• No results found

Explorations of the therapeutic potential of influencing metabolism during critical illness - Thesis

N/A
N/A
Protected

Academic year: 2021

Share "Explorations of the therapeutic potential of influencing metabolism during critical illness - Thesis"

Copied!
222
0
0

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

Hele tekst

(1)UvA-DARE (Digital Academic Repository). Explorations of the therapeutic potential of influencing metabolism during critical illness Aslami, H. Publication date 2013 Document Version Final published version. Link to publication Citation for published version (APA): Aslami, H. (2013). Explorations of the therapeutic potential of influencing metabolism during critical illness.. General rights 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), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.. UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl) Download date:21 Jun 2021.

(2) Explorations of the therapeutic potential of influencing metabolism during critical illness. UITNODIGING. Explorations of the therapeutic potential of influencing metabolism during critical illness Hamid Aslami. Voor het bijwonen van de openbare verdediging van het proefschrift. Explorations of the therapeutic potential of influencing metabolism during critical illness van. Hamid Aslami Op vrijdag 1 maart 2013 om 13:00 uur Locatie Aula van de Universiteit van Amsterdam, Oude Lutherse kerk, Singel 411 Amsterdam Receptie ter plaatse na afloop van de promotie Hamid Aslami h.aslami@amc.uva.nl 06-24703463 Paranimfen Azar Aslami azari.aslami@gmail.com 06- 39 11 63 61. Hamid Aslami. Friso de Beer f.m.debeer@amc.uva.nl 06- 51 71 34 74.

(3) Explorations of the therapeutic potential of influencing metabolism during critical illness. Hamid Aslami.

(4) Explorations of the therapeutic potential of influencing metabolism during critical illness Thesis, University of Amsterdam, The Netherlands Copyright © 2013 by Hamid Aslami. Lay-out: Gildeprint Drukkerijen - Enschede, The Netherlands Printed by: Gildeprint Drukkerijen - Enschede, The Netherlands ISBN: 978-94-6108-395-1. Financial support for printing this thesis was provided by: Chipsoft, Academisch Medisch Centrum, Longfonds, Maquet Nederland b.v., MI consultantie en Stichting Gilles Hondius Foundation..

(5) Explorations of the therapeutic potential of influencing metabolism during critical illness. ACADEMISCH PROEFSCHRIFT. ter verkrijging van de graad van doctor aan de Universiteit van Amsterdam op gezag van de Rector Magnificus prof. dr. D.C. van den Boom ten overstaan van een door het college voor promoties ingestelde commissie, in het openbaar te verdedigen in de Aula der Universiteit op vrijdag 1 maart 2013, te 13:00 uur. door Hamid Aslami geboren te Kabul, Afghanistan.

(6) Promotor: Copromotor:. Prof. Dr. M.J. Schultz Dr. N.P. Juffermans. Overige leden:. Prof. Dr. R.J.A. Wanders Prof. Dr. A.R.J. Girbes Prof. Dr. T.M. van Gulik Prof. Dr. M. Hollmann Prof. Dr. J.G. van der Hoeven Prof. Dr. P. Pelosi Prof. Dr. L. Aarts. Faculteit der Geneeskunde.

(7) ‫تقدیم بو‪:‬‬ ‫پدرم بو استواری کوه‬ ‫مادرم بو زاللی چشمو‬ ‫برادرم بو صمیمیت باران‬ ‫خواىرم بو طراوت شبنم‬.

(8)

(9) Contents Chapter 1. General outline of the thesis . 9. Chapter 2. Targeting mitochondrial function in sepsis Netherlands Journal of Critical Care, August 2012. 17. Chapter 3. Induction of a hypometabolic state during critical illness – a new concept in the ICU? The Netherlands Journal of Medicine, May 2010. 35. Chapter 4. Potential applications of hydrogen sulfide–induced suspended animation Current Medicinal Chemistry, June 2009. 53. Chapter 5 . Mild hypothermia reduces Ventilator−Induced Lung Injury, irrespective of reducing respiratory rate Translational Research, February 2012. 75. Chapter 6. Induced hypothermia is protective in a rat model of pneumococcal pneumonia associated with increased adenosine triphosphate availability and turnover Critical Care Medicine, March 2012. 89. Chapter 7. The effect of induced hypothermia on respiratory parameters in mechanically ventilated patients Resuscitation, December 2010. 107. Chapter 8. Induced hypothermia reduces the level of circulating mitochondrial DNA in survivors of a cardiac arrest Submitted. 115. Chapter 9. Suspended animation inducer hydrogen sulfide is protective in an in vivo model of ventilator–induced lung injury Intensive Care Medicine, November 2010. 125.

(10) Chapter 10. Hydrogen sulfide donor NaHS reduces organ injury in pneumosepsis, 141 associated with improved bio–energetic status Submitted. Chapter 11. A short course of hydrogen sulfide reduces organ injury in endotoxemia, with no additional protection from prolonged infusion Cytokine, December 2012. 167. Chapter 12. Pharmacological induction of a hypo–metabolic state with 3–iodothyronamine is not protective in a mouse model of LPS induced lung injury Submitted. 189. Chapter 13. Summary and general discussion. 203. Appendix A Nederlandse samenvatting Appendix B Dankwoord. 209 213.

(11) I General outline of this thesis. Hamid Aslami and Nicole P. Juffermans.

(12) Chapter I. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39 10.

(13) General outline of this thesis. General outline of this thesis. I. Multiple organ failure is a common complication which can occur in the course of sepsis or systemic inflammatory response syndrome (SIRS) (1). The host response to invading micro–organisms or to a sterile inflammation can cause endothelial damage, microvascular dysfunction and vasodilatation, ultimately resulting in impaired tissue oxygenation and organ injury (2). Although an adequate host response may be needed to combat infection, it is thought that the hyper–inflammatory response seen during sepsis (and SIRS) may also contribute to ‘collateral damage’. The lungs and kidneys are the first failing organs, resulting in acute respiratory distress syndrome (ARDS) (3) and acute kidney injury respectively (4), significantly contributing to adverse outcome in these patients (5–7). Increased inflammation and coagulation play a role in the pathogenesis of multiple organ failure, obstructing microcirculation and hampering organ perfusion (8). However, the pathogenesis of organ failure is not entirely understood. Even during adequate organ perfusion, there is a failure of mitochondria to live up to increased ATP demand, thereby causing bio–energetic failure, eventually leading to organ failure (9). Mitochondrial dysfunction is thought to occur as a result of damage inflicted by proinflammatory mediators. Most studies in sepsis have shown a decrease in mitochondrial respiration and ATP generation. Alternatively, it has been hypothesized that reduced cellular metabolism may potentially be a functional, protective mechanism. A metabolic shut–down may thereby increase the chances of survival of cells, and thus organs, during overwhelming inflammation. Regardless the cause, mitochondrial dysfunction is increasingly recognized as an important key player in the occurrence of organ failure in the critically ill. Treatment of organ failure traditionally consists of goal–directed supportive care (10), ensuring adequate tissue perfusion and oxygenation to meet the high metabolic demands of severe inflammation. Although mechanical ventilation is a life–saving treatment, it can also cause additional lung injury, termed ventilator–induced lung injury (VILI) (11;12). Mechanisms of VILI include repetitive opening and closing as well as overstretching of alveoli, resulting in a pro–inflammatory response with enhanced pulmonary coagulation and complement activation (13–15). Mechanical ventilation not only contributes to lung injury, but can also affect distant organs (11). Thereby, the management of ARDS is a double edged sword. In the past decade, much knowledge has been gained about how we should ventilate our critically ill patients. Application of low tidal volumes has proven to be an effective therapy to reduce mortality in the critically ill (16). However, even in the era of protective ventilation, mortality in ARDS patients remains high (17). Taken together, physicians treat the underlying cause, they try to inflict ‘no further harm’ by their supportive care and wait for the disease to abate. Specific treatment options for these critically ill patients are not available. This has led us to hypothesize about a new perspective.. 11. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(14) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter I. Instead of enhancing oxygen delivery, an alternative approach may be to reduce energy consumption. The regulated induction of a hypo–metabolic state, analogous to hibernation, may be beneficial in the imbalance between oxygen delivery and demand, thereby protecting the cells from severe bio–energetic failure and a critical fall in ATP. In naturally hibernating animals, a stepwise decrease in metabolism is followed by a reduction in body temperature when temperatures reach far below zero (18). It might be argued that humans do not hibernate and that we have only limited tolerance to hypoxia. Nevertheless, the human myocardium for example is able to adapt to chronic hypoxia in patients with ischemic heart disease, termed myocardial hibernation. Regulated down–regulation of myocardial contractions may thereby be an adaptation to reduced energy supply in the myocardium (19). Also, survivors of prolonged deep accidental hypothermia suggest that humans have an ability to reduce metabolism when oxygen delivery is compromised (20). We hypothesize that regulated induction of a hypo–metabolic state may reduce organ injury in critically ill patients by restoring the imbalance between oxygen delivery and demand, thereby protecting the cells from severe bio–energetic failure and a critical fall in ATP supply.. Introduction In chapter 2 and 3, we discuss the mechanisms advancing organ failure. Special emphasis is put on the inability of mitochondria to use oxygen due to dysfunction, leading to a bio– energetic failure. Interventions aimed at decreasing metabolic rate are discussed, including hypothermia and hydrogen sulfide (H2S) induced suspended animation, as novel strategies to reduce organ failure in the critically ill. The effects of H2S on various regulatory systems including metabolism, circulation and inflammation, are described in detail in chapter 4. We further discuss potential therapeutic possibilities of a H2S–induced suspended animation– like state. Induced hypothermia Induced hypothermia is applied in patients admitted to the intensive care unit following cardiac arrest because it improves neurological outcome (21). The beneficial effect of hypothermia is thought to occur from reducing excessive inflammation and by reducing cerebral energy demands (22). In this thesis, we investigated whether induced hypothermia was also beneficial in reducing lung injury in animal models of ARDS. In chapter 5, we studied the effect of hypothermia on lung injury inflicted by mechanical ventilation. We hypothesized that hypothermia would allow for lower respiratory rates required for adequate gas exchange, thereby reducing the ‘repetitive strain injury’ from repeated opening and closing of alveoli. We found that in a physiological rat VILI model, mild hypothermia (32°C) not only. 12.

(15) General outline of this thesis. reduced lung injury markers, but also allowed for lower respiratory rates while maintaining normal acid–base balance compared to animals kept at 37°C body temperature. In chapter 6, we induced hypothermia in mechanically ventilated rats with established S. pneumoniae pneumosepsis. Hypothermia was associated with diminished bacterial dissemination to the spleen and reduced levels of lung injury markers, as well as with improved mitochondrial function, as reflected by a high ATP availability and mitochondrial respiration in the liver and muscle. Next we studied the effect of hypothermia on respiratory parameters in patients following an out of hospital cardiac arrest. In these patients, applied hypothermia reduced arterial CO2, at unchanged alveolar dead space ventilation, minute volume ventilation and static compliance, thereby improving ventilation (chapter 7). As a prospective sub–analysis of a randomized controlled trial on the effect of hypothermia after cardiac arrest, we were able to dissect the effect of hypothermia on levels of circulating mtDNA. These parts of mitochondria reflect the amount of tissue damage and are thought to act as ‘alarmins’, which can strongly drive the host inflammatory response and is associated with adverse outcome in SIRS (23). We found that applied hypothermia reduced circulating mtDNA levels in cardiac arrest patients (chapter 8). Hydrogen sulfide Inhalation of H2S causes a reversible hypo–metabolic state, termed suspended animation like state in mice, (24). Besides a rapid reduction in metabolism, H2S also has anti–inflammatory effects and improves mitochondrial function and integrity (25). In a rat VILI model, infusion of NaHS, which is a H2S donor, reduced body temperature, heart rate and exhaled CO2 levels, thereby resembling a suspended animation like state (chapter 9). NaHS protected against lung injury, an effect which was independent of ensuing hypothermia. NaHS infusion also improved cardiac performance by increasing stroke volume at unchanged cardiac output. Of note, after cessation of NaHS infusion, rats showed no behavioural or neurological damage. Next, we hypothesized that H2S–induced hypo–metabolism is associated with reduced lung injury and maintainment of mitochondrial function in rats with established pneumosepsis (chapter 10). We again observed a significant reduction in lung and kidney injury markers, associated with improved mitochondrial respiration with increased ATP availability and less oxidative damage. Improved mitochondrial function was associated with upregulation of regulators of VDAC, such as α–tubulin and phophorylated PKC–ε. VDAC is a voltage dependent porin involved in ATP and ADP exchange between mitochondria and the cytosol. Not only was mitochondrial function better preserved, H2S also improved mitochondrial biogenesis. As prolonged hypo–metabolism may be more beneficial, we observed surprising results in animals with LPS induced SIRS with ARDS, during short and prolonged course of NaHS infusion (chapter 11).. 13. I. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(16) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter I. While short course of NaHS infusion offered organ protection, prolonged infusion did not enhance protection in a rat endotoxemia model. In this model, intravenous NaHS caused both a pro–inflammatory as well as an anti–inflammatory response, with increased TNF–α and IL–10 levels. In LPS stimulated whole blood co incubated with NaHS and neutralizing IL–10 antibody, NaHS abrogated inflammation. These results suggest that NaHS offers protection possibly via IL–10 production. 3–iodothyronamine (T1am) Pharmacological induction of a hypometabolic state may have less side effects then hypothermia and H2S. In chapter 12 we hypothesized that T1am, a thyroid hormone derivate known to rapidly reduce metabolism, would protect against lung injury caused by LPS in spontaneously breathing mice. After T1am injection the animals stopped moving and their O2 consumption and CO2 production decreased. Reduced metabolism was however not associated with a reduction in lung injury. Reversely, inflammation was even enhanced, associated with elevated plasma T3 levels. This thesis closes with a summary and general discussion, followed by a summary in Dutch (appendix A) and acknowledgements (appendix B).. 14.

(17) General outline of this thesis. References. I. (1) Balk RA. Pathogenesis and management of multiple organ dysfunction or failure in severe sepsis and septic shock. Crit Care Clin 2000 April;16(2):337–52, vii. (2) Landry DW, Oliver JA. The pathogenesis of vasodilatory shock. N Engl J Med 2001 August 23;345(8):588–95. (3) Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin Definition. JAMA 2012 June 20;307(23):2526–33. (4) American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med 1992 June;20(6):864–74. (5) Brivet FG, Kleinknecht DJ, Loirat P, Landais PJ. Acute renal failure in intensive care units––causes, outcome, and prognostic factors of hospital mortality; a prospective, multicenter study. French Study Group on Acute Renal Failure. Crit Care Med 1996 February;24(2):192–8. (6) Sprung CL, Sakr Y, Vincent JL, Le Gall JR, Reinhart K, Ranieri VM, Gerlach H, Fielden J, Groba CB, Payen D. An evaluation of systemic inflammatory response syndrome signs in the Sepsis Occurrence In Acutely Ill Patients (SOAP) study. Intensive Care Med 2006 March;32(3):421–7. (7) Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, Schetz M, Tan I, Bouman C, Macedo E, Gibney N, Tolwani A, Ronco C. Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA 2005 August 17;294(7):813–8. (8) Abraham E, Singer M. Mechanisms of sepsis–induced organ dysfunction. Crit Care Med 2007 October;35(10):2408–16. (9) Brealey D, Singer M. Mitochondrial Dysfunction in Sepsis. Curr Infect Dis Rep 2003 October;5(5):365–71. (10) Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, Peterson E, Tomlanovich M. Early goal–directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med 2001 November 8;345(19):1368–77. (11) Plotz FB, Slutsky AS, van Vught AJ, Heijnen CJ. Ventilator–induced lung injury and multiple system organ failure: a critical review of facts and hypotheses. Intensive Care Med 2004 October;30(10):1865–72. (12) Frank JA, Matthay MA. Science review: mechanisms of ventilator–induced injury. Crit Care 2003 June;7(3):233–41. (13) Belperio JA, Keane MP, Lynch JP, III, Strieter RM. The role of cytokines during the pathogenesis of ventilator–associated and ventilator–induced lung injury. Semin Respir Crit Care Med 2006 August;27(4):350–64. (14) Schultz MJ, Haitsma JJ, Zhang H, Slutsky AS. Pulmonary coagulopathy as a new target in therapeutic studies of acute lung injury or pneumonia––a review. Crit Care Med 2006 March;34(3):871–7. (15) Takahashi K, Saha D, Shattino I, Pavlov VI, Stahl GL, Finnegan P, Melo MF. Complement 3 is involved with ventilator–induced lung injury. Int Immunopharmacol 2011 December;11(12):2138–43. (16) Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. The Acute Respiratory Distress Syndrome Network. N Engl J Med 2000 May 4;342(18):1301–8. (17) Phua J, Badia JR, Adhikari NK, Friedrich JO, Fowler RA, Singh JM, Scales DC, Stather DR, Li A, Jones A, Gattas DJ, Hallett D, Tomlinson G, Stewart TE, Ferguson ND. Has mortality from acute respiratory distress syndrome decreased over time?: A systematic review. Am J Respir Crit Care Med 2009 February 1;179(3):220–7. (18) Carey HV, Andrews MT, Martin SL. Mammalian hibernation: cellular and molecular responses to depressed metabolism and low temperature. Physiol Rev 2003 October;83(4):1153–81. (19) Ferrari R, Ferrari F, Benigno M, Pepi P, Visioli O. Hibernating myocardium: its pathophysiology and clinical role. Mol Cell Biochem 1998 September;186(1–2):195–9. (20) Aslami H, Schultz MJ, Juffermans NP. Potential applications of hydrogen sulfide–induced suspended animation. Curr Med Chem 2009;16(10):1295–303. (21) Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Gutteridge G, Smith K. Treatment of comatose survivors of out–of–hospital cardiac arrest with induced hypothermia. N Engl J Med 2002 February 21;346(8):557–63.. 15. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(18) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter I. (22) Polderman KH. Mechanisms of action, physiological effects, and complications of hypothermia. Crit Care Med 2009 July;37(7 Suppl):S186–S202. (23) Arnalich F, Codoceo R, Lopez–Collazo E, Montiel C. Circulating cell–free mitochondrial DNA: a better early prognostic marker in patients with out–of–hospital cardiac arrest. Resuscitation 2012 July;83(7):e162–e163. (24) Blackstone E, Morrison M, Roth MB. H2S induces a suspended animation–like state in mice. Science 2005 April 22;308(5721):518. (25) Szabo C. Hydrogen sulphide and its therapeutic potential. Nat Rev Drug Discov 2007 November;6(11):917–35.. 16.

(19) II Targeting mitochondrial function in sepsis. Hamid Aslami1, Nicole P. Juffermans2 1. Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Academic Medical Center, Amsterdam, The Netherlands. 2. Department of Intensive Care Medicine, Academic Medical Center, Amsterdam, The Netherlands.. Netherlands Journal of Critical Care, August 2012 16:117–124.

(20) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II. Abstract Sepsis is characterized by an exaggerated inflammatory response, with mobilization of bodily energy reserves to combat the invading microorganism. Despite increasing knowledge on pathophysiology and improved treatment, mortality of sepsis is increasing and calling for new therapeutic strategies. Mitochondria may be a target in reducing organ failure in sepsis, as the severity of mitochondrial dysfunction has been linked to the severity of sepsis. Pre– clinical and clinical data suggest that mitochondrial ‘resuscitation’ is beneficial in sepsis, by improving substrate utilization resulting in an improvement in local energy expenditures. In this review, we will discuss metabolic changes occurring during the course of sepsis and discuss possible strategies that target mitochondria which may be a novel way of reducing organ failure and thereby mortality in sepsis.. 18.

(21) Targeting mitochondrial function in sepsis. Introduction Sepsis is the systemic inflammatory response in the presence of infection (1). Current treatment for sepsis is supportive and includes antibiotics, fluid resuscitation and vasopressor agents to maintain adequate organ perfusion, as well as the use of organ replacement therapy. Despite extensive research and the exploration of various therapeutic possibilities, sepsis is still the leading cause of death in the critically ill patient and the incidence of sepsis– related death is rising (2) which calls for new therapeutic targets. The inflammatory response to infection is initiated by the binding of bacterial microbial products to recognition receptors on immune cells, with activation of nuclear transcription factors leading to the production of tumour necrosis factor and various other acute phase cytokines. Together with the activation of the complement system, a pro–coagulant state is present (3). In response to invading micro–organisms, circulating immune cells, epithelial and endothelial cells are stimulated to produce reactive oxygen species (ROS) (4). ROS not only damages the invading micro–organism, but also healthy tissue. With the pro–inflammatory response, a compensatory anti–inflammatory response is simultaneously initiated (3). A dysregulated immune response, in which the pro–inflammatory response overwhelms the anti–inflammatory response, can lead to multiple organ failure (3). At the later phase of sepsis, when anti–inflammatory responses ensue, a state of “immune paralysis” develops, in which patients are more vulnerable to secondary infections (5). Furthermore, metabolic (6) and endocrine (7) disturbances are frequently found in sepsis and are thought to contribute to organ failure. The mechanisms of sepsis–induced organ failure remain controversial. The correlation between high plasma lactate levels and mortality in septic shock patients has led to the conclusion that oxygen debt was likely to contribute to organ failure. Indeed, adequate oxygenation as part of early goal directed therapy was found to improve outcome of sepsis (8). However, increasing oxygen delivery failed to improve outcome in sepsis. On the contrary, boosting oxygen delivery may even be detrimental in sepsis patients (9), suggesting a failure in oxygen utilization rather than impairment in delivery. Mitochondria utilize 90% of total bodily oxygen need and have been shown to play a crucial role in the so–called “cytopathic hypoxia”. Indeed, recent research indicates that mitochondrial dysfunction is a feature of organ injury in sepsis (10;11), even in the presence of adequate tissue oxygenation (12). It has been hypothesized that a decrease in mitochondrial oxygen consumption is a functional response to the overwhelming inflammatory response in sepsis (13;14). In this view, mitochondrial ‘shut–down’ with concomitant hypo–metabolism may increase the chance of survival of cells or recovery of mitochondria after sustaining an intense inflammatory insult, thereby contributing to recovery of organ failure in sepsis. This hypothesis is strengthened by the. 19. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(22) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II. finding that cellular hypoxia during sepsis is absent, oxygen consumption is reduced (15) and cell death in failing organs is practically not observed (16;17). In particular, histological examination of failing organs is often remarkably normal, with minimal or no apoptosis or necrosis, even in those who die. Alternatively, reduced mitochondrial activity may be due to mitochondrial damage. In line with this, increased levels of cytokines and the generation of ROS have been found to induce damage of mitochondrial respiratory complexes as well as other vital mitochondrial proteins. In addition, increased lactate levels are found in sepsis, associated with adverse outcome (18). This may suggest an ATP demand at the cellular level. Of note, survival of critically ill patients improved by increasing intracellular glucose levels by insulin, which resulted in increased activity of mitochondrial respiratory complexes (19), the product of which is ATP. Also, mitochondria in muscle of sepsis patients who progress to death appear much more swollen compared to mitochondria in those who survive (20). Thereby, mitochondrial dysfunction may not be an adaptive process but rather a result of injury inflicted by excessive inflammation. Regardless the hypothesis, it is clear that mitochondria are important players in the progression of sepsis. Several clinical studies have suggested that mitochondrial structure (19;21;22) as well as mitochondrial function can be improved by modulating mitochondrial substrate (23). Thereby, mitochondria may be a promising target in combating sepsis. In the following section, we will discuss mitochondrial abnormalities in sepsis and possible strategies to improve mitochondrial function.. Methods The following keywords or MeSH terms were used to obtain papers published in the Medline database: Mitochondria OR mitochondrial dysfunction OR mitochondrial therapy OR metabolism OR oxidative phosphorylation were combined with sepsis OR severe sepsis OR multiple organ failure OR shock OR critical illness OR antioxidants OR hypothermia OR suspended animation like state OR hydrogen sulfide. The relevance of each paper was assessed using the online abstracts. In addition, the reference list of the retrieved papers was screened for potentially important papers. Mitochondrial energy production in health and disease Mitochondria are membrane–bound organelles found in most cells. They consist of an inner membrane, an outer membrane, an inter–membrane space, cristae and the matrix. The inner membrane has a large surface area, which contains the 5 respiratory complexes involved in the generation of energy in the form of ATP through oxidative phosphorylation (Figure 1). Nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FADH2), two highly energetic molecules produced in the citric acid cycle, deliver electrons. 20.

(23) Targeting mitochondrial function in sepsis. to respiratory complexes I and II respectively. The electrons pass through from one respiratory complex to the other, thereby generating a force which is used by the complexes to pump H+ from the matrix into the inter–membrane space. The generated mitochondrial current is used by complex V to generate ATP by coupling ADP with phosphate (Figure 1). The generated ATP exit mitochondria through ATP–transporters and is available for cellular metabolism. Daily mitochondrial ATP production and consumption is incredibly high and has been estimated to equal the body weight of a grown man (24). Mitochondrial function can be estimated in vitro by stimulating mitochondrial enzymes involved in the citric acid cycle. Also, mitochondrial respiration can be measured in isolated mitochondria, as well as ATP, ADP, phosphate, creatinine phosphate and lactate levels as markers of mitochondrial functionality in vivo. The number and structure of mitochondria, as well as their function, are regulated by mitochondrial biogenesis (25). This is a complex cellular program between mitochondrial DNA and the nucleus of the cell, which regulates cellular energy production. Synthesis of mitochondrial proteins and components is enhanced when energy demand is high. Synthesis is counterbalanced by mitophagy, a process which involves selective removal of mitochondria when energy demand is low or when mitochondria are damaged (26). In animal models of sepsis caused by endotoxin (27) or live bacteria (28), functional and structural mitochondrial abnormalities have been described. Also in patients with sepsis, electron microscopy images of mitochondria in muscle tissue appear to be swollen with destructed cristae, associated with organ failure (10). It is not entirely clear how mitochondrial abnormalities are initiated, but nitric oxide (NO) may be a main pathway (Figure 1) (29). In sepsis, NO is produced in excess, due to pro–inflammatory cytokines which promote NO formation via inducible nitric oxide synthase (iNOS) (30). NO has been shown to competitively block mitochondrial complex I (31) and IV (32), resulting in diminished oxygen consumption. In septic patients, elevated muscle oxygen levels can be observed (9). When intracellular oxygen levels increase, non–enzymatic production of ROS at complex I and III can occur (Figure 1). The increased ROS can react and thereby directly damage mitochondrial proteins, lipids and DNA (11;27;28). Mitochondrial abnormalities, reflected by reduced tissue ATP/ADP ratios, may inhibit ATP–dependent reactions, in particular an adequate host response to invading organisms. Decreased ATP content has been shown to be negatively associated with resolution of organ dysfunction in a long term rat model of sepsis (11) as well as with increased mortality in muscle biopsies taken from septic patients (11). Besides the inflammatory response, a number of medications that are frequently administered during sepsis may decrease mitochondrial respiration, including antibiotics (33). Data regarding mitochondrial abnormalities in sepsis are, however, not all consistent. In some models of sepsis, damage to muscle mitochondria or alteration in ATP or creatinine phosphate concentrations were not observed (34;35). Contrasting results may be explained. 21. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(24) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II. by methodological differences with regard to sepsis severity, sepsis duration and species used in the models. The metabolic responses in sepsis which aim to mobilize bodily energy reserves suggest a compensatory response to local low ATP levels. The response may, however, be exaggerated, as suggested by both clinical and preclinical studies, with concomitant organ failure and increased mortality when left untreated. Whether this response is effective, contributing to improved host response, or exaggerated, contributing to adverse outcome, is discussed in the following section. Changes in energy and substrate metabolism in sepsis In sepsis, energy expenditure is usually increased by mobilizing bodily energy reserves, in order to maintain an adequate host response (36). Plasma glucose levels increase due to insulin resistance and increased gluconeogenesis, initiated by pro–inflammatory cytokines (37). The release of glucagon, cortisol, epinephrine and growth hormone further contribute to an increase in plasma glucose levels. Amino acids and fatty acid levels are increased by the breakdown of muscle and fat respectively. The amino acids are essentially used in the liver to fuel the synthesis of acute phase proteins and for gluconeogenesis, while fatty acids are important substrates for formation of prostaglandins and leukotrienes at the site of infection. The mobilization of body reserves and the breakdown of proteins can be seen as a general response to stress, which, in a broad way, releases substrates to organs to support vital functions. From an evolutionary standpoint, re–allocating energy reserves to combat the infection has more priority than storage and growth. In line with this view, insulin resistance is also seen during starvation and growth (e.g. pregnancy and puberty) (38). Thereby, it can be hypothesized that insulin resistance may be a beneficial adaptation that secures survival or growth of the organism. In sepsis however, this response may be exaggerated, leading to enhanced wasting. Insulin resistance in sepsis is supposedly an unwanted change in glucose metabolism and attempts are then made to correct hyperglycaemia (39), albeit with conflicting results on outcome(40). It seems clear, however, that gross hyperglycaemia is associated with adverse outcome in sepsis (39;41). Similarly to the detrimental effects of hyperglycaemia, enhanced lipid breakdown could also be damaging. The enhanced lipid breakdown observed in sepsis results in an increase in triglyceride levels, with high levels of very–low–density lipoprotein (VLDL) and reduced levels of high density lipoprotein (HDL), thereby not only promoting atherogenesis, but also aggravating the course of sepsis (42), as HDL is considered to contribute to scavenging of bacterial toxins. Furthermore, low cholesterol levels have also been associated with increased risk of infection, leading to prolonged hospital stay (43). Enhanced protein breakdown results in muscle wasting and fatigue. Muscle weakness acquired in the ICU not only increases length of stay, but also time on the mechanical ventilator, and is associated with increased mortality (44;45). Thereby, mobilization of body reserves may be detrimental in sepsis. 22.

(25) Targeting mitochondrial function in sepsis. Contrary to the belief that organ dysfunction is a result of a failure to increase energy expenditure, is the hypothesis that a decrease in mitochondrial activity and subsequent cellular processes is a functional response to overwhelming infection. An observation that supports this notion is that organ damage is mostly reversible, including organs with poor regenerative capacity, such as the liver. Both decreased mitochondrial functionality as well as the regenerative capacity of mitochondria have been found to be associated with improved survival in patients with sepsis (10;20). However, whereas these observations point out the important role of mitochondria in the pathogenesis of organ failure, they do not determine whether recovery of mitochondrial function is a result of a functional mitochondrial ‘shut– down’ or of resolving mitochondrial damage. Direct measurement of ATP turnover and thus cellular metabolism in patients which may provide more insight into this issue, remains a challenge. As the body copes with altered energy levels, influencing mitochondrial substrate utilization, or more general, improving mitochondrial function, may represent a candidate mechanism of improvement or resolution of sepsis–induced organ failure. Influencing mitochondrial substrate utilization in sepsis Carbohydrates The most efficient pathway to produce ATP is through oxidative phosphorylation in the mitochondria. An alternative pathway is anaerobic glycolysis, thereby forming lactate. As discussed, plasma levels of lactate are increased in sepsis even in the absence of hypoxia. Mitochondrial complex I deficiency is common, while complex II is well preserved in sepsis (10;23). Thus, when electrons flow through complex I is compromised, the addition of complex II substrate might be a strategy to improve electron flow and thus enhance ATP production in sepsis. In line with this thought, adding succinate as substrate reversed the decline in mitochondrial oxygen consumption in a rat model of abdominal sepsis (23) by approximately 40% (Figure 1). In this model, mitochondrial oxygen consumption stimulated with glutamate and malate was reduced, suggesting a complex I deficiency, which correlated with sepsis severity. Similarly, infusion of succinate prevented a decrease in liver ATP content and prolonged survival in murine endotoxemia as well as in a model of abdominal sepsis (46;47). Also, normoglycemia achieved by infusion of insulin was found to improve mitochondrial integrity in the liver of patients with sepsis (19). On electron microscopy imaging, liver mitochondria from patients who were assigned intensive insulin therapy showed less morphological abnormalities when compared to patient’s assigned to conventional therapy (19). Also, mitochondrial complexes I and IV showed significantly higher activity in the intensive insulin therapy group compared to conventional therapy (Figure 1) (19). The effects on outcome are however conflicting (40;48;49).. 23. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(26) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II. Figure 1: Schematic representations of mitochondrial ATP production through oxidative phosphorylation starting with production of highly energetic molecules in the Crebs cycles in healthy mitochondria. Sepsis associated damage and potential strategies to resuscitate mitochondria are shown in red and blue color respectively in the right section of the figure. See text for a more detailed description.. Amino acids Experimental data suggest impairment of enzymes involved in the citric acid cycle in sepsis, which catalyze degradation of carbohydrates to form NADH and FADH2 (50) (Figure 1). A drop in NADH and FADH2 can lead to reduction in electron flow through the respiratory complexes, thereby diminishing ATP production. Glutamine, the most abundant circulatory amino acid, is converted to glutamate and α–ketogluterate, thereby entering the citric acid cycle at a different location than carbohydrates and boosting up the formation of electron donating molecules resulting in enhanced ATP production through oxidative phosphorylation. This was proven in a rat model of sepsis–induced by cecal ligation and puncture. Administration of glutamine in this model resulted in increased oxygen extraction and mitochondrial function, associated with improvement in cytochrome c oxidase activity (51). Beside glutamine, arginine has also been studied extensively in the septic patient. Arginine is a non–essential amino acid. In sepsis, levels of arginine are decreased, presumably due to enhanced demand caused by increased formation of NO from arginine (52) and by starvation due to limited nutritional supply. Supplementing arginine was shown to improve immunologic response by enhancing macrophage phagocytic activity and ROS production in animal studies (53), but also to contribute to mortality in a canine model of peritonitis (54). In clinical trials, the aggregated outcome data of septic patients who received supplemental arginine were unfavourably affected. Thereby, the use of arginine during sepsis was discouraged. Recently, however, it was found that low arginine levels in relation to its inhibitor are associated with disease severity and higher mortality (55). To date, it is undecided whether improving arginine bioavailability is beneficial in sepsis. Also,. 24.

(27) Targeting mitochondrial function in sepsis. it is not known whether the effects of glutamine are manifested by altering mitochondrial function. Targeting oxidative stress in sepsis Sepsis is associated with massive oxidative stress, defined as an excessive production of ROS (Figure 1). ROS is the most important antimicrobial defence mechanism (56). However, ROS can also exacerbate organ injury (57). Nicotinamide–adenine dinucleotide phosphate (NADPH) oxidase, xantine oxidase and proton leak across the inner mitochondrial membrane are the main pathways for ROS formation. ROS which is formed by a leakage of protons across the inner mitochondrial membrane is presumably a result of damage to the respiratory complexes (56). Tissue damage due to ROS is counter balanced by antioxidants and by ROS scavenging enzymes (57). In sepsis, however, this balance is disturbed, due to increased ROS production and exhausted antioxidant pools. Alterations in these systems are associated with disease severity (4;58). In septic patients, low plasma levels of antioxidants are found, especially selenium and ascorbic acid. Also, lipid peroxidation levels, which are a marker for oxidative damage, are high in sepsis, correlating with organ failure (58). As the degree of oxidative stress correlates with disease severity (4), a supplement of antioxidants or ROS scavengers as a therapeutic strategy to reduce ROS damage in sepsis is a logical approach. Vitamins Uncontrolled ROS formation in the mitochondria can be scavenged by increasing the levels of antioxidants in sepsis. Vitamin A has been known for its antioxidant and immunomodulatory properties. When given to septic animals, a reduction in inflammation was observed (59). Vitamin E and C are other antioxidants, which are depleted in sepsis (60). Vitamin C acts on vascular endothelial cells through an inhibitory effect on iNOS, thereby reducing levels of NO in sepsis (61). Additionally, supplementation with vitamin C was shown to induce apoptosis in circulating neutrophils in patients with abdominal sepsis (62). The clinical benefit of vitamin C and E supplementation was shown in double–blind, placebo–controlled trials, showing a reduction in mortality, organ failure, duration of mechanical ventilation and length of ICU stay (21;63). Similarly, early enteral supplementation with key pharmaco– nutrients, including vitamins and glutamine, was associated with faster recovery of organ function compared to control (22). Selenium Selenium is an important component of enzymes involved in ROS degradation (64). Selenium plasma levels tend to be low in sepsis and to correlate with disease severity (65). In a double blind, randomized placebo–controlled multicenter trial in septic patients,. 25. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(28) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II. continuous infusion of selenium reduced the rate of new infections, as well as mortality (65;66). However, these beneficial effects were not confirmed in another randomized trial (67). Probably, dose and timing of selenium supplementation played an important role. In the positive trial, lower doses were infused for a longer period, while in the negative trial, higher doses were administered during a shorter period. Importantly, selenium in combination with glutamine possibly increased the number of mitochondria, measured as an increase in mitochondrial DNA copy number (68). Delivery of antioxidants in the mitochondria Direct delivery of antioxidants at the site of ROS generation in the mitochondria can be achieved with MitoQ (69). In an animal model of sepsis, administration of MitoQ reduced organ dysfunction (70). Thus far, phase II clinical trials have been performed in chronic hepatitis and in Parkinson’s disease (71), but not yet in sepsis patients. Taken together, antioxidant therapy in the early phase of sepsis seems promising to prevent or attenuate the progression of organ failure in sepsis. However, studies of antioxidant administration in patients with sepsis have not been convincing to date and more research on this issue is warranted (72). Inducing a hypo–metabolic state in sepsis Sepsis is characterized by hypermetabolism, in which bodily fuel reserves are mobilized in order to cope with the overwhelming insult, as described in the previous sections. Thus far, therapies are supportive, aimed at supplementing substrates or at increasing oxygen delivery to meet high demands (3). An alternative way of reducing organ failure and thereby mortality rate would be to reduce metabolic demand at the cellular level, a state as observed in naturally hibernating mammals. Humans do not hibernate and have only a limited tolerance to hypoxia. Nevertheless, myocardial hibernation, which can occur in patients with myocardial contractile dysfunction due to ischemic heart disease, is thought to provide an adaptive response to hypoxia. Down regulation of myocardial contraction may result in reduced energy expenditure, thereby preserving cellular integrity and viability (73). Hypothermia Hypothermia is a well–known therapeutic strategy to reduce organ injury. This approach is used during cardiothoracic surgery and organ transplantation (74). Also, hypothermia reduces brain injury in patients after a cardiac arrest (75). Hypothermia reduces metabolism by 7% per grade reduction, thereby reducing oxygen requirement and carbon dioxide production, leading to decreasing ATP requirements. In patients with severe ARDS associated with sepsis, induced hypothermia applied as a last resort reduced mortality (76). Also, hypothermia is able to reduce inflammation and prevent the production of superoxide and subsequent. 26.

(29) Targeting mitochondrial function in sepsis. formation of reactive oxygen and nitrogen species during ischemia (74). In a model of ventilator–induced lung injury (77), we found increased mitochondrial oxygen consumption compared to animals subjected to lung protective mechanical ventilation (Figure 2). As injurious mechanical ventilation is characterized by increased production of inflammatory cytokines (78), increase in oxygen consumption suggests increased ATP demand to maintain the pro–inflammatory state. When the body temperature was reduced to 32°C, oxidative phosphorylation decreased (Figure 2). This may reflect less oxygen consumption in vivo, in line with a previous report (79). Also, respiratory control ratio, a parameter of mitochondrial coupling between respiration and oxidative phosphorylation, was decreased, while state 4 respiration increased, which suggests mitochondrial uncoupling during hypothermia, as seen before (79). Uncoupling induced by hypothermia prevents the production of ROS that mediates organ damage (80). Of note, detrimental effects of hypothermia are described, in particular increased risk for infection, coagulation disorders and arrhythmia (74). Therefore, besides hypothermia, other novel strategies to reduce metabolism are needed. Suspended animation like state Hydrogen sulfide (H2S), commonly considered to be an environmental hazard, has been shown to induce a hibernation–like state in naturally non–hibernating animals (81), characterized by reduction in body temperature, carbon dioxide production and oxygen consumption. Similar physiological changes were observed, when NaHS, a H2S donor, was infused in rats (82). Physiological changes were associated with reduced lung injury inflicted by mechanical ventilation. Furthermore, H2S protected against ischemia reperfusion injury in an animal model of myocardial ischemia by preservation of mitochondrial structure and function (83). Other mechanisms of protection are attributed via a vaso–relaxant effect, attenuation of inflammation and reduction of apoptosis (84). In addition, an antioxidant effect of H2S was observed (85). However, pro–inflammatory and pro–apoptotic effects have also been described (84;86;87). These contrasting results may depend on differences in dose, route of application and on tissue. Despite the disadvantages related to H2S toxicity, its use as a potential therapy in battle wounds has raised the attention of the US military forces. Battle wounds are associated with extreme haemorrhagic shock, multiple organ failure and high mortality (88). Survival from battle wounds depends on rapid damage control surgery. The hypothesis that treatment with H2S could buy time to reach a hospital by lowering metabolism and tissue oxygen demand in the wounded soldier is currently under investigation (89;90).. 27. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(30)  '&*,%(+"'&&%'$%"&% .  

(31) . . ')%'+!)%" .('+!)%". . .     .

(32). .  . . .     .  ,& &#,)"',* %!&"$-&+"$+"'&.

(33) .

(34).  )+"'. . ++  '&*,%(+"'&&%'$%"&% . .   . R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II.  ,& )'++"- %!&"$-&+"$+"'&. ,& &#,)"',* %!&"$-&+"$+"'&. ,& )'++"- %!&"$-&+"$+"'&. Figure 2: The effect of hypothermia on mitochondrial oxygen consumption (A), state 4 respiration (B), respiratory control ratio (RCR) (C) and ADP/O2 ratio (D) during oxidative phosphorylation of mitochondrial complex II substrate succinate and complex I blocker rotenone in animals after lung protective or injurious mechanical ventilation. State 4 respiration, RCR (mitochondrial oxygen consumption / state 4) and ADP/O2 ratios are functional parameters for mitochondrial uncoupling, coupling between respiration and oxidative phosphorylation and mitochondrial efficiency respectively. Data are means ± SEM.. Conclusion Despite advances in understanding the pathophysiology of sepsis–induced organ failure, mortality and morbidity remain high. Impaired mitochondrial function leading to a fall in organ ATP levels is probably a main pathway. Thus far, strategies targeting mitochondrial function seem promising, as well as reducing collateral damage caused by excessive ROS formation by antioxidants in sepsis. Combining antioxidant therapy with mitochondrial substrate is worth investigating in future trials. Timing and dosing of these interventions need further investigation.. 28.

(35) Targeting mitochondrial function in sepsis. References (1) Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, Cohen J, Opal SM, Vincent JL, Ramsay G. 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Crit Care Med 2003 April;31(4):1250–6. (2) Dombrovskiy VY, Martin AA, Sunderram J, Paz HL. Rapid increase in hospitalization and mortality rates for severe sepsis in the United States: a trend analysis from 1993 to 2003. Crit Care Med 2007 May;35(5):1244–50. (3) Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med 2003 January 9;348(2):138–50. (4) Alonso de Vega JM, Diaz J, Serrano E, Carbonell LF. Plasma redox status relates to severity in critically ill patients. Crit Care Med 2000 June;28(6):1812–4. (5) Hotchkiss RS, Opal S. Immunotherapy for sepsis––a new approach against an ancient foe. N Engl J Med 2010 July 1;363(1):87–9. (6) Carre JE, Singer M. Cellular energetic metabolism in sepsis: the need for a systems approach. Biochim Biophys Acta 2008 July;1777(7–8):763–71. (7) Nylen ES, Muller B. Endocrine changes in critical illness. J Intensive Care Med 2004 March;19(2):67–82. (8) Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, Peterson E, Tomlanovich M. Early goal–directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med 2001 November 8;345(19):1368–77. (9) Hayes MA, Timmins AC, Yau EH, Palazzo M, Hinds CJ, Watson D. Elevation of systemic oxygen delivery in the treatment of critically ill patients. N Engl J Med 1994 June 16;330(24):1717–22. (10) Brealey D, Brand M, Hargreaves I, Heales S, Land J, Smolenski R, Davies NA, Cooper CE, Singer M. Association between mitochondrial dysfunction and severity and outcome of septic shock. Lancet 2002 July 20;360(9328):219–23. (11) Brealey D, Karyampudi S, Jacques TS, Novelli M, Stidwill R, Taylor V, Smolenski RT, Singer M. Mitochondrial dysfunction in a long–term rodent model of sepsis and organ failure. Am J Physiol Regul Integr Comp Physiol 2004 March;286(3):R491–R497. (12) VanderMeer TJ, Wang H, Fink MP. Endotoxemia causes ileal mucosal acidosis in the absence of mucosal hypoxia in a normodynamic porcine model of septic shock. Crit Care Med 1995 July;23(7):1217–26. (13) Abraham E, Singer M. Mechanisms of sepsis–induced organ dysfunction. Crit Care Med 2007 October;35(10):2408–16. (14) Singer M, De S, V, Vitale D, Jeffcoate W. Multiorgan failure is an adaptive, endocrine–mediated, metabolic response to overwhelming systemic inflammation. Lancet 2004 August 7;364(9433):545– 8. (15) Boekstegers P, Weidenhofer S, Kapsner T, Werdan K. Skeletal muscle partial pressure of oxygen in patients with sepsis. Crit Care Med 1994 April;22(4):640–50. (16) Hotchkiss RS, Swanson PE, Freeman BD, Tinsley KW, Cobb JP, Matuschak GM, Buchman TG, Karl IE. Apoptotic cell death in patients with sepsis, shock, and multiple organ dysfunction. Crit Care Med 1999 July;27(7):1230–51. (17) Mongardon N, Dyson A, Singer M. Is MOF an outcome parameter or a transient, adaptive state in critical illness? Curr Opin Crit Care 2009 October;15(5):431–6. (18) Nguyen HB, Rivers EP, Knoblich BP, Jacobsen G, Muzzin A, Ressler JA, Tomlanovich MC. Early lactate clearance is associated with improved outcome in severe sepsis and septic shock. Crit Care Med 2004 August;32(8):1637–42. (19) Vanhorebeek I, De VR, Mesotten D, Wouters PJ, De Wolf–Peeters C, Van den BG. Protection of hepatocyte mitochondrial ultrastructure and function by strict blood glucose control with insulin in critically ill patients. Lancet 2005 January 1;365(9453):53–9. (20) Carre JE, Orban JC, Re L, Felsmann K, Iffert W, Bauer M, Suliman HB, Piantadosi CA, Mayhew TM, Breen P, Stotz M, Singer M. Survival in critical illness is associated with early activation of mitochondrial biogenesis. Am J Respir Crit Care Med 2010 September 15;182(6):745–51.. 29. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(36) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II. (21) Crimi E, Liguori A, Condorelli M, Cioffi M, Astuto M, Bontempo P, Pignalosa O, Vietri MT, Molinari AM, Sica V, Della CF, Napoli C. The beneficial effects of antioxidant supplementation in enteral feeding in critically ill patients: a prospective, randomized, double–blind, placebo–controlled trial. Anesth Analg 2004 September;99(3):857–63, table. (22) Beale RJ, Sherry T, Lei K, Campbell–Stephen L, McCook J, Smith J, Venetz W, Alteheld B, Stehle P, Schneider H. Early enteral supplementation with key pharmaconutrients improves Sequential Organ Failure Assessment score in critically ill patients with sepsis: outcome of a randomized, controlled, double–blind trial. Crit Care Med 2008 January;36(1):131–44. (23) Protti A, Carre J, Frost MT, Taylor V, Stidwill R, Rudiger A, Singer M. Succinate recovers mitochondrial oxygen consumption in septic rat skeletal muscle. Crit Care Med 2007 September;35(9):2150–5. (24) Tornroth–Horsefield S, Neutze R. Opening and closing the metabolite gate. Proc Natl Acad Sci U S A 2008 December 16;105(50):19565–6. (25) Kelly DP, Scarpulla RC. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes Dev 2004 February 15;18(4):357–68. (26) Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol 2011 January;12(1):9–14. (27) Crouser ED, Julian MW, Blaho DV, Pfeiffer DR. Endotoxin–induced mitochondrial damage correlates with impaired respiratory activity. Crit Care Med 2002 February;30(2):276–84. (28) Gellerich FN, Trumbeckaite S, Hertel K, Zierz S, Muller–Werdan U, Werdan K, Redl H, Schlag G. Impaired energy metabolism in hearts of septic baboons: diminished activities of Complex I and Complex II of the mitochondrial respiratory chain. Shock 1999 May;11(5):336–41. (29) Bolanos JP, Heales SJ, Peuchen S, Barker JE, Land JM, Clark JB. Nitric oxide–mediated mitochondrial damage: a potential neuroprotective role for glutathione. Free Radic Biol Med 1996;21(7):995– 1001. (30) Okamoto I, Abe M, Shibata K, Shimizu N, Sakata N, Katsuragi T, Tanaka K. Evaluating the role of inducible nitric oxide synthase using a novel and selective inducible nitric oxide synthase inhibitor in septic lung injury produced by cecal ligation and puncture. Am J Respir Crit Care Med 2000 August;162(2 Pt 1):716–22. (31) Brown GC, Borutaite V. Inhibition of mitochondrial respiratory complex I by nitric oxide, peroxynitrite and S–nitrosothiols. Biochim Biophys Acta 2004 July 23;1658(1–2):44–9. (32) Clementi E, Brown GC, Feelisch M, Moncada S. Persistent inhibition of cell respiration by nitric oxide: crucial role of S–nitrosylation of mitochondrial complex I and protective action of glutathione. Proc Natl Acad Sci U S A 1998 June 23;95(13):7631–6. (33) Duewelhenke N, Krut O, Eysel P. Influence on mitochondria and cytotoxicity of different antibiotics administered in high concentrations on primary human osteoblasts and cell lines. Antimicrob Agents Chemother 2007 January;51(1):54–63. (34) Geller ER, Jankauskas S, Kirkpatrick J. Mitochondrial death in sepsis: a failed concept. J Surg Res 1986 May;40(5):514–7. (35) Hotchkiss RS, Song SK, Neil JJ, Chen RD, Manchester JK, Karl IE, Lowry OH, Ackerman JJ. Sepsis does not impair tricarboxylic acid cycle in the heart. Am J Physiol 1991 January;260(1 Pt 1):C50–C57. (36) Tappy L, Chiolero R. Substrate utilization in sepsis and multiple organ failure. Crit Care Med 2007 September;35(9 Suppl):S531–S534. (37) Plomgaard P, Bouzakri K, Krogh–Madsen R, Mittendorfer B, Zierath JR, Pedersen BK. Tumor necrosis factor–alpha induces skeletal muscle insulin resistance in healthy human subjects via inhibition of Akt substrate 160 phosphorylation. Diabetes 2005 October;54(10):2939–45. (38) Hadden DR, McLaughlin C. Normal and abnormal maternal metabolism during pregnancy. Semin Fetal Neonatal Med 2009 April;14(2):66–71. (39) Van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R. Intensive insulin therapy in the critically ill patients. N Engl J Med 2001 November 8;345(19):1359–67. (40) Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, Bellomo R, Cook D, Dodek P, Henderson WR, Hebert PC, Heritier S, Heyland DK, McArthur C, McDonald E, Mitchell I, Myburgh JA, Norton R, Potter J, Robinson BG, Ronco JJ. Intensive versus conventional glucose control in critically ill patients. N Engl J Med 2009 March 26;360(13):1283–97.. 30.

(37) Targeting mitochondrial function in sepsis. (41) Van den BG, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, Van WE, Bobbaers H, Bouillon R. Intensive insulin therapy in the medical ICU. N Engl J Med 2006 February 2;354(5):449– 61. (42) Chien JY, Jerng JS, Yu CJ, Yang PC. Low serum level of high–density lipoprotein cholesterol is a poor prognostic factor for severe sepsis. Crit Care Med 2005 August;33(8):1688–93. (43) Shor R, Wainstein J, Oz D, Boaz M, Matas Z, Fux A, Halabe A. Low HDL levels and the risk of death, sepsis and malignancy. Clin Res Cardiol 2008 April;97(4):227–33. (44) Sharshar T, Bastuji–Garin S, Stevens RD, Durand MC, Malissin I, Rodriguez P, Cerf C, Outin H, de JB. Presence and severity of intensive care unit–acquired paresis at time of awakening are associated with increased intensive care unit and hospital mortality. Crit Care Med 2009 December;37(12):3047–53. (45) de JB, Bastuji–Garin S, Durand MC, Malissin I, Rodrigues P, Cerf C, Outin H, Sharshar T. Respiratory weakness is associated with limb weakness and delayed weaning in critical illness. Crit Care Med 2007 September;35(9):2007–15. (46) Malaisse WJ, Nadi AB, Ladriere L, Zhang TM. Protective effects of succinic acid dimethyl ester infusion in experimental endotoxemia. Nutrition 1997 April;13(4):330–41. (47) Ferreira FL, Ladriere L, Vincent JL, Malaisse WJ. Prolongation of survival time by infusion of succinic acid dimethyl ester in a caecal ligation and perforation model of sepsis. Horm Metab Res 2000 August;32(8):335–6. (48) Brunkhorst FM, Engel C, Bloos F, Meier–Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U, John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M, Hartog C, Natanson C, Loeffler M, Reinhart K. Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med 2008 January 10;358(2):125–39. (49) Arabi YM, Dabbagh OC, Tamim HM, Al–Shimemeri AA, Memish ZA, Haddad SH, Syed SJ, Giridhar HR, Rishu AH, Al–Daker MO, Kahoul SH, Britts RJ, Sakkijha MH. Intensive versus conventional insulin therapy: a randomized controlled trial in medical and surgical critically ill patients. Crit Care Med 2008 December;36(12):3190–7. (50) Mason KE, Stofan DA. Endotoxin challenge reduces aconitase activity in myocardial tissue. Arch Biochem Biophys 2008 January 15;469(2):151–6. (51) Groening P, Huang Z, La Gamma EF, Levy RJ. Glutamine restores myocardial cytochrome C oxidase activity and improves cardiac function during experimental sepsis. JPEN J Parenter Enteral Nutr 2011 March;35(2):249–54. (52) Luiking YC, Poeze M, Ramsay G, Deutz NE. Reduced citrulline production in sepsis is related to diminished de novo arginine and nitric oxide production. Am J Clin Nutr 2009 January;89(1):142–52. (53) Kalil AC, Sevransky JE, Myers DE, Esposito C, Vandivier RW, Eichacker P, Susla GM, Solomon SB, Csako G, Costello R, Sittler KJ, Banks S, Natanson C, Danner RL. Preclinical trial of L–arginine monotherapy alone or with N–acetylcysteine in septic shock. Crit Care Med 2006 November;34(11):2719–28. (54) Kalil AC, Danner RL. L–Arginine supplementation in sepsis: beneficial or harmful? Curr Opin Crit Care 2006 August;12(4):303–8. (55) Gough MS, Morgan MA, Mack CM, Darling DC, Frasier LM, Doolin KP, Apostolakos MJ, Stewart JC, Graves BT, Arning E, Bottiglieri T, Mooney RA, Frampton MW, Pietropaoli AP. The ratio of arginine to dimethylarginines is reduced and predicts outcomes in patients with severe sepsis. Crit Care Med 2011 June;39(6):1351–8. (56) Fang FC. Antimicrobial reactive oxygen and nitrogen species: concepts and controversies. Nat Rev Microbiol 2004 October;2(10):820–32. (57) Crimi E, Sica V, Slutsky AS, Zhang H, Williams–Ignarro S, Ignarro LJ, Napoli C. Role of oxidative stress in experimental sepsis and multisystem organ dysfunction. Free Radic Res 2006 July;40(7):665–72. (58) Cowley HC, Bacon PJ, Goode HF, Webster NR, Jones JG, Menon DK. Plasma antioxidant potential in severe sepsis: a comparison of survivors and nonsurvivors. Crit Care Med 1996 July;24(7):1179–83. (59) Carlson D, Maass DL, White DJ, Tan J, Horton JW. Antioxidant vitamin therapy alters sepsis–related apoptotic myocardial activity and inflammatory responses. Am J Physiol Heart Circ Physiol 2006 December;291(6):H2779–H2789. (60) Borrelli E, Roux–Lombard P, Grau GE, Girardin E, Ricou B, Dayer J, Suter PM. Plasma concentrations of cytokines, their soluble receptors, and antioxidant vitamins can predict the development of multiple organ failure in patients at risk. Crit Care Med 1996 March;24(3):392–7.. 31. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(38) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter II. (61) Wu F, Wilson JX, Tyml K. Ascorbate protects against impaired arteriolar constriction in sepsis by inhibiting inducible nitric oxide synthase expression. Free Radic Biol Med 2004 October 15;37(8):1282–9. (62) Ferron–Celma I, Mansilla A, Hassan L, Garcia–Navarro A, Comino AM, Bueno P, Ferron JA. Effect of vitamin C administration on neutrophil apoptosis in septic patients after abdominal surgery. J Surg Res 2009 May 15;153(2):224–30. (63) Nathens AB, Neff MJ, Jurkovich GJ, Klotz P, Farver K, Ruzinski JT, Radella F, Garcia I, Maier RV. Randomized, prospective trial of antioxidant supplementation in critically ill surgical patients. Ann Surg 2002 December;236(6):814–22. (64) Rayman MP. The importance of selenium to human health. Lancet 2000 July 15;356(9225):233–41. (65) Angstwurm MW, Engelmann L, Zimmermann T, Lehmann C, Spes CH, Abel P, Strauss R, Meier– Hellmann A, Insel R, Radke J, Schuttler J, Gartner R. Selenium in Intensive Care (SIC): results of a prospective randomized, placebo–controlled, multiple–center study in patients with severe systemic inflammatory response syndrome, sepsis, and septic shock. Crit Care Med 2007 January;35(1):118–26. (66) Andrews PJ, Avenell A, Noble DW, Campbell MK, Croal BL, Simpson WG, Vale LD, Battison CG, Jenkinson DJ, Cook JA. Randomised trial of glutamine, selenium, or both, to supplement parenteral nutrition for critically ill patients. BMJ 2011;342:d1542. (67) Forceville X. Effects of high doses of selenium, as sodium selenite, in septic shock patients a placebo– controlled, randomized, double–blind, multi–center phase II study––selenium and sepsis. J Trace Elem Med Biol 2007;21 Suppl 1:62–5. (68) Heyland DK, Dhaliwalm R, Day A, Drover J, Cote H, Wischmeyer P. Optimizing the dose of glutamine dipeptides and antioxidants in critically ill patients: a phase I dose–finding study. JPEN J Parenter Enteral Nutr 2007 March;31(2):109–18. (69) Smith RA, Murphy MP. Animal and human studies with the mitochondria–targeted antioxidant MitoQ. Ann N Y Acad Sci 2010 July;1201:96–103. (70) Lowes DA, Thottakam BM, Webster NR, Murphy MP, Galley HF. The mitochondria–targeted antioxidant MitoQ protects against organ damage in a lipopolysaccharide–peptidoglycan model of sepsis. Free Radic Biol Med 2008 December 1;45(11):1559–65. (71) Snow BJ, Rolfe FL, Lockhart MM, Frampton CM, O’Sullivan JD, Fung V, Smith RA, Murphy MP, Taylor KM. A double–blind, placebo–controlled study to assess the mitochondria–targeted antioxidant MitoQ as a disease–modifying therapy in Parkinson’s disease. Mov Disord 2010 August 15;25(11):1670–4. (72) Mishra V. Oxidative stress and role of antioxidant supplementation in critical illness. Clin Lab 2007;53(3–4):199–209. (73) Ferrari R, Ferrari F, Benigno M, Pepi P, Visioli O. Hibernating myocardium: its pathophysiology and clinical role. Mol Cell Biochem 1998 September;186(1–2):195–9. (74) Polderman KH. Mechanisms of action, physiological effects, and complications of hypothermia. Crit Care Med 2009 July;37(7 Suppl):S186–S202. (75) Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med 2002 February 21;346(8):549–56. (76) Villar J, Slutsky AS. Effects of induced hypothermia in patients with septic adult respiratory distress syndrome. Resuscitation 1993 October;26(2):183–92. (77) Aslami H, Kuipers MT, Beurskens CJ, Roelofs JJ, Schultz MJ, Juffermans NP. Mild hypothermia reduces ventilator–induced lung injury, irrespective of reducing respiratory rate. Transl Res 2012 February;159(2):110–7. (78) Chu EK, Whitehead T, Slutsky AS. Effects of cyclic opening and closing at low– and high–volume ventilation on bronchoalveolar lavage cytokines. Crit Care Med 2004 January;32(1):168–74. (79) Bravo C, Vargas–Suarez M, Rodriguez–Enriquez S, Loza–Tavera H, Moreno–Sanchez R. Metabolic changes induced by cold stress in rat liver mitochondria. J Bioenerg Biomembr 2001 August;33(4):289–301. (80) Shao ZH, Sharp WW, Wojcik KR, Li CQ, Han M, Chang WT, Ramachandran S, Li J, Hamann KJ, Vanden Hoek TL. Therapeutic hypothermia cardioprotection via Akt– and nitric oxide–mediated attenuation of mitochondrial oxidants. Am J Physiol Heart Circ Physiol 2010 June;298(6):H2164–H2173.. 32.

(39) Targeting mitochondrial function in sepsis. (81) Blackstone E, Morrison M, Roth MB. H2S induces a suspended animation–like state in mice. Science 2005 April 22;308(5721):518. (82) Aslami H, Heinen A, Roelofs JJ, Zuurbier CJ, Schultz MJ, Juffermans NP. Suspended animation inducer hydrogen sulfide is protective in an in vivo model of ventilator–induced lung injury. Intensive Care Med 2010 November;36(11):1946–52. (83) Elrod JW, Calvert JW, Morrison J, Doeller JE, Kraus DW, Tao L, Jiao X, Scalia R, Kiss L, Szabo C, Kimura H, Chow CW, Lefer DJ. Hydrogen sulfide attenuates myocardial ischemia–reperfusion injury by preservation of mitochondrial function. Proc Natl Acad Sci U S A 2007 September 25;104(39):15560– 5. (84) Aslami H, Schultz MJ, Juffermans NP. Potential applications of hydrogen sulfide–induced suspended animation. Curr Med Chem 2009;16(10):1295–303. (85) Wei HL, Zhang CY, Jin HF, Tang CS, Du JB. Hydrogen sulfide regulates lung tissue–oxidized glutathione and total antioxidant capacity in hypoxic pulmonary hypertensive rats. Acta Pharmacol Sin 2008 June;29(6):670–9. (86) Li L, Bhatia M, Zhu YZ, Zhu YC, Ramnath RD, Wang ZJ, Anuar FB, Whiteman M, Salto–Tellez M, Moore PK. Hydrogen sulfide is a novel mediator of lipopolysaccharide–induced inflammation in the mouse. FASEB J 2005 July;19(9):1196–8. (87) Yang G, Wu L, Wang R. Pro–apoptotic effect of endogenous H2S on human aorta smooth muscle cells. FASEB J 2006 March;20(3):553–5. (88) Holcomb JB, McMullin NR, Pearse L, Caruso J, Wade CE, Oetjen–Gerdes L, Champion HR, Lawnick M, Farr W, Rodriguez S, Butler FK. Causes of death in U.S. Special Operations Forces in the global war on terrorism: 2001–2004. Ann Surg 2007 June;245(6):986–91. (89) Morrison ML, Blackwood JE, Lockett SL, Iwata A, Winn RK, Roth MB. Surviving blood loss using hydrogen sulfide. J Trauma 2008 July;65(1):183–8. (90) Roth MB, Nystul T. Buying time in suspended animation. Sci Am 2005 June;292(6):48–55.. 33. II. R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39.

(40)

(41) III Induction of a hypo–metabolic state during critical illness – a new concept in the ICU?. Hamid Aslami1, Nicole P. Juffermans1,2 1 2. Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.). Department of Intensive Care Medicine, Academic Medical Centre, Amsterdam, the Netherlands.. Netherlands Journal of Medicine 2010 May, 68:190–8.

(42) R1 R2 R3 R4 R5 R6 R7 R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39. Chapter III. Abstract Induced hypothermia after cardiopulmonary resuscitation provides organ protection and is currently considered standard of care in clinical practice. An increasing amount of reports indicate that induced hypothermia is also beneficial in other conditions of hypoxia–induced organ injury, including brain injury, intestinal ischemia–reperfusion injury and acute lung injury. The mechanism of the protective effect is thought to be caused by a reduction in metabolism. A hibernation–like state, characterized by hypothermia, bradypnea and a reduction in metabolic rate, was induced in animals that normally do not hibernate, after inhalation of hydrogen sulfide. This state was termed a ´suspended animation like state´. In critically ill patients, an exaggerated systemic inflammatory response is common, which often results in multiple organ injury. Inducing a hypo–metabolic state during critical illness may limit organ injury by reducing oxygen consumption, constituting a fascinating new therapeutic perspective for the treatment of critically ill patients. In this manuscript, we describe mitochondrial dysfunction during critical illness and preclinical data that suggest a potential therapeutic possibility of lowering metabolism. In addition, we discuss issues that warrant further research before clinical applicability. Key words: critical illness, hypothermia, metabolism and suspended animation like state.. 36.

Referenties

GERELATEERDE DOCUMENTEN

Vascular occlusion techniques that can be applied during liver resection to reduce blood loss or hepatic IRI Inflow occlusion In- and outflow occlusion Hepatic vascular

Many authors reported that conservative management is the preferred treatment in patients with postoperative bile leakage after liver resection without HJ, citing high success

After 6 hours of insufflation (14 or 25 mmHg) blood samples were collected for determination of liver injury parameters, and measurements of liver function using the indocyanine

The following procedures were performed: total laparoscopic right hemi-colectomy with tumourectomy of segment 2 (extraction via umbilical incision), total laparoscopic

Based upon additional findings, SL may change treatment strategy in patients with HCC, and in patients found to be unresectable, avoids an unnecessary laparotomy, thereby

We assessed tumor growth rate (TGR) and liver regeneration after PVE in a rabbit hepatic tumor model in chapter 9, and compared the results with a tumor control group,.. in which

Deze parameters kunnen dus gebruikt worden voor het bepalen van het tijdsinterval tussen VPE en resectie.. Serum ApoA-V veranderde niet door VPE, maar steeg kortdurend

Na bijna anderhalf jaar klinisch werk, werd zij aangenomen voor een promotie-onderzoek in het Chirurgisch Laboratorium van het Academisch Medisch Centrum te Amsterdam, onder