• No results found

University of Groningen Targeting brain death-induced injury van Erp, Anne Cornelie

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Targeting brain death-induced injury van Erp, Anne Cornelie"

Copied!
15
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Targeting brain death-induced injury

van Erp, Anne Cornelie

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

van Erp, A. C. (2018). Targeting brain death-induced injury. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Introduction

1

(3)

1

Organ transplantation remains the preferred life-saving treatment for most patients with

ORGAN DONATION, PRESERVATION AND TRANSPLANTATION

end-stage liver and kidney disease, as it can improve patients’ quality of life as well as their survival rates1-3. Unfortunately, not every patient on the waiting list can receive an

organ because of a global shortage of suitable organ grafts. As a result, 12 people die each day while waiting for an organ transplant in Europe alone4,5. This problem underlines the

need for a concerted action within the transplant community to increase the number of available organs. This pertains mostly to organs obtained from deceased donors to avoid ethical problems related to living organ donation as well as illegal organ trafficking. On an international level, this effort has resulted in the establishment of organizations such as the Eurotransplant International Foundation, a non-profit organization established to optimize the allocation and distribution of organs amongst a number of European countries5. On

a national level, several countries including The Netherlands have adopted legislation that automatically enters their civilians as potential organ donors unless they personally object. Another approach to increase the donor pool is to increase the use of organs of suboptimal quality. However, this approach mandates an improvement of organ quality prior to transplantation in order not to risk poor transplantation outcomes. As the quality of the organ graft may be challenged during each step of the transplantation process, this approach should encompass strategies and treatments in the donor, but also afterwards during organ preservation and in the recipient. To facilitate organ-improving strategies, the first step is to understand which injuries the graft endures during the transplantation process.

ORGAN DONATION

Both chronic (aging, pre-existing medical conditions) and acute injuries (donor death, organ retrieval strategies) in the donor are risk factors for impaired graft survival after transplantation6. Of these injuries, donor age is considered a classical risk factor that is

associated with both short-term and long-term graft survival7,8. However, recent studies

suggest that biological organ age and not donor age is a better predictor of transplantation outcomes9-11. Halloran et al. postulated that the accumulation of aging combined with

exposure to injury and stressors impairs the ability of an organ to repair and remodel, thereby hindering graft survival6. This might explain why we observe a clear difference

in survival dependent on the type of organ donor used. Of all donor types, organ grafts obtained from living donors are superior when compared to those obtained from deceased donors. However, the number of living organ grafts is limited, which means that the majority of organs used are obtained from decreased donors. Deceased donors can be classified into deceased brain-dead (DBD) donors or deceased cardiac death (DCD) donors. Of the two, most organs worldwide are obtained from DBD donors. Nevertheless, the number of DCD

(4)

1

organs continues to increase, particularly since more countries are accepting organs from these donors12. In addition, many countries have started using suboptimal organs obtained

from so-called extended criteria donors (ECD)13. ECD are over 60 years of age, or between

50-60 years with comorbidities including hypertension, impaired estimated GFR based on plasma creatinine (> 1.5 mg/dL), age, and gender, or death by a cerebrovascular incident. When compared to living donation, deceased donation is associated with allograft dysfunction or delayed graft function, impaired survival or even higher mortality rates following transplantation14-16. This inferiority of deceased donors is caused by pathophysiological

changes of death. In DBD donors, most commonly cerebrovascular incidents or traumatic brain injuries cause increased intracranial pressure. When the body fails to maintain cerebral perfusion, this results in ischemia of the brain, and brain stem. As a result, large amounts of catecholamines are released and a cascade of derailments commences that includes inflammation, hemodynamic instability, and hormonal and metabolic changes17-19. In DCD

donors, not a cerebral event but cardiopulmonary arrest causes hypoxia, which poses a great ischemic insult to the organs. The subsequent prolonged period of warm ischemia adds additional ischemic damage to the organs and is considered another risk factor associated with impaired graft function and higher mortality rates following transplantation20,21.

Altogether, this suggests that minimizing and targeting the accumulation of injuries in the donor could reduce cellular injury. This strategy could improve graft function and survival after transplantation and even allow more organs to be suitable for transplantation.

Organ preservation

Following this period of donor-related injuries, organ grafts suffer additional injury during the preservation period. For decades, organs have been statically preserved on ice. This strategy was based on the rate-of-life theory, which states that aging and longevity are regulated by the rate of cellular metabolism22. According to this principle, hypothermic preservation

intends to lower the metabolic rate, thereby reducing hypoxic injury and preserving cellular function23. To ensure tolerance of organs to hypothermia, preservation solutions have been

developed to counter negative side effects of hypothermic preservation including edema, accumulation of reactive oxygen species (ROS) production, ATP depletion, mitochondrial damage, a switch to anaerobic glycolytic metabolism, and microvascular changes23-25. With

this in mind, prolonged duration of the cold ischemia is considered an independent risk factor for a nonfunctioning or dysfunctioning transplant, particularly in marginal or high-risk donors26,27.

These negative side effects of cold storage have recently led to the first clinical implementation of hypothermic machine perfusion (HMP). This technique allows continuous, pulsatile perfusion of the organ at 4 to 10°C, while blood and other components are flushed out

(5)

1

and the organ equilibrates with the perfusion solution23. HMP has clear benefits over static

cold storage in kidney transplantation, evidenced by improved graft function and survival rates23,28. Despite these benefits, HMP has only recently begun to be clinically implemented

in the field of kidney, liver, heart, and lung transplantation. Static cold storage currently remains the only clinical preservation method of the pancreas26. Adaptations of HMP that are

currently being investigated pre-clinically are the addition of oxygen and other additives to the perfusion fluid, as well as perfusion at subnormothermic (25°C) or normothermic (37°C) temperatures. Especially normothermic temperatures would add the additional benefit of allowing viability testing of organs prior to transplantation. The key to all these preservation methods is the improvement of graft quality by limiting (the duration of) ischemic injury, preventing abrupt reperfusion and preserving or restoring cellular energy supplies.

Organ transplantation

Optimizing graft quality prior to transplantation is especially important given that the reintroduction of warm oxygenated blood in the recipient will only amplify injury to the organ graft. After a period of cold ischemia, graft reperfusion causes immediate ROS production by the vascular cells of the donor allograft, followed by a subsequent hit of ROS production originating from the recipient’s phagocytes29. Interestingly, the initial ROS produced by the

donor’s vascular cells trigger the subsequent second burst of ROS, together reducing the capacity of the anti-oxidant machinery. Hence, this phenomenon has been described as a vicious cycle with excess ROS production as its end result29. One of the main contributors

to this cycle are the mitochondria, which are known to increase their ROS production in response to hypoxia30. This ROS production in turn facilitates the initiation of various cellular

death pathways, including necrosis, apoptosis, and autophagy24,31,32. It is suggested that the

amount and duration of the oxidative injury related to first ischemia and then reperfusion, so-called ischemia-reperfusion injury (IRI), is responsible for the ischemic damage and immune activation immediately after transplantation. This makes oxidative stress an important factor which can ultimately result in rejection of the organ graft24. Furthermore,

the level of IRI subsequently determines the extent to which cellular death pathways such as autophagy and apoptosis become activated. Their level of activation and interplay seems to determine whether the end result is pro-survival or pro-death33,34.

(6)

1

TARGETING BRAIN DEATH-INDUCED INJURY

The potential benefit for patients with organ failure together with the global organ shortage is the driving force behind the body of research that has tried to elucidate what damages the graft during each step of the transplantation process. Recently, this has led to the clinical implementation of ex vivo hypothermic kidney perfusion as the preferred preservation method. However, the great potential of targeted treatments during ex vivo organ perfusion and the sometimes ethical difficulty of treating deceased donors, should not be a reason to refrain from investigating or targeting organ grafts in deceased donors. Even though each step of the transplantation process may harm the organ graft, a very substantial amount of damage already occurs within the deceased organ donor. Given that many intracellular pathways such as oxidative stress and autophagy (introduced in more detail later) can be either detrimental or beneficial depending on the level of injury or stress, particularly early treatment of donor-related injuries could be of great benefit. Besides expanding on the current knowledge on donor pathophysiology, research should investigate novel injury mechanisms in the brain-dead donor, such as metabolic changes and autophagy dysregulation. Understanding what damages the organ prior to transplantation is essential to optimize organ-specific treatments in the donor but also during later stages of the transplantation process. Finally, if the outcome of a transplantation can already be predicted in the donor or during organ preservation, this would greatly benefit the success of transplantations while minimizing the impact on recipients.

Brain death physiology

Most organs world-wide are obtained from DBD donors12. Brain death is defined as a state with irreversible absence of brain and brainstem function, in which mechanical ventilation is required to prevent apneas while the systemic circulation remains intact35. Several common etiologies of brain death include cerebrovascular accidents, traumatic brain injury, and diffuse hypoxia. The common denominator in each of these injuries is a rise in intracranial pressure. When the risen cerebral pressure cannot be overcome by a rise in blood pressure, this results in hypoperfusion of the brain and subsequent progressive ischemia of the brain, brainstem, and ultimately spinal cord. Herniation of the ischemic brain stem results in sympathetic hyperactivity that is characterized by an immediate increase in systemic blood pressure and peripheral vasoconstriction due to the release of endogenous catecholamines, also called the catecholamine storm17,36. This catecholamine storm has been described as the body’s final attempt to overcome the rise in intracranial pressure. This vasoconstrictive response results in decreased blood flow through peripheral organs such as the liver and kidneys37. The systemic hypertension also stimulates (to a lesser extent) parasympathetic activity via the baroreceptors, resulting in subsequent bradycardia. These changes, together called the Cushing response, are characteristic for the brain death condition.

(7)

1

This hypertensive period is followed by a decline in sympathetic tone, which marks the beginning of the hemodynamic instability in the DBD donor35. In addition to these changes in hemodynamics, ischemia of the brain results in failure of the hypothalamus and pituitary axis. As a consequence, a depletion of antidiuretic hormone is evident in the majority of the brain-dead donors38. The resulting diabetes insipidus causes increased diuresis and risk of hypovolemia and further contributes to the hemodynamically unstable condition. In addition, plasma levels of free thyroid hormone, thyroid stimulating hormone, and cortisol generally decline39,40. Besides hemodynamic and hormonal changes, BD is characterized as a systemic inflammatory state, seen by a rise in circulating cytokines, including interleukin (IL)-6, IL-10, MCP-1, and TNF-α41-44.This systemic inflammatory environment triggers a local response in the kidneys, liver, and lungs. The inflammatory and apoptotic response in these organs is the results of activation of the vascular endothelium, the complement and coagulation system, as well as the innate and adaptive immune response45. What triggers this pro-inflammatory environment during brain death is not well understood. Nevertheless, cerebral cytokines from the dying cerebrum46, complement activation47, translocation of bacteria from the intestines48, and organ-specific inflammation49 have all been implicated. Altogether, brain death-induced pathophysiology negatively affects organ quality prior to transplantation and predisposes the recipients of these grafts to a higher risk of acute rejection, delayed graft function and lower survival rates after transplantation16,42,50. Targeting brain death-induced injury is, therefore, essential.

AIM OF THIS THESIS

The aim of this thesis is to review and expand on the current knowledge required to target brain death-induced injury (see Fig 1). The first part of the thesis (Chapters 1 – 4) focuses on interventions in the donor that target brain death-induced pathophysiological changes. The second part of this thesis (Chapters 5 – 8) expands on this knowledge by investigating novel injury mechanisms pertaining metabolic changes and autophagy (dys)regulation during brain death.

Interventions in the brain-dead donor

Currently, brain-dead donor management protocols are aimed at targeting brain death-related pathophysiological changes. Despite considerable differences per individual center, protocols have focused mainly on providing hemodynamic support, suppressing the immune system, optimizing donor ventilation management, controlling donor body temperature, and administrating hormone replacement therapy51. Providing hemodynamic

support and optimizing organ oxygenation is important, given the hemodynamic instability that is evident within the brain-dead donor. Immunosuppressive therapy to counteract brain death-induced inflammation is also clinically relevant, as donor plasma levels of pro-inflammatory cytokine IL-6 were inversely correlated to recipient six-month

(8)

hospital-1

free survival54 and an increased risk of recipient death following lung transplantation52-54.

However, despite promising animal studies56,57, immunosuppressive therapy in brain-dead

donors has not been able to improve the outcomes of liver and kidney transplantations58-61

(see Fig 2). Finally, despite mixed reports about the effectiveness of thyroid hormone replacement therapies, its use has increased over the years40,62-64. Altogether, this highlights

that despite the extensive body of preclinical and clinical research on interventions in the brain-dead donor, we are still lacking a universally accepted, optimized brain-dead donor management protocol.

Figure 1. Targeting brain death-induced injury. Brain death results in hormonal changes, hemodynamic

instability, inflammation, and oxidative stress. It is unclear how these changes affect organ-specific metabolism, perfusion, mitochondrial function, and autophagy. Together these changes negatively affect organ quality, which impacts transplantation outcomes and patient and graft survival. Targeting brain death-induced injury is essential to optimize organ quality prior to transplantation, particularly given the subsequent injuries the graft endures during organ preservation and transplantation, as well as the global organ shortage.

As a starting point for this thesis in Chapter 2, we provided an update on all systematically tested, clinical interventions that have been tested in brain-dead organ donors thus far (see Fig 2). Only those studies were included that focused on the effects on organ quality and graft and/or patient survival after transplantation. In Chapter 3, we tested whether EA230, an oligopeptide of human chorionic gonadotropin with promising anti-inflammatory properties in models of septic and haemorrhage shock, would attenuate brain death-induced inflammation. Finally, in Chapter 4, we studied whether pre-conditioning of brain-dead animals with free thyroid hormone T3 could improve liver function and reduce apoptosic cell death.

(9)

1

Figure 2. Overview of all systematically tested, clinical treatments or interventions in the brain-dead donor

with outcome parameters pertaining the lungs, liver, heart, and kidneys. Treatments or interventions denoted in black did not affect graft function or graft and patient survival. Treatments or interventions denoted in green were beneficial, those in red were detrimental, and those in orange showed mixed results.

NOVEL INJURY MECHANISMS

Previous studies have indicated that brain death alters the plasma metabolite profile and shifts the balance from aerobic to anaerobic metabolism65. To explain these metabolic

changes, several theories have been provided. Firstly, these changes have been attributed to mitochondrial impairment, as this was previously observed in the muscles of brain-dead patients66 and the hearts of brain-dead pigs67. Alternatively, the hemodynamic instability

and initial organ hypoperfusion immediately following the catecholamine storm have been implicated as the underlying cause. Regardless, the metabolic status of organs prior to

(10)

1

transplantation is of clinical importance, as these changes have been linked to transplantation outcomes following both kidney68,69 and liver transplantation70,71. However, effects of donor

brain death on metabolism in the individual organs has not previously been investigated. Therefore, in Chapter 5, we investigated how brain death affects metabolism, both systemically as well as in the liver and kidney. Besides studying major metabolic pathways, we investigated whether mitochondrial function and organ perfusion were altered during brain death. In Chapter 6, we used a non-invasive imaging tool to study in vivo, metabolic pathways in the liver and kidney during and following brain death. Using hyperpolarized magnetic resonance imaging with MRI-active pyruvate molecules, we were able to visualize metabolic pathways in real time during brain death. Afterwards, we visualized glucose metabolism during ex vivo organ reperfusion using radioactively labelled glucose.

Oxidative stress, hemodynamic instability, inflammation, and hormonal perturbances induced during brain death are each influencers of autophagy72-74. Autophagy is an intracellular

degradation pathway that removes, degrades, and recycles cellular constituents75. Autophagy

normally occurs at a basal level within the cells and serves as a cellular housekeeper that removes damaged or unwanted organelles or cellular constituents73,75. Several types

of autophagy exist, but all types of autophagy involve the transportation of intracellular compounds to the lysosomes for degradation. In the presence of cellular stressors such as hypoxia, inflammation, or energy depletion, autophagy becomes stimulated and protects the cell by removing damaged or toxic cellular products76,77. In this way, autophagy is

generally considered to be a protective, stress-adaptation pathway that can counter cellular death pathways such as apoptosis77,78. Conversely, excessive autophagy stimulation can be

detrimental and can push the balance away from a protective towards a detrimental role for autophagy, as is seen in disease states such as cancer, neurodegeneration, aging, and IRI79,80.

This is of particular importance, given the current increased use of older and extended criteria donors. Even though autophagy has been implicated to play a role during oxidative stress and IRI, the exact role that autophagy plays during the transplantation process is only beginning to be understood81. In Chapter 7, we reviewed all the available knowledge on

the role that autophagy and oxidative stress play during each step of the transplantation process. Furthermore, we covered the complex interdependency of these two pathways and discussed several compounds that target each of these pathways. Finally, in Chapter

8 we investigated how brain death affects autophagy in the liver and kidney. Furthermore,

we studied whether stimulation of autophagy with mTOR-inhibitor rapamycin affected autophagy, organ quality, and apoptosis.

(11)

1

1. Wolfe, R. A. et al. Comparison of mortality

REFERENCES

in all patients on dialysis, patients on dialy-sis awaiting transplantation, and recipients of a first cadaveric transplant. N. Engl. J. Med. 341, 1725–1730 (1999).

2. Singer, L. G. Cost-effectiveness and qual-ity of life: benefits of lung transplanta-tion. Respir Care Clin N Am 10, 449–57– v (2004).

3. Yang, L. S., Shan, L. L., Saxena, A. & Morris, D. L. Liver transplantation: a systematic re-view of long-term quality of life. Liver Int. 34, 1298–1313 (2014).

4. European Directorate for the Quality of Medicines and Healthcare. (2018). Avail-able at: https://www.edqm.eu/en/events/ european-day-organ-donation-and-trans-plantation. (Accessed: 20 February 2018)

5. Eurotransplant. Available at: http://statis-tics.eurotransplant.org/index.php?search_ type=transplants&search_period=by+-year+chart. (Accessed: 6 July 2017) 6. Halloran, P. F., Melk, A. & Barth, C.

Rethink-ing chronic allograft nephropathy: the con-cept of accelerated senescence. J. Am. Soc. Nephrol. 10, 167–181 (1998).

7. Slegtenhorst, B. R. et al. Mechanisms and Consequences of Injury and Repair in Old-er Organ Transplants. Transplantation 97, 1091–1099 (2014).

8. Moers, C., Kornmann, N. S. S., Leuvenink, H. G. D. & Ploeg, R. J. The influence of de-ceased donor age and old-for-old allocation on kidney transplant outcome. Transplanta-tion 88, 542–552 (2009).

9. Koppelstaetter, C. et al. Markers of cellular senescence in zero hour biopsies predict outcome in renal transplantation. Aging Cell 7, 491–497 (2008).

10. Aini, W. et al. Telomere shortening and karyotypic alterations in hepatocytes in long-term transplanted human liv-er allografts. Transpl. Int. 25, 956–966 (2012).

11. McGlynn, L. M. et al. Cellular senescence in pretransplant renal biopsies predicts postoperative organ function. Aging Cell 8, 45–51 (2009).

12. Bendorf, A. et al. An international compar-ison of the effect of policy shifts to organ donation following cardiocirculatory death (DCD) on donation rates after brain death (DBD) and transplantation rates. PLoS ONE 8, e62010 (2013).

13. MD, D. S. C. et al. Trends in Organ Donor Management: 2002 to 2012. J. Am. Coll. Surg. 219, 752–756 (2014).

14. Aubert, O. et al. Long term outcomes of transplantation using kidneys from expand-ed criteria donors: prospective, popula-tion based cohort study. BMJ 351, h3557 (2015).

15. Lué, A. et al. How important is donor age in liver transplantation? World J. Gastroenter-ol. 22, 4966–4976 (2016).

16. Terasaki, P. I., Cecka, J. M., Gjertson, D. W. & Takemoto, S. High survival rates of kidney transplants from spousal and living unrelat-ed donors. N. Engl. J. Munrelat-ed. 333, 333–336 (1995).

17. Floerchinger, B., Oberhuber, R. & Tullius, S. G. Effects of brain death on organ quality and transplant outcome. Transplant Rev (Orlando) 26, 54–59 (2012).

18. Weiss, S. et al. Brain death activates do-nor organs and is associated with a worse I/R injury after liver transplantation. Am J Transplant 7, 1584–1593 (2007).

(12)

1

19. Olinga, P. et al. The influence of brain death on liver function. Liver Int. 25, 109–116 (2005).

20. Tennankore, K. K., Kim, S. J., Alwayn, I. P. J. & Kiberd, B. A. Prolonged warm ischemia time is associated with graft failure and mortality after kidney transplantation. Kid-ney International 89, 648–658 (2016). 21. Marasco, S. F., Kras, A., Schulberg, E., Vale,

M. & Lee, G. A. Impact of warm ischemia time on survival after heart transplan-tation. Transplantation Proceedings 44, 1385–1389 (2012).

22. Wang, Y. & Hekimi, S. Mitochondrial dys-function and longevity in animals: Untan-gling the knot. Science 350, 1204–1207 (2015).

23. Maathuis, M.-H. J., Leuvenink, H. G. D. & Ploeg, R. J. Perspectives in organ preser-vation. Transplantation 83, 1289–1298 (2007).

24. 2Salvadori, M., Rosso, G. & Bertoni, E. Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J Transplant 5, 52–67 (2015).

25. 2Southard, J. H. & Belzer, F. O. Organ pres-ervation. Annu. Rev. Med. 46, 235–247 (1995).

26. 2Guibert, E. E. et al. Organ Preservation: Current Concepts and New Strategies for the Next Decade. Transfus Med Hemother 38, 125–142 (2011).

27. Ploeg, R. J. et al. Risk factors for primary dysfunction after liver transplantation--a multivariate analysis. Transplantation 55, 807–813 (1993).

28. Moers, C., Pirenne, J., Paul, A., Ploeg, R. J.Machine Preservation Trial Study Group. Machine perfusion or cold storage in de-ceased-donor kidney transplantation. N. Engl. J. Med. 366, 770–771 (2012).

29. Land, W. G. Emerging role of innate immu-nity in organ transplantation: part I: evolu-tion of innate immunity and oxidative al-lograft injury. Transplant Rev (Orlando) 26, 60–72 (2012).

30. Granger, D. N. & Kvietys, P. R. Redox Biology. Redox Biology 6, 524–551 (2015).

31. Sureshbabu, A., Ryter, S. W. & Choi, M. E. Oxidative stress and autophagy: crucial modulators of kidney injury. Redox Biology 4, 208–214 (2015).

32. Li, C. & Jackson, R. M. Reactive species mechanisms of cellular hypoxia-reoxygen-ation injury. Am. J. Physiol., Cell Physiol. 282, C227–41 (2002).

33. Nikoletopoulou, V., Markaki, M., Palikaras, K. & Tavernarakis, N. Biochimica et Bio-physica Acta. BBA - Molecular Cell Research 1833, 3448–3459 (2013).

34. Mizushima, N. & Komatsu, M. Autophagy: renovation of cells and tissues. Cell 147, 728–741 (2011).

35. Novitzky, D. & Cooper, D. K. C. The Brain-Dead Organ Donor. (Springer Science & Business Media, 2012).

36. Novitzky, D., Cooper, D. K. & Reichart, B. Hemodynamic and metabolic responses to hormonal therapy in brain-dead potential organ donors. Transplantation 43, 852–854 (1987).

37. Herijgers, P., Leunens, V., Tjandra-Maga, T. B., Mubagwa, K. & Flameng, W. Chang-es in organ perfusion after brain death in the rat and its relation to circulating cate-cholamines. Transplantation 62, 330–335 (1996).

38. Smith, M. Physiologic changes during brain stem death--lessons for management of the organ donor. J. Heart Lung Transplant. 23, S217–22 (2004).

(13)

1

Maurette, P. Thyroid function in brain-dead donors. Transpl. Int. 3, 226–233 (1990). 40. Novitzky, D., Cooper, D. K. C., Rosendale, J.

D. & Kauffman, H. M. Hormonal therapy of the brain-dead organ donor: experimental and clinical studies. Transplantation 82, 1396–1401 (2006).

41. Barklin, A. Systemic inflammation in the brain-dead organ donor. Acta Anaesthesiol Scand 53, 425–435 (2009).

42. Bos, E. M., Leuvenink, H. G. D., van Goor, H. & Ploeg, R. J. Kidney grafts from brain dead donors: Inferior quality or opportunity for improvement? Kidney International 72, 797–805 (2007).

43. Nijboer, W. N. et al. Effects of brain death on stress and inflammatory response in the human donor kidney. Transplantation Pro-ceedings 37, 367–369 (2005).

44. Saat, T. C. et al. A Comparison of Inflam-matory, Cytoprotective and Injury Gene Expression Profiles in Kidneys From Brain Death and Cardiac Death Donors. Trans-plantation 98, 15–21 (2014).

45. Floerchinger, B. et al. Graft-specific immune cells communicate inflammatory immune responses after brain death. J. Heart Lung Transplant. 31, 1293–1300 (2012).

46. Higashida, T. et al. The role of hypoxia-in-ducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumat-ic brain injury. J. Neurosurg. 114, 92–101 (2011).

47. Damman, J., Schuurs, T. A., Ploeg, R. J. & Seelen, M. A. Complement and renal trans-plantation: from donor to recipient. Trans-plantation 85, 923–927 (2008).

48. Koudstaal, L. G. et al. Brain death induces inflammation in the donor intestine. Trans-plantation 86, 148–154 (2008).

49. Skrabal, C. A. et al. Organ-specific regulation of pro-inflammatory molecules in heart, lung, and kidney following brain death. J. Surg. Res. 123, 118–125 (2005).

50. Westendorp, W. H., Leuvenink, H. G. & Ploeg, R. J. Brain death induced renal inju-ry. Curr Opin Organ Transplant 16, 151–156 (2011).

51. Kumar, L. Brain death and care of the organ donor. J Anaesthesiol Clin Pharmacol 32, 146–152 (2016).

52. Molnar, M. Z. et al. Inflammatory Markers and Outcomes in Kidney Transplant Re-cipients. Transplantation 101, 2152–2164 (2017).

53. Dahle, D. O. et al. Inflammation-associat-ed graft loss in renal transplant recipients. Nephrol. Dial. Transplant. 26, 3756–3761 (2011).

54. Murugan, R. et al. Increased plasma inter-leukin-6 in donors is associated with lower recipient hospital-free survival after cadav-eric organ transplantation. Critical Care Medicine 36, 1810–1816 (2008).

55. Kaneda, H. et al. Pre-implantation multiple cytokine mRNA expression analysis of do-nor lung grafts predicts survival after lung transplantation in humans. Am J Transplant 6, 544–551 (2006).

56. Rebolledo, R. et al. Prednisolone has a pos-itive effect on the kidney but not on the liver of brain dead rats: a potencial role in complement activation. J Transl Med 12, 111 (2014).

57. Pratschke, J. et al. Improvements in early behavior of rat kidney allografts after treat-ment of the brain-dead donor. Annals of Surgery 234, 732–740 (2001).

58. Kainz, A. et al. Steroid pretreatment of or-gan donors to prevent postischemic renal allograft failure: a randomized, controlled trial. Ann. Intern. Med. 153, 222–230

(14)
(15)

Referenties

GERELATEERDE DOCUMENTEN

Altogether, future donor selection and management strategies might shift from a general to a more customized approach, in which baseline factors of the lung donor such as cause

Bovendien lijkt de lactaat productie tijdens EVLP door vrouwelijke donor longen hoger dan door mannelijke donor longen, wat zou kunnen duiden op een meer uitgesproken

Department of Internal Medicine University Medical Center Groningen Groningen, The Netherlands..

Brain death-induced lung injury is a consequence of the brain accepting the approach of death, while the heart insists upon immortality. Fast induction of brain death is

Junior Scientific Masterclass, Faculty of Medicine University of Groningen Research Institute GUIDE The printing of this thesis was kindly supported by: Chipsoft..

Plasma creatinine, AST, total bilirubin, and IL-6; renal and hepatic gene expression levels of IL-6 and MCP-1; and polymorphonuclear influx in the liver and kidney were

gene Bax and anti-apoptotic gene Bcl-2, and the Bax/Bcl2 mRNA ratio, following 4 h of BD in the BD groups and 0.5 h of ventilation in the sham groups. T 3 pre-treatment

In contrast, the reduction in apoptosis and injury markers in the liver does appear to be associated with increased hepatic autophagy, suggesting an important role for