Skip to main content
HPB : The Official Journal of the International Hepato Pancreato Biliary Association logoLink to HPB : The Official Journal of the International Hepato Pancreato Biliary Association
. 2007;9(3):174–182. doi: 10.1080/13651820601175926

Hyperbaric oxygen therapy and liver transplantation

Vijayaragavan Muralidharan 1,, Chris Christophi 1
PMCID: PMC2063597  PMID: 18333218

Abstract

Liver transplantation is the treatment of choice for end stage liver disease and is often used for primary liver malignancies. The main limitation of its wider application is the availability of suitable donor organs. The use of marginal donor organs, split-liver transplantation and living-related liver transplantation techniques contribute to increase the donor pool. However, the use of these techniques is associated with a higher risk of post transplantation organ dysfunction, predominantly due to ischaemia, preservation and reperfusion injury (IPRI). A number of studies have demonstrated that hyperbaric oxygen (HBO) therapy influences IPRI and consequential acute cellular rejection. This article reviews the rationale of HBO therapy in the field of transplantation with particular emphasis on liver transplantation.

Keywords: acute rejection, ischaemia reperfusion injury, hyperbaric oxygen, liver failure, liver support

Introduction

Liver transplantation is an established therapeutic modality for acute and chronic liver diseases. The improvement in outcome of liver transplantation is related to advances in patient selection criteria, organ preservation, operative techniques, perioperative care and efficacy of immunosuppressive agents. The resultant increase in demand for donor organs, widely estimated to be between 15 and 80 per million population, has led to the development of many strategies 1. These include the use of split-liver transplantation 2,3, living-related transplantation 4,5, domino transplantation 6, non-heart beating donor transplantation and the use of marginal donor organs. These techniques result in the procurement of an organ that potentially has suboptimal function compared with normal organs, either due to the reduction in mass or the functional quality of the tissue 7. Such organs are more susceptible to ischaemia, preservation and reperfusion injury (IPRI), which accounts for the majority of graft loss.

A further cause for donor organ damage is acute cellular rejection (ACR), which affects approximately 30–40% of patients after transplantation. Although ACR is effectively treated with modern immunosuppressive agents, this adds significantly to the postoperative morbidity and overall cost of transplantation. Recent evidence suggests a correlation between the severity of IPRI and ACR. Limiting the severity of IPRI may reduce the incidence of primary non-function (PNF) as well as ACR after liver transplantation. Hyperbaric oxygen (HBO) therapy has been shown to reduce the severity of IPRI as well as modulate both humoral and cellular immune response. This paper provides an overview of the relationship between IPRI and ACR and the potential therapeutic implications of HBO therapy in liver transplantation.

Pathophysiology of the interaction of IPRI and ACR

The cumulative effect of warm ischaemia and cold preservation is an energy deficiency within the Kupffer and endothelial cells leading to intracellular oedema 8. The early stages of reperfusion are characterized by a depletion of L-arginine, a precursor of the potent vasodilator nitric oxide (NO) and an elevation of endothlin-1 (ET-1). The latter stimulates hepatic stellate cell contraction, leading to sinusoidal narrowing, congestion and impaired flow. The low velocity within the sinusoids progressively promotes leucostasis, further hindering sinusoidal blood flow and exacerbating the hypoxic injury to the donor organ 9. Activation of Kupffer cells follows with the release of potent inflammatory cytokines such as tumour necrosis factor (TNF)-α and interleukin-1 (IL-1) as well as the production of reactive oxygen species (ROS) 10. The proinflammatory activity of these cytokines manifests through many pathways, including the increased expression of adhesion molecules. These further increase the leukocyte–endothelial cell interactions and the induction of IL-8 synthesis 11,12,13,14.

An additional effect of IPRI is its potential to stimulate host response in the form of acute cellular rejection. Many antigens that are not normally present on vascular endothelium and biliary epithelium are expressed in response to IPRI. This exposure results in an innate immune response that involves humoral components such as cytokines, complement and coagulation proteases as well as cellular components made up of macrophages and neutrophils. This initial response is followed by an adaptive immune response which leads to the activation of T and B cells acting specifically against exposed allo-antigens. Apart from antigen exposure, IPRI-induced tissue injury also results in the increased expression of adhesion molecules. These include P and E selectins as well as vascular cell adhesion molecule (VCAM). Chemokines such as macrophage chemoattractant protein 1 (MCP-1) and IL-8 lead to the mobilization of macrophages and neutrophils towards the injured tissue. Integrins are then expressed allowing leukocytes to adhere and transmigrate to the target tissues. IPRI also leads to the activation of the complement system by both the classical and alternate pathways. Activation of the latter pathway leads to the release of further IL-8 and platelet activating factors (PAFs), which aggravates the inflammatory response at the site of injury.

Accumulating evidence indicates an association between IPRI and the onset and severity of acute rejection. The significance of the innate immune response and its relationship to the adaptive immune response has become clearer. The initial signal activation for the innate response is mainly provided through dendritic cells. A number of stress proteins that are expressed as danger signals, namely the heat shock protein family (HSP), act as endogenous ligands to Toll-like receptors (TLRs) 15. Extracellular release of HSPs resulting from cell injury is thus able to act through TLRs to induce activation and maturation of antigen-presenting cells (APCs), particularly dendritic cells 16,17.Other studies have not only confirmed the role of ROS in inducing the endogenous ligands of TLRs but also the central role played by TLR4 to establish the full impact of hepatic IPRI 18.

Tissue injury that follows due to IPRI leads to activation of donor APCs within the graft and recipient APCs that enter the graft. Activated APCs then migrate to secondary lymphoid tissue where they interact with T cells and induce the allo response. Activation of APCs by TLRs also overcomes the suppression of T helper cells by CD4+ CD25+ T cells, allowing progression of the adaptive response 19. It may be hypothesized that reducing IPRI and minimizing the activation of TLRs may leave the APCs in an inactivated state, leading to antigen-specific tolerance 20. Absence of previous evidence supporting the relationship between IPRI and the severity of allo rejection have been attributed to the use of potent immunosuppressive agents 21. More recent clinical studies have shown a closer association of IPRI and ACR 22 and the important role played by the innate response 23,24.

The pathophysiology underlying acute cellular rejection is complex, multi-factorial and not fully understood. The host response to donor allo-antigens is centred on the major histocompatibility complex (MHC) antigens, while donor dendritic cells within the organ are thought to exert the most potent stimulus for the onset of acute rejection 25,26,27. Antigen recognition may occur by direct or indirect allo-recognition. Antigen specificity is maintained by T-cell receptor (TCR) complex while CD4 and CD8 act as co-receptors, binding to class II and class I MHC antigens, respectively. Once the TCR is engaged to an allo-antigen a series of intracellular events leads to the production of IL-2 28,29,30. Maximal IL-2 production by activated T cells is dependent on co-stimulation through a number of molecules, the most important of which seems to be CD28. The majority of CD4+ T cells and approximately 50% of CD8+ T cells express CD28 on their surface, the absence of which fails to induce an immune response from such T cells 31.

Activation of IL-2 and IL-2 receptors is followed by IL-3, IL-4, IL-5, IL-6 and interferon-γ. These activities lead to the transformation of the T cells to take on differentiated functions, such as immunoregulation and cytotoxicity. The exact roles of various cytokines remain unclear, although there is cross-regulation between the two types of CD4+ cells and some cytokines have been shown to have both immunostimulatory and immunomodulatory effects. Activated CD8+ T cells achieve destruction of target cells by the secretion of perforins and granzymes. Perforins create large pores in the target cell membrane leading to cytolysis, while granzymes activate apoptosis in the target population 32.

Mechanisms of the immunomodulatory effects of hyperbaric oxygen

The focus of HBO therapy has been directed towards its effect on IPRI. Researchers have also identified a significant immunomodulatory effect in animal studies by treating grafts in culture 33 as well as the recipients 34,35. These studies have demonstrated a suppressive effect on both humoral 36 and cell-mediated immunity 35. As the mechanisms of allograft rejection became better understood, the potential mechanisms of action of HBO were further investigated. Most of these studies were aimed at understanding the effects of HBO on the release of cytokines and modulation of leukocyte function and have been conducted via in vitro or in vivo studies in normal human volunteers.

The effect of both HBO and its components (normobaric hyperoxia and hyperbaric normoxaemia) on the release of TNF-α has been studied in detail 37. In vitro studies have shown that the release of TNF-α from unstimulated macrophages requires both the hyerbaric and hyperoxaemic components. In contrast, macrophages exposed to bacterial lipopolysaccharide (LPS) only require the hyperbaric component. Some investigators demonstrated an initial increase in TNF-α and IL-1 secretion in patients with Crohn's disease 38 and normal volunteers 39 during HBO therapy but a subsequent reduction with prolonged HBO therapy. Whether prolonged exposure to HBO inhibits production or if it was due to initial depletion of these cytokines is uncertain 38. The study of IFN-γ in human volunteers also further supports the immunomodulatory effect of HBO therapy. Production of IFN-γ by peripheral blood lymphocytes was significantly inhibited for 24 h following exposure to HBO therapy. Further studies have demonstrated this effect to be attributed predominantly to the hyperbaric effect rather than to hyperoxaemia 40. Animal studies have further supported the depressive effect on IFN-γ by hyperbaric therapy in the absence of hyperoxaemia 41. This has led to the conclusion that the hyperbaric effect acts on the cellular cytoskeleton, leading to a reduction in IFN-γ rather than the increase in oxygen tension.

Early studies reporting an immunosuppressive effect from HBO therapy implicated a defect in the macrophage–monocyte system 34,42. Recent studies investigating leukocyte function in response to HBO therapy have focused mainly on in vitro functions of neutrophils (PMNs) and lymphocytes. PMN function, particularly bactericidal ability, is related to the oxygen tension of its environment 43. HBO therapy was shown to not impede the ability of PMNs to phagocytose or develop the oxidative burst by Gadd et al., demonstrating that PMN phagocytosis was possibly independent of the oxygen tension 44. PMNs have the ability to inhibit T-lymphocyte function in their vicinity due to the release of myeloperoxidase and H2O245,46,47. This appears to be one of the mechanisms by which immunomodulation of T cells seem to occur after HBO therapy, as demonstrated by Gadd et al. where inhibition of T-lymphocyte proliferation was noted in response to HBO therapy. Despite this finding, there appear to be subpopulations of T lymphocytes that are resistant to the inhibitory effects of PMN-induced H2O246.

An in vitro study by MacKenzie et al. demonstrated the failure of peripheral blood monocytes treated with HBO therapy to activate lymphocytes when exposed to allogenic stimuli 48. In this study the investigators elegantly demonstrated the inhibition of cytotoxic T lymphocyte activation by HBO therapy. The main focus was shown to be due to its effect on CD4+ cells. The addition of the T helper 1 cytokine IFN-γ to the culture reversed this effect. The administration of anti-IL-4 and anti-IL-10 blocking antibodies which are T helper 2-related cytokines also restored normalcy to the T lymphocytes. However, when the CD4+ cells were removed prior to re-stimulation, the CTL activity was restored. Interestingly, the addition of IL-2 to cells treated by HBO did not stimulate CTL activity, suggesting that the mechanism of HBO immunomodulation was other than the creation of anergy. It is more likely, based on the above results, that HBO therapy acts by modulating the CD4+ cells which cross-regulate each other. An increase in the activity of T helper 2 cells and suppression of T helper 1 cells is likely to be one mechanism of its action. HBO therapy may also act through the modulation of the relative concentration of high affinity and low affinity IL-2 receptors on T lymphocytes. Binding to the high affinity receptors promotes proliferation while the reverse is true of the low affinity ones. HBO may lead to a change in the proportion of the two receptors leading to reduced T-cell proliferation 49,50.

HBO therapy has also been shown to alter cell surface MHC class I antigen expression. The binding of MHC class I specific antibodies showed reduced affinity after thyroid and human fetal pancreas allografts were cultured with exposure to HBO therapy 51,52,53,54. The expression of CD4 was slightly decreased when treated by HBO in culture, while that of CD28, CD80 and CD8 remained unchanged. Altered peptide ligands interacting with the TCR/CD3 complex may partially activate T cells leading to different signal transduction pathways 55. Such altered peptide ligands have the capacity to turn a potentially harmful T helper 1 response into a protective T helper 2 cell response 56,57. Recognition of this phenomenon and the understanding that such T cells may then undergo phenotypical changes has led to further insights into the potential action of HBO therapy. In addition to the potential direct immunomodulatory effects, HBO therapy may also significantly affect ACR by modulating the severity of IPRI. The effects of HBO therapy on IPRI have been studied in detail in many clinical and experimental situations.

Mechanisms of action of HBO on IPRI

HBO has been used with beneficial therapeutic effect in a wide variety of models of IPRI including myocardium, skeletal muscle, small intestine and the liver 58. Despite the theoretical adverse effects of generating ROS during reperfusion, the majority of in vivo studies have shown a reduction in the tissue injury when reperfusion is performed in the presence of HBO. HBO interacts with the effects of IPRI at numerous interfaces, which include the endothelium, neutrophils, mediators of inflammation, microvascular blood flow 59, lipid peroxidation and cellular energy levels.

The earliest demonstrable effect of HBO is on adhesion of neutrophils within the microcirculation. A significant reduction in rolling and adhesion on neutrophils is noted 59. Recently, Kihara et al. demonstrated a significant reduction in the accumulation of neutrophils and severity of IPRI in a rat model when HBO was delivered within 3 h. This resulted in a significant reduction in neutrophil accumulation, suppression of malondialdehyde (MDA) within the affected tissue and reduced congestion within sinusoids 60. Furthermore, HBO therapy resulted in the specific reduction in the expression of CD11/18 adhesion molecule which acts as a ligand to ICAM-1, thus leading to the above effect 61. Neutrophil adhesion also occurs through the up-regulation of selectins (P and E selectins) and the immunoglobulin superfamily (ICAM-1), all of which are expressed on endothelial surfaces. P-selectin is present in its pre-made form within cells and therefore up-regulates and binds very rapidly to its ligand CD15 on the neutrophils. E-selectin up-regulation requires new synthesis and is thus slower and binds to the sialyl-Lewis molecule on neutrophils. As previously mentioned, ICAM-1 molecules bind to CD18 on neutrophils and are induced by IL-1, TNF-α and bacterial LPS. In vitro experiments have shown that single doses of HBO have a significant effect on IPRI-injured endothelial cells and neutrophil adhesion, by reducing not only CD18 and CD11expression but also endothelial ICAM-1 expression 62,63. The latter effect is thought to act through the induction of endothelial nitric oxide synthase (eNOS) 64. This contradicts other studies which demonstrated increased expression of ICAM-1 with prolonged HBO therapy. However, the latter studies were conducted on normal tissue not exposed to the deleterious effects of IPRI, which may explain the different outcomes.

NO is a free radical which plays a major role in various aspects of IPRI. It is produced by three types of NOS. eNOS is constitutively expressed in endothelial cells and plays a role in vasodilatation of the microcirculation in response to hypoxia. Inducible NOS (iNOS) is produced by neutrophils and macrophages in response to a number of stimuli including IL-1, TNF-α and bacterial LPS. The NO produced by iNOS activation results in increased vascular leak and tissue injury, unlike the beneficial effects of eNOS. This toxicity is likely to be due to the greater production of superoxide radicals rather than toxicity from NO itself. Normobaric oxygen therapy has been shown to induce eNOS production while HBO therapy reduces serum nitrite/nitrate concentration in a rat shock model, where the likely source of nitrate is from iNOS. A number of in vitro and in vivo studies have subsequently confirmed the ability of HBO to selectively induce eNOS while inhibiting iNOS production 65.

Lipid peroxidation occurs in the presence of IPRI, due to the release of ROS leading to cell membrane and organelle injury. Exposure to normobaric 100% oxygen increases the degree of such injury during reperfusion. Although HBO has been noted to increase ROS, a number of studies on IPRI have confirmed the reduction of lipid peroxidation when exposed to HBO 66,67. The underlying mechanism of this effect remains ill understood. Similarly the interaction between HBO therapy and cytokine release remains contradictory. Spontaneous TNF-α production from normal tissue increases with HBO therapy in animal studies 68. However, initial HBO exposure inhibits TNF-α production following subsequent exposure to bacterial LPS. Other studies have shown that stimulus-induced synthesis of TNF-α and IL-1 is inhibited transiently. In an intestinal IPRI model HBO was shown to inhibit the production of TNF-α 69. Prolonged exposure to HBO reverses this effect leading to an increased production of IL-1 70. In other animal studies, once shock had been induced HBO tended to reduce the production of TNF-α, suggesting a possible role in the treatment of IPRI. Yet another mode of its effect may be through the inhibition of apoptosis. Specific inhibition of the apoptosis executor caspases 3 and 7 has been shown to reduce graft injury from IPRI 71. Studies on cerebral ischaemia have further confirmed the ability of HBO to inhibit apoptosis leading to a reduction in IPRI 72. Furthermore, a number of in vitro and in vivo studies have confirmed the ability of HBO therapy to stimulate hepatocyte regeneration 66,67,73.

In vivo studies of HBO therapy in transplantation

Despite extensive studies on the in vitro effects of HBO therapy on allografts, few studies have been conducted on its effects in vivo, most of which concentrate on the effect on ischaemia reperfusion and amelioration of tissue hypoxia. Several studies on bone and thyroid allografts have demonstrated accelerated union and immunomodulatory effect of HBO therapy 74. The main finding was a reduction in the expression of MHC class I antigen on the allografts 51,53. HBO therapy has a number of different roles in pancreatic islet cell transplantation, as it also improves the initial allograft hypoxia that is associated with graft failure. A more detailed study of islet allograft transplantation was preformed by Juang et al., who demonstrated a significant advantage in using HBO therapy 75. Better outcome was noted with pre-transplantation HBO therapy to the donor as well as twice-daily HBO therapy in the post transplantation period. The use of syngenic animals as donor and recipient indicates that the graft survival advantage in this study is related to non-immunological mechanisms 75. Rat fetal spinal transplantation into adult Wistar rats also demonstrated similar, non-immunogenic advantages from HBO therapy. These were due to a combination of reduction in tissue hypoxia and tissue oedema and an increase in microvascular proliferation at the graft interface leading to better graft survival 76.

A rat model of liver transplantation was used by Chen et al. to demonstrate the effect of a single episode of pre-transplantation HBO therapy 77. Post sinusoidal blood flow rate was increased and leukocyte adherence reduced. Tissue levels of MDA were reduced while tissue ATP levels were increased 77. However, no in vivo studies on liver transplantation have specifically investigated the immunomodulatory effect of HBO therapy.

Another potential role for HBO therapy is its use in acute liver failure. Liver failure induced by carbon tetrachloride (CCL4) poisoning in rats was shown to improve survival from 31% to 96% when applied immediately after ingestion of the substance. When applied 24 h later, the survival only improved from 36% to 50% 78. This effect seems to be specific against the actions of CCL4 and more study is required in its application for other causes of acute liver failure. HBO therapy seems to be protective against hepatotoxic agents that undergo reductive biotransformation through the cytochrome P450 system and aggravates the injury caused by those that undergo oxidative transformation 79.

Clinical experience of HBO therapy in liver transplantation

A handful of clinical studies have been published on the use of HBO therapy in relation to liver transplantation. All such studies relate to either its use in the post transplantation period for the management of hepatic artery thrombosis (HAT) 80,81 or its use in acute liver failure prior to transplantation 82,83. A single case report of a 3-year-old with fulminant hepatic failure did not show any advantage to HBO therapy. The clinical improvement was related to episodes of plasma exchange. However, HBO therapy was instituted only on day 18 and as such may have missed the window of effectiveness 82. Blum et al. reported four children with hepatic coma treated by exchange transfusion of whom three underwent HBO therapy 84. Exchange transfusion was noted to be clinically effective while HBO therapy did not contribute to an improvement in their neurological status 84. HBO therapy was used to treat early post transplantation HAT in children by Mazariegos et al. 81. This study investigated 375 patients who underwent liver transplantation between 1989 and 1998, of whom 31 developed HAT (7.5%) at a mean time of 8.2 days. Fourteen patients (between 1989 and 1994) were treated conventionally and served as the historical control group. Seventeen patients underwent HBO therapy within 24 h of HAT between 1994 and 1998. Eight patients in each group underwent re-transplantation, at a mean time of 12.7 days for the control group and 157 days for the HBO group. None of the re-transplantations in the HBO group was due to hepatic necrosis. There was one death in the HBO group and five in the control group, although the latter had a much longer follow-up. Hepatic arterial collaterals were noted on Doppler ultrasound at a mean time of 14 days in the HBO group while the control group demonstrated such development at a mean time of 31 days. This may in part explain the lack of hepatic necrosis in the HBO group 81.

HBO therapy in liver transplantation – discussion

Since its inception in 1662 and practical application in 1930, application of HBO therapy has continued to elicit controversy 85. Its use is well established in the treatment of decompression sickness and generally accepted for the treatment of carbon monoxide poisoning, gas embolism and radio-necrosis 86. Its remains controversial in the management of chronic wounds, refractory osteomyelitis, burns, ischaemic flaps and multiple sclerosis. The fact that HBO therapy increases the oxygen supply to the target tissue is unquestioned. This is more so in the liver with its dual blood supply. In addition, the effect of HBO therapy in ameliorating IPRI has been investigated to a great depth 87. However, its potential role in immunomodulation remains uncertain 37,88. Liver transplantation and HBO therapy have some common limitations. Both require extensive financial, medical and supportive infrastructure to function adequately. This simple commonality may allow both specialties to be situated within the same facility or within easy supportive range, thus providing an advantage in their clinical application. As the demand for liver transplantation grows and innovation forces the clinician to accept grafts of smaller volumes (split-liver, living-related OLTx) and those with less viable hepatocytes (marginal donor liver, steatotic livers) it becomes imperative to develop methods of minimizing donor organ injury and loss due to the IPRI and the process of rejection. In particular, the use of marginal donor organs is associated with an increased risk of PNF and primary poor function (PPF) of the graft, leading to longer stays in intensive care units, morbidity and mortality. PNF is a relative emergency often requiring urgent re-transplantation. Based on the available scientific evidence, HBO seems to have the capacity to influence the outcomes of liver transplantation at multiple levels.

The effect of HBO therapy on IPRI is the one that has been most investigated, defined and applied in clinical practice, although not in the field of liver transplantation. HBO therapy seems to reduce the effect of IPRI by various mechanisms depending on where it is applied, including exposure to the donor organ prior to re-implantation as well as the recipient both before and after transplantation 89,90. It has the potential to reduce IPRI and improve graft function in the post transplantation period. It may be of particular relevance when marginal or reduced size donor organs are used. HBO therapy also has a role to play in the management of post transplantation complications. Clinical evidence suggests that HBO therapy may delay or even eliminate hepatic necrosis following HAT 81. It has also been used in acute fulminant hepatic failure 82,84. In both situations it may provide badly needed time for an urgent donor organ to be made available. In the latter situation it may act as a hepatic assist device in conjunction with haemofiltration and plasma exchange procedures 83.

Another important role for which there is much animal and in vitro evidence but no clinical studies is its role in immunomodulation. If the laboratory studies were proven in the clinical situation, it may provide the opportunity to minimize drug-based immunosuppression, particularly in the first week following transplantation. The effect of minimizing IPRI is also known to reduce immune activation as it prevents the up-regulation of MHC class II molecules in the donor organ 23,91. The effect of HBO therapy in ultimately inhibiting the transcription of immunomodulatory cytokines, especially IL-2 along with its ability to reduce the expression of MHC class I antigens, has potential implications for a further, low dose regime of immunosuppression. Finally, the proven effect on pancreatic islet cell uptake and graft survival may allow a similar improvement in the newly developing technique of hepatocyte transplantation.

Most strategies aimed at minimizing IPRI act through very specific pathways. This includes the use of preservation solutions, hepatic microcirculatory expansion 92, continuous ex vivo hepatic perfusion, inactivation of Kupffer cells 93 and inhibition of complement activation 94,95. Similarly the mainstay of controlling ACR remains with immunosuppressive therapies targeting specific channels such as inhibition of calcineurin 96, use of antimetabolites 97 or more recently the use of interleukin receptor blockade 98. One of the major and unique potential advantages of HBO therapy is that it affects the processes of IPRI and ACR through numerous cellular and molecular mechanisms. In addition it may play potential roles as a liver support adjunct for fulminant hepatic failure, post transplantation graft dysfunction and acute hepatic artery thrombosis. The difficulty of moving critically ill patients in and out of the chambers and the high infrastructure requirements have been major obstacles to the delivery of HBO therapy in the clinical setting. However, the technology relating to the delivery of HBO therapy has advanced significantly in the past decades and overcome most of such impediments. The older generation of single chamber units were small with limited space and cumbersome access which increased the difficulty of managing complex patients. Modern multi-chamber units, especially the rectangular hyperbaric systems, are large by comparison, with adequate internal space for multiple patients, active medical intervention and easy drive-in access for bed-bound critically ill patients and supporting equipment. Almost all liver transplantation activity occurs at major referral centres which have the capacity, staff and infrastructure required for the functioning of an HBO chamber and the safe delivery of HBO therapy to critically ill patients. Thus, HBO therapy, often described as ‘a treatment looking for a disease’ has the potential to intervene and influence the outcomes of liver transplantation through multiple avenues. However, more research is required on the influence of HBO therapy in liver transplantation, specifically in its effect on immune response and liver support prior to clinical applicability.

References

  • 1.Modan B, Shpilberg O, Baruch Y, Sikuler E, Anis E, Ashur Y, et al. The need for liver transplantation: a nationwide estimate based on consensus review. Lancet. 1995;346:660–2. doi: 10.1016/s0140-6736(95)92279-2. [DOI] [PubMed] [Google Scholar]
  • 2.Malago M, Hertl M, Testa G, Rogiers X. Split liver transplantation: future use of scarce donor organs? World J Surg. 2002;26:275–82. doi: 10.1007/s00268-001-0216-z. [DOI] [PubMed] [Google Scholar]
  • 3.Azoulay D, Astarcioglu I, Bismuth H, Castaing D, Majno P, Adam R, et al. Split-liver transplantation: the Paul Brousse policy. Ann Surg. 1996;224:737–48. doi: 10.1097/00000658-199612000-00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Emre S. Living donor liver transplantation: a critical review. Transplant Proc. 2001;33:3456–7. doi: 10.1016/s0041-1345(01)02489-7. [DOI] [PubMed] [Google Scholar]
  • 5.Fan ST, Lo CM, Liu CL, Yong BH, Chan JK, Ng IO. Safety of donors in live donor liver transplantation using right lobe grafts. Arch Surg. 2000;135:336–40. doi: 10.1001/archsurg.135.3.336. [DOI] [PubMed] [Google Scholar]
  • 6.Azoulay D, Samuel D, Castaing D, Adam R, Adams D, Said G, et al. Domino liver transplants for metabolic disorders: experience with familial amyloidotic polyneuropathy. J Am Coll Surg. 1999;189:584–93. doi: 10.1016/s1072-7515(99)00208-2. [DOI] [PubMed] [Google Scholar]
  • 7.Mor E, Klintmalm GB, Gonwa TA, Solomon H, Holman MJ, Gibbs JF, et al. The use of marginal donors for liver transplantation. A retrospective study of 365 liver donors. Transplantation. 1992;53:383–6. doi: 10.1097/00007890-199202010-00022. [DOI] [PubMed] [Google Scholar]
  • 8.Vollmar B, Glasz J, Leiderer R, Post S, Menger MD. Hepatic microcirculatory perfusion failure is a determinant of liver dysfunction in warm ischemia-reperfusion. Am J Pathol. 1994;145:1421–31. [PMC free article] [PubMed] [Google Scholar]
  • 9.Vollmar B, Richter S, Menger MD. Leukocyte stasis in hepatic sinusoids. Am J Physiol. 1996;270(Pt 1):G798–G803. doi: 10.1152/ajpgi.1996.270.5.G798. [DOI] [PubMed] [Google Scholar]
  • 10.Colletti LM, Kunkel SL, Walz A, Burdick MD, Kunkel RG, Wilke CA, et al. The role of cytokine networks in the local liver injury following hepatic ischemia/reperfusion in the rat. Hepatology. 1996;23:506–14. doi: 10.1002/hep.510230315. [DOI] [PubMed] [Google Scholar]
  • 11.Yoshidome H, Kato A, Edwards MJ, Lentsch AB. Interleukin-10 suppresses hepatic ischemia/reperfusion injury in mice: implications of a central role for nuclear factor kappaB. Hepatology. 1999;30:203–8. doi: 10.1002/hep.510300120. [DOI] [PubMed] [Google Scholar]
  • 12.Yoshidome H, Kato A, Edwards MJ, Lentsch AB. Interleukin-10 inhibits pulmonary NF-kappaB activation and lung injury induced by hepatic ischemia-reperfusion. Am J Physiol. 1999;277(5 Pt 1):L919–923. doi: 10.1152/ajplung.1999.277.5.L919. [DOI] [PubMed] [Google Scholar]
  • 13.Todo S, Zhu Y, Zhang S, Jin MB, Ishizaki N, Tanaka H, et al. Attenuation of ischemic liver injury by augmentation of endogenous adenosine. Transplantation. 1997;63:213–17. doi: 10.1097/00007890-199701270-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zhang S, Jin MB, Zhu Y, Ishizaki N, Tanaka H, Subbotin VM, et al. Effect of endogenous adenosine augmentation on ischemia and reperfusion injury to the liver. Transplant Proc. 1997;29:1336–7. doi: 10.1016/s0041-1345(96)00584-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Schnare M, Barton GM, Holt AC, Takeda K, Akira S, Medzhitov R. Toll-like receptors control activation of adaptive immune responses. Nat Immunol. 2001;2:947–50. doi: 10.1038/ni712. [DOI] [PubMed] [Google Scholar]
  • 16.Ohashi K, Burkart V, Flohe S, Kolb H. Cutting edge: heat shock protein 60 is a putative endogenous ligand of the toll-like receptor-4 complex. J Immunol. 2000;164:558–61. doi: 10.4049/jimmunol.164.2.558. [DOI] [PubMed] [Google Scholar]
  • 17.Kosieradzki M, Kuczynska J, Piwowarska J, Wegrowicz-Rebandel I, Kwiatkowski A, Lisik W, et al. Prognostic significance of free radicals: mediated injury occurring in the kidney donor. Transplantation. 2003;75:1221–7. doi: 10.1097/01.TP.0000065282.46425.87. [DOI] [PubMed] [Google Scholar]
  • 18.Wu HS, Zhang JX, Wang L, Tian Y, Wang H, Rotstein O. Toll-like receptor 4 involvement in hepatic ischemia/reperfusion injury in mice. Hepatobiliary Pancreat Dis Int. 2004;3:250–3. [PubMed] [Google Scholar]
  • 19.Pasare C, Medzhitov R. Toll pathway-dependent blockade of CD4 + CD25+ T cell-mediated suppression by dendritic cells. Science. 2003;299:1033–6. doi: 10.1126/science.1078231. [DOI] [PubMed] [Google Scholar]
  • 20.Coates PT, Duncan FJ, Colvin BL, Wang Z, Zahorchak AF, Shufesky WJ, et al. In vivo-mobilized kidney dendritic cells are functionally immature, subvert alloreactive T-cell responses, and prolong organ allograft survival. Transplantation. 2004;77:1080–9. doi: 10.1097/01.tp.0000122183.60680.c9. [DOI] [PubMed] [Google Scholar]
  • 21.Katz E, Mor E, Schwartz ME, Theise N, Patel T, Miller CM. Preservation injury in clinical liver transplantation: incidence and effect on rejection and survival. Clin Transpl. 1994;8:492–6. [PubMed] [Google Scholar]
  • 22.Bjorkbacka H, Kunjathoor VV, Moore KJ, Koehn S, Ordija CM, Lee MA, et al. Reduced atherosclerosis in MyD88-null mice links elevated serum cholesterol levels to activation of innate immunity signaling pathways. Nat Med. 2004;10:416–21. doi: 10.1038/nm1008. [DOI] [PubMed] [Google Scholar]
  • 23.Kouwenhoven EA, de Bruin RW, Bajema IM, Marquet RL, Ijzermans JN. Cold ischemia augments allogeneic-mediated injury in rat kidney allografts. Kidney Int. 2001;59:1142–8. doi: 10.1046/j.1523-1755.2001.0590031142.x. [DOI] [PubMed] [Google Scholar]
  • 24.Lu CY, Penfield JG, Kielar ML, Vazquez MA, Jeyarajah DR. Hypothesis: is renal allograft rejection initiated by the response to injury sustained during the transplant process? Kidney Int. 1999;55:2157–68. doi: 10.1046/j.1523-1755.1999.00491.x. [DOI] [PubMed] [Google Scholar]
  • 25.Zinkernagel RM, Doherty PC. Immunological surveillance against altered self components by sensitised T lymphocytes in lymphocytic choriomeningitis. Nature. 1974;251:547–8. doi: 10.1038/251547a0. [DOI] [PubMed] [Google Scholar]
  • 26.Zinkernagel RM, Doherty PC. Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneic system. Nature. 1974;248:701–2. doi: 10.1038/248701a0. [DOI] [PubMed] [Google Scholar]
  • 27.Pober JS, Orosz CG, Rose ML, Savage CO. Can graft endothelial cells initiate a host anti-graft immune response? Transplantation. 1996;61:343–9. doi: 10.1097/00007890-199602150-00001. [DOI] [PubMed] [Google Scholar]
  • 28.Weiss A, Littman DR. Signal transduction by lymphocyte antigen receptors. Cell. 1994;76:263–74. doi: 10.1016/0092-8674(94)90334-4. [DOI] [PubMed] [Google Scholar]
  • 29.Izquierdo M, Leevers SJ, Williams DH, Marshall CJ, Weiss A, Cantrell D. The role of protein kinase C in the regulation of extracellular signal-regulated kinase by the T cell antigen receptor. Eur J Immunol. 1994;24:2462–8. doi: 10.1002/eji.1830241031. [DOI] [PubMed] [Google Scholar]
  • 30.Clipstone NA, Crabtree GR. Calcineurin is a key signaling enzyme in T lymphocyte activation and the target of the immunosuppressive drugs cyclosporin A and FK506. Ann N Y Acad Sci. 1993;696:20–30. doi: 10.1111/j.1749-6632.1993.tb17138.x. [DOI] [PubMed] [Google Scholar]
  • 31.Linsley PS, Ledbetter JA. The role of the CD28 receptor during T cell responses to antigen. Annu Rev Immunol. 1993;11:191–212. doi: 10.1146/annurev.iy.11.040193.001203. [DOI] [PubMed] [Google Scholar]
  • 32.Lipman ML, Stevens AC, Strom TB. Heightened intragraft CTL gene expression in acutely rejecting renal allografts. J Immunol. 1994;152:5120–7. [PubMed] [Google Scholar]
  • 33.Talmage DW, Dart GA. Effect of oxygen pressure during culture on survival of mouse thyroid allografts. Science. 1978;200:1066–7. doi: 10.1126/science.653355. [DOI] [PubMed] [Google Scholar]
  • 34.Warren J, Sacksteder MR, Thuning CA. Oxygen immunosuppression: modification of experimental allergic encephalomyelitis in rodents. J Immunol. 1978;121:315–20. [PubMed] [Google Scholar]
  • 35.Jacobs BB, Thuning CA, Sacksteder MR, Warren J. Extended skin allograft survival in mice during prolonged exposure to hyperbaric oxygen. Transplantation. 1979;28:70–2. doi: 10.1097/00007890-197907000-00017. [DOI] [PubMed] [Google Scholar]
  • 36.Saito K, Tanaka Y, Ota T, Eto S, Yamashita U. Suppressive effect of hyperbaric oxygenation on immune responses of normal and autoimmune mice. Clin Exp Immunol. 1991;86:322–7. doi: 10.1111/j.1365-2249.1991.tb05817.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.van den Blink B, van der Kleij AJ, Versteeg HH, Peppelenbosch MP. Immunomodulatory effect of oxygen and pressure. Comp Biochem Physiol A Mol Integr Physiol. 2002;132:193–7. doi: 10.1016/s1095-6433(01)00547-5. [DOI] [PubMed] [Google Scholar]
  • 38.Weisz G, Lavy A, Adir Y, Melamed Y, Rubin D, Eidelman S, et al. Modification of in vivo and in vitro TNF-alpha, IL-1, and IL-6 secretion by circulating monocytes during hyperbaric oxygen treatment in patients with perianal Crohn's disease. J Clin Immunol. 1997;17:154–9. doi: 10.1023/a:1027378532003. [DOI] [PubMed] [Google Scholar]
  • 39.Rocco M, Antonelli M, Letizia V, Alampi D, Spadetta G, Passariello M, et al. Lipid peroxidation, circulating cytokine and endothelin 1 levels in healthy volunteers undergoing hyperbaric oxygenation. Minerva Anestesiol. 2001;67:393–400. [PubMed] [Google Scholar]
  • 40.Granowitz EV, Skulsky EJ, Benson RM, Wright J, Garb JL, Cohen ER, et al. Exposure to increased pressure or hyperbaric oxygen suppresses interferon-gamma secretion in whole blood cultures of healthy humans. Undersea Hyperb Med. 2002;29:216–25. [PubMed] [Google Scholar]
  • 41.Huang KY, Gordon FB. Production of interferon in mice: effect of altered gaseous environments. Appl Microbiol. 1968;16:1551–6. doi: 10.1128/am.16.10.1551-1556.1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hansborough JF, Piacentine JG, Eiseman B. Immunosuppression by hyperbaric oxygen. Surgery. 1980;87:662–7. [PubMed] [Google Scholar]
  • 43.Hohn DC, MacKay RD, Halliday B, Hunt TK. Effect of O2 tension on microbicidal function of leukocytes in wounds and in vitro. Surg Forum. 1967;27:18–20. [PubMed] [Google Scholar]
  • 44.Gadd MA, McClellan DS, Neuman TS, Hansborough JF. Effect of hyperbaric oxygen on murine neutrophil and T-lymphocyte functions. Crit Care Med. 1990;18:974–9. doi: 10.1097/00003246-199009000-00014. [DOI] [PubMed] [Google Scholar]
  • 45.Zoschke DC, Staite ND. Suppression of human lymphocyte proliferation by activated neutrophils or H2O2: surviving cells have an altered T helper/T suppressor ratio and an increased resistance to secondary oxidant exposure. Clin Immunol Immunopathol. 1987;42:160–70. doi: 10.1016/0090-1229(87)90003-1. [DOI] [PubMed] [Google Scholar]
  • 46.Staite ND, Messner RP, Zoschke DC. Inhibition of human T lymphocyte E rosette formation by neutrophils and hydrogen peroxide. Differential sensitivity between helper and suppressor T lymphocytes. J Immunol. 1987;139:2424–30. [PubMed] [Google Scholar]
  • 47.Gougerot-Pocidalo MA, Fay M, Pocidalo JJ. In vivo normobaric oxygen exposure depresses spleen cell in vitro Con A response. Effects of 2-mercaptoethanol and peritoneal cells. Clin Exp Immunol. 1984;58:428–35. [PMC free article] [PubMed] [Google Scholar]
  • 48.MacKenzie DA, Sollinger HA, Hullett DA. Role of CD4 regulatory T cells in hyperbaric oxygen mediated immune non-responsiveness. Hum Immunol. 2000;61:1320–31. doi: 10.1016/s0198-8859(00)00214-7. [DOI] [PubMed] [Google Scholar]
  • 49.Gaulton GN, Markmann JF. Regulation of lymphocyte growth by antagonists of interleukin-2 or its cellular receptor. Immunol Res. 1988;7:113–35. doi: 10.1007/BF02918096. [DOI] [PubMed] [Google Scholar]
  • 50.Smith KA. Interleukin-2: inception, impact, and implications. Science. 1988;240:1169–76. doi: 10.1126/science.3131876. [DOI] [PubMed] [Google Scholar]
  • 51.Hullett DA, Landry AS, Leonard DK, Sollinger HW. Enhancement of thyroid allograft survival following organ culture. Tranplantation. 1989;47:24–7. doi: 10.1097/00007890-198901000-00006. [DOI] [PubMed] [Google Scholar]
  • 52.Hullett DA, Landry AS, Leonard DK, Sollinger HW. Improved human fetal pancreatic tissue survival following hyperbaric oxygen culture. Transplant Proc. 1989;21:2659–60. [PubMed] [Google Scholar]
  • 53.Hullett DA, Landry AS, Leonard DK, Sollinger HW. Regulation of class I antigens responsible for prolonged graft survival after organ culture. Transplant Proc. 1989;21:240–1. [PubMed] [Google Scholar]
  • 54.MacKenzie DA, Sollinger HW, Hullett DA. Decreased immunogenicity of human fetal pancreas allografts following hyperbaric oxygen culture. Transplant Proc. 2003;35:1499–502. doi: 10.1016/s0041-1345(03)00362-2. [DOI] [PubMed] [Google Scholar]
  • 55.Sloan-Lancaster J, Allen PM. Altered peptide ligand-induced partial T cell activation: molecular mechanisms and role in T cell biology. Annu Rev Immunol. 1996;14:1–27. doi: 10.1146/annurev.immunol.14.1.1. [DOI] [PubMed] [Google Scholar]
  • 56.Sloan-Lancaster J, Steinberg TH, Allen PM. Selective loss of the calcium ion signalling pathway in T cells maturing towards a T helper 2 phenotype. J Immunol. 1997;159:1160–8. [PubMed] [Google Scholar]
  • 57.Nicholson LB, Greer JM, Sobel RA, Lees MB, Kuchroo VK. An altered peptide ligand mediates immune deviation and prevents autoimmune encephalomyelitis. Immunity. 1995;3:397–405. doi: 10.1016/1074-7613(95)90169-8. [DOI] [PubMed] [Google Scholar]
  • 58.Piper HM, Garcia-Dorado D, Ovize M. A fresh look at reperfusion injury. Cardiovasc Res. 1998;38:291–300. doi: 10.1016/s0008-6363(98)00033-9. [DOI] [PubMed] [Google Scholar]
  • 59.Zamboni WA, Roth AC, Russell RC, Graham B, Suchy H, Kucan JO. Morphologic analysis of the microcirculation during reperfusion of ischemic skeletal muscle and the effects of hyperbaric oxygen. Plast Reconstr Surg. 1993;91:1110–23. doi: 10.1097/00006534-199305000-00022. [DOI] [PubMed] [Google Scholar]
  • 60.Kihara K, Ueno S, Sakoda M, Aikou T. Effects of hyperbaric oxygen exposure on experimental hepatic ischemia reperfusion injury: relationship between its timing and neutrophil sequestration. Liver Transpl. 2005;11:1574–80. doi: 10.1002/lt.20533. [DOI] [PubMed] [Google Scholar]
  • 61.Thom S. Functional inhibition of leucocyte [beta] integrins by hyperbaric oxygen in carbon monoxide mediated brain injury in rats. Toxicol Appl Pharm. 1993;123:248–56. doi: 10.1006/taap.1993.1243. [DOI] [PubMed] [Google Scholar]
  • 62.Larson JL, Stephenson LL, Zamboni WA. Effect of hyperbaric oxygen on neutrophil CD18 expression. Plast Reconstr Surg. 2000;105:1375–81. doi: 10.1097/00006534-200004040-00017. [DOI] [PubMed] [Google Scholar]
  • 63.Labrouche S, Javorschi S, Leroy D, Gbikpi-Benissan G, Freyburger G. Influence of hyperbaric oxygen on leukocyte functions and haemostasis in normal volunteer divers. Thromb Res. 1999;96:309–15. doi: 10.1016/s0049-3848(99)00107-3. [DOI] [PubMed] [Google Scholar]
  • 64.Buras JA, Stahl GL, Svoboda KK, Reenstra WR. Hyperbaric oxygen downregulates ICAM-1 expression induced by hypoxia and hypoglycemia: the role of NOS. Am J Physiol Cell Physiol. 2000;278:C292–C302. doi: 10.1152/ajpcell.2000.278.2.C292. [DOI] [PubMed] [Google Scholar]
  • 65.Sakoda M, Ueno S, Kihara K, Arikawa K, Dogomori H, Nuruki K, et al. A potential role of hyperbaric oxygen exposure through intestinal nuclear factor-kappaB. Crit Care Med. 2004;32:1722–9. doi: 10.1097/01.ccm.0000132898.27101.6c. [DOI] [PubMed] [Google Scholar]
  • 66.Ozden TA, Uzun H, Bohloli M, Toklu AS, Paksoy M, Simsek G, et al. The effect of hyperbaric oxygen treatment on oxidant and antioxidant levels during liver regeneration in rats. Tohoku J Exp Med. 2004;203:253–65. doi: 10.1620/tjem.203.253. [DOI] [PubMed] [Google Scholar]
  • 67.Kurir TT, Markotic A, Katalinic V, Bozanic D, Cikes V, Zemunik T, et al. Effect of hyperbaric oxygenation on the regeneration of the liver after partial hepatectomy in rats. Braz J Med Biol Res. 2004;37:1231–7. doi: 10.1590/s0100-879x2004000800014. [DOI] [PubMed] [Google Scholar]
  • 68.Lahat N, Bitterman H, Yaniv N, Kinarty A, Bitterman N. Exposure to hyperbaric oxygen induces tumour necrosis factor-alpha (TNF-alpha) secretion from rat macrophages. Clin Exp Immunol. 1995;102:655–9. doi: 10.1111/j.1365-2249.1995.tb03867.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yang ZJ, Bosco G, Montante A, Ou XI, Camporesi EM. Hyperbaric O2 reduces intestinal ischemia-reperfusion-induced TNF-alpha production and lung neutrophil sequestration. Eur J Appl Physiol. 2001;85:96–103. doi: 10.1007/s004210100391. [DOI] [PubMed] [Google Scholar]
  • 70.Benson RM, Minter LM, Osborne BA, Granowitz EV. Hyperbaric oxygen inhibits stimulus induced proinflammatory cytokine synthesis by human blood derived monocyte macrophages. Clin Exp Immunol. 2003;134:57–62. doi: 10.1046/j.1365-2249.2003.02248.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Mueller TH, Kienle K, Beham A, Geissler EK, Jauch KW, Rentsch M. Caspase 3 inhibition improves survival and reduces early graft injury after ischemia and reperfusion in rat liver transplantation. Transplantation. 2004;78:1267–73. doi: 10.1097/01.tp.0000141095.06273.10. [DOI] [PubMed] [Google Scholar]
  • 72.Yin D, Zhou C, Kusaka I, Calvert JW, Parent AD, Nanda A, et al. Inhibition of apoptosis by hyperbaric oxygen in a rat focal cerebral ischemic model. J Cereb Blood Flow Metab. 2003;23:855–64. doi: 10.1097/01.WCB.0000073946.29308.55. [DOI] [PubMed] [Google Scholar]
  • 73.Mizuguchi T, Oshima H, Imaizumi H, Kohara H, Kawamoto M, Nobuoka T, et al. Hyperbaric oxygen stimulates cell proliferation and normalizes multidrug resistance protein-2 protein localization in primary rat hepatocytes. Wound Repair Regen. 2005;13:551–7. doi: 10.1111/j.1524-475X.2005.00077.x. [DOI] [PubMed] [Google Scholar]
  • 74.Sawai T, Niimi A, Takahashi H, Ueda M. Histologic study of the effect of hyperbaric oxygen therapy on autogenous free bone grafts. J Oral Maxillofac Surg. 1996;54:975–81. doi: 10.1016/s0278-2391(96)90396-1. [DOI] [PubMed] [Google Scholar]
  • 75.Juang J-H, Hsu BR-S, Kuo C-H, Ueng SW-N. Beneficial effects of hyperbaric oxygen therapy on islet transplantation. Cell Transplant. 2002;11:95–101. doi: 10.3727/096020198389825. [DOI] [PubMed] [Google Scholar]
  • 76.Kutlay M, Colak A, Demircan N, Akin ON, Kibici K, Dundar K, et al. Effect of hyperbaric oxygen therapy on fetal spinal grafts: an experimental study. Undersea Hyperb Med. 2000;27:205–13. [PubMed] [Google Scholar]
  • 77.Chen MF, Chen HM, Ueng SW, Shyr MH. Hyperbaric oxygen pretreatment attenuates hepatic reperfusion injury. Liver. 1998;18:110–16. doi: 10.1111/j.1600-0676.1998.tb00135.x. [DOI] [PubMed] [Google Scholar]
  • 78.Burk RF, Reiter R, Lane JM. Hyperbaric oxygen protection against carbon tetrachloride hepatotoxicity in the rat. Association with altered metabolism. Gastroenterology. 1986;90:812–18. doi: 10.1016/0016-5085(86)90856-5. [DOI] [PubMed] [Google Scholar]
  • 79.Marzella L, Muhvich K, Myers RA. Effect of hyperoxia on liver necrosis induced by hepatotoxins. Virchows Arch B Cell Pathol Incl Mol Pathol. 1986;51:497–507. doi: 10.1007/BF02899055. [DOI] [PubMed] [Google Scholar]
  • 80.Dubost T, Goubaux B, Duhalde M, Raucoules-Aime M, Wolkiewiez J, Gugenheim J. Use of hyperbaric oxygen for hepatic artery thrombosis following adult orthotopic liver transplantation. Eur J Anaesthesiol. 2002;19:223–4. doi: 10.1017/s0265021502210388. [DOI] [PubMed] [Google Scholar]
  • 81.Mazariegos GV, O'Toole K, Mieles LA, Dvorchik I, Meza MP, Briassoulis G, et al. Hyperbaric oxygen therapy for hepatic artery thrombosis after liver transplantation in children. Liver Transpl Surg. 1999;5:429–6. doi: 10.1002/lt.500050518. [DOI] [PubMed] [Google Scholar]
  • 82.Ponikvar R, Buturovic J, Cizman M, Mekjavic I, Kandus A, Premru V, et al. Hyperbaric oxygenation, plasma exchange, and hemodialysis for treatment of acute liver failure in a 3-year-old child. Artif Organs. 1998;22:952–7. doi: 10.1046/j.1525-1594.1998.06239.x. [DOI] [PubMed] [Google Scholar]
  • 83.Mizuguchi K, Nose Y. Hyperbaric oxygenation: is it an hepatic assist device? Artif Organs. 1994;18:545–6. doi: 10.1111/j.1525-1594.1994.tb03376.x. [DOI] [PubMed] [Google Scholar]
  • 84.Blum D, Viart P, Szliwowski HB, Thys JP, Dubois J. Exchange transfusion and hyperbaric oxygen in the treatment of children with acute hepatic failure. Helv Paediatr Acta. 1972;27:425–36. [PubMed] [Google Scholar]
  • 85.Gill A, Bell CN. Hyperbaric oxygen: its uses, mechanisms of action and outcomes. Q J Med. 2004;97:385–95. doi: 10.1093/qjmed/hch074. [DOI] [PubMed] [Google Scholar]
  • 86.Pasquier D, Hoelscher T, Schmutz J, Dische S, Mathieu D, Baumann M, et al. Hyperbaric oxygen therapy in the treatment of radio-induced lesions in normal tissues: a literature review. Radiother Oncol. 2004;72:1–13. doi: 10.1016/j.radonc.2004.04.005. [DOI] [PubMed] [Google Scholar]
  • 87.Buras J. Basic mechanisms of hyperbaric oxygen in the treatment of ischemia-reperfusion injury. Int Anaesthesiol Clin. 2000;38:91–109. doi: 10.1097/00004311-200001000-00007. [DOI] [PubMed] [Google Scholar]
  • 88.Hansborough JF, Eiseman B. Immunosuppression with hyperbaric oxygen. Surg Forum. 1979;30:313–15. [PubMed] [Google Scholar]
  • 89.Hong JP, Kwon H, Chung YK, Jung SH. The effect of hyperbaric oxygen on ischaemia-reperfusion injury. Ann Plast Surg. 2003;51:478–87. doi: 10.1097/01.sap.0000095651.05156.0f. [DOI] [PubMed] [Google Scholar]
  • 90.Kolski JM, Mazolewski PJ, Stephenson LL, Texter J, Grigoriev VE, Zamboni WA. Effect of hyperbaric oxygen therapy on testicular ischemia-reperfusion injury. J Urol. 1998;160:601–4. [PubMed] [Google Scholar]
  • 91.Serracio-Inglott F, Habib NA, Mathie RT. Hepatic ischaemia reperfusion. Am J Surg. 2001;181:160–6. doi: 10.1016/s0002-9610(00)00573-0. [DOI] [PubMed] [Google Scholar]
  • 92.Astarcioglu H, Karademir S, Atila K, Sagol O, Kocdor H, Coker A, et al. The effects of vascular bed expansion in steatotic rat liver graft viability. Transpl Int. 2004;17:188–94. doi: 10.1007/s00147-004-0692-9. [DOI] [PubMed] [Google Scholar]
  • 93.Frankenberg MV, Weimann J, Fritz S, Fiedler J, Mehrabi A, Buchler MW, et al. Gadolinium chloride-induced improvement of postischemic hepatic perfusion after warm ischemia is associated with reduced hepatic endothelin secretion. Transpl Int. 2005;18:429–36. doi: 10.1111/j.1432-2277.2004.00058.x. [DOI] [PubMed] [Google Scholar]
  • 94.Bellamy MC, Gedney JA, Buglass H, Gooi JH, Leeds Liver Group. Complement membrane attack complex and hemodynamic changes during human orthotopic liver transplantation. Liver Transpl 2004;10:273–8. [DOI] [PubMed] [Google Scholar]
  • 95.Arumugam TV, Woodruff TM, Stocks SZ, Proctor LM, Pollitt S, Shiels IA, et al. Protective effect of a human C5a receptor antagonist against hepatic ischaemia-reperfusion injury in rats. J Hepatol. 2004;40:934–41. doi: 10.1016/j.jhep.2004.02.017. [DOI] [PubMed] [Google Scholar]
  • 96.Levy GA. Neoral is superior to FK 506 in liver transplantation. Transplant Proc. 1998;30:1812–15. doi: 10.1016/s0041-1345(98)00442-4. [DOI] [PubMed] [Google Scholar]
  • 97.Raimondo ML, Dagher L, Papatheodoridis GV, Rolando N, Patch DW, Davidson BR, et al. Long-term mycophenolate mofetil monotherapy in combination with calcineurin inhibitors for chronic renal dysfunction after liver transplantation. Transplantation. 2003;75:186–90. doi: 10.1097/01.TP.0000041702.31262.CD. [DOI] [PubMed] [Google Scholar]
  • 98.Calmus Y, Scheele JR, Gonzalez-Pinto I, Jaurrieta EJ, Klar E, Pageaux GP, et al. Immunoprophylaxis with basiliximab, a chimeric anti-interleukin-2 receptor monoclonal antibody, in combination with azathioprine-containing triple therapy in liver transplant recipients. Liver Transpl. 2002;8:123–31. doi: 10.1053/jlts.2002.30882. [DOI] [PubMed] [Google Scholar]

Articles from HPB : The Official Journal of the International Hepato Pancreato Biliary Association are provided here courtesy of Elsevier

RESOURCES