Review Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Aug 7, 2013; 19(29): 4638-4650
Published online Aug 7, 2013. doi: 10.3748/wjg.v19.i29.4638
Strategies to rescue steatotic livers before transplantation in clinical and experimental studies
Qiang Liu, Maria-Louisa Izamis, Hongzhi Xu, Tim Berendsen, Martin Yarmush, Korkut Uygun, Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, and Shriners Burns Hospital, Boston, MA 02114, United States
Martin Yarmush, Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, United States
Author contributions: Liu Q, Izamis ML, Xu H, Berendsen T, Yarmush M and Uygun K contributed to collect literatures and write the paper.
Supported by Grants from the United States National Institutes of Health, R01DK59766 to Yarmush M; R00DK080942 and R01DK096075 to Uygun K
Correspondence to: Martin Yarmush, MD, PhD, Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, 51 Blossom Street, Boston, MA 02114, United States. ireis@sbi.org
Telephone: +1-617-3174882 Fax: +1-617-5739471
Received: October 19, 2012
Revised: December 10, 2012
Accepted: December 15, 2012
Published online: August 7, 2013

Abstract

The shortage of donor livers has led to an increased use of organs from expanded criteria donors. Included are livers with steatosis, a metabolic abnormality that increases the likelihood of graft complications post-transplantation. After a brief introduction on the etiology, pathophysiology, categories and experimental models of hepatic steatosis, we herein review the methods to rescue steatotic donor livers before transplantation applied in clinical and experimental studies. The methods span the spectrum of encouraging donor weight loss, employing drug therapy, heat shock preconditioning, ischemia preconditioning and selective anesthesia on donors, and the treatment on isolated grafts during preservation. These methods work at different stages of transplantation process, although share similar molecular mechanisms including lipid metabolism stimulation through enzymes or nuclear receptor e.g., peroxisomal proliferator-activated receptor, or anti-inflammation through suppressing cytokines e.g., tumor necrosis factor-α, or antioxidant therapies to alleviate oxidative stress. This similarity of molecular mechanisms implies possible future attempts to reinforce each approach by repeating the same treatment approach at several stages of procurement and preservation, as well as utilizing these alternative approaches in tandem.

Key Words: Liver transplantation, Steatosis, Donor liver, Clinical, Experimental



INTRODUCTION

Liver transplantation (LTx) is a successful therapy for end-stage liver disease, but it is severely restricted by the donor organ shortage[1-4]. In an effort to increase the size of the donor pool, livers from expanded criteria donors (ECD), including steatotic livers, are increasingly being used[3]. The current contribution of steatotic livers is marginal however, since the majority has an increased risk of ischemia-reperfusion injury (IRI) after LTx[3]. Here we review the clinical and experimental attempts to minimize this risk by modifying the quality of livers at different stages, on donors and isolated liver grafts during preservation, before LTx. We begin by briefly introducing the etiology, pathophysiology, categories and experimental models of hepatic steatosis, provide a summary of the techniques applied on donors and liver grafts, and conclude by providing future perspectives for these experimental approaches.

ETIOLOGY OF HEPATIC STEATOSIS

Livers are defined as steatotic or fatty when they have excessive (above 5% of wet liver weight) accumulation of lipids, mainly triglycerides. Steatosis occurs when lipid ingestion and synthesis exceed export and consumption in livers[5]. Based on the patient’s alcohol consumption[5,6] fatty liver disease is classified as either alcoholic or non-alcoholic in origin (AFLD and NAFLD). Alcohol can decrease fatty acid oxidation and lipoprotein excretion, and increases the esterification of fatty acid to triglycerides via alpha-glycerophosphate[6]. Several factors contribute to NAFLD including dietary lipid overload; insulin resistance, which results in abnormal lipid metabolism; and ingestion of drugs or toxins such as carbon tetrachloride, which induces a decrease in apoprotein synthesis and lipid export[5-8].

PATHOPHYSIOLOGY OF HEPATIC STEATOSIS

A complicated pathophysiology has been exposed although the exact pathways have not been completely elucidated. Briefly, a “two-hit” theory is the current consensus, with the first “hit” being the initial abnormal fat accumulation and the second “hit” being the consequent inflammation (“steatohepatitis”) leading to fibrosis and cirrhosis[6]. In the first “hit”, excessive fat accumulates in vacuoles within hepatocytes, increasing cell volume and narrowing sinusoidal lumens[5,9]. This impairs microcirculation and decreases nutrient, oxygen and waste exchange. Excessive non-esterified fatty acids in hepatocytes inhibit β-oxidation thereby reduce acetyl-coenzyme A production[10]. Mitochondria uncoupling protein-2 is upregulated and associated with a dysfunction of adenosine triphosphate (ATP) synthesis[11-13]. Fat-induced hyperactivity of cytochrome P-450 enzymes increases the production of reactive oxygen species (ROS)[6,14]. ROS in turn lead to lipid peroxidation, phospholipid depletion and membrane dysfunction[6,14,15] as well as the release of inflammatory cytokines such as tumor necrosis factor (TNF)-α[14,16,17]. Inflammation occurs gradually and marks a significant downturn in disease progression as the second “hit”[18,19]. Alcohol can exacerbate oxidative injury and Kupffer cell activation[20], though alcoholic and non-alcoholic steatohepatitis (ASH and NASH) are thought to progress similarly[18].

Steatotic livers have reduced tolerance to ischemia due to low ATP stores, and are thus prone to early onset of acidosis and cellular edema during standard liver preservation method, static cold storage (SCS)[21]. Edema significantly impairs hepatic microcirculation further than the preceded impairment induced by excessive fat. Moreover, steatosis-induced inflammation is not addressed in the present liver SCS preservation solutions (Table 1).

Table 1 Intrinsic composition of preservation solutions in this review.
UWHTKCelsiorIGL-1UW-gluconateKreb-HenseleitMEM
ElectrolyteNa2515100125125143143.4
(mmol/L)K12091530255.95.4
Mg5413551.20.8
Ca0.00150.250.51.251.4
Cl2032421125.2124
SO4551.20.8
BufferPhosphate2525151.21
(mmol/L)Bicarbonate252526.2
HEPES1020
Histidine198300.27
AntioxidantGlutathione3333
(mmol/L)Allopurinol111
Mannitol3860
NAC5
Vit C0.25
Glucose1055.5
Metabolic SubstratesAdenosine555
(mmol/L)Adenine5
Ribose5
Tryptophan2
Ketoglutarate1
Glutamate20
Amino acid0.7
ImpermeantsLactobionate10080100
(mmol/L)Gluconate95
Raffinose303030
ColloidHES50
(g/L)PEG150
Other intrinsic compoundsInsulin100 U
Dexamethason8 mg
Penicillin40 U
Phenol-red (mmol/L)0.03
Osmolarity340300363330360320310

Upon reperfusion, a complex inflammatory response involving Kupffer cells, lymphocytes, neutrophils, numerous cytokines[22] and nuclear factor kappa-B (NFκB)[23], is inevitably worse in steatotic livers compared to non-steatotic livers. Oxidative stress is also exacerbated e.g., through xanthine oxidase[24,25]. The microcirculation is deteriorated further due to adherence of platelets in the sinusoids[22]. Strategies to minimize fat content and ameliorate the inflammatory and oxidative injury of steatotic livers are essential for improving these organs for transplantation.

CATEGORIES OF HEPATIC STEATOSIS

Besides AFLD and NAFLD in etiology, steatosis is also classified as “macro-” or “micro-” in histology based on the size and number of the fat vacuoles and on the location of the nucleus in the hepatocytes[5,26]. Macrosteatosis has a single fat vacuole larger than the nucleus filling the majority of the cell and pushing the nucleus to the periphery. Microsteatosis has many small fat vacuoles surrounding the nucleus in the central zone of the hepatocytes, and has more LTx success than macrosteatosis[3,27]. Steatosis can also be classified based on the proportion of hepatocytes affected, being mild (< 30%), moderate (30%-60%), or severe (> 60%), with incremental risk of graft dysfunction after LTx[5,26].

CLINICALLY APPLIED STRATEGIES

Approaches to improve steatotic livers before LTx have been tested in a handful of pilot clinical studies on living donors or donors after brain death (DBD) (Table 2). The interventions focused on reducing excessive fat (the first “hit”) through limiting lipid intake and increasing lipolysis, or stimulating factors likely to be protective against inflammation and oxidative stress (the second “hit”) of steatohepatitis. Living donors, though a minority in western countries[1,2], are used extensively in Asia[28] and are theoretically amenable to therapies before procurement after ethical concern is taken into account[29]. DBD livers, which comprise the majority of donor organs for LTx in western countries, could be treated between brain death declaration and organ procurement since circulation is maintained until procurement. Livers from donors after cardiac death (DCD) are seldom utilized when they have steatosis because they experience a period of warm ischemia (WI) before procurement[3] and thus were normally considered as unacceptable with two defects (steatosis and WI). Currently there are no attempts to rescue steatotic DCD livers.

Table 2 Overview of the clinical and experimental strategies in this review.
Clinically appliedExperimentally applied
DonorsDietaryPharmacological
PharmacologicalHeat shock
Ischemic (except remote ischemic)
Anesthetic
Liver graftsSCS preservation
New solution
Pharmacological additives
Additional oxygen (VSOP)
MP preservation
MP solely
MP + pharmacological additives
Flushing
Pharmacological additives
Physical exercise and dietary intervention

Physical exercise and dietary restriction are general therapies for NAFLD patients, independent of whether or not they are organ donors[30,31]. But this treatment normally needs several months and might be risky to increase the mortality of recipients during the waiting time for treating donors[32]. An intensive protocol might be a solution, which was already reported to successfully reduce macrosteatosis on obese human living donors in 2-8 wk, through exercise burning 600 kcal/d, a protein-rich diet of 1000 kcal/d, and bezafibrate, an anti-hyperlipidaemia drug, at 400 mg/d[33]. However, supplementary glucose to donors a few hours before living donor liver transplantation was recommended to supply additional energy reserves, since fasting before procurement can induce glycogen depletion, decrease glycolytic ATP generation, and compromise graft transplantability[34].

The lack of omega-3 polyunsaturated fatty acids (PUFA) has been recognized in the development of NAFLD because they can activate peroxisomal proliferator-activated receptor (PPAR)-α, suppress sterol regulatory element-binding protein-1, improve microcirculation, and reduce Kupffer cell activity and inflammation[35-43]. The mechanism on microcirculation might work through reducing TXA2 synthesis after manipulating the composition of hepatic lipid (omega-3: omega-6 PUFA ratio)[43]. Based on experimental success on rodents[35-43], omega-3 PUFA was shown to be effective on NAFLD clinically after treatment at 1-2 g/d for 6-12 mo[44-46]. This has not been applied specifically on living donors but is expected to be a safe and promising approach.

Pharmacological preconditioning

Many drugs are being used clinically to treat NAFLD by decreasing lipid intake[47-49], stimulating lipid metabolism[50-53], or improving insulin sensitivity[54-57]. Ursodeoxycholic acid, a natural bile acid, was used as a non-specific hepato-protector to treat NAFLD in a pilot clinical study[58], but afterward was revealed to be controversial[59]. Pentoxifylline was used against NASH and ASH in patients[60,61] due to the effect of reducing TNF-α by inhibiting phosphodiesterase[62] and lessening oxidative stress by increasing glutathione[63]. But to date, only bezafibrate was used to treat human living donors for LTx[33]. This drug works through activating PPAR-α and β/δ to stimulate lipid metabolism and decrease fat content in livers[64,65]. While there are other candidate drugs that could potentially be taken by living donors, concerns of significant side effects are limiting their use[66,67].

Ischemic preconditioning

Though extended ischemia is deleterious to organs, it has been recognized since the 1980s that a short period of ischemia with subsequent reperfusion triggers natural defense mechanisms against future ischemic insults and protects the organ against IRI[68]. Ischemic preconditioning (IP) was first observed in kidneys and hearts[69,70], and then employed for clinical liver resections and transplantation[68]. It can be applied intermittently[71], or as a single short period (5-10 min) of ischemia followed by 10-15 min reperfusion before cold flush during liver procurement[72,73]. Franchello et al[73] have used the technique clinically on marginal DBD livers including steatotic livers, and observed a reduction of hepatocyte swelling and enzyme release in recipients after LTx.

IP is protective because ATP consumption during the short ischemic period increases endogenous adenosine and nitric oxide[74]. Adenosine protects sinusoidal endothelial cells through adenosine A2 receptor[75]. Cyclic adenosine monophosphate (cAMP) worked as the second messenger, but whether increasing or blocking cAMP would be beneficial was still controversial[75,76]. Nitric oxide is a vasodilator, and further it attenuates the release of TNF-α, decreases the injurious interleukin (IL)-1β and increases the anti-inflammatory IL-10[77]. Another effect of the intermittent ATP consumption is to increase the level of adenosine monophosphate (AMP), which stimulates AMP-activated protein kinase (AMPK). AMPK can regulate an energy-conserving state, decrease inflammation through inhibiting NFκB, and induce the synthesis of nitric oxide as well[78-80]. Overall, the advantages of IP were an improved microcirculation[81,82], mitochondrial permeability transition and mitochondrial function[83], cytochrome oxidase C activity and tissue oxygenation[82,84], and the reduction of oxidative stress such as the xanthine accumulation and xanthine oxidase activity[85,86].

Interestingly, IP can work remotely, e.g., liver IP decreased lung IRI[25,87] and limb IP decreased liver IRI[88-90]. This possibly works through some protective agents, e.g., heme oxygenase-1, endothelial nitric oxide synthase, and nuclear protein High Mobility Group-Box 1[88-90]. But foreseeable ethical concerns exist with the logistics of implementing this technique in human donors.

Anesthesia selection

During clinical liver resection, Beck-Schimmer et al[91] observed the superiority of volatile anesthesia using sevoflurane in the prevention of hepatic injury after reperfusion compared to intravenous anesthesia with propofol. The mechanism was suggested to be the increased synthesis of nitric oxide with sevoflurane, which alleviates some of the effects of IRI as discussed above. Moreover they observed that patients with steatosis did benefit more[91]. This method could be easily applied during the procurement of steatotic livers from both DBD and living donors.

EXPERIMENTAL STRATEGIES
Induction of steatosis in animal models

Steatosis in animal models are established with or without alcohol and classified also as micro- vs macro- or as mild, moderate, vs severe. The timeline to develop the different classifications largely depends on strain and method used (Table 3).

Table 3 Animal models of hepatic steatosis for liver transplantation.
DiseaseApproachesDescriptionAnimalsTreatment time
NAFLDGeneticCerebral leptin receptor deficiencyRodent
DietaryHigh fatRodent7 d
High cholesterolRodent, rabbits8-12 wk
Cafeteria dietRodent4-15 wk
Choline/methionine-deficientRodent7 d-6 wk
High starch and fat free after fastingRodent4 d
High fat and choline deficiencyDog8-12 wk
High fat and high cholesterol, plus choline deficiency or notMiniature pig24 wk
High fat and carbohydrate with streptozotocin for a diabetic stateLandrace pig5 wk
AFLDDietaryEthanol in liquid diet, intragastric infusion or gavageRodent20 h-9 wk
Ethanol and high fat dietRodent6 wk
Ethanol and deficient folate dietMicropig12 wk

A common rodent NAFLD model induces cerebral deficiency of the leptin receptor through a genetic mutation that causes the animals to become obese through overeating[92-98]. The methods inducing lipid overload with high fat or cholesterol diets are also quite common. A high fat diet (50% dextrose, 18% casein, 25% lipids, 4% minerals, 1% cholesterol, 0.5% sodium cholate, 0.2% choline, and 1% vitamins) for 7 d induces severe steatosis in rats[99]. A high-cholesterol (2%) diet for 12 wk in rats[82] and 8 wk in rabbits results in moderate steatosis[84]. A cafeteria diet (65% of fat) for 4-15 wk was also used to create NAFLD on rats[100,101]. Recent studies suggested it reflects human metabolic syndrome better than high-fat diet[100,101]. A choline/methionine-deficient diet (CMDD) was another rather common method to develop steatosis, for 4-6 wk or short as 7 d in rodents[5,27,102,103]. Choline is essential for the formation of phosphatidyl-choline and very low density lipoprotein needed for lipid export, while methionine is a good source of methyl groups for the endogenous synthesis of choline. This model could be criticized for not being clinically accurate because NAFLD patients due to CMDD are rather unrealistic. The most rapid induction of NAFLD in rats was with a high-starch, fat-free diet [saccharose (40%), starch (40%), casein (16%), and a mineral and vitamin mix (4%)] administered for 2 d after fasting for 2 d, which can lead to mild to moderate steatosis[104-106]. While none of these models are unanimously agreed to be ideal in replicating clinical NAFLD, the high fat or cholesterol diet model was the most widely used to mimic steatosis in humans.

There are fewer large animal NAFLD models, which usually combine more than one method described above to achieve steatosis. Takahashi et al[107] established a dog model using a diet rich in fat and deficient in choline, which produced moderate to severe macrosteatosis after 8-12 wk feeding. Lee et al[108] used a high fat and high cholesterol diet (20% kcal from fructose, 46% kcal from fat, 2% cholesterol and 0.7% cholate by weight) for 24 wk to develop microsteatosis on miniature pigs. When using lowered choline content simultaneously, severe steatosis with fibrosis was observed with increased TNF-α and oxidative stress. A recent study on Landrace pigs used a diet rich in fat (20% in volume) for 5 wk together with intravenous streptozotocin (125 mg/kg) to induce a diabetic state in the last 2 wk; but this treatment led only to mild steatosis[109].

In rodent AFLD models, ethanol was provided in a liquid diet[110-114] or via intragastric infusion[115,116]. Different degrees of alcohol exposure have been reported: 5%-8% in the concentration of liquid diet; 35%-40% of total energy consumption; 8-16 g/kg per day; or 150-300 mg/dL of blood ethanol[110-116]. Normally several days are needed for the animals to adapt to the alcoholic diet, and an additional 4-9 wk to observe steatosis. Acute responses to ethanol have also been reported as early as 20 h after feeding 6 g/kg ethanol by gavage on rats[117]. Combined ethanol with high-fat diet to develop steatosis on rats was also reported[118]. Large animal AFLD models are uncommon though micropigs have been fed a diet with ethanol and a deficiency of folate, as a substrate for methionine synthase, with some efficacy[119,120].

Experimental strategies applied to donors

Pharmacological preconditioning: Reduction of oxidative and inflammatory activity with heme oxygenase-1, a microsomal enzyme[121-123], was used intravenously or intraperitoneally on AFLD and NAFLD rats 24 h before liver procurement. It decreased macrophage infiltration, improved portal venous blood flow, bile production, and survival rate after LTx[121-123]. Bortezomib, a NFκB inhibitor, was used intravenously on obese donor rats and reduced IRI after LTx[23]. N-acetylcysteine, a precursor of glutathione, was injected through the mesenteric vein of CMDD rats 15 min before liver procurement and showed a protective effect on IRI in an isolated reperfusion system[124]. The subcutaneous injection of IL-6 for 10 d was observed to be protective against IRI after in situ partial ischemia-reperfusion on NAFLD and ALFD mice[92]. The mechanism might be the prevention of cell death and the reduction of TNF-α[125], in addition to stimulating PPAR-α, β-oxidation of fatty acids, and the export of triglycerides and cholesterol[92,125]. Theaflavin, a polyphenol substance extracted from black tea, was tested on CMDD mice and observed to have antioxidant, anti-inflammatory, and anti-apoptotic effect[126]. A multi-drug approach was reported by von Heesen et al[127] including N-acetylcysteine as an antioxidant, pentoxifylline for anti-inflammation, glycine to stabilize Kupffer cells, deferoxamine as an iron chelator to reduce ROS, and erythropoietin, melatonin and simvastatin to protect against IRI. In the treated rats they observed no inflammatory response with significantly reduced parenchymal dysfunction and injury compared to the untreated rats.

Heat shock preconditioning: An intriguing experimental method to improve the quality of steatotic donor livers has been to induce protective heat shock proteins (HSPs) endogenously by exposure to heat. Termed “heat shock preconditioning” and applied at 3-48 h before organ procurement by exposing anesthetized donor animals to warm (42 °C) bath water for 10-15 min[128-132], obese and CMDD rats showed an increased expression of HSP-32 (heme oxygenase-1), -72 and -90[128,129]. These HSPs can decrease TNF-α production[129], improve microcirculation through producing carbon monoxide, and inhibit platelet aggregation[62,64]. Our group has also reported the inhibition of CD4+ T lymphocytes in CMDD rats after LTx with heat shock preconditioning[130]. Other factors might be involved in the treatment since studies on normal and WI rat livers showed IL-6, inter-cellular adhesion molecule-1, and some neutrophil chemo-attractants were also impacted[131,132].

Strategies applied on liver grafts during ex vivo preservation

Obviously, strategies to improve steatotic liver quality during preservation are more desirable than those on donors, as they have no effect on the donor’s other organs, and are practical when it’s not possible to work on the donor. The clinical standard for liver graft preservation has been SCS with University of Wisconsin (UW) solution for more than 20 years[3,21]. In the past decade, histidine-trypotphan-ketoglutarate (HTK) solution[133-135] and Celsior solution[136,137] were recognized as having similar efficacy and safety as UW solution; Institut Georges Lopez-1 (IGL-1) was reported to be comparable to UW for healthy human livers[138], and better for steatotic rat livers[139]. Besides the arising new solutions, many adaptations have been suggested through enriching the intrinsic composition of the solutions with additives, or replacing SCS by machine perfusion (MP) preservation in experimental studies to rescue steatotic livers.

Pharmacological additives during SCS preservation: Liver preservation solutions normally comprise electrolytes, pH buffers, antioxidants, metabolic substrates, impermeants with or without colloid, insulin, dexamethasone, and antibiotics (Table 1)[139-143]. The additives for improving steatotic liver preservation were intended to ameliorate metabolism or suppress oxidative injury and inflammation. They were reported to be effective during SCS, despite the reduced metabolic rate of liver grafts during hypothermic preservation.

Addition of IL-6 into UW solution for donor liver flushing and SCS was tested by Sun et al[144], leading to improvement in microcirculation and reduced IRI after LTx of NAFLD and AFLD rats. Arnault et al[99] added pentoxifylline into UW solution and also observed a benefit to the microcirculation, but the exact mechanism is yet to be identified.

Tolba et al[106] added L-carnitine into HTK solution for SCS preservation of steatotic rat livers and observed a reduction of IRI in an isolated reperfusion system. L-carnitine is a nonessential amino acid but is essential for transporting fatty acids through the inner mitochondrial membrane and for β-oxidation[106]. It has also been reported to function as an antioxidant and to stabilize the membrane fluidity and stability in vitro and in vivo[145].

Ben Mosbah et al[146] added carvedilol, a cardiologic drug to block α- and β-adrenergic receptor, into UW solution for SCS of obese rat livers, and reduced oxidative stress and mitochondrial damage after isolated reperfusion. The mechanism might be an enhanced release of nitric oxide that facilitates vasodilatation and ROS scavenging[147]. AMPK activators, trimetazidine and aminoimidazole-4-carboxamide ribonucleoside (AICAR) were also tested as UW additives on obese rat livers by this group. Increased bile production, decreased enzyme release and vascular resistance, and reduced oxidative stress after isolated reperfusion were observed. It was noted that combination of trimetazidine and AICAR was not necessary[95].

Zaouali et al[148] tested the use of epidermal growth factor and insulin-like growth factor-I as UW additives and observed that each additive resulted in the improvement of fatty rat liver function after LTx. The mechanisms are suggested to be upregulation of Akt, a cytoprotector[149], and the subsequent over-expression of PPAR-γ. They also tested melatonin as additive in IGL solution and reported its protective role through generating nitric oxide and decreasing oxidative stress and inflammation[150].

Venous systemic oxygen persufflation during SCS preservation: In 1990s, Minor et al[151] developed a new method, called venous systemic oxygen persufflation (VSOP) to supply gaseous oxygen to livers during SCS preservation. The oxygen was introduced into hepatic vasculature via the suprahepatic vena cava and allowed to exit via several small pin pricks on the liver capsule made using an acupuncture needle. This technique was employed on steatotic rat livers for 24 h, and resulted in improved preservation of mitochondria and sinusoidal endothelial linings, less Kupffer cell activation and reduced hepatocellular enzyme release compared to SCS preservation[105]. Recently, by assessing the enzyme release, energy storage, bile production, and cell death during isolated reperfusion, it was demonstrated that application of VSOP for 90 min may rescue steatotic livers after extended (18 h) SCS preservation[152].

MP preservation: MP is an alternative preservation method to SCS[153], which can be further categorized based on the temperature employed[154]. Hypothermic (4 °C) machine perfusion (HMP) preservation has proven to be superior to SCS for human kidneys[155], and feasible for normal human livers[156]. Normothermic (32  °C-37  °C) and sub-normothermic (20  °C-30  °C) machine perfusion (NMP and subNMP) preservation have been reported in experimental studies on livers, but mostly on their advantages for DCD models[143,157-162]. MP preservation of steatotic livers is limited, but also reported to be beneficial on preserving energy content and liver function experimentally[109,143,163,164]. The advantages of MP preservation result from continuously supplying nutrients, removing waste products, and maintaining microcirculation[154]. Because MP, especially NMP, provides a physiologically-relevant environment to the isolated donor organ, the quality of liver grafts can be manipulated more efficiently than those simply stored in an ice-box during SCS. Another advantage of MP is the considerable convenience for non-invasively evaluating liver viability, a key issue when ECD livers are used[153].

Bessems et al[163] employed HMP preservation with UW-gluconate solution on steatotic rat livers for 24 h and alleviated IRI compared to SCS. Vairetti et al[143] preserved steatotic rat livers by subNMP (20 °C) with Kreb-Henseleit solution for 6 h and obtained similar results. The longest preservation of steatotic livers was the NMP preservation for 48 h in a pig model by Jamieson et al[109], who employed blood containing additional insulin and vasodilators as perfusate, and observed a mild reduction of steatosis from 28% to 15%. This NMP setting provided the most physiological environment to liver grafts and lead to an activated function of the isolated organs with sufficient oxygen and nutritional support. This is expected to be the best preservation method in spite of the highest logistic restriction.

Recently our group has combined NMP and pharmacological preconditioning for decreasing steatosis[96]. A “defatting cocktail” was developed with 6 compounds to activate nuclear receptors such as PPARs, pregnane X receptor, and constitutive androstane receptor, to exert an insulin mimetic effect and to stimulate intracellular cAMP. This cocktail was added into Minimum Essential cell culture medium as a perfusate to stimulate lipid metabolism of obese rat liver grafts preserved at NMP. A significant decrease (50%) in steatosis was observed after 3 h NMP[96].

Other experimental approaches

The solution used for SCS preservation needs to be flushed out of the donor organ prior to implantation to remove possible air bubbles, and preservative components such as high potassium, which are deleterious to the recipient. This provides an opportunity for treatment of ischemic injury, although it has been very little explored. In one study, polyphenols, an antioxidant extracted from Camellia sinensis (green tea), was added to the flushing solution and improved the hepatic injury and survival rate after LTx in a steatotic rat model[117].

Another option is applying preconditioning at several phases. While it appears little explored, in one study Ye et al[118] injected glutathione intraperitoneally in rats with hepatic steatosis 2 d before procurement and preserved the livers by SCS with VSOP and additional glutathione. Remarkable improvement on survival rate, liver function, and oxidative stress in liver tissues after transplantation was observed.

SUMMARY AND FUTURE PERSPECTIVES

Based on the understanding on pathophysiology, current strategies to rescue steatotic donor livers work through ameliorating the abnormal lipid metabolism (the first “hit”), and the oxidative stress and inflammation (the second “hit”). Each approach employs various methods at different phases of organ recovery and preservation but generally targets similar molecular mechanisms. Notably, it may be possible to attack the same mechanism but reinforce the effects by applying the treatment at multiple points of organ recovery and preservation process, thereby producing a stacking effect. The use of glutathione on donors and in SCS solution by Ye et al[118] is a good example in this direction that demonstrates the therapeutic effects may stack. Stacking other medications, e.g., IL-6 and pentoxifylline could also be promising as these could be given to the donors prior to recovery or readily added into preservation solutions to treat liver grafts. Whether the stacking approach works for different targets in lipid metabolism, oxidative stress or inflammation remain to be elucidated.

Similar to stacking the same/similar pharmacological agents at different stages, it is a reasonable idea to attack the two “hits” simultaneously by stacking medications targeting different pathways. Development of such combinations is usually shunned because it exponentially increases the complexity of development and clinical testing, but given that the disease itself is a very complex phenomenon spanning multiple pathways, it may be unavoidable, and a single silver bullet to treat steatosis simply nonexistent.

An intriguing alternative in development is the efforts to use MP for liver preservation. Especially in near normothermic conditions, MP provides a combined opportunity to improve energy storage, maintain microcirculation, and support pharmacological approaches to decrease fat content and treat IRI. While machine perfusion by definition is more complex than simple storage on ice, it is a very promising approach available in the near future and could be the ultimate solution to rescue steatotic as well as ischemic livers.

Both steatotic livers and DCD livers are highly susceptible to IRI. Therefore, potentially, a method to rescue DCD livers could be also applicable to steatotic livers even with the fat content intact. For instance, MP preservation was able to rescue DCD livers and steatotic livers[143,157-164]. Similarly, perfluorocarbon as an artificial oxygen carrier to improve SCS preservation of DCD livers[165] could also be tested on steatotic livers. If successful, we would then secure both DCD and fatty livers for transplantation, which would boost the organ availability dramatically and resolve donor liver shortage for a decade or more.

ACKNOWLEDGMENTS

We thank the support of the Shriners Hospitals for Children.

Footnotes

P- Reviewers Hatipoglu S, Unal B, Vairetti M S- Editor Gou SX L- Editor A E- Editor Zhang DN

References
1.  Wolfe RA, Merion RM, Roys EC, Port FK. Trends in organ donation and transplantation in the United States, 1998-2007. Am J Transplant. 2009;9:869-878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
2.  Oosterlee A, Rahmel A. Eurotransplant International Foundation: annual report 2010.  Available from: http://www.eurotransplant.org/cms/mediaobject.php?file=ar_2010.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Durand F, Renz JF, Alkofer B, Burra P, Clavien PA, Porte RJ, Freeman RB, Belghiti J. Report of the Paris consensus meeting on expanded criteria donors in liver transplantation. Liver Transpl. 2008;14:1694-1707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 199]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
4.  Berg CL, Steffick DE, Edwards EB, Heimbach JK, Magee JC, Washburn WK, Mazariegos GV. Liver and intestine transplantation in the United States 1998-2007. Am J Transplant. 2009;9:907-931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 142]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
5.  Imber CJ, St Peter SD, Handa A, Friend PJ. Hepatic steatosis and its relationship to transplantation. Liver Transpl. 2002;8:415-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 129]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
6.  Veteläinen R, van Vliet A, Gouma DJ, van Gulik TM. Steatosis as a risk factor in liver surgery. Ann Surg. 2007;245:20-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 221]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
7.  Chalasani N, Deeg MA, Persohn S, Crabb DW. Metabolic and anthropometric evaluation of insulin resistance in nondiabetic patients with nonalcoholic steatohepatitis. Am J Gastroenterol. 2003;98:1849-1855.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
8.  Capeau J. Insulin resistance and steatosis in humans. Diabetes Metab. 2008;34:649-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 92]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
9.  Ijaz S, Yang W, Winslet MC, Seifalian AM. Impairment of hepatic microcirculation in fatty liver. Microcirculation. 2003;10:447-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
10.  Selzner M, Clavien PA. Fatty liver in liver transplantation and surgery. Semin Liver Dis. 2001;21:105-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 323]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
11.  Uchino S, Yamaguchi Y, Furuhashi T, Wang FS, Zhang JL, Okabe K, Kihara S, Yamada S, Mori K, Ogawa M. Steatotic liver allografts up-regulate UCP-2 expression and suffer necrosis in rats. J Surg Res. 2004;120:73-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
12.  Evans ZP, Ellett JD, Schmidt MG, Schnellmann RG, Chavin KD. Mitochondrial uncoupling protein-2 mediates steatotic liver injury following ischemia/reperfusion. J Biol Chem. 2008;283:8573-8579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
13.  Evans ZP, Palanisamy AP, Sutter AG, Ellett JD, Ramshesh VK, Attaway H, Schmidt MG, Schnellmann RG, Chavin KD. Mitochondrial uncoupling protein-2 deficiency protects steatotic mouse hepatocytes from hypoxia/reoxygenation. Am J Physiol Gastrointest Liver Physiol. 2012;302:G336-G342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
14.  Koneru B, Dikdan G. Hepatic steatosis and liver transplantation current clinical and experimental perspectives. Transplantation. 2002;73:325-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 89]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
15.  Berthiaume F, Barbe L, Mokuno Y, MacDonald AD, Jindal R, Yarmush ML. Steatosis reversibly increases hepatocyte sensitivity to hypoxia-reoxygenation injury. J Surg Res. 2009;152:54-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 39]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
16.  Diehl AM. Nonalcoholic steatosis and steatohepatitis IV. Nonalcoholic fatty liver disease abnormalities in macrophage function and cytokines. Am J Physiol Gastrointest Liver Physiol. 2002;282:G1-G5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 135]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
17.  Nagata K, Suzuki H, Sakaguchi S. Common pathogenic mechanism in development progression of liver injury caused by non-alcoholic or alcoholic steatohepatitis. J Toxicol Sci. 2007;32:453-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 146]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
18.  Gyamfi MA, Damjanov I, French S, Wan YJ. The pathogenesis of ethanol versus methionine and choline deficient diet-induced liver injury. Biochem Pharmacol. 2008;75:981-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
19.  Tevar AD, Clarke C, Wang J, Rudich SM, Woodle ES, Lentsch AB, Edwards ML. Clinical review of nonalcoholic steatohepatitis in liver surgery and transplantation. J Am Coll Surg. 2010;210:515-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
20.  Beier JI, McClain CJ. Mechanisms and cell signaling in alcoholic liver disease. Biol Chem. 2010;391:1249-1264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 126]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
21.  Pirenne J. Time to think out of the (ice) box. Curr Opin Organ Transplant. 2010;15:147-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
22.  Abu-Amara M, Yang SY, Tapuria N, Fuller B, Davidson B, Seifalian A. Liver ischemia/reperfusion injury: processes in inflammatory networks--a review. Liver Transpl. 2010;16:1016-1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 260]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
23.  Ramachandran S, Liaw JM, Jia J, Glasgow SC, Liu W, Csontos K, Upadhya GA, Mohanakumar T, Chapman WC. Ischemia-reperfusion injury in rat steatotic liver is dependent on NFκB P65 activation. Transpl Immunol. 2012;26:201-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
24.  Pesonen EJ, Linder N, Raivio KO, Sarnesto A, Lapatto R, Höckerstedt K, Mäkisalo H, Andersson S. Circulating xanthine oxidase and neutrophil activation during human liver transplantation. Gastroenterology. 1998;114:1009-1015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
25.  Fernández L, Heredia N, Grande L, Gómez G, Rimola A, Marco A, Gelpí E, Roselló-Catafau J, Peralta C. Preconditioning protects liver and lung damage in rat liver transplantation: role of xanthine/xanthine oxidase. Hepatology. 2002;36:562-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 81]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
26.  McCormack L, Dutkowski P, El-Badry AM, Clavien PA. Liver transplantation using fatty livers: always feasible? J Hepatol. 2011;54:1055-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 205]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
27.  Selzner N, Selzner M, Jochum W, Amann-Vesti B, Graf R, Clavien PA. Mouse livers with macrosteatosis are more susceptible to normothermic ischemic injury than those with microsteatosis. J Hepatol. 2006;44:694-701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
28.  Kaido T, Uemoto S. Does living donation have advantages over deceased donation in liver transplantation? J Gastroenterol Hepatol. 2010;25:1598-1603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
29.  Strong RW. Living-donor liver transplantation: an overview. J Hepatobiliary Pancreat Surg. 2006;13:370-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
30.  Hwang S, Lee SG, Jang SJ, Cho SH, Kim KH, Ahn CS, Moon DB, Ha TY. The effect of donor weight reduction on hepatic steatosis for living donor liver transplantation. Liver Transpl. 2004;10:721-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 67]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
31.  Chan HL, de Silva HJ, Leung NW, Lim SG, Farrell GC. How should we manage patients with non-alcoholic fatty liver disease in 2007? J Gastroenterol Hepatol. 2007;22:801-808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 46]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
32.  Oshita A, Tashiro H, Amano H, Kobayashi T, Onoe T, Ide K, Takaki S, Takahashi S, Arihiro K, Chayama K. Safety and feasibility of diet-treated donors with steatotic livers at the initial consultation for living-donor liver transplantation. Transplantation. 2012;93:1024-1030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
33.  Nakamuta M, Morizono S, Soejima Y, Yoshizumi T, Aishima S, Takasugi S, Yoshimitsu K, Enjoji M, Kotoh K, Taketomi A. Short-term intensive treatment for donors with hepatic steatosis in living-donor liver transplantation. Transplantation. 2005;80:608-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 80]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
34.  Caraceni P, Domenicali M, Maria Pertosa A, Maiolini E, Grattagliano I, Principe A, Palasciano G, Trevisani F, Bernardi M. The nutritional status modulates preservation-reperfusion injury in rat fatty liver. J Surg Res. 2005;127:190-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
35.  El-Badry AM, Moritz W, Contaldo C, Tian Y, Graf R, Clavien PA. Prevention of reperfusion injury and microcirculatory failure in macrosteatotic mouse liver by omega-3 fatty acids. Hepatology. 2007;45:855-863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 87]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
36.  Masterton GS, Plevris JN, Hayes PC. Review article: omega-3 fatty acids - a promising novel therapy for non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2010;31:679-692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 130]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
37.  Vuppalanchi R, Cummings OW, Saxena R, Ulbright TM, Martis N, Jones DR, Bansal N, Chalasani N. Relationship among histologic, radiologic, and biochemical assessments of hepatic steatosis: a study of human liver samples. J Clin Gastroenterol. 2007;41:206-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 43]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
38.  Sekiya M, Yahagi N, Matsuzaka T, Najima Y, Nakakuki M, Nagai R, Ishibashi S, Osuga J, Yamada N, Shimano H. Polyunsaturated fatty acids ameliorate hepatic steatosis in obese mice by SREBP-1 suppression. Hepatology. 2003;38:1529-1539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 249]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
39.  Schmöcker C, Weylandt KH, Kahlke L, Wang J, Lobeck H, Tiegs G, Berg T, Kang JX. Omega-3 fatty acids alleviate chemically induced acute hepatitis by suppression of cytokines. Hepatology. 2007;45:864-869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 127]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
40.  Stulnig TM. Immunomodulation by polyunsaturated fatty acids: mechanisms and effects. Int Arch Allergy Immunol. 2003;132:310-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 140]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
41.  Levy JR, Clore JN, Stevens W. Dietary n-3 polyunsaturated fatty acids decrease hepatic triglycerides in Fischer 344 rats. Hepatology. 2004;39:608-616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 117]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
42.  Alwayn IP, Andersson C, Zauscher B, Gura K, Nosé V, Puder M. Omega-3 fatty acids improve hepatic steatosis in a murine model: potential implications for the marginal steatotic liver donor. Transplantation. 2005;79:606-608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 58]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
43.  El-Badry AM, Jang JH, Elsherbiny A, Contaldo C, Tian Y, Raptis DA, Laczko E, Moritz W, Graf R, Clavien PA. Chemical composition of hepatic lipids mediates reperfusion injury of the macrosteatotic mouse liver through thromboxane A(2). J Hepatol. 2011;55:1291-1299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
44.  Araya J, Rodrigo R, Videla LA, Thielemann L, Orellana M, Pettinelli P, Poniachik J. Increase in long-chain polyunsaturated fatty acid n - 6/n - 3 ratio in relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin Sci (Lond). 2004;106:635-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 499]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
45.  Capanni M, Calella F, Biagini MR, Genise S, Raimondi L, Bedogni G, Svegliati-Baroni G, Sofi F, Milani S, Abbate R. Prolonged n-3 polyunsaturated fatty acid supplementation ameliorates hepatic steatosis in patients with non-alcoholic fatty liver disease: a pilot study. Aliment Pharmacol Ther. 2006;23:1143-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 293]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
46.  Zhu FS, Liu S, Chen XM, Huang ZG, Zhang DW. Effects of n-3 polyunsaturated fatty acids from seal oils on nonalcoholic fatty liver disease associated with hyperlipidemia. World J Gastroenterol. 2008;14:6395-6400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 137]  [Cited by in F6Publishing: 136]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
47.  Banasch M, Goetze O, Schmidt WE, Meier JJ. Rimonabant as a novel therapeutic option for nonalcoholic steatohepatitis. Liver Int. 2007;27:1152-1155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
48.  Roth JD, Trevaskis JL, Wilson J, Lei C, Athanacio J, Mack C, Kesty NC, Coffey T, Weyer C, Parkes DG. Antiobesity effects of the beta-cell hormone amylin in combination with phentermine or sibutramine in diet-induced obese rats. Int J Obes (Lond). 2008;32:1201-1210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
49.  Harrison SA, Fecht W, Brunt EM, Neuschwander-Tetri BA. Orlistat for overweight subjects with nonalcoholic steatohepatitis: A randomized, prospective trial. Hepatology. 2009;49:80-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 313]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
50.  Wierzbicki AS, Mikhailidis DP, Wray R, Schacter M, Cramb R, Simpson WG, Byrne CB. Statin-fibrate combination: therapy for hyperlipidemia: a review. Curr Med Res Opin. 2003;19:155-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 98]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
51.  Argo CK, Loria P, Caldwell SH, Lonardo A. Statins in liver disease: a molehill, an iceberg, or neither? Hepatology. 2008;48:662-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 84]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
52.  Merat S, Aduli M, Kazemi R, Sotoudeh M, Sedighi N, Sohrabi M, Malekzadeh R. Liver histology changes in nonalcoholic steatohepatitis after one year of treatment with probucol. Dig Dis Sci. 2008;53:2246-2250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 25]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
53.  Abdelmalek MF, Angulo P, Jorgensen RA, Sylvestre PB, Lindor KD. Betaine, a promising new agent for patients with nonalcoholic steatohepatitis: results of a pilot study. Am J Gastroenterol. 2001;96:2711-2717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 307]  [Cited by in F6Publishing: 283]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
54.  Marchesini G, Brizi M, Bianchi G, Tomassetti S, Zoli M, Melchionda N. Metformin in non-alcoholic steatohepatitis. Lancet. 2001;358:893-894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 502]  [Cited by in F6Publishing: 469]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
55.  Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, Balas B, Gastaldelli A, Tio F, Pulcini J. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med. 2006;355:2297-2307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1307]  [Cited by in F6Publishing: 1235]  [Article Influence: 68.6]  [Reference Citation Analysis (0)]
56.  Yokohama S, Yoneda M, Haneda M, Okamoto S, Okada M, Aso K, Hasegawa T, Tokusashi Y, Miyokawa N, Nakamura K. Therapeutic efficacy of an angiotensin II receptor antagonist in patients with nonalcoholic steatohepatitis. Hepatology. 2004;40:1222-1225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 342]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
57.  Tushuizen ME, Bunck MC, Pouwels PJ, van Waesberghe JH, Diamant M, Heine RJ. Incretin mimetics as a novel therapeutic option for hepatic steatosis. Liver Int. 2006;26:1015-1017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
58.  Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, Rakela J, McGill DB. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology. 1996;23:1464-1467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 396]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
59.  Lindor KD, Kowdley KV, Heathcote EJ, Harrison ME, Jorgensen R, Angulo P, Lymp JF, Burgart L, Colin P. Ursodeoxycholic acid for treatment of nonalcoholic steatohepatitis: results of a randomized trial. Hepatology. 2004;39:770-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 516]  [Cited by in F6Publishing: 463]  [Article Influence: 23.2]  [Reference Citation Analysis (0)]
60.  Akriviadis E, Botla R, Briggs W, Han S, Reynolds T, Shakil O. Pentoxifylline improves short-term survival in severe acute alcoholic hepatitis: a double-blind, placebo-controlled trial. Gastroenterology. 2000;119:1637-1648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 584]  [Cited by in F6Publishing: 485]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
61.  Satapathy SK, Garg S, Chauhan R, Sakhuja P, Malhotra V, Sharma BC, Sarin SK. Beneficial effects of tumor necrosis factor-alpha inhibition by pentoxifylline on clinical, biochemical, and metabolic parameters of patients with nonalcoholic steatohepatitis. Am J Gastroenterol. 2004;99:1946-1952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 145]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
62.  Day CP. Treatment of alcoholic liver disease. Liver Transpl. 2007;13:S69-S75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 35]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
63.  Koppe SW, Sahai A, Malladi P, Whitington PF, Green RM. Pentoxifylline attenuates steatohepatitis induced by the methionine choline deficient diet. J Hepatol. 2004;41:592-598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 100]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
64.  Peters JM, Aoyama T, Burns AM, Gonzalez FJ. Bezafibrate is a dual ligand for PPARalpha and PPARbeta: studies using null mice. Biochim Biophys Acta. 2003;1632:80-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 79]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
65.  Nagasawa T, Inada Y, Nakano S, Tamura T, Takahashi T, Maruyama K, Yamazaki Y, Kuroda J, Shibata N. Effects of bezafibrate, PPAR pan-agonist, and GW501516, PPARdelta agonist, on development of steatohepatitis in mice fed a methionine- and choline-deficient diet. Eur J Pharmacol. 2006;536:182-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 169]  [Article Influence: 9.4]  [Reference Citation Analysis (1)]
66.  Florentin M, Liberopoulos EN, Mikhailidis DP, Elisaf MS. Fibrate-associated adverse effects beyond muscle and liver toxicity. Curr Pharm Des. 2008;14:574-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
67.  Chalasani N. Statins and hepatotoxicity: focus on patients with fatty liver. Hepatology. 2005;41:690-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 200]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
68.  Desai KK, Dikdan GS, Shareef A, Koneru B. Ischemic preconditioning of the liver: a few perspectives from the bench to bedside translation. Liver Transpl. 2008;14:1569-1577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
69.  Zager RA, Baltes LA, Sharma HM, Jurkowitz MS. Responses of the ischemic acute renal failure kidney to additional ischemic events. Kidney Int. 1984;26:689-700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 99]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
70.  Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74:1124-1136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5406]  [Cited by in F6Publishing: 5434]  [Article Influence: 143.0]  [Reference Citation Analysis (0)]
71.  Petrowsky H, McCormack L, Trujillo M, Selzner M, Jochum W, Clavien PA. A prospective, randomized, controlled trial comparing intermittent portal triad clamping versus ischemic preconditioning with continuous clamping for major liver resection. Ann Surg. 2006;244:921-928; discussion 921-928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 170]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
72.  Clavien PA, Selzner M, Rüdiger HA, Graf R, Kadry Z, Rousson V, Jochum W. A prospective randomized study in 100 consecutive patients undergoing major liver resection with versus without ischemic preconditioning. Ann Surg. 2003;238:843-850; discussion 851-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 391]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
73.  Franchello A, Gilbo N, David E, Ricchiuti A, Romagnoli R, Cerutti E, Salizzoni M. Ischemic preconditioning (IP) of the liver as a safe and protective technique against ischemia/reperfusion injury (IRI). Am J Transplant. 2009;9:1629-1639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
74.  Peralta C, Hotter G, Closa D, Gelpí E, Bulbena O, Roselló-Catafau J. Protective effect of preconditioning on the injury associated to hepatic ischemia-reperfusion in the rat: role of nitric oxide and adenosine. Hepatology. 1997;25:934-937.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 271]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
75.  Arai M, Thurman RG, Lemasters JJ. Contribution of adenosine A(2) receptors and cyclic adenosine monophosphate to protective ischemic preconditioning of sinusoidal endothelial cells against Storage/Reperfusion injury in rat livers. Hepatology. 2000;32:297-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 94]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
76.  Jimenez-Castro MB, Casillas-Ramirez A, Massip-Salcedo M, Elias-Miro M, Serafin A, Rimola A, Rodes J, Peralta C. Cyclic adenosine 3’,5’-monophosphate in rat steatotic liver transplantation. Liver Transpl. 2011;17:1099-1110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
77.  Serafín A, Roselló-Catafau J, Prats N, Gelpí E, Rodés J, Peralta C. Ischemic preconditioning affects interleukin release in fatty livers of rats undergoing ischemia/reperfusion. Hepatology. 2004;39:688-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 88]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
78.  Peralta C, Bartrons R, Serafin A, Blázquez C, Guzmán M, Prats N, Xaus C, Cutillas B, Gelpí E, Roselló-Catafau J. Adenosine monophosphate-activated protein kinase mediates the protective effects of ischemic preconditioning on hepatic ischemia-reperfusion injury in the rat. Hepatology. 2001;34:1164-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 148]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
79.  Carrasco-Chaumel E, Roselló-Catafau J, Bartrons R, Franco-Gou R, Xaus C, Casillas A, Gelpí E, Rodés J, Peralta C. Adenosine monophosphate-activated protein kinase and nitric oxide in rat steatotic liver transplantation. J Hepatol. 2005;43:997-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
80.  Bouma HR, Ketelaar ME, Yard BA, Ploeg RJ, Henning RH. AMP-activated protein kinase as a target for preconditioning in transplantation medicine. Transplantation. 2010;90:353-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
81.  Cutrn JC, Perrelli MG, Cavalieri B, Peralta C, Rosell Catafau J, Poli G. Microvascular dysfunction induced by reperfusion injury and protective effect of ischemic preconditioning. Free Radic Biol Med. 2002;33:1200-1208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 124]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
82.  Koti RS, Yang W, Glantzounis G, Quaglia A, Davidson BR, Seifalian AM. Effect of ischaemic preconditioning on hepatic oxygenation, microcirculation and function in a rat model of moderate hepatic steatosis. Clin Sci (Lond). 2005;108:55-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
83.  Rolo AP, Teodoro JS, Peralta C, Rosello-Catafau J, Palmeira CM. Prevention of I/R injury in fatty livers by ischemic preconditioning is associated with increased mitochondrial tolerance: the key role of ATPsynthase and mitochondrial permeability transition. Transpl Int. 2009;22:1081-1090.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
84.  Hafez TS, Glantzounis GK, Fusai G, Taanman JW, Wignarajah P, Parkes H, Fuller B, Davidson BR, Seifalian AM. Intracellular oxygenation and cytochrome oxidase C activity in ischemic preconditioning of steatotic rabbit liver. Am J Surg. 2010;200:507-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
85.  Fernández L, Carrasco-Chaumel E, Serafín A, Xaus C, Grande L, Rimola A, Roselló-Catafau J, Peralta C. Is ischemic preconditioning a useful strategy in steatotic liver transplantation? Am J Transplant. 2004;4:888-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 60]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
86.  Peralta C, Bulbena O, Xaus C, Prats N, Cutrin JC, Poli G, Gelpi E, Roselló-Catafau J. Ischemic preconditioning: a defense mechanism against the reactive oxygen species generated after hepatic ischemia reperfusion. Transplantation. 2002;73:1203-1211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 85]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
87.  Peralta C, Prats N, Xaus C, Gelpí E, Roselló-Catafau J. Protective effect of liver ischemic preconditioning on liver and lung injury induced by hepatic ischemia-reperfusion in the rat. Hepatology. 1999;30:1481-1489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 118]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
88.  Lai IR, Chang KJ, Chen CF, Tsai HW. Transient limb ischemia induces remote preconditioning in liver among rats: the protective role of heme oxygenase-1. Transplantation. 2006;81:1311-1317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 88]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
89.  Wang F, Birch SE, He R, Tawadros P, Szaszi K, Kapus A, Rotstein OD. Remote ischemic preconditioning by hindlimb occlusion prevents liver ischemic/reperfusion injury: the role of High Mobility Group-Box 1. Ann Surg. 2010;251:292-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
90.  Abu-Amara M, Yang SY, Quaglia A, Rowley P, Fuller B, Seifalian A, Davidson B. Role of endothelial nitric oxide synthase in remote ischemic preconditioning of the mouse liver. Liver Transpl. 2011;17:610-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 53]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
91.  Beck-Schimmer B, Breitenstein S, Urech S, De Conno E, Wittlinger M, Puhan M, Jochum W, Spahn DR, Graf R, Clavien PA. A randomized controlled trial on pharmacological preconditioning in liver surgery using a volatile anesthetic. Ann Surg. 2008;248:909-918.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 186]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
92.  Hong F, Radaeva S, Pan HN, Tian Z, Veech R, Gao B. Interleukin 6 alleviates hepatic steatosis and ischemia/reperfusion injury in mice with fatty liver disease. Hepatology. 2004;40:933-941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 153]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
93.  Hasegawa T, Ito Y, Wijeweera J, Liu J, Malle E, Farhood A, McCuskey RS, Jaeschke H. Reduced inflammatory response and increased microcirculatory disturbances during hepatic ischemia-reperfusion injury in steatotic livers of ob/ob mice. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1385-G1395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
94.  Soltys K, Dikdan G, Koneru B. Oxidative stress in fatty livers of obese Zucker rats: rapid amelioration and improved tolerance to warm ischemia with tocopherol. Hepatology. 2001;34:13-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 87]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
95.  Ben Mosbah I, Massip-Salcedo M, Fernández-Monteiro I, Xaus C, Bartrons R, Boillot O, Roselló-Catafau J, Peralta C. Addition of adenosine monophosphate-activated protein kinase activators to University of Wisconsin solution: a way of protecting rat steatotic livers. Liver Transpl. 2007;13:410-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
96.  Nagrath D, Xu H, Tanimura Y, Zuo R, Berthiaume F, Avila M, Yarmush R, Yarmush ML. Metabolic preconditioning of donor organs: defatting fatty livers by normothermic perfusion ex vivo. Metab Eng. 2009;11:274-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 111]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
97.  Amersi F, Shen XD, Moore C, Melinek J, Busuttil RW, Kupiec-Weglinski JW, Coito AJ. Fibronectin-alpha 4 beta 1 integrin-mediated blockade protects genetically fat Zucker rat livers from ischemia/reperfusion injury. Am J Pathol. 2003;162:1229-1239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
98.  Hayashi K, Aoki T, Jin Z, Wang H, Nishino N, Kusano T, Yasuda D, Koizumi T, Enami Y, Odaira M. Hepatocyte transplantation from steatotic liver in a rat model. J Surg Res. 2007;142:104-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
99.  Arnault I, Bao YM, Sebagh M, Anjo A, Dimicoli JL, Lemoine A, Delvart V, Adam R. Beneficial effect of pentoxifylline on microvesicular steatotic livers submitted to a prolonged cold ischemia. Transplantation. 2003;76:77-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
100.  Sampey BP, Vanhoose AM, Winfield HM, Freemerman AJ, Muehlbauer MJ, Fueger PT, Newgard CB, Makowski L. Cafeteria diet is a robust model of human metabolic syndrome with liver and adipose inflammation: comparison to high-fat diet. Obesity (Silver Spring). 2011;19:1109-1117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 391]  [Cited by in F6Publishing: 411]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
101.  Pasarín M, La Mura V, Gracia-Sancho J, García-Calderó H, Rodríguez-Vilarrupla A, García-Pagán JC, Bosch J, Abraldes JG. Sinusoidal endothelial dysfunction precedes inflammation and fibrosis in a model of NAFLD. PLoS One. 2012;7:e32785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 164]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
102.  Ghoshal AK, Farber E. The induction of liver cancer by dietary deficiency of choline and methionine without added carcinogens. Carcinogenesis. 1984;5:1367-1370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 178]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
103.  Takeda Y, Arii S, Kaido T, Niwano M, Moriga T, Mori A, Hanaki K, Gorrin-Rivas MJ, Ishii T, Sato M. Morphologic alteration of hepatocytes and sinusoidal endothelial cells in rat fatty liver during cold preservation and the protective effect of hepatocyte growth factor. Transplantation. 1999;67:820-828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
104.  Delzenne NM, Hernaux NA, Taper HS. A new model of acute liver steatosis induced in rats by fasting followed by refeeding a high carbohydrate-fat free diet. Biochemical and morphological analysis. J Hepatol. 1997;26:880-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
105.  Minor T, Akbar S, Tolba R, Dombrowski F. Cold preservation of fatty liver grafts: prevention of functional and ultrastructural impairments by venous oxygen persufflation. J Hepatol. 2000;32:105-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 86]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
106.  Tolba RH, Pütz U, Decker D, Dombrowski F, Lauschke H. L-carnitine ameliorates abnormal vulnerability of steatotic rat livers to cold ischemic preservation. Transplantation. 2003;76:1681-1686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
107.  Takahashi K, Hakamada K, Totsuka E, Umehara Y, Sasaki M. Warm ischemia and reperfusion injury in diet-induced canine fatty livers. Transplantation. 2000;69:2028-2034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
108.  Lee L, Alloosh M, Saxena R, Van Alstine W, Watkins BA, Klaunig JE, Sturek M, Chalasani N. Nutritional model of steatohepatitis and metabolic syndrome in the Ossabaw miniature swine. Hepatology. 2009;50:56-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 157]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
109.  Jamieson RW, Zilvetti M, Roy D, Hughes D, Morovat A, Coussios CC, Friend PJ. Hepatic steatosis and normothermic perfusion-preliminary experiments in a porcine model. Transplantation. 2011;92:289-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 115]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
110.  Lieber CS, DeCarli LM. The feeding of ethanol in liquid diets. Alcohol Clin Exp Res. 1986;10:550-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 194]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
111.  Banerjee A, Russell WK, Jayaraman A, Ramaiah SK. Identification of proteins to predict the molecular basis for the observed gender susceptibility in a rat model of alcoholic steatohepatitis by 2-D gel proteomics. Proteomics. 2008;8:4327-4337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
112.  Song BJ, Moon KH, Olsson NU, Salem N. Prevention of alcoholic fatty liver and mitochondrial dysfunction in the rat by long-chain polyunsaturated fatty acids. J Hepatol. 2008;49:262-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 86]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
113.  Park SH, Choi MS, Park T. Changes in the hepatic gene expression profile in a rat model of chronic ethanol treatment. Food Chem Toxicol. 2008;46:1378-1388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
114.  Tipoe GL, Liong EC, Leung TM, Nanji AA. A voluntary oral-feeding rat model for pathological alcoholic liver injury. Methods Mol Biol. 2008;447:11-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
115.  Polavarapu R, Spitz DR, Sim JE, Follansbee MH, Oberley LW, Rahemtulla A, Nanji AA. Increased lipid peroxidation and impaired antioxidant enzyme function is associated with pathological liver injury in experimental alcoholic liver disease in rats fed diets high in corn oil and fish oil. Hepatology. 1998;27:1317-1323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 234]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
116.  Sharma MR, Polavarapu R, Roseman D, Patel V, Eaton E, Kishor PB, Nanji AA. Increased severity of alcoholic liver injury in female verses male rats: a microarray analysis. Exp Mol Pathol. 2008;84:46-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
117.  Zhong Z, Connor HD, Froh M, Lind H, Bunzendahl H, Mason RP, Thurman RG, Lemasters JJ. Polyphenols from Camellia sinenesis prevent primary graft failure after transplantation of ethanol-induced fatty livers from rats. Free Radic Biol Med. 2004;36:1248-1258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
118.  Ye S, Dong J, Han B. Protective effect of reduced glutathione and venous systemic oxygen persufflation on rat steatotic graft following liver transplantation. J Surg Res. 2010;158:138-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
119.  Villanueva JA, Esfandiari F, White ME, Devaraj S, French SW, Halsted CH. S-adenosylmethionine attenuates oxidative liver injury in micropigs fed ethanol with a folate-deficient diet. Alcohol Clin Exp Res. 2007;31:1934-1943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
120.  Zivkovic AM, Bruce German J, Esfandiari F, Halsted CH. Quantitative lipid metabolomic changes in alcoholic micropigs with fatty liver disease. Alcohol Clin Exp Res. 2009;33:751-758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
121.  Katori M, Busuttil RW, Kupiec-Weglinski JW. Heme oxygenase-1 system in organ transplantation. Transplantation. 2002;74:905-912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 168]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
122.  Kim SJ, Park JG, Lee SM. Protective effect of heme oxygenase-1 induction against hepatic injury in alcoholic steatotic liver exposed to cold ischemia/reperfusion. Life Sci. 2012;90:169-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
123.  Coito AJ, Buelow R, Shen XD, Amersi F, Moore C, Volk HD, Busuttil RW, Kupiec-Weglinski JW. Heme oxygenase-1 gene transfer inhibits inducible nitric oxide synthase expression and protects genetically fat Zucker rat livers from ischemia-reperfusion injury. Transplantation. 2002;74:96-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 123]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
124.  Nakano H, Nagasaki H, Barama A, Boudjema K, Jaeck D, Kumada K, Tatsuno M, Baek Y, Kitamura N, Suzuki T. The effects of N-acetylcysteine and anti-intercellular adhesion molecule-1 monoclonal antibody against ischemia-reperfusion injury of the rat steatotic liver produced by a choline-methionine-deficient diet. Hepatology. 1997;26:670-678.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Selzner M, Graf R, Clavien PA. IL-6: a magic potion for liver transplantation? Gastroenterology. 2003;125:256-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
126.  Luo XY, Takahara T, Hou J, Kawai K, Sugiyama T, Tsukada K, Takemoto M, Takeuchi M, Zhong L, Li XK. Theaflavin attenuates ischemia-reperfusion injury in a mouse fatty liver model. Biochem Biophys Res Commun. 2012;417:287-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
127.  von Heesen M, Seibert K, Hülser M, Scheuer C, Wagner M, Menger MD, Schilling MK, Moussavian MR. Multidrug donor preconditioning protects steatotic liver grafts against ischemia-reperfusion injury. Am J Surg. 2012;203:168-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
128.  Yamagami K, Enders G, Schauer RJ, Leiderer R, Hutter J, Yamamoto Y, Yamaoka Y, Hammer C, Messmer K. Heat-shock preconditioning protects fatty livers in genetically obese Zucker rats from microvascular perfusion failure after ischemia reperfusion. Transpl Int. 2003;16:456-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
129.  Mokuno Y, Berthiaume F, Tompkins RG, Balis UJ, Yarmush ML. Technique for expanding the donor liver pool: heat shock preconditioning in a rat fatty liver model. Liver Transpl. 2004;10:264-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
130.  Mokuno Y, Berthiaume F, Tanimura Y, Yarmush ML. Heat shock preconditioning inhibits CD4+ T lymphocyte activation in transplanted fatty rat livers. J Surg Res. 2006;135:92-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
131.  Watanabe J, Kushihata F, Matsumoto K, Honda K, Matsuda S, Kobayashi N. Downregulation of cytokine release by heat preconditioning of livers used for transplantation in rats. Dig Dis Sci. 2005;50:1823-1828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
132.  Yonezawa K, Yamamoto Y, Yamamoto H, Ishikawa Y, Uchinami H, Taura K, Nakajima A, Yamaoka Y. Suppression of tumor necrosis factor-alpha production and neutrophil infiltration during ischemia-reperfusion injury of the liver after heat shock preconditioning. J Hepatol. 2001;35:619-627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
133.  Mangus RS, Tector AJ, Agarwal A, Vianna R, Murdock P, Fridell JA. Comparison of histidine-tryptophan-ketoglutarate solution (HTK) and University of Wisconsin solution (UW) in adult liver transplantation. Liver Transpl. 2006;12:226-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 86]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
134.  Pokorny H, Rasoul-Rockenschaub S, Langer F, Windhager T, Rosenstingl A, Lange R, Königsrainer A, Ringe B, Mühlbacher F, Steininger R. Histidine-tryptophan-ketoglutarate solution for organ preservation in human liver transplantation-a prospective multi-centre observation study. Transpl Int. 2004;17:256-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
135.  Feng L, Zhao N, Yao X, Sun X, Du L, Diao X, Li S, Li Y. Histidine-tryptophan-ketoglutarate solution vs. University of Wisconsin solution for liver transplantation: a systematic review. Liver Transpl. 2007;13:1125-1136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 63]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
136.  Lopez-Andujar R, Deusa S, Montalvá E, San Juan F, Moya A, Pareja E, DeJuan M, Berenguer M, Prieto M, Mir J. Comparative prospective study of two liver graft preservation solutions: University of Wisconsin and Celsior. Liver Transpl. 2009;15:1709-1717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
137.  García-Gil FA, Serrano MT, Fuentes-Broto L, Arenas J, García JJ, Güemes A, Bernal V, Campillo A, Sostres C, Araiz JJ. Celsior versus University of Wisconsin preserving solutions for liver transplantation: postreperfusion syndrome and outcome of a 5-year prospective randomized controlled study. World J Surg. 2011;35:1598-1607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
138.  Dondéro F, Paugam-Burtz C, Danjou F, Stocco J, Durand F, Belghiti J. A randomized study comparing IGL-1 to the University of Wisconsin preservation solution in liver transplantation. Ann Transplant. 2010;15:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Ben Mosbah I, Roselló-Catafau J, Franco-Gou R, Abdennebi HB, Saidane D, Ramella-Virieux S, Boillot O, Peralta C. Preservation of steatotic livers in IGL-1 solution. Liver Transpl. 2006;12:1215-1223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 77]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
140.  Belzer FO, Southard JH. Principles of solid-organ preservation by cold storage. Transplantation. 1988;45:673-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 986]  [Cited by in F6Publishing: 932]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
141.  Maathuis MH, Leuvenink HG, Ploeg RJ. Perspectives in organ preservation. Transplantation. 2007;83:1289-1298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 158]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
142.  Bessems M, Doorschodt BM, van Marle J, Vreeling H, Meijer AJ, van Gulik TM. Improved machine perfusion preservation of the non-heart-beating donor rat liver using Polysol: a new machine perfusion preservation solution. Liver Transpl. 2005;11:1379-1388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
143.  Vairetti M, Ferrigno A, Carlucci F, Tabucchi A, Rizzo V, Boncompagni E, Neri D, Gringeri E, Freitas I, Cillo U. Subnormothermic machine perfusion protects steatotic livers against preservation injury: a potential for donor pool increase? Liver Transpl. 2009;15:20-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 86]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
144.  Sun Z, Klein AS, Radaeva S, Hong F, El-Assal O, Pan HN, Jaruga B, Batkai S, Hoshino S, Tian Z. In vitro interleukin-6 treatment prevents mortality associated with fatty liver transplants in rats. Gastroenterology. 2003;125:202-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 110]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
145.  Atila K, Coker A, Sagol O, Coker I, Topalak O, Astarcioglu H, Karademir S, Astarcioglu I. Protective effects of carnitine in an experimental ischemia-reperfusion injury. Clin Nutr. 2002;21:309-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
146.  Ben Mosbah I, Roselló-Catafau J, Alfany-Fernandez I, Rimola A, Parellada PP, Mitjavila MT, Lojek A, Ben Abdennebi H, Boillot O, Rodés J. Addition of carvedilol to University Wisconsin solution improves rat steatotic and nonsteatotic liver preservation. Liver Transpl. 2010;16:163-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
147.  Oliveira PJ, Gonçalves L, Monteiro P, Providencia LA, Moreno AJ. Are the antioxidant properties of carvedilol important for the protection of cardiac mitochondria? Curr Vasc Pharmacol. 2005;3:147-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
148.  Zaouali MA, Padrissa-Altés S, Ben Mosbah I, Alfany-Fernandez I, Massip-Salcedo M, Casillas-Ramirez A, Bintanel-Morcillo M, Boillot O, Serafin A, Rimola A. Improved rat steatotic and nonsteatotic liver preservation by the addition of epidermal growth factor and insulin-like growth factor-I to University of Wisconsin solution. Liver Transpl. 2010;16:1098-1111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
149.  Mullonkal CJ, Toledo-Pereyra LH. Akt in ischemia and reperfusion. J Invest Surg. 2007;20:195-203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 132]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
150.  Zaoualí MA, Reiter RJ, Padrissa-Altés S, Boncompagni E, García JJ, Ben Abnennebi H, Freitas I, García-Gil FA, Rosello-Catafau J. Melatonin protects steatotic and nonsteatotic liver grafts against cold ischemia and reperfusion injury. J Pineal Res. 2011;50:213-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
151.  Minor T, Saad S, Nagelschmidt M, Kötting M, Fu Z, Paul A, Isselhard W. Successful transplantation of porcine livers after warm ischemic insult in situ and cold preservation including postconditioning with gaseous oxygen. Transplantation. 1998;65:1262-1264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
152.  Minor T, Stegemann J, Hirner A, Koetting M. Impaired autophagic clearance after cold preservation of fatty livers correlates with tissue necrosis upon reperfusion and is reversed by hypothermic reconditioning. Liver Transpl. 2009;15:798-805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
153.  St Peter SD, Imber CJ, Friend PJ. Liver and kidney preservation by perfusion. Lancet. 2002;359:604-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 157]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
154.  Monbaliu D, Brassil J. Machine perfusion of the liver: past, present and future. Curr Opin Organ Transplant. 2010;15:160-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 71]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
155.  Moers C, Smits JM, Maathuis MH, Treckmann J, van Gelder F, Napieralski BP, van Kasterop-Kutz M, van der Heide JJ, Squifflet JP, van Heurn E. Machine perfusion or cold storage in deceased-donor kidney transplantation. N Engl J Med. 2009;360:7-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 758]  [Cited by in F6Publishing: 712]  [Article Influence: 47.5]  [Reference Citation Analysis (0)]
156.  Guarrera JV, Henry SD, Samstein B, Odeh-Ramadan R, Kinkhabwala M, Goldstein MJ, Ratner LE, Renz JF, Lee HT, Brown RS. Hypothermic machine preservation in human liver transplantation: the first clinical series. Am J Transplant. 2010;10:372-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 387]  [Cited by in F6Publishing: 390]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
157.  Brockmann J, Reddy S, Coussios C, Pigott D, Guirriero D, Hughes D, Morovat A, Roy D, Winter L, Friend PJ. Normothermic perfusion: a new paradigm for organ preservation. Ann Surg. 2009;250:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 238]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
158.  Schön MR, Kollmar O, Wolf S, Schrem H, Matthes M, Akkoc N, Schnoy NC, Neuhaus P. Liver transplantation after organ preservation with normothermic extracorporeal perfusion. Ann Surg. 2001;233:114-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 201]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
159.  Fondevila C, Hessheimer AJ, Maathuis MH, Muñoz J, Taurá P, Calatayud D, Leuvenink H, Rimola A, Ploeg RJ, García-Valdecasas JC. Superior preservation of DCD livers with continuous normothermic perfusion. Ann Surg. 2011;254:1000-1007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 157]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
160.  Tolboom H, Pouw RE, Izamis ML, Milwid JM, Sharma N, Soto-Gutierrez A, Nahmias Y, Uygun K, Berthiaume F, Yarmush ML. Recovery of warm ischemic rat liver grafts by normothermic extracorporeal perfusion. Transplantation. 2009;87:170-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 73]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
161.  Tolboom H, Izamis ML, Sharma N, Milwid JM, Uygun B, Berthiaume F, Uygun K, Yarmush ML. Subnormothermic machine perfusion at both 20°C and 30°C recovers ischemic rat livers for successful transplantation. J Surg Res. 2012;175:149-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 83]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
162.  Berendsen TA, Bruinsma BG, Lee J, D’Andrea V, Liu Q, Izamis ML, Uygun K, Yarmush ML. A simplified subnormothermic machine perfusion system restores ischemically damaged liver grafts in a rat model of orthotopic liver transplantation. Transplant Res. 2012;1:6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
163.  Bessems M, Doorschodt BM, Kolkert JL, Vetelainen RL, van Vliet AK, Vreeling H, van Marle J, van Gulik TM. Preservation of steatotic livers: a comparison between cold storage and machine perfusion preservation. Liver Transpl. 2007;13:497-504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 79]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
164.  Boncompagni E, Gini E, Ferrigno A, Milanesi G, Gringeri E, Barni S, Cillo U, Vairetti M, Freitas I. Decreased apoptosis in fatty livers submitted to subnormothermic machine-perfusion respect to cold storage. Eur J Histochem. 2011;55:e40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
165.  Bezinover D, Ramamoorthy S, Uemura T, Kadry Z, McQuillan PM, Mets B, Falcucci O, Rannels S, Ruiz-Velasco V, Spiess B. Use of a third-generation perfluorocarbon for preservation of rat DCD liver grafts. J Surg Res. 2012;175:131-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]