Skip to main content
Erschienen in: European Surgery 2/2016

Open Access 01.04.2016 | Review

Ischemia reperfusion-facilitated sinusoidal endothelial cell injury in liver transplantation and the resulting impact of extravasated platelet aggregation

verfasst von: T. Miyashita, MD, PhD, S. Nakanuma, MD, PhD, A.K. Ahmed, I. Makino, MD, PhD, H. Hayashi, MD, PhD, K. Oyama, MD, PhD, H. Nakagawara, MD, PhD, H. Tajima, MD, PhD, H. Takamura, MD, PhD, I. Ninomiya, MD, PhD, S. Fushida, MD, PhD, J.W. Harmon, MD, T. Ohta, MD, PhD

Erschienen in: European Surgery | Ausgabe 2/2016

Summary

Background

The exact sequence of events leading to ultimate hepatocellular damage following ischemia/reperfusion (I/R) is incompletely understood. In this article, we review a mechanism of organ dysfunction after hepatic I/R or immunosuppressive treatment, in addition to the potential of liver sinusoidal endothelial cell (LSEC) protection and antiplatelet treatment for the suppression of hepatocellular damage.

Methods

A review of the literature, utilizing PubMed-NCBI, was used to provide information on the components necessary for the development of hepatocellular damage following I/R.

Results

It is well-established that LSECs damage following hepatic I/R or immunosuppressive treatment followed by extravasated platelet aggregation (EPA) is the root cause of organ dysfunction in liver transplantation. We have classified three phases, from LSECs damage to organ dysfunction, utilizing the predicted pathogenic mechanism of sinusoidal obstruction syndrome. The first phase is detachment of LSECs and sinusoidal wall destruction after LSECs injury by hepatic I/R or immunosuppressive treatment. The second phase is EPA, accomplished by sinusoidal wall destruction. The various growth factors, including thromboxane A2, serotonin, transforming growth factor-beta and plasminogen activator inhibitor-1, released by EPA in the Disse’s space of zone three, induce portal hypertension and the progression of hepatic fibrosis. The third phase is organ dysfunction following portal hypertension, hepatic fibrosis, and suppressed liver regeneration through various growth factors secreted by EPA.

Conclusion

We suggest that EPA in the space of Disse, initiated by LSECs damage due to hepatic I/R or immunosuppressive treatment, and activated platelets may primarily contribute to liver damage in liver transplantation. Endothelial protective therapy or antiplatelet treatment may be useful in the treatment of hepatic I/R following EPA.

Introduction

Ischemia/reperfusion (I/R) injury to the liver is a major complication of hemorrhagic shock, liver resection, and transplantation [1]. Although the sequence of events that leads to extent of hepatocellular damage after hepatic I/R is incompletely understood, liver sinusoidal endothelial cells (LSECs) may loosen from their tetherings to the space of Disse in zone 3 or even detach completely [2].
Hepatic I/R is considered as a biphasic phenomenon. Cellular damage due to hypoxia and a lack of biomechanical stimulus is exacerbated upon the restoration of oxygen delivery and shear stress [3]. Although the initial ischemic insult to the liver is tolerable, this first step triggers essential molecules in the induction of the more devastating reperfusion injury. Early phases of reperfusion are characterized pathologically by endothelial cell swelling, vasoconstriction, neutrophil entrapment, and platelet aggregation within the sinusoids–resulting in failure of the microcirculation [4]. Simultaneously, nitric oxide (NO) levels are markedly reduced and there is an imbalance between endothelin-1 and NO production from NO synthase (NOS). This leads to vasoconstriction of the sinusoids [5]. As such, vasoconstriction of the sinusoids gives rise to narrowing of the sinusoidal lumen with consequential decreased leukocyte velocity. The frequency of leukocyte-endothelial cell contact is elevated, promoting leukostasis. Flow is hindered in the sinusoidal network of the hepatic microcirculation due to stagnant leukocytes, unable to completely occlude the sinusoidal lumen [6]. Microcirculatory failure leads to aggravated and prolonged ischemia. Hypoxic regions of the liver heighten the degree of necrosis, Kupffer cell activation, and induce further cytokine and reactive oxygen species release. This creates a cycle of excessive inflammatory response, reactive oxygen and nitrogen species production, and further oxidative tissue injury [7].
LSECs, which lack an organized basal membrane, form the vascular wall of the hepatic sinusoid. Fenestrations penetrate the cytoplasm of these flattened cells forming clusters, called sieve plates, which render the hepatic microvascular endothelium discontinuous [8]. LSECs play an integral protective role in maintaining vascular homeostasis, inflammation, vascular tone, and toxicant clearance. Thus, the preservation of a healthy LSEC phenotype is fundamental to minimize any type of liver injury [3]. Damage to the endothelium, following hepatic I/R injury, is apparent within LSECs and hepatocytes, indicated by the deposition of fibrinogen and erythrocyte congestion resulting in enlarged sinusoids. Sloughed LSECs, red cells, and stellate cells embolize downstream, leading to venous occlusions that progress to disrupt the normal liver architecture and achieve centrilobular necrosis [9]. In the late phases of the disease, fibrosis and occlusion of the terminal venules develop, leading to hepatic failure and possibly death [10].
Sinusoidal obstruction syndrome (SOS), previously known as veno-occlusive disease, commences with sustained LSECs injury, resulting in bleeding in the space of Disse and centrilobular hemorrhagic necrosis. The fundamental cause, moreover, is damage around the centrilobular area, including the sinusoid, by acute cellular rejection, antibody-mediated rejection or hepatic I/R injury [11]. SOS is a life-threatening syndrome that results from sinusoidal congestion and is characterized by hepatomegaly, ascites, portal hypertension, weight gain and jaundice [12].
Platelets play an important role in hepatocellular damage. Furthermore, platelets have been suggested to be involved in the inflammatory response of hepatic I/R injury in various organs [13]. They are able to roll and adhere to postreperfusion endothelium in a P-selectin-dependent manner [14, 15]. Platelets accumulate in the postischemic microvasculature early after reperfusion via P-selectin-ligand interactions. Platelet recruitment and subsequent activation might play an important role in the pathogenesis of hepatic I/R injury [14]. Platelet aggregation also correlated with reperfusion injury, thrombocytopenia and early graft dysfunction in liver transplantation [16]. As such, persistent thrombocytopenia after reperfusion is an unfavorable indicator for early liver graft dysfunction [17].
We previously reported that platelet aggregation in the space of Disse along with the sinusoid and platelets phagocytosis by hepatocytes were observed in the allograft tissue of a living donor liver transplantation recipient with thrombocytopenia, who encountered a complication of SOS [18]. Therefore, LSECs damage after hepatic I/R or immunosuppressive treatment followed by extravasated platelet aggregation (EPA) is the root cause of organ dysfunction in liver transplantation.
This manuscript will review the role of platelet aggregation and possible prevention for hepatocyte injury in hepatic I/R. The PubMed database was utilized in searching for published literature in this area.

Platelets and hepatic ischemia/reperfusion injury

Whereas many of the mechanisms underlying the hepatic I/R-induced inflammatory response still remain unknown, growing evidence suggests a role for platelets in the pathogenesis of postischemia reperfusion injury [14]. Ischemia leads to the accumulation and activation of platelets within vascular beds early after reperfusion [19].
We reported that CD42b expression, as a platelet marker, was apparent along with the sinusoid in Zone 3 but not in Zone 1 in liver allograft tissue (Fig. 1a).
Remarkably, superior adenosine diphosphate (ADP)-dependent aggregative levels of platelets prior to graft reperfusion were demonstrated to be positively correlated with serum markers for hepatocellular reperfusion damage. Schulte et al. reported that a certain increase in the ADP-dependent functional status of platelets in the pediatric recipients of liver grafts may result in a critical promotion of platelet adherence and activation in liver graft LSECs following reperfusion [16, 20]. Platelet accumulation in the postischemic microvasculature might significantly contribute to the manifestation of hepatic I/R injury [14].
Extracellular nucleotides are released in a regulated manner either by platelets, a variety of vascular and hepatic cells in response to inflammatory stress, through cellular swelling, or with exocytosis. Levels of extracellular nucleotides are, in turn, regulated by CD39 (ectonucleoside triphosphate diphosphohydrolase-1/ENTPD1), the dominant vascular ectonucleotidase. CD39, expressed only on the luminal surface of proliferating or activated LSECs and absent in hepatocytes [21, 22], hydrolyzes the terminal phosphate of adenosine triphosphate(ATP) and ADP in an enzymatic cascade that generates adenosine monophosphate(AMP) [23]. Under normal conditions, in the absence of stressors such as hypoxia/ischemia, high shear stress, and triggers for inflammation (cytokines/chemokines), CD39 helps to maintain a homeostatic vascular environment, maintain blood fluidity and inhibit inflammation [24]. ADP, one of the most potent signals for platelet aggregation, as well as proinflammatory signals such as ATP, are released into the extracellular environment in the presence of adverse conditions, such as hepatic I/R. When this occurs, the extracellular concentration of ADP and ATP increases markedly [25]. When platelets contact collagen, they produce ADP, which further promotes platelet aggregation.
Platelets release potent proinflammatory chemokines and modulate leukocyte function [26]. Activated platelets release growth factors, such as thromboxane (TX) A2, serotonin, vascular endothelial growth factor (VEGF)-A, transforming growth factor (TGF)-β and plasminogen activator inhibitor (PAI)-1. TXA2 is a strong vasoactive metabolite of arachidonic acid with powerful proaggregatory and proinflammatory properties. TXA2 is a vasoconstrictor that increases portal venous resistance [27] and causes portal hypertension. Serotonin is well-known released from platelets on damage to the blood vessels walls. It acts as a potent vasoconstrictor. Although VEGF-A acts as a vasodilator under ordinary circumstances, it acts, paradoxically, as a vasoconstrictor in patients with endothelial failure [28]. Bevacizumab, an antibody against VEGF-A, protects against liver injury associated with SOS[29]. PAI-1 suppresses fibrinolysis and the progression to fibrosis in the tissue microenvironment. In addition, PAI-1 acts as a negative regulator of hepatocyte proliferation by inhibiting urokinase-type plasminogen activator (u-PA), which activates hepatocyte growth factor [30, 31]. TGF-β, a major antiproliferative factor for hepatocytes, stimulates collagen synthesis through activated hepatic stellate cells (HSCs) [32].
LSEC apoptosis also play an important role in hepatic I/R injury during liver transplantation [33]. Platelet sequestration occurs after transplantation with possibly deleterious effects. In an isolated perfused rat liver model, single platelets were adherent to sinusoidal lining without morphological or dynamic evidence of impairment in microcirculation with increase in the number of apoptotic LSECs. Platelets cause LSECs apoptosis upon reperfusion of liver grafts. These results indicate that the prevention of adhesion plays a protective role [15].

Extravasated platelet aggregation and organ dysfunction

The liver can be subjected to three forms of ischemia, namely cold (or hypothermic), warm (or normothermic), and rewarming [34]. In transplanted liver ischemia, disruption of the endothelial wall leads to leukocyte [35] and platelet adhesion [15, 36], which induces microcirculatory disturbances [37]. Transplanted liver damage is slightly different from liver resection, due to the duration of sustained ischemia.
The subendothelial space of Disse, located between the hepatocytes and the sinusoid, contains HSCs (myofibroblasts) and a network of reticular fibers holding the hepatocytes together. Large amounts of albumin and other plasma proteins enter the space of Disse, pass through hepatocyte junctions, and form ascites containing a high concentration of protein and a low serum to ascites albumin gradient. In normal liver, the space of Disse contains a matrix of basement-membrane constituents that is not electron dense [38]; in injured tissue this may be replaced by matrix filled with fibril-forming collagens and fibronectin [39].
In the normal liver, collagen type III are concentrated in the portal tracts and around terminal hepatic veins, with occasional bundles located between hepatocytes and endothelial cells in the space of Disse. Hepatic ischemia causes endothelial cell activation following HSCs activation, increasing hepatic parenchymal and portal tract fibrosis in ischemic liver tissues [40]. Following a fibrogenic stimulus, HSCs undergo a complex process of activation in which they become transformed from quiescent to activated myofibroblast-like cells [4143]. Activated HSCs are the primary cell type responsible for the production of collagen I. This subendothelial accumulation of collagen, termed “capillarization” of the sinusoid, is associated with clinical liver disease [44]. Fibrosis may also impede the rapid exchange of solutes between the sinusoidal space and hepatocytes [45].
Following the destruction of endothelial wall and resulting LSECs damage caused by hepatic I/R or immunosuppressive treatment, platelets enter the space of Disse (Fig. 1b) and aggregate by activated HSCs (Fig. 2). Furthermore, platelets produce platelet-derived growth factor TGF-β to activate HSCs and promote fibrosis [46]. Therefore, liver fibrosis may be caused by EPA in this pathway (Fig. 3).
The production of collagen I may be associated with increased serum alanine transaminase level and be utilized as a marker for hepatic I/R injury.

Possible treatment

No standard method for treating hepatic dysfunction by hepatic I/R has yet been established. Systemic anticoagulation and thrombolytic therapies have been tested extensively [47]. Defibrotide, a polydeoxyribonucleic acid, was recently shown to have a promising response rate in patients with severe SOS [48]. Conditional to the extent of LSECs damage and EPA in the space of Disse, prophylactic administration of endothelial protective and antiplatelet agents may be effective prior to the development of irreversible damage.
Additionally, antiapoptosis induced by ishchemic preconditioning can improve liver function, as well as protect LSECs [49]. Ischemic preconditioning directly protected hepatocytes after warm I/R, not via suppression of alterations in sinusoidal cells, as is present in cold hepatic I/R injury [50].

Reinforcement and protection of LSECs

Hyperglycemia, indicative of diabetes mellitus, is a major risk factor for endothelial dysfunction and vascular complications. In recent years, significant advances have been made in understanding endothelial cell dysfunction, triggered by high glucose concentration [51]. In order to prevent graft injury during perioperatively tight glycemic control, it is necessary to reduce the risk of microangiopathic organ injury by preventing endothelial cell injury [52]. The artificial pancreas might have beneficial effects in liver transplant recipients due to the hepatoprotective effects of insulin, including the stimulation of regeneration and endothelial cell protection [53]. Additionally, insulin has an antiinflammatory effect through the suppression of inflammatory cytokines (e.g, nuclear factor kappa B) [54]. To ascertain this, we applied a closed-loop glycemic control system with an artificial pancreas (STG-55, NIKKISO CO., LTD., Tokyo, Japan) to the liver transplant recipient.
In obese patients with type 2 diabetes, insulin delivery to and insulin-dependent glucose uptake by skeletal muscle are delayed and impaired. Therefore, it is pivotal to impair insulin resistance for endothelial cells. Kubota et al. [55] demonstrated that impaired insulin signaling in endothelial cells, due to reduced insulin receptor substrate two expression and insulin-induced eNOS phosphorylation, causes attenuation of insulin-induced capillary recruitment and insulin delivery, reducing glucose uptake by skeletal muscle. The use of agents such as Beraprost, a stable prostacyclin analogue (PGI2), capable of improving insulin resistance and resulting vascular endothelial function, may ultimately contribute to increasing the life expectancy of patients with peripheral artery disease [56]. To improve insulin resistance, we administer Beraprost sodium during the perioperative state.

Antiplatelet agents therapy

Phosphodiesterase 3 (PDE3) inhibitors Cilostazol and Milrinone may be appropriate, owing to their antiplatelet properties, ability to increase tolerance to hepatic I/R injury [57], and induction of immune tolerance via an enhanced regulatory T-cell response [58]. Furthermore, PDE3 inhibition regulates endothelial CD39 at a posttranslational level [25]. To reaffirm such a treatment, a few experimental studies in rat models demonstrated that PDE3 inhibitors protected SOS [59], and attenuated graft injury using an orthotopic liver transplant model [60].
In clinical practice, we administered a PDE3 inhibitor with the use of local infusion therapy to living donor liver transplant recipients or patients with small remnant liver volume after a hepatectomy such as in tri-segmentectomy, with favorable outcomes[18].
Beraprost also prevents platelet aggregation by increasing cyclic AMP to effectively reduce the amount of TXA2, which has coagulant properties and is made by platelets. Beraprost is also administered as an antiplatelet agent.

Conclusion

We suggest that EPA in the space of Disse, initiated by LSECs damage due to hepatic I/R or immunosuppressive treatment, and activated platelets may primarily contribute to liver damage in liver transplantation. Endothelial protective therapy or antiplatelet treatment may be useful in the treatment of hepatic I/R following EPA.
Conflict of interest
The authors declares that there is no conflict of interests regarding the publication of this paper.
Funding
The authors received no financial support for the research, author-ship, and/or publication of this article.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Clarke CN, Kuboki S, Tevar A, Lentsch AB, Edwards M. Cxc chemokines play a critical role in liver injury, recovery, and regeneration. Am J Surg. 2009;198:415–9.CrossRefPubMedPubMedCentral Clarke CN, Kuboki S, Tevar A, Lentsch AB, Edwards M. Cxc chemokines play a critical role in liver injury, recovery, and regeneration. Am J Surg. 2009;198:415–9.CrossRefPubMedPubMedCentral
2.
3.
Zurück zum Zitat Peralta C, Jiménez-Castro MB, Gracia-Sancho J. Hepatic ischemia and reperfusion injury: effects on the liver sinusoidal milieu. J Hepatol. 2013;59:1094–106.CrossRefPubMed Peralta C, Jiménez-Castro MB, Gracia-Sancho J. Hepatic ischemia and reperfusion injury: effects on the liver sinusoidal milieu. J Hepatol. 2013;59:1094–106.CrossRefPubMed
4.
Zurück zum Zitat Serracino-Inglott F, Habib NA, Mathie RT. Hepatic ischemia-reperfusion injury. Am J Surg. 2001;181:160–6.CrossRefPubMed Serracino-Inglott F, Habib NA, Mathie RT. Hepatic ischemia-reperfusion injury. Am J Surg. 2001;181:160–6.CrossRefPubMed
5.
Zurück zum Zitat Marzi I, Takei Y, Rücker M, Kawano S, Fusamoto H, Walcher F, Kamada T. Endothelin-1 is involved in hepatic sinusoidal vasoconstriction after ischemia and reperfusion. Transpl Int. 1994;7(Suppl 1):S503–S6.CrossRefPubMed Marzi I, Takei Y, Rücker M, Kawano S, Fusamoto H, Walcher F, Kamada T. Endothelin-1 is involved in hepatic sinusoidal vasoconstriction after ischemia and reperfusion. Transpl Int. 1994;7(Suppl 1):S503–S6.CrossRefPubMed
6.
Zurück zum Zitat Vollmar B, Richter S, Menger MD. Leukocyte stasis in hepatic sinusoids. Am J Physiol. 1996;270:G798–803.PubMed Vollmar B, Richter S, Menger MD. Leukocyte stasis in hepatic sinusoids. Am J Physiol. 1996;270:G798–803.PubMed
7.
Zurück zum Zitat Nastos C, Kalimeris K, Papoutsidakis N, Tasoulis MK, Lykoudis PM, Theodoraki K, Nastou D, Smyrniotis V, Arkadopoulos N. Global consequences of liver ischemia/reperfusion injury. Oxid Med Cell Longev. 2014;2014:906965.CrossRefPubMedPubMedCentral Nastos C, Kalimeris K, Papoutsidakis N, Tasoulis MK, Lykoudis PM, Theodoraki K, Nastou D, Smyrniotis V, Arkadopoulos N. Global consequences of liver ischemia/reperfusion injury. Oxid Med Cell Longev. 2014;2014:906965.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Wisse E. An ultrastructural characterization of the endothelial cell in the rat liver sinusoid under normal and various experimental conditions, as a contribution to the distinction between endothelial and kupffer cells. J Ultrastruct Res. 1972;38:528–62.CrossRefPubMed Wisse E. An ultrastructural characterization of the endothelial cell in the rat liver sinusoid under normal and various experimental conditions, as a contribution to the distinction between endothelial and kupffer cells. J Ultrastruct Res. 1972;38:528–62.CrossRefPubMed
9.
Zurück zum Zitat Shulman HM, Fisher LB, Schoch HG, Henne KW, McDonald GB. Veno-occlusive disease of the liver after marrow transplantation: histological correlates of clinical signs and symptoms. Hepatology. 1994;19:1171–81.CrossRefPubMed Shulman HM, Fisher LB, Schoch HG, Henne KW, McDonald GB. Veno-occlusive disease of the liver after marrow transplantation: histological correlates of clinical signs and symptoms. Hepatology. 1994;19:1171–81.CrossRefPubMed
11.
Zurück zum Zitat Takamura H, Nakanuma S, Hayashi H, Tajima H, Kakinoki K, Kitahara M, Sakai S, Makino I, Nakagawara H, Miyashita T, Okamoto K, Nakamura K, Oyama K, Inokuchi M, Ninomiya I, Kitagawa H, Fushida S, Fujimura T, Onishi I, Kayahara M, Tani T, Arai K, Yamashita T, Kitamura H, Ikeda H, Kaneko S, Nakanuma Y, Matsui O, Ohta T. Severe veno-occlusive disease/sinusoidal obstruction syndrome after deceased-donor and living-donor liver transplantation. Transplant Proc. 2014;46:3523–35.CrossRefPubMed Takamura H, Nakanuma S, Hayashi H, Tajima H, Kakinoki K, Kitahara M, Sakai S, Makino I, Nakagawara H, Miyashita T, Okamoto K, Nakamura K, Oyama K, Inokuchi M, Ninomiya I, Kitagawa H, Fushida S, Fujimura T, Onishi I, Kayahara M, Tani T, Arai K, Yamashita T, Kitamura H, Ikeda H, Kaneko S, Nakanuma Y, Matsui O, Ohta T. Severe veno-occlusive disease/sinusoidal obstruction syndrome after deceased-donor and living-donor liver transplantation. Transplant Proc. 2014;46:3523–35.CrossRefPubMed
12.
Zurück zum Zitat Campos-Varela I, Castells L, Dopazo C, Pérez-Lafuente M, Allende H, Len O, Llopart L, Vargas V, Charco R. Transjugular intrahepatic portosystemic shunt for the treatment of sinusoidal obstruction syndrome in a liver transplant recipient and review of the literature. Liver Transpl. 2012;18:201–5.CrossRefPubMed Campos-Varela I, Castells L, Dopazo C, Pérez-Lafuente M, Allende H, Len O, Llopart L, Vargas V, Charco R. Transjugular intrahepatic portosystemic shunt for the treatment of sinusoidal obstruction syndrome in a liver transplant recipient and review of the literature. Liver Transpl. 2012;18:201–5.CrossRefPubMed
13.
14.
Zurück zum Zitat Massberg S, Enders G, Leiderer R, Eisenmenger S, Vestweber D, Krombach F, Messmer K. Platelet-endothelial cell interactions during ischemia/reperfusion: the role of P-selectin. Blood. 1998;92:507–15.PubMed Massberg S, Enders G, Leiderer R, Eisenmenger S, Vestweber D, Krombach F, Messmer K. Platelet-endothelial cell interactions during ischemia/reperfusion: the role of P-selectin. Blood. 1998;92:507–15.PubMed
15.
Zurück zum Zitat Sindram D, Porte RJ, Hoffman MR, Bentley RC, Clavien PA. Platelets induce sinusoidal endothelial cell apoptosis upon reperfusion of the cold ischemic rat liver. Gastroenterology. 2000;118:183–91.CrossRefPubMed Sindram D, Porte RJ, Hoffman MR, Bentley RC, Clavien PA. Platelets induce sinusoidal endothelial cell apoptosis upon reperfusion of the cold ischemic rat liver. Gastroenterology. 2000;118:183–91.CrossRefPubMed
16.
Zurück zum Zitat Schulte am Esch J, Akyildiz A, Tustas RY, Ganschow R, Schmelzle M, Krieg A, Robson SC, Topp SA, Rogiers X, Knoefel WT, Fischer L. Adp-dependent platelet function prior to and in the early course of pediatric liver transplantation and persisting thrombocytopenia are positively correlated with ischemia/reperfusion injury. Transpl Int. 2010;23:745–52.CrossRefPubMed Schulte am Esch J, Akyildiz A, Tustas RY, Ganschow R, Schmelzle M, Krieg A, Robson SC, Topp SA, Rogiers X, Knoefel WT, Fischer L. Adp-dependent platelet function prior to and in the early course of pediatric liver transplantation and persisting thrombocytopenia are positively correlated with ischemia/reperfusion injury. Transpl Int. 2010;23:745–52.CrossRefPubMed
17.
Zurück zum Zitat McCaughan GW, Herkes R, Powers B, Rickard K, Gallagher ND, Thompson JF, Sheil AG. Thrombocytopenia post liver transplantation. Correlations with pre-operative platelet count, blood transfusion requirements, allograft function and outcome. J Hepatol. 1992;16:16–22.CrossRefPubMed McCaughan GW, Herkes R, Powers B, Rickard K, Gallagher ND, Thompson JF, Sheil AG. Thrombocytopenia post liver transplantation. Correlations with pre-operative platelet count, blood transfusion requirements, allograft function and outcome. J Hepatol. 1992;16:16–22.CrossRefPubMed
18.
Zurück zum Zitat Nakanuma S, Miyashita T, Hayashi H, Tajima H, Takamura H, Tsukada T, Okamoto K, Sakai S, Makino I, Kinoshita J, Nakamura K, Oyama K, Inokuchi M, Nakagawara H, Ninomiya I, Kitagawa H, Fushida S, Fujimura T, Ohta T. Extravasated platelet aggregation in liver zone 3 may correlate with the progression of sinusoidal obstruction syndrome following living donor liver transplantation: a case report. Exp Ther Med. 2015;9:1119–24.PubMedPubMedCentral Nakanuma S, Miyashita T, Hayashi H, Tajima H, Takamura H, Tsukada T, Okamoto K, Sakai S, Makino I, Kinoshita J, Nakamura K, Oyama K, Inokuchi M, Nakagawara H, Ninomiya I, Kitagawa H, Fushida S, Fujimura T, Ohta T. Extravasated platelet aggregation in liver zone 3 may correlate with the progression of sinusoidal obstruction syndrome following living donor liver transplantation: a case report. Exp Ther Med. 2015;9:1119–24.PubMedPubMedCentral
19.
Zurück zum Zitat Chintala MS, Bernardino V, Chiu PJ. Cyclic gmp but not cyclic amp prevents renal platelet accumulation after ischemia-reperfusion in anesthetized rats. J Pharmacol Exp Ther. 1994;271:1203–8.PubMed Chintala MS, Bernardino V, Chiu PJ. Cyclic gmp but not cyclic amp prevents renal platelet accumulation after ischemia-reperfusion in anesthetized rats. J Pharmacol Exp Ther. 1994;271:1203–8.PubMed
20.
Zurück zum Zitat Plevak DJ, Halma GA, Forstrom LA, Dewanjee MK, O’Connor MK, Moore SB, Krom RA, Rettke SR. Thrombocytopenia after liver transplantation. Transplant Proc. 1988;20:630–3.PubMed Plevak DJ, Halma GA, Forstrom LA, Dewanjee MK, O’Connor MK, Moore SB, Krom RA, Rettke SR. Thrombocytopenia after liver transplantation. Transplant Proc. 1988;20:630–3.PubMed
21.
Zurück zum Zitat Enjyoji K, Sévigny J, Lin Y, Frenette PS, Christie PD, Esch JS, Imai M, Edelberg JM, Rayburn H, Lech M, Beeler DL, Csizmadia E, Wagner DD, Robson SC, Rosenberg RD. Targeted disruption of cd39/ATP diphosphohydrolase results in disordered hemostasis and thromboregulation. Nat Med. 1999;5:1010–7.CrossRefPubMed Enjyoji K, Sévigny J, Lin Y, Frenette PS, Christie PD, Esch JS, Imai M, Edelberg JM, Rayburn H, Lech M, Beeler DL, Csizmadia E, Wagner DD, Robson SC, Rosenberg RD. Targeted disruption of cd39/ATP diphosphohydrolase results in disordered hemostasis and thromboregulation. Nat Med. 1999;5:1010–7.CrossRefPubMed
22.
Zurück zum Zitat Goepfert C, Sundberg C, Sévigny J, Enjyoji K, Hoshi T, Csizmadia E, Robson S. Disordered cellular migration and angiogenesis in cd39-null mice. Circulation. 2001;104:3109–15.CrossRefPubMed Goepfert C, Sundberg C, Sévigny J, Enjyoji K, Hoshi T, Csizmadia E, Robson S. Disordered cellular migration and angiogenesis in cd39-null mice. Circulation. 2001;104:3109–15.CrossRefPubMed
23.
Zurück zum Zitat Koziak K, Sévigny J, Robson SC, Siegel JB, Kaczmarek E. Analysis of CD39/ATP diphosphohydrolase (ATPDase) expression in endothelial cells, platelets and leukocytes. Thromb Haemost. 1999;82:1538–44.PubMed Koziak K, Sévigny J, Robson SC, Siegel JB, Kaczmarek E. Analysis of CD39/ATP diphosphohydrolase (ATPDase) expression in endothelial cells, platelets and leukocytes. Thromb Haemost. 1999;82:1538–44.PubMed
24.
Zurück zum Zitat Eltzschig HK, Köhler D, Eckle T, Kong T, Robson SC, Colgan SP. Central role of Sp1-regulated CD39 in hypoxia/ischemia protection. Blood. 2009;113:224–32.CrossRefPubMedPubMedCentral Eltzschig HK, Köhler D, Eckle T, Kong T, Robson SC, Colgan SP. Central role of Sp1-regulated CD39 in hypoxia/ischemia protection. Blood. 2009;113:224–32.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Baek AE, Kanthi Y, Sutton NR, Liao H, Pinsky DJ. Regulation of ecto-apyrase cd39 (ENTPD1) expression by phosphodiesterase III (PDE3). FASEB J. 2013;27:4419–28.CrossRefPubMedPubMedCentral Baek AE, Kanthi Y, Sutton NR, Liao H, Pinsky DJ. Regulation of ecto-apyrase cd39 (ENTPD1) expression by phosphodiesterase III (PDE3). FASEB J. 2013;27:4419–28.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Ghasemzadeh M, Hosseini E. Platelet-leukocyte crosstalk: linking proinflammatory responses to procoagulant state. Thromb Res. 2013;131:191–7.CrossRefPubMed Ghasemzadeh M, Hosseini E. Platelet-leukocyte crosstalk: linking proinflammatory responses to procoagulant state. Thromb Res. 2013;131:191–7.CrossRefPubMed
27.
Zurück zum Zitat Ruan Z, Shibamoto T, Shimo T, Koizumi T, Tsuchida H, Kurata Y, Ogura T, Kubo K. Effects of platelet-activating factor and thromboxane A2 on isolated perfused guinea pig liver. Prostaglandins Other Lipid Mediat. 2004;73:73–85.CrossRefPubMed Ruan Z, Shibamoto T, Shimo T, Koizumi T, Tsuchida H, Kurata Y, Ogura T, Kubo K. Effects of platelet-activating factor and thromboxane A2 on isolated perfused guinea pig liver. Prostaglandins Other Lipid Mediat. 2004;73:73–85.CrossRefPubMed
28.
Zurück zum Zitat Parenti A, Brogelli L, Filippi S, Donnini S, Ledda F. Effect of hypoxia and endothelial loss on vascular smooth muscle cell responsiveness to VEGF-A: role of flt-1/VEGF-receptor-1. Cardiovasc Res. 2002;55:201–12.CrossRefPubMed Parenti A, Brogelli L, Filippi S, Donnini S, Ledda F. Effect of hypoxia and endothelial loss on vascular smooth muscle cell responsiveness to VEGF-A: role of flt-1/VEGF-receptor-1. Cardiovasc Res. 2002;55:201–12.CrossRefPubMed
29.
Zurück zum Zitat Ribero D, Wang H, Donadon M, Zorzi D, Thomas MB, Eng C, Chang DZ, Curley SA, Abdalla EK, Ellis LM, Vauthey JN. Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer. 2007;110:2761–7.CrossRefPubMed Ribero D, Wang H, Donadon M, Zorzi D, Thomas MB, Eng C, Chang DZ, Curley SA, Abdalla EK, Ellis LM, Vauthey JN. Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer. 2007;110:2761–7.CrossRefPubMed
30.
Zurück zum Zitat Mars WM, Zarnegar R, Michalopoulos GK. Activation of hepatocyte growth factor by the plasminogen activators uPA and tPA. Am J Pathol. 1993;143:949–58.PubMedPubMedCentral Mars WM, Zarnegar R, Michalopoulos GK. Activation of hepatocyte growth factor by the plasminogen activators uPA and tPA. Am J Pathol. 1993;143:949–58.PubMedPubMedCentral
31.
Zurück zum Zitat Watanabe K, Togo S, Takahashi T, Matsuyama R, Yamamoto H, Shimizu T, Makino H, Matsuo K, Morioka D, Kubota T, Nagashima Y, Shimada H. PAI-1 plays an important role in liver failure after excessive hepatectomy in the rat. J Surg Res. 2007;143:13–9.CrossRefPubMed Watanabe K, Togo S, Takahashi T, Matsuyama R, Yamamoto H, Shimizu T, Makino H, Matsuo K, Morioka D, Kubota T, Nagashima Y, Shimada H. PAI-1 plays an important role in liver failure after excessive hepatectomy in the rat. J Surg Res. 2007;143:13–9.CrossRefPubMed
32.
Zurück zum Zitat Ueda S, Yamanoi A, Hishikawa Y, Dhar DK, Tachibana M, Nagasue N. Transforming growth factor-beta1 released from the spleen exerts a growth inhibitory effect on liver regeneration in rats. Lab Invest. 2003;83:1595–603.CrossRefPubMed Ueda S, Yamanoi A, Hishikawa Y, Dhar DK, Tachibana M, Nagasue N. Transforming growth factor-beta1 released from the spleen exerts a growth inhibitory effect on liver regeneration in rats. Lab Invest. 2003;83:1595–603.CrossRefPubMed
33.
Zurück zum Zitat Cursio R, Miele C, Filippa N, Van Obberghen E, Gugenheim J. Liver HIF-1 alpha induction precedes apoptosis following normothermic ischemia-reperfusion in rats. Transplant Proc. 2008;40:2042–5.CrossRefPubMed Cursio R, Miele C, Filippa N, Van Obberghen E, Gugenheim J. Liver HIF-1 alpha induction precedes apoptosis following normothermic ischemia-reperfusion in rats. Transplant Proc. 2008;40:2042–5.CrossRefPubMed
34.
Zurück zum Zitat Selzner N, Rudiger H, Graf R, Clavien PA. Protective strategies against ischemic injury of the liver. Gastroenterology. 2003;125:917–36.CrossRefPubMed Selzner N, Rudiger H, Graf R, Clavien PA. Protective strategies against ischemic injury of the liver. Gastroenterology. 2003;125:917–36.CrossRefPubMed
35.
Zurück zum Zitat Takei Y, Marzi I, Gao WS, Gores GJ, Lemasters JJ, Thurman RG. Leukocyte adhesion and cell death following orthotopic liver transplantation in the rat. Transplantation. 1991;51:959–65.CrossRefPubMed Takei Y, Marzi I, Gao WS, Gores GJ, Lemasters JJ, Thurman RG. Leukocyte adhesion and cell death following orthotopic liver transplantation in the rat. Transplantation. 1991;51:959–65.CrossRefPubMed
36.
Zurück zum Zitat Cywes R, Packham MA, Tietze L, Sanabria JR, Harvey PR, Phillips MJ, Strasberg SM. Role of platelets in hepatic allograft preservation injury in the rat. Hepatology. 1993;18:635–47.CrossRefPubMed Cywes R, Packham MA, Tietze L, Sanabria JR, Harvey PR, Phillips MJ, Strasberg SM. Role of platelets in hepatic allograft preservation injury in the rat. Hepatology. 1993;18:635–47.CrossRefPubMed
37.
Zurück zum Zitat Marzi I, Knee J, Menger MD, Harbauer G, Bühren V. Hepatic microcirculatory disturbances due to portal vein clamping in the orthotopic rat liver transplantation model. Transplantation. 1991;52:432–6.CrossRefPubMed Marzi I, Knee J, Menger MD, Harbauer G, Bühren V. Hepatic microcirculatory disturbances due to portal vein clamping in the orthotopic rat liver transplantation model. Transplantation. 1991;52:432–6.CrossRefPubMed
38.
Zurück zum Zitat Bissell DM, Arenson DM, Maher JJ, Roll FJ. Support of cultured hepatocytes by a laminin-rich gel. Evidence for a functionally significant subendothelial matrix in normal rat liver. J Clin Invest. 1987;79:801–12.CrossRefPubMedPubMedCentral Bissell DM, Arenson DM, Maher JJ, Roll FJ. Support of cultured hepatocytes by a laminin-rich gel. Evidence for a functionally significant subendothelial matrix in normal rat liver. J Clin Invest. 1987;79:801–12.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Martinez-Hernandez A. The hepatic extracellular matrix. II. Electron immunohistochemical studies in rats with CCl4-induced cirrhosis. Lab Invest. 1985;53:166–86.PubMed Martinez-Hernandez A. The hepatic extracellular matrix. II. Electron immunohistochemical studies in rats with CCl4-induced cirrhosis. Lab Invest. 1985;53:166–86.PubMed
40.
Zurück zum Zitat Cheng F, Li Y, Feng L, Li S. Hepatic stellate cell activation and hepatic fibrosis induced by ischemia/reperfusion injury. Transplant Proc. 2008;40:2167–70.CrossRefPubMed Cheng F, Li Y, Feng L, Li S. Hepatic stellate cell activation and hepatic fibrosis induced by ischemia/reperfusion injury. Transplant Proc. 2008;40:2167–70.CrossRefPubMed
41.
Zurück zum Zitat Friedman SL. Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injury. J Biol Chem. 2000;275:2247–50.CrossRefPubMed Friedman SL. Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injury. J Biol Chem. 2000;275:2247–50.CrossRefPubMed
43.
Zurück zum Zitat Vera M, Nieto N. Hepatic stellate cells and alcoholic liver disease. Rev Esp Enferm Dig. 2006;98:674–84.CrossRefPubMed Vera M, Nieto N. Hepatic stellate cells and alcoholic liver disease. Rev Esp Enferm Dig. 2006;98:674–84.CrossRefPubMed
44.
Zurück zum Zitat Schaffner F, Poper H. Capillarization of hepatic sinusoids in man. Gastroenterology. 1963;44:239–42.PubMed Schaffner F, Poper H. Capillarization of hepatic sinusoids in man. Gastroenterology. 1963;44:239–42.PubMed
45.
Zurück zum Zitat Friedman SL. Seminars in medicine of the Beth Israel Hospital, Boston. The cellular basis of hepatic fibrosis. Mechanisms and treatment strategies. N Engl J Med. 1993;328:1828–35.CrossRefPubMed Friedman SL. Seminars in medicine of the Beth Israel Hospital, Boston. The cellular basis of hepatic fibrosis. Mechanisms and treatment strategies. N Engl J Med. 1993;328:1828–35.CrossRefPubMed
46.
Zurück zum Zitat Yoshida S, Ikenaga N, Liu SB, Peng ZW, Chung J, Sverdlov DY, Miyamoto M, Kim YO, Ogawa S, Arch RH, Schuppan D, Popov Y. Extrahepatic platelet-derived growth factor-β, delivered by platelets, promotes activation of hepatic stellate cells and biliary fibrosis in mice. Gastroenterology. 2014;147:1378–92.CrossRefPubMed Yoshida S, Ikenaga N, Liu SB, Peng ZW, Chung J, Sverdlov DY, Miyamoto M, Kim YO, Ogawa S, Arch RH, Schuppan D, Popov Y. Extrahepatic platelet-derived growth factor-β, delivered by platelets, promotes activation of hepatic stellate cells and biliary fibrosis in mice. Gastroenterology. 2014;147:1378–92.CrossRefPubMed
47.
Zurück zum Zitat Yamada N, Urahashi T, Ihara Y, Sanada Y, Wakiya T, Okada N, Mizuta K. Veno-occlusive disease/sinusoidal obstruction syndrome associated with potential antibody-mediated rejection after pediatric living donor liver transplantation: a case report. Transplant Proc. 2012;44:810–3.CrossRefPubMed Yamada N, Urahashi T, Ihara Y, Sanada Y, Wakiya T, Okada N, Mizuta K. Veno-occlusive disease/sinusoidal obstruction syndrome associated with potential antibody-mediated rejection after pediatric living donor liver transplantation: a case report. Transplant Proc. 2012;44:810–3.CrossRefPubMed
48.
Zurück zum Zitat Zhang L, Wang Y, Huang H. Defibrotide for the prevention of hepatic veno-occlusive disease after hematopoietic stem cell transplantation: a systematic review. Clin Transplant. 2012;26:511–9.CrossRefPubMed Zhang L, Wang Y, Huang H. Defibrotide for the prevention of hepatic veno-occlusive disease after hematopoietic stem cell transplantation: a systematic review. Clin Transplant. 2012;26:511–9.CrossRefPubMed
49.
Zurück zum Zitat Sindram D, Rüdiger HA, Upadhya AG, Strasberg SM, Clavien PA. Ischemic preconditioning protects against cold ischemic injury through an oxidative stress dependent mechanism. J Hepatol. 2002;36:78–84.CrossRefPubMed Sindram D, Rüdiger HA, Upadhya AG, Strasberg SM, Clavien PA. Ischemic preconditioning protects against cold ischemic injury through an oxidative stress dependent mechanism. J Hepatol. 2002;36:78–84.CrossRefPubMed
50.
Zurück zum Zitat Tejima K, Arai M, Ikeda H, Tomiya T, Yanase M, Inoue Y, Nagashima K, Nishikawa T, Watanabe N, Omata M, Fujiwara K. Ischemic preconditioning protects hepatocytes via reactive oxygen species derived from kupffer cells in rats. Gastroenterology. 2004;127:1488–96.CrossRefPubMed Tejima K, Arai M, Ikeda H, Tomiya T, Yanase M, Inoue Y, Nagashima K, Nishikawa T, Watanabe N, Omata M, Fujiwara K. Ischemic preconditioning protects hepatocytes via reactive oxygen species derived from kupffer cells in rats. Gastroenterology. 2004;127:1488–96.CrossRefPubMed
51.
Zurück zum Zitat Georgescu A, Popov D, Dragan E, Dragomir E, Badila E. Protective effects of nebivolol and reversal of endothelial dysfunction in diabetes associated with hypertension. Eur J Pharmacol. 2007;570:149–58.CrossRefPubMed Georgescu A, Popov D, Dragan E, Dragomir E, Badila E. Protective effects of nebivolol and reversal of endothelial dysfunction in diabetes associated with hypertension. Eur J Pharmacol. 2007;570:149–58.CrossRefPubMed
52.
Zurück zum Zitat Hanazaki K, Kitagawa H, Yatabe T, Munekage M, Dabanaka K, Takezaki Y, Tsukamoto Y, Asano T, Kinoshita Y, Namikawa T. Perioperative intensive insulin therapy using an artificial endocrine pancreas with closed-loop glycemic control system: the effects of no hypoglycemia. Am J Surg. 2014;207:935–41.CrossRefPubMed Hanazaki K, Kitagawa H, Yatabe T, Munekage M, Dabanaka K, Takezaki Y, Tsukamoto Y, Asano T, Kinoshita Y, Namikawa T. Perioperative intensive insulin therapy using an artificial endocrine pancreas with closed-loop glycemic control system: the effects of no hypoglycemia. Am J Surg. 2014;207:935–41.CrossRefPubMed
53.
Zurück zum Zitat Hayashi H, Takamura H, Nakanuma S, Makino I, Tajima H, Fushida S, Hanazaki K, Ohta T. Application of an artificial pancreas for a liver transplant recipient. Exp Clin Transplant. 2014;12:572–3.PubMed Hayashi H, Takamura H, Nakanuma S, Makino I, Tajima H, Fushida S, Hanazaki K, Ohta T. Application of an artificial pancreas for a liver transplant recipient. Exp Clin Transplant. 2014;12:572–3.PubMed
54.
Zurück zum Zitat Wang S, Moustaid-Moussa N, Chen L, Mo H, Shastri A, Su R, Bapat P, Kwun I, Shen CL. Novel insights of dietary polyphenols and obesity. J Nutr Biochem. 2014;25:1–18.CrossRefPubMedPubMedCentral Wang S, Moustaid-Moussa N, Chen L, Mo H, Shastri A, Su R, Bapat P, Kwun I, Shen CL. Novel insights of dietary polyphenols and obesity. J Nutr Biochem. 2014;25:1–18.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Kubota T, Kubota N, Kumagai H, Yamaguchi S, Kozono H, Takahashi T, Inoue M, Itoh S, Takamoto I, Sasako T, Kumagai K, Kawai T, Hashimoto S, Kobayashi T, Sato M, Tokuyama K, Nishimura S, Tsunoda M, Ide T, Murakami K, Yamazaki T, Ezaki O, Kawamura K, Masuda H, Moroi M, Sugi K, Oike Y, Shimokawa H, Yanagihara N, Tsutsui M, Terauchi Y, Tobe K, Nagai R, Kamata K, Inoue K, Kodama T, Ueki K, Kadowaki T. Impaired insulin signaling in endothelial cells reduces insulin-induced glucose uptake by skeletal muscle. Cell Metab. 2011;13:294–307.CrossRefPubMed Kubota T, Kubota N, Kumagai H, Yamaguchi S, Kozono H, Takahashi T, Inoue M, Itoh S, Takamoto I, Sasako T, Kumagai K, Kawai T, Hashimoto S, Kobayashi T, Sato M, Tokuyama K, Nishimura S, Tsunoda M, Ide T, Murakami K, Yamazaki T, Ezaki O, Kawamura K, Masuda H, Moroi M, Sugi K, Oike Y, Shimokawa H, Yanagihara N, Tsutsui M, Terauchi Y, Tobe K, Nagai R, Kamata K, Inoue K, Kodama T, Ueki K, Kadowaki T. Impaired insulin signaling in endothelial cells reduces insulin-induced glucose uptake by skeletal muscle. Cell Metab. 2011;13:294–307.CrossRefPubMed
56.
Zurück zum Zitat Utsunomiya K. Treatment strategy for type 2 diabetes from the perspective of systemic vascular protection and insulin resistance. Vasc Health Risk Manag. 2012;8:429–36.PubMedPubMedCentral Utsunomiya K. Treatment strategy for type 2 diabetes from the perspective of systemic vascular protection and insulin resistance. Vasc Health Risk Manag. 2012;8:429–36.PubMedPubMedCentral
57.
Zurück zum Zitat Iba T, Kidokoro A, Fukunaga M, Takuhiro K, Ouchi M, Ito Y. Comparison of the protective effects of type III phosphodiesterase (PDE3) inhibitor (cilostazol) and acetylsalicylic acid on intestinal microcirculation after ischemia reperfusion injury in mice. Shock. 2006;26:522–6.CrossRefPubMed Iba T, Kidokoro A, Fukunaga M, Takuhiro K, Ouchi M, Ito Y. Comparison of the protective effects of type III phosphodiesterase (PDE3) inhibitor (cilostazol) and acetylsalicylic acid on intestinal microcirculation after ischemia reperfusion injury in mice. Shock. 2006;26:522–6.CrossRefPubMed
58.
Zurück zum Zitat Wang S, Yan C, Xu H, Zhao X, Han Y. Suppression of encephalitogenic T-cell responses by cilostazol is associated with upregulation of regulatory T cells. Neuroreport. 2010;21:629–35.CrossRefPubMed Wang S, Yan C, Xu H, Zhao X, Han Y. Suppression of encephalitogenic T-cell responses by cilostazol is associated with upregulation of regulatory T cells. Neuroreport. 2010;21:629–35.CrossRefPubMed
59.
Zurück zum Zitat Narita M, Hatano E, Ikai I, Miyagawa-Hayashino A, Yanagida A, Nagata H, Asechi H, Taura K, Uemoto S. A phosphodiesterase III inhibitor protects rat liver from sinusoidal obstruction syndrome through heme oxygenase-1 induction. Ann Surg. 2009;249:806–13.CrossRefPubMed Narita M, Hatano E, Ikai I, Miyagawa-Hayashino A, Yanagida A, Nagata H, Asechi H, Taura K, Uemoto S. A phosphodiesterase III inhibitor protects rat liver from sinusoidal obstruction syndrome through heme oxygenase-1 induction. Ann Surg. 2009;249:806–13.CrossRefPubMed
60.
Zurück zum Zitat Ikegami T, Nishizaki T, Hiroshige S, Ohta R, Yanaga K, Sugimachi K. Experimental study of a type 3 phosphodiesterase inhibitor on liver graft function. Br J Surg. 2001;88:59–64.CrossRefPubMed Ikegami T, Nishizaki T, Hiroshige S, Ohta R, Yanaga K, Sugimachi K. Experimental study of a type 3 phosphodiesterase inhibitor on liver graft function. Br J Surg. 2001;88:59–64.CrossRefPubMed
Metadaten
Titel
Ischemia reperfusion-facilitated sinusoidal endothelial cell injury in liver transplantation and the resulting impact of extravasated platelet aggregation
verfasst von
T. Miyashita, MD, PhD
S. Nakanuma, MD, PhD
A.K. Ahmed
I. Makino, MD, PhD
H. Hayashi, MD, PhD
K. Oyama, MD, PhD
H. Nakagawara, MD, PhD
H. Tajima, MD, PhD
H. Takamura, MD, PhD
I. Ninomiya, MD, PhD
S. Fushida, MD, PhD
J.W. Harmon, MD
T. Ohta, MD, PhD
Publikationsdatum
01.04.2016
Verlag
Springer Vienna
Erschienen in
European Surgery / Ausgabe 2/2016
Print ISSN: 1682-8631
Elektronische ISSN: 1682-4016
DOI
https://doi.org/10.1007/s10353-015-0363-3

Weitere Artikel der Ausgabe 2/2016

European Surgery 2/2016 Zur Ausgabe

Book Review

Acute abdomen