonary interleukin-1 beta assay. After transplantation, there was a significant decrease in the levels of interleukin-1 beta in the methylprednisolone group compared with the control group (*p = 0.0155). The values are expressed as the mean ± standard error of the mean.

The severity of the histological damage, as measured by the histology score (HIS), was similar between the CONTROL and MET groups. The predominant finding was mild focal alveolar collapse and congestion (Figure 3).

Figure 3
The main histological findings in the control and methylprednisolone groups after lung transplantation. The lungs from the methylprednisolone group (A) showed the recruitment of septal neutrophils (N) and edema (E). The lungs from the control group (B) showed areas of hemorrhage (H) (200× magnification).

DISCUSSION

While the early survival rates of patients after lung transplantation have improved (1), their long-term survival remains challenging (22. Fernandes PM, Samano MN, Junqueira JJ, Waisberg DR, Jatene FB, et al. Lung donor profile in the State of Sao Paulo, Brazil, in 2006. J Bras Pneumol. 2008;34(7):497-505, http://dx.doi.org/10.1590/S1806-37132008000700010.
http://dx.doi.org/10.1590/S1806-37132008...
,33. Organ and tissue donation and transplantation (update 2000). Canadian Medical Association. Cmaj. 2000;163(2):206-11.). Immunological events are considered to be key factors in this scenario, and these events may be aggravated by the alterations triggered by brain death (1717. Pratschke J, Wilhelm MJ, Kusaka M, Cooper DK, Hancock WW, et al. Brain death and its influence on donor organ quality and outcome after transplantation. Transplantation. 1999;67(3):343-8, http://dx.doi.org/10.1097/00007890-199902150-00001.
http://dx.doi.org/10.1097/00007890-19990...
). The intensive care of potential donors may reflect the outcome after transplantation, and the administration of methylprednisolone has been suggested to be beneficial in promoting organ maintenance in brain-dead donors (1818. Dhar R, Cotton C, Coleman J, Brockmeier D, Marklin G, et al. Comparison of high- and low-dose corticosteroid regimens for organ donor management. J Crit Care. 2012;28(1):111.e1-7.).

Currently, the use of corticosteroids in potential donors remains controversial, and there is no consensus regarding the time of the first administration or the ideal dose required to achieve an adequate anti-inflammatory effect (1919. Kutsogiannis DJ, Pagliarello G, Doig C, Ross H, Shemie SD. Medical management to optimize donor organ potential: review of the literature. Can J Anaesth. 2006;53(8):820-30.). While several studies have shown that resuscitative hormonal protocols are promising, to date, no standard protocols for lung retrieval have been accepted (1919. Kutsogiannis DJ, Pagliarello G, Doig C, Ross H, Shemie SD. Medical management to optimize donor organ potential: review of the literature. Can J Anaesth. 2006;53(8):820-30.-2020. Venkateswaran RV, Patchell VB, Wilson IC, Mascaro JG, Quinn DW, et al. Early donor management increases the retrieval rate of lungs for transplantation. Ann Thorac Surg. 2008;85(1):278-86, http://dx.doi.org/10.1016/j.athoracsur.2007.07.092.
http://dx.doi.org/10.1016/j.athoracsur.2...
). The use of corticosteroids in brain-dead donors is justified by two potential beneficial effects, namely a reduction in the inflammatory response and a replacement of the blood hormonal levels. Faropoulos et al. observed that in a baboon model, cortisol levels were reduced 15-45 min after the induction of brain death and almost disappeared after approximately 4 h (2121. Faropoulos K, Apostolakis E. Brain death and its influence on the lungs of the donor: how is it prevented? Transplant Proc. 2009;41(10):4114-9, http://dx.doi.org/10.1016/j.transproceed.2009.09.087.
http://dx.doi.org/10.1016/j.transproceed...
).

In our previous study, we found that both the early (5 min) and late (60 min) administration of methylprednisolone reduced TNF-α to levels similar to those present 2 h after brain death. In the present study, we administered methylprednisolone after 60 min of brain death to simulate the clinical scenario, in which the prompt diagnosis of brain death is usually delayed. We retrieved donor lungs after 2 h of brain death, under the assumption that there was no harmful impact on lung function until 5 h following brain death (2222. Avlonitis VS, Kirby JA, Dark JH. The effect of time from donor brain death to retrieval on reperfusion injury after lung transplantation. The Journal of heart and lung transplantation: the official publication of the International Society for Heart Transplantation. 2005;24(2):S121, http://dx.doi.org/10.1016/j.healun.2004.11.264.
http://dx.doi.org/10.1016/j.healun.2004....
). However, other studies have shown that 6 h is the minimum elapsed time following brain death necessary for the analysis of inflammatory mediators (2323. Zweers N, Petersen AH, van der Hoeven JA, de Haan A, de Leij LF, et al. Donor brain death aggravates chronic rejection after lung transplantation in rats. Transplantation. 2004;78(9):1251-8, http://dx.doi.org/10.1097/01.TP.0000142679.45418.96.
http://dx.doi.org/10.1097/01.TP.00001426...
).

TNF-α is the main inflammatory mediator after brain death. In addition to its direct participation in the inflammatory process, it is also responsible for the activation of apoptotic processes (2424. Van Der Hoeven JA, Moshage H, Schuurs T, Nijboer M, Ploeg RJ, et al. Brain death induces apoptosis in donor liver of the rat. Transplantation. 2003;76(8):1150-4, http://dx.doi.org/10.1097/01.TP.0000080983.14161.95.
http://dx.doi.org/10.1097/01.TP.00000809...
). Zhou HC et al. showed that carbon monoxide inhalation reduced the expression of TNF-α, IL-6, adhesion molecules, and pro-apoptotic pathways, improving PaO2/FiO2 and the base excess and acidosis generated by brain death (2525. Zhou HC, Ding WG, Cui XG, Zhang B, Li WZ, et al. Carbon monoxide inhalation ameliorates conditions of lung grafts from rat brain death donors. Chin Med J (Engl). 2008;121(15):1411-9.). Our study showed that the administration of a corticosteroid 1 h after brain death can reduce the concentration of TNF-α, even after lung transplantation, thus demonstrating its benefit in this scenario.

Several studies have measured IL-1β levels in models of acute lung injury (2626. Strieter RM, Kunkel SL. Acute lung injury: the role of cytokines in the elicitation of neutrophils. J Investig Med. 1994;42(4):640-51.), brain death (2727. Skrabal CA, Thompson LO, Potapov EV, Southard RE, Youker KA, et al. Organ-specific regulation of pro-inflammatory molecules in heart, lung, and kidney following brain death. J Surg Res. 2005;123(1):118-25, http://dx.doi.org/10.1016/j.jss.2004.07.245.
http://dx.doi.org/10.1016/j.jss.2004.07....
), and lung transplantation (2828. Blocher S, Wilker S, Sucke J, Pfeil U, Weimer R, et al. Acute rejection of experimental lung allografts: characterization of intravascular mononuclear leukocytes. Clin Immunol. 2007;124(1):98-108, http://dx.doi.org/10.1016/j.clim.2007.04.005.
http://dx.doi.org/10.1016/j.clim.2007.04...
). High levels of IL-1β have previously been associated with acute lung injury and rejection (2626. Strieter RM, Kunkel SL. Acute lung injury: the role of cytokines in the elicitation of neutrophils. J Investig Med. 1994;42(4):640-51.). In our study, we observed a significant decrease in IL-1β in the transplanted lungs of brain-dead donors treated with methylprednisolone, reflecting its potential protective anti-inflammatory effect after lung reperfusion, which would consequently reduce ischemia-reperfusion injury.

Cytokines released after brain injury are likely crucial factors associated with the inflammatory activation that takes place in peripheral organs. Severe cerebral injury leads to the release of cytokines, such as TNF-α, IL-1β, and IL-6, from astrocytes and microglial cells (2929. Woiciechowsky C, Schoning B, Daberkow N, Asche K, Docke WD, et al. Brain IL-1beta increases neutrophil and decreases lymphocyte counts through stimulation of neuroimmune pathways. Neurobiol Dis. 1999 Jun;6(3):200-8.). These cytokines cross the blood-brain barrier and reach peripheral organs and tissues (3030. Ott L, McClain CJ, Gillespie M, Young B. Cytokines and metabolic dysfunction after severe head injury. J Neurotrauma. 1994;11(5):447-72, http://dx.doi.org/10.1089/neu.1994.11.447.
http://dx.doi.org/10.1089/neu.1994.11.44...
), resulting in the expression of IL-1, IL-2, IL-6, TNF-α, and INF-γ in these organs, as well as the lungs (3131. Takada M, Nadeau KC, Hancock WW, Mackenzie HS, Waaga AM, et al. Effects of explosive brain death on cytokine activation of peripheral organs in the rat. Transplantation. 1998;65(12):1533-42, http://dx.doi.org/10.1097/00007890-199806270-00001.
http://dx.doi.org/10.1097/00007890-19980...
). An experimental study that performed cross circulation between controls and rats that underwent sudden brain death revealed an elevation of levels of lymphocytic and leukocyte inflammatory molecules (3131. Takada M, Nadeau KC, Hancock WW, Mackenzie HS, Waaga AM, et al. Effects of explosive brain death on cytokine activation of peripheral organs in the rat. Transplantation. 1998;65(12):1533-42, http://dx.doi.org/10.1097/00007890-199806270-00001.
http://dx.doi.org/10.1097/00007890-19980...
).

Focal or generalized cerebral ischemia promotes the release of IL-1β and TNF-α in the brain (3232. McKeating EG, Andrews PJ, Signorini DF, Mascia L. Transcranial cytokine gradients in patients requiring intensive care after acute brain injury. Br J Anaesth. 1997;78(5):520-3.), which induces the production of IL-6. It has been shown that patients in the intensive care unit for cerebral trauma had higher IL-6 levels in their jugular venous blood than their arterial blood (3333. Martin TR. Lung cytokines and ARDS: Roger S. Mitchell Lecture. Chest. 1999;116(1 Suppl):2S-8S, http://dx.doi.org/10.1378/chest.116.suppl_1.2S.
http://dx.doi.org/10.1378/chest.116.supp...
). A clinical study showed that patients with stroke exhibited an elevated expression of IL-1β, IL-8, and IL-17 in peripheral mononuclear cells and concluded that cerebral ischemia stimulates the production of inflammatory cytokines in peripheral leukocytes (3434. Kostulas N, Pelidou SH, Kivisakk P, Kostulas V, Link H. Increased IL-1beta, IL-8, and IL-17 mRNA expression in blood mononuclear cells observed in a prospective ischemic stroke study. Stroke. 1999;30(10):2174-9, http://dx.doi.org/10.1161/01.STR.30.10.2174.
http://dx.doi.org/10.1161/01.STR.30.10.2...
).

After brain death, hemodynamic events occur concomitantly with systemic inflammatory events. This systemic inflammatory response has been associated with the production of cytokines by the brain. However, the precise role of these cytokines and whether the inflammatory response is due to central or peripheral factors are unclear (3535. Reynolds RW. Pulmonary edema as a consequence of hypothalamic lesions in rats. Science. 1963;141(3584):930-2, http://dx.doi.org/10.1126/science.141.3584.930.
http://dx.doi.org/10.1126/science.141.35...
).

In this study, the administration of methylprednisolone did not change hemodynamics and blood exchange prior to and after transplantation, indicating that its administration does not add any additional detrimental effect, as was previously shown by Pilla et al. (99. Pilla E, Pereira RB, Forgiarini Jr LA, Forgiarini LF, Paludo AO, Kulczynski JMU, Cardoso PFG, Andrade CF. Effects of methylprednisolone on inflammatory activity and oxidative stress in the lungs of brain-dead rats. J Bras Pneumol. 2013;39(2):173-180, http://dx.doi.org/10.1590/S1806-37132013000200008.
http://dx.doi.org/10.1590/S1806-37132013...
). In contrast, Wigfield et al. showed that in a rat transplantation model, methylprednisolone treatment resulted in better graft function and less inflammatory activity after transplantation (88. Wigfield C, Golledge H, Shenton B, Kirby J, Dark J. Ameliorated reperfusion injury in lung transplantation after reduction of brain death induced inflammatory graft damage in the donor. J Heart Lung Transplant. 2002;21(1):57, http://dx.doi.org/10.1016/S1053-2498(01)00440-5.
http://dx.doi.org/10.1016/S1053-2498(01)...
).

The present study has some limitations, and extrapolation of our results to clinical scenarios should be carefully performed. This study used a small, standardized animal model that involved the induction of brain death by a sudden increase in intracranial pressure. These conditions do not always match the clinical situation because intracranial pressure sometimes rises at a slow rate until brain death is established. Thus, a brain death model that employs a gradual rise of intracranial pressure may be more appropriate. Additionally, the interval between the establishment of brain death and lung harvest was shorter than is common in clinical practice. However, in our experimental model, we tried to reduce all of the confounding factors that were related to the time of preservation and that were detrimental to lung function due to long periods of ventilation after brain death.

We conclude that the administration of methylprednisolone after 60 min of brain death in donor rats reduces the inflammatory response after lung transplantation and has no effect on lipid peroxidation activity. Further studies with larger animals and models that better simulate the clinical scenarios, such as a longer period of brain death, a longer period prior to intervention, and a gradual increase in the intracranial pressure, are needed to confirm the beneficial effects of this treatment method. We suggest that the administration of methylprednisolone in potential multi-organ donors may be a useful strategy for reducing the dangerous effects of reperfusion after lung transplantation.

This research was supported by grants from FIPE/HCPA (Hospital de Clínicas de Porto Alegre Institutional Research Fund) and National Counsel of Technological and Scientific Development (CNPq). We express our gratitude to American Journal Experts for proofreading this manuscript.

REFERENCES

  • 1
    Trulock EP, Christie JD, Edwards LB, Boucek MM, Aurora P, et al. Registry of the International Society for Heart and Lung Transplantation: twenty-fourth official adult lung and heart-lung transplantation report-2007. J Heart Lung Transplant. 2007;26(8):782-95, http://dx.doi.org/10.1016/j.healun.2007.06.003.
    » http://dx.doi.org/10.1016/j.healun.2007.06.003
  • 2
    Fernandes PM, Samano MN, Junqueira JJ, Waisberg DR, Jatene FB, et al. Lung donor profile in the State of Sao Paulo, Brazil, in 2006. J Bras Pneumol. 2008;34(7):497-505, http://dx.doi.org/10.1590/S1806-37132008000700010.
    » http://dx.doi.org/10.1590/S1806-37132008000700010
  • 3
    Organ and tissue donation and transplantation (update 2000). Canadian Medical Association. Cmaj. 2000;163(2):206-11.
  • 4
    Avlonitis VS, Fisher AJ, Kirby JA, Dark JH. Pulmonary transplantation: the role of brain death in donor lung injury. Transplantation. 2003;75(12):1928-33, http://dx.doi.org/10.1097/01.TP.0000066351.87480.9E.
    » http://dx.doi.org/10.1097/01.TP.0000066351.87480.9E
  • 5
    de Perrot M, Liu M, Waddell TK, Keshavjee S. Ischemia-reperfusion-induced lung injury. Am J Respir Crit Care Med. 2003;167(4):490-511, http://dx.doi.org/10.1164/rccm.200207-670SO.
    » http://dx.doi.org/10.1164/rccm.200207-670SO
  • 6
    Pratschke J, Neuhaus P, Tullius SG. What can be learned from brain-death models? Transpl Int. 2005;18(1):15-21, http://dx.doi.org/10.1111/j.1432-2277.2004.00018.x.
    » http://dx.doi.org/10.1111/j.1432-2277.2004.00018.x
  • 7
    Follette DM, Rudich SM, Babcock WD. Improved oxygenation and increased lung donor recovery with high-dose steroid administration after brain death. J Heart Lung Transplant. 1998;17(4):423-9.
  • 8
    Wigfield C, Golledge H, Shenton B, Kirby J, Dark J. Ameliorated reperfusion injury in lung transplantation after reduction of brain death induced inflammatory graft damage in the donor. J Heart Lung Transplant. 2002;21(1):57, http://dx.doi.org/10.1016/S1053-2498(01)00440-5.
    » http://dx.doi.org/10.1016/S1053-2498(01)00440-5
  • 9
    Pilla E, Pereira RB, Forgiarini Jr LA, Forgiarini LF, Paludo AO, Kulczynski JMU, Cardoso PFG, Andrade CF. Effects of methylprednisolone on inflammatory activity and oxidative stress in the lungs of brain-dead rats. J Bras Pneumol. 2013;39(2):173-180, http://dx.doi.org/10.1590/S1806-37132013000200008.
    » http://dx.doi.org/10.1590/S1806-37132013000200008
  • 10
    Sanchez PG, Martins LK, Martins FK, Cardoso PF, Andrade CF, et al. Technical modification of unilateral lung transplantation in rats. J Bras Pneumol. 2007;33(4):448-53, http://dx.doi.org/10.1590/S1806-37132007000400015.
    » http://dx.doi.org/10.1590/S1806-37132007000400015
  • 11
    Bittner HB, Kendall SW, Campbell KA, Montine TJ, Van Trigt P. A valid experimental brain death organ donor model. J Heart Lung Transplant. 1995;14(2):308-17.
  • 12
    Herijgers P, Leunens V, Tjandra-Maga TB, Mubagwa K, Flameng W. Changes in organ perfusion after brain death in the rat and its relation to circulating catecholamines. Transplantation. 1996;62(3):330-5, http://dx.doi.org/10.1097/00007890-199608150-00005.
    » http://dx.doi.org/10.1097/00007890-199608150-00005
  • 13
    Pearce ML, Yamashita J, Beazell J. Measurement of Pulmonary Edema. Circulation research. 1965;16:482-8, http://dx.doi.org/10.1161/01.RES.16.5.482.
    » http://dx.doi.org/10.1161/01.RES.16.5.482
  • 14
    Buege JA, Aust SD. Microsomal lipid peroxidation. Methods Enzymol. 1978;52:302-10.
  • 15
    Misra HP, Fridovich I. The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J Biol Chem. 1972;247(10):3170-5.
  • 16
    Fujino Y, Goddon S, Chiche JD, Hromi J, Kacmarek RM. Partial liquid ventilation ventilates better than gas ventilation. Am J Respir Crit Care Med. 2000;162(2 Pt 1):650-7.
  • 17
    Pratschke J, Wilhelm MJ, Kusaka M, Cooper DK, Hancock WW, et al. Brain death and its influence on donor organ quality and outcome after transplantation. Transplantation. 1999;67(3):343-8, http://dx.doi.org/10.1097/00007890-199902150-00001.
    » http://dx.doi.org/10.1097/00007890-199902150-00001
  • 18
    Dhar R, Cotton C, Coleman J, Brockmeier D, Marklin G, et al. Comparison of high- and low-dose corticosteroid regimens for organ donor management. J Crit Care. 2012;28(1):111.e1-7.
  • 19
    Kutsogiannis DJ, Pagliarello G, Doig C, Ross H, Shemie SD. Medical management to optimize donor organ potential: review of the literature. Can J Anaesth. 2006;53(8):820-30.
  • 20
    Venkateswaran RV, Patchell VB, Wilson IC, Mascaro JG, Quinn DW, et al. Early donor management increases the retrieval rate of lungs for transplantation. Ann Thorac Surg. 2008;85(1):278-86, http://dx.doi.org/10.1016/j.athoracsur.2007.07.092.
    » http://dx.doi.org/10.1016/j.athoracsur.2007.07.092
  • 21
    Faropoulos K, Apostolakis E. Brain death and its influence on the lungs of the donor: how is it prevented? Transplant Proc. 2009;41(10):4114-9, http://dx.doi.org/10.1016/j.transproceed.2009.09.087.
    » http://dx.doi.org/10.1016/j.transproceed.2009.09.087
  • 22
    Avlonitis VS, Kirby JA, Dark JH. The effect of time from donor brain death to retrieval on reperfusion injury after lung transplantation. The Journal of heart and lung transplantation: the official publication of the International Society for Heart Transplantation. 2005;24(2):S121, http://dx.doi.org/10.1016/j.healun.2004.11.264.
    » http://dx.doi.org/10.1016/j.healun.2004.11.264
  • 23
    Zweers N, Petersen AH, van der Hoeven JA, de Haan A, de Leij LF, et al. Donor brain death aggravates chronic rejection after lung transplantation in rats. Transplantation. 2004;78(9):1251-8, http://dx.doi.org/10.1097/01.TP.0000142679.45418.96.
    » http://dx.doi.org/10.1097/01.TP.0000142679.45418.96
  • 24
    Van Der Hoeven JA, Moshage H, Schuurs T, Nijboer M, Ploeg RJ, et al. Brain death induces apoptosis in donor liver of the rat. Transplantation. 2003;76(8):1150-4, http://dx.doi.org/10.1097/01.TP.0000080983.14161.95.
    » http://dx.doi.org/10.1097/01.TP.0000080983.14161.95
  • 25
    Zhou HC, Ding WG, Cui XG, Zhang B, Li WZ, et al. Carbon monoxide inhalation ameliorates conditions of lung grafts from rat brain death donors. Chin Med J (Engl). 2008;121(15):1411-9.
  • 26
    Strieter RM, Kunkel SL. Acute lung injury: the role of cytokines in the elicitation of neutrophils. J Investig Med. 1994;42(4):640-51.
  • 27
    Skrabal CA, Thompson LO, Potapov EV, Southard RE, Youker KA, et al. Organ-specific regulation of pro-inflammatory molecules in heart, lung, and kidney following brain death. J Surg Res. 2005;123(1):118-25, http://dx.doi.org/10.1016/j.jss.2004.07.245.
    » http://dx.doi.org/10.1016/j.jss.2004.07.245
  • 28
    Blocher S, Wilker S, Sucke J, Pfeil U, Weimer R, et al. Acute rejection of experimental lung allografts: characterization of intravascular mononuclear leukocytes. Clin Immunol. 2007;124(1):98-108, http://dx.doi.org/10.1016/j.clim.2007.04.005.
    » http://dx.doi.org/10.1016/j.clim.2007.04.005
  • 29
    Woiciechowsky C, Schoning B, Daberkow N, Asche K, Docke WD, et al. Brain IL-1beta increases neutrophil and decreases lymphocyte counts through stimulation of neuroimmune pathways. Neurobiol Dis. 1999 Jun;6(3):200-8.
  • 30
    Ott L, McClain CJ, Gillespie M, Young B. Cytokines and metabolic dysfunction after severe head injury. J Neurotrauma. 1994;11(5):447-72, http://dx.doi.org/10.1089/neu.1994.11.447.
    » http://dx.doi.org/10.1089/neu.1994.11.447
  • 31
    Takada M, Nadeau KC, Hancock WW, Mackenzie HS, Waaga AM, et al. Effects of explosive brain death on cytokine activation of peripheral organs in the rat. Transplantation. 1998;65(12):1533-42, http://dx.doi.org/10.1097/00007890-199806270-00001.
    » http://dx.doi.org/10.1097/00007890-199806270-00001
  • 32
    McKeating EG, Andrews PJ, Signorini DF, Mascia L. Transcranial cytokine gradients in patients requiring intensive care after acute brain injury. Br J Anaesth. 1997;78(5):520-3.
  • 33
    Martin TR. Lung cytokines and ARDS: Roger S. Mitchell Lecture. Chest. 1999;116(1 Suppl):2S-8S, http://dx.doi.org/10.1378/chest.116.suppl_1.2S.
    » http://dx.doi.org/10.1378/chest.116.suppl_1.2S
  • 34
    Kostulas N, Pelidou SH, Kivisakk P, Kostulas V, Link H. Increased IL-1beta, IL-8, and IL-17 mRNA expression in blood mononuclear cells observed in a prospective ischemic stroke study. Stroke. 1999;30(10):2174-9, http://dx.doi.org/10.1161/01.STR.30.10.2174.
    » http://dx.doi.org/10.1161/01.STR.30.10.2174
  • 35
    Reynolds RW. Pulmonary edema as a consequence of hypothalamic lesions in rats. Science. 1963;141(3584):930-2, http://dx.doi.org/10.1126/science.141.3584.930.
    » http://dx.doi.org/10.1126/science.141.3584.930
  • No potential conflict of interest was reported.

Publication Dates

  • Publication in this collection
    Feb 2014

History

  • Received
    21 June 2013
  • Reviewed
    23 July 2013
  • Accepted
    14 Aug 2013
Faculdade de Medicina / USP Rua Dr Ovídio Pires de Campos, 225 - 6 and., 05403-010 São Paulo SP - Brazil, Tel.: (55 11) 2661-6235 - São Paulo - SP - Brazil
E-mail: clinics@hc.fm.usp.br