Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Trisulfate Disaccharide Decreases Calcium Overload and Protects Liver Injury Secondary to Liver Ischemia/Reperfusion

Abstract

Background

Ischemia and reperfusion (I/R) causes tissue damage and intracellular calcium levels are a factor of cell death. Sodium calcium exchanger (NCX) regulates calcium extrusion and Trisulfated Disaccharide (TD) acts on NCX decreasing intracellular calcium through the inhibition of the exchange inhibitory peptide (XIP).

Objectives

The aims of this research are to evaluate TD effects in liver injury secondary to I/R in animals and in vitro action on cytosolic calcium of hepatocytes cultures under calcium overload.

Methods

Wistar rats submitted to partial liver ischemia were divided in groups: Control: (n = 10): surgical manipulation with no liver ischemia; Saline: (n = 15): rats receiving IV saline before reperfusion; and TD: (n = 15): rats receiving IV TD before reperfusion. Four hours after reperfusion, serum levels of AST, ALT, TNF-α, IL-6, and IL-10 were measured. Liver tissue samples were collected for mitochondrial function and malondialdehyde (MDA) content. Pulmonary vascular permeability and histologic parameters of liver were determined. TD effect on cytosolic calcium was evaluated in BRL3A hepatic rat cell cultures stimulated by thapsigargin pre and after treatment with TD.

Results

AST, ALT, cytokines, liver MDA, mitochondrial dysfunction and hepatic histologic injury scores were less in TD group when compared to Saline Group (p<0.05) with no differences in pulmonary vascular permeability. In culture cells, TD diminished the intracellular calcium raise and prevented the calcium increase pre and after treatment with thapsigargin, respectively.

Conclusion

TD decreases liver cell damage, preserves mitochondrial function and increases hepatic tolerance to I/R injury by calcium extrusion in Ca2+ overload situations.

Introduction

Ischemia-reperfusion (I/R) injury remains a major problem in various clinical situations, including trauma, shock, hemorrhage, liver resection, and transplantation [1, 2]. Current therapeutic procedures, such as ischemic pre or post conditioning and intermittent vascular occlusion, can minimize but not prevent hepatic I/R injury [36]. Up to the present time, although many compounds have been tested, only a few drugs have been shown to ameliorate or diminish liver I/R injury [78].

The pathophysiology of hepatic damage after I/R is not entirely understood, but the changes in intracellular calcium behavior has emerged as an important mechanism in the development of cell damage [9, 10]. It has been shown that inhibition of cellular calcium accumulation prevents hepatic I/R damage [11]. Furthermore, expression of calcium-sensing receptors plays a vital role in apoptosis induced by I/R injury [12] and a decrease in mitochondrial Ca2+ levels are associated with decreased apoptosis and cellular changes [13]. In addition, inhibition of intracellular calcium overloading effectively minimizes organ damage in models of liver I/R [14]. The intracellular calcium concentration is controlled by the membrane sodium-calcium exchanger (NCX), which decreases calcium by exchanging calcium for extracellular sodium. An endogenous regulatory peptide named Exchange Inhibitor Peptide (XIP) modulates the activity of NCX [15]. In contrast, the in vitro stimulation of lipid peroxidation in isolated rat hepatocytes increases cytosolic calcium levels by the influx of extracellular calcium especially in the absence of sodium. When sodium is added to the incubation medium, the influx of calcium induced by lipid peroxidation diminishes, preventing cell death, this process being mediated by NCX [16].

Trisulfated disaccharide (TD) affects NCX action in rabbit endothelial aortic cells by decreasing intracellular calcium through the inhibition of XIP [17]. However, the TD effect in the liver has never been previously investigated. Thus, the purposes of this study were to investigate the effects of the reduction of intracellular calcium concentration, determined by TD administration, on the hepatic injury of rats submitted to liver I/R and to demonstrate TD effect in vitro on hepatocyte cultures under calcium overload.

Materials and Methods

In vivo experiments

Animals.

Forty adult male Wistar rats weighing 250-300g housed in individual cages in a 12h dark-light controlled environment were used for the experimental protocol. Rats had free access to standard rat chow and water. The experimental protocol was approved by the Ethics Committee for Animal Research of the Medical School of São Paulo University (research protocol n.138/13 in 04/24/2013) and received humanized care according to the criteria outlined in the Guide for the Care and Use of Laboratory Animals (Institute of Laboratory Animal Resources, Commission on Life Sciences and National Research Council. National Academic Press, Washington, D.C., 1996).

Experimental design.

The animals were randomly submitted to the following experimental protocols:

  1. Control Group (CONTR): 10 animals submitted to surgical liver manipulation with no hepatic ischemia induction.
  2. Saline Group (SAL): 15 animals submitted to hepatic ischemia for one hour that received 0.4 ml of saline solution (0.9% NaCl) injected via the dorsal penile vein 10 minutes before liver reperfusion.
  3. Trisulfated disaccharide Group (TD): 15 animals submitted to hepatic ischemia for one hour that received 0.2 mg/kg of trisulfated disaccharide (UNIFESP, Molecular Biology Laboratory, São Paulo, Brazil) diluted in 0.4 ml of 0.9% NaCl, injected via the dorsal penile vein 10 minutes before liver reperfusion.

Surgical procedure.

Animals were anesthetized intraperitoneally with 30mg/kg of ketamine hydrochloride (Cristália, São Paulo, Brazil), and 30 mg/kg of xylazine (Bayer, São Paulo, Brazil), and kept under mechanical ventilation (Small Animal Ventilator model 683, Harvard Apparatus, Holliston, MA, USA) at a frequency of 60 cycles per minute and a tidal volume of 0.08 ml/kg. Animal’s body temperature was monitored throughout the whole procedure by a digital rectal thermometer (YSI Precision 4000A Thermometer, USA) and kept between 35 and 37°C through external heating by a halogen lamp (45 W and 127 V). The surgical procedure was performed through a 4 cm laparotomy extending caudally from the xiphoid process with dissection of the medium and lateral common hepatic pedicle in all the three experimental groups. Partial liver ischemia (70% of liver parenchyma) was produced in the SAL and TD groups for one hour by clamping the pedicles of the median and left lateral hepatic lobes. The abdominal incision was closed and reopened after 60 min for clamp removal and liver reperfusion [18].

Sample collection.

At 4 h after reperfusion, the animals were re-anesthetized as described previously and submitted to laparotomy for collection of blood and tissues according to the study design. The 4-hour period after the reperfusion has been chosen based on the results of several previous studies from our laboratory that could demonstrate a significant major hepatic damage at this time period compared to more prolonged time observation periods [19, 20]. Blood was collected by transdiaphragmatic cardiac puncture for determinations of AST, ALT, TNF-α, IL-6 and IL-10. Euthanasia was performed by transecting the abdominal aorta and inferior vena cava. Liver tissue was removed and samples of the ischemic lobes were obtained for assessments of mitochondrial oxidation and phosphorylation, malondialdehyde (MDA) contents, and histologic analysis. Pulmonary vascular permeability was determined in lung tissue by Evans blue (EB) dye extravasation.

Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels.

The extent of hepatocellular injury was evaluated by determination of serum AST and ALT levels. Quantification of enzyme activities was performed by optimized ultraviolet method (COBAS MIRA, Roche) and results expressed in U/L.

Liver mitochondrial oxidation and phosphorylation activities.

Liver mitochondrial function was studied in ischemic liver lobes separated after liver removal and immersed in homogenization solution for mitochondrial extraction as described previously [21].

Mitochondrial oxygen consumption (activated state, S3 and basal state, S4) was measured polarographically [22] using a Gilson 5/6H Oxygraph (Gilson Medical Electronics, Inc., Middleton, WI) in a closed reaction vessel fitted with a Clark oxygen electrode (Yellow Springs Instruments Co., Yellow Springs, OH) at 28°C, using succinate as respiratory substrate. The ratio of oxygen consumption in the presence of ADP to that in the absence of ADP (respiratory control rate, RCR) and the ADP/O ratio were calculated as indices of oxidative and phosphorylative mitochondrial functions. [23]

RCR = oxygen consumption in state 3 / oxygen consumption in state 4.

ADP/O = moles of ATP formed from ADP per atom of oxygen consumed.

Respiratory states 3 and 4 were measured and reported as nanogram atoms of oxygen per milligram mitochondrial protein per minute. Mitochondria protein content was determined by the method of Lowry et al [24].

Lipid peroxidation analysis.

Malondialdehyde (MDA) formation was used as indicative of the occurrence of lipid peroxidation in the tissues and was estimated as thiobarbituric acid-reactive substances (TBARS). Liver tissues (100 mg/ml) were homogenized in 1.15% KCl buffer and centrifuged at 14,000×g for 20 min. An aliquot of the supernatant was then added to a reaction mixture consisting of 1.5 ml 0.8% thiobarbituric acid, 200 μl 8.1% (v/v) sodium dodecyl sulfate, 1.5 ml 20% acetic acid (pH 3.5), and 600 μl distilled water. The mixture was then heated at 90°C for 45 min. After cooling to room temperature, samples were cleared by centrifugation (10,000×g for 10 min), and the absorbance was measured at 532 nm using malondialdehyde bis (dimethyl acetal) as external standard. The content of lipid peroxides was expressed as nmol MDA per mg of protein [25].

Determination of inflammatory mediators.

Serum levels of TNF-α, IL-6, and IL-10 were determined by ELISA using commercial kits (Invitrogen, CA, USA).

Lung tissue microvascular permeability analysis.

Lung microvascular permeability was quantified by the Evans blue dye (EBD) extravasation technique as described previously [26]. The results are expressed as microgram of EBD per gram of dry weight tissue. Expression of results as a function of dry weight avoids under evaluation due to edema formation.

Histologic analysis of the liver.

Liver samples were fixed in 10% buffered formalin for standard hematoxylin and eosin staining. Histologic evaluation of the liver sections was performed by the same blinded pathologist. The severity of histologic injury was analyzed according to the scoring system proposed by Quireze et al [27].

Trisulfated Dissacharide.

Enzymes of adapted Flavobacterium heparinun degraded heparin and its degradation products were separated by combined Sephadex-gel filtration, paper chromatography and chemically analysed. These products were identified as saturated disaccharides constituted of uronic acid and glucosamine and containing three sulphate residues with the same basic disaccharide units [28].

In vitro studies

Culture cells and TD.

BRL3A culture cells from Rattusnorvegicus liver (cell line that express NCX protein, the suggested target of TD) were grown in supplemented Dulbecco’s modified Eagle’s medium for 24h [29]. Then, were loaded with the fluorescent Ca2+ indicator fura-2/AM (4 μM) for 30 min at 25°C in a 5% CO2 atmosphere. Changes in cytosolic calcium levels were measured kinetically at 37°C for 15 min. The ratio images were obtained using Calcium Imaging mode in Leica Application Suite 3D Deconvolution & 3D Visualization software. Thapsigargin (4 μM) was added to the culture medium to promote a sustained raise in the cytosolic calcium level from endoplasmic reticulum. Calcium ionophore ionomycin (0.25 μM) was used at the end of each experiment to reach the maximal increase in cytosolic calcium. To evaluate the TD effect on cytosolic calcium level, 30 μM of TD was added to the medium 120 seconds before and after the thapsigargin addition. The experiment was repeated for three times under the same experimental conditions.

Statistical analysis.

Results are presented as mean values ± SEM. Continuous variables were compared using analysis of variance and post hoc testing with Tukey-Kramer multiple comparison test. Results from the histologic analysis were compared using the Kruskal-Wallis test and Mann-Whitney’s test. The GraphPad 6 Prism Software (GraphPad Software, San Diego, CA) was used for statistical analysis. The level of p<0.05 was considered as statistically significant.

Results

Animal experiments

There were no experimental animal deaths in the present study and all animals finished the experiments successfully.

Hepatic enzymes (Serum AST and ALT).

Four hours after reperfusion, serum levels of both AST and ALT were significantly increased in the SAL and TD groups when compared to the CONTR group. However, in the TD group AST and ALT serum levels were significantly lower than in the SAL group (Fig 1A and 1B).

thumbnail
Fig 1. Aminotransferases four hours after liver reperfusion.

1A –AST—aspartate aminotransferase; 1B –ALT—alanine aminotransferase CONTR—Control Group (n = 10); SAL—Saline Group (n = 15); TD—Trisulfated Disaccharide Group (n = 15). Data are expressed as mean values * (SAL vs TD and SAL vs CONTR) p<0.05.

https://doi.org/10.1371/journal.pone.0149630.g001

Liver mitochondrial oxidation and phosphorylation activities.

Four hours after reperfusion, there was a decrease in the oxygen consumption rate by liver mitochondria in state 3 (S3), in respiratory control ratio (RCR), and in ADP/O ratio in the SAL group. This decrease was not observed in TD and CONTR groups. No differences were found in state 4 (S4) between SAL and TD groups (Fig 2A, 2B, 2C and 2D).

thumbnail
Fig 2. Liver mitochondrial function.

2A –RCR—Respiratory control rate * (TD vs SAL) p<0.05; # (CONTR vs SAL) p<0,05; 2B—ADP/O—ADP/Oxygen ratio * (SAL vs TD and SAL vs CONTR) p<0.05; 2C –S3—State 3 respiration * (SAL vs TD and SAL vs CONTR) p<0.05; 2D –S4—State 4 respiration NS: No Significance. CONTR—Control Group (n = 10); SAL—Saline Group; TD (n = 15)—Trisulfated Disaccharide Group (n = 15). Data are expressed as mean values.

https://doi.org/10.1371/journal.pone.0149630.g002

Liver MDA and lung permeability.

At four hours after reperfusion, MDA content was significantly increased in the ischemic liver in the SAL group. MDA content was also increased in TD group but in a much lower extent, showing that TD significantly decreased the liver peroxidation induced by the I/R injury (Fig 3A). Lung microvascular permeability, as evaluated by EBD extravasation, was increased 4 h after reperfusion in the SAL and TD groups compared to the CONTR group, without significant difference between TD and SAL groups. (Fig 3B).

thumbnail
Fig 3. Liver MDA and Lung microvascular permeability (EBD).

3A –Liver MDA. Data are expressed as mean values * (SAL vs TD and SAL vs CONTR) p<0.05; 3B—Lung microvascular permeability (EBD) *(CONT vs SAL and CONTR vs TD) p<0.05. (TD vs SAL) NS. CONTR—Control Group; SAL (n = 10)–Saline Group (n = 15); TD—Trisulfated Disaccharide Group (n = 15). NS: No Significance.

https://doi.org/10.1371/journal.pone.0149630.g003

Inflammatory mediators.

A significant increase in serum levels of TNF-α, IL-6, and IL-10 was observed in the SAL and TD group compared to the CONTR group. However, serum levels of all three mediators were significantly lower in TD group when compared to the SAL group (Fig 4A, 4B and 4C). As expected, TNF-αcould not be detected in the serum of animals from CONTR group.

thumbnail
Fig 4. Serum levels of inflammatory mediators.

4A—TNF-α; 4B—IL-6; 4C—IL-10 * (TD vs SAL) p<0.05; # (CONTR vs SAL) p<0.05. CONTR—Control Group (n = 10); SAL—Saline Group (n = 15); TD—Trisulfated Disaccharide Group (n = 15). Data are expressed as mean values.

https://doi.org/10.1371/journal.pone.0149630.g004

Liver histologic analysis.

The histologic injury scores at 4 hours after liver reperfusion was decreased in TD group compared to the SAL group. Additionally, liver cell necrosis was significantly lower in the TD group when compared to the SAL group (Table 1 and Fig 5A, 5B and 5C).

thumbnail
Fig 5. Liver histology.

Liver cell necrosis was significantly lower in the TD group when compared to the SAL group. 5A– Control Group (n = 10); 5B –Saline Group (n = 15); 5C –Trisulfated Disaccharide Group (n = 15).

https://doi.org/10.1371/journal.pone.0149630.g005

In vitro studies

Cytosolic calcium levels under TD action in liver cell cultures.

Representative ratio images of BRL3A cells loaded with fura-2 are presented in Fig 6, panel A, with a scale color varying from blue (minimum) to pink (maximum). In panel B, the kinetics of the cytosolic calcium variation of each sample is shown. As expected, the addition of 4 μM thapsigargin promoted a sustained raise in the cytosolic calcium level in BRL3A liver cells (Fig 6, panel A). The addition of TD to the culture medium after the thapsigargin-induced cytosolic calcium increase transiently decreased intracellular calcium levels for about 250 seconds (Fig 6, panel B). In contrast, when TD was added prior to thapsigargin, just a slight decrease of intracellular calcium basal level was observed. However, TD addition prior thapsigargin resulted in a lesser extend of cytosolic calcium increase determined by thapsigargin. These results show that TD was able to decrease the intracellular calcium raise promoted by thapsigargin and to prevent it when added prior to the inducer.

thumbnail
Fig 6. TD effect on thapsigargin-induced cytosolic calcium increase in liver cells (n = 3).

Panel A—Images of 340/380 nm fluorescence ratio using a non-true color scale. Scale bars—50 μm; Time shown in white inside each frame; Panel B—Relative quantification of changes in calcium levels. Arrows indicate additions. THAP—4 μM thapsigargin; IONO—0.25 μM ionomycin; TD—30 μM trisulfated disaccharide.

https://doi.org/10.1371/journal.pone.0149630.g006

Discussion

Multiple clinical situations expose the liver to I/R injury. The pathogenesis of I/R damage is multifactorial [3032]. Altered cellular calcium handling is known to be of vital importance in the I/R injury. Many investigators agree that modulation of cellular calcium homeostasis represents a key strategy for preventing I/R injury [33].

Heparin compounds have been studied in I/R injury. Heparin administration in high doses has no beneficial effects and is associated with bleeding complications in liver and kidney injury on the rat experimental model of I/R [34]. In contrast, protective effects of heparin were demonstrated on hepatic I/R lesions in rabbits [35]. The heparin fragment Dalteparin reduces I/R-induced liver injury in rats by attenuating the inflammatory response due to its capacity to enhance endothelial production of prostacyclin via cyclooxygenase-1 activation, independently of its anticoagulant activity [36]. N-desulfated heparin prevents hepatic/renal damage induced by I/R injury without significant anticoagulant activity [37]. Enoxaparin has been shown to decrease hepatic necrosis in a rat model of cholestatic liver injury, but this same effect was not observed with heparin or tinzaparin [38]. The effect of different heparins on calcium extrusion depends on the correlation between the fragments molecular weight and calcium modulation [17]. Therefore, heparin compounds might be useful in liver protection but bleeding represents a limiting factor in the eventual clinical use.

TD is the smallest heparin fragment, 585 Da weight, with no anticoagulant effect and higher affinity to XIP than other heparin fragments [17]. It probably increases the activity of NCX in the liver tissue by inhibition of XIP, thus accelerating calcium extrusion in situations of calcium overload in a similar way as observed in muscle cell lines [17]. Therefore, it appears that XIP, a part of NCX structure, is the target for the effect of TD in the toxic accumulation of calcium that occurs during ischemia. Thus, it may have an eventual application in the clinical setting to protect cells in calcium overload situations.

The present study, based on a model of partial warm hepatic I/R injury, showed that TD pretreatment decreases hepatic I/R injury as evidenced by diminished AST and ALT levels as well as by attenuated histopathological alterations. Furthermore, TD pre-treatment suppressed the increases of inflammation serum markers (TNF-α, IL-6, IL-10) and hepatic lipid peroxidation associated with hepatic lesion after I/R. In addition, mitochondrial function was preserved under this situation. The protective mechanism involves the action of TD on intracellular calcium extrusion, as demonstrated in the Fura-2 in vitro experiments, since TD was able to decrease the intracellular calcium raise promoted by thapsigargin and to prevent it when added prior to the inducer, suggesting a better protection in the pretreatment setting. These findings are in agreement with the results observed in vivo, when liver protection was observed when TD infusion was performed 10 minutes before reperfusion.

In smooth muscle cell and, in other cell lines with NCX expression, TD decreases intracellular calcium in a dose-dependent manner via its binding to NCX [17]. In a human hepatoma cell line, it was proposed that tert-butyl hydroperoxide induced apoptosis and increased intracellular calcium concentration through the activation of an inhibitory mechanism on NCX [39].

The Inflammatory cytokines TNF-α and IL-6 and, the anti-inflammatory IL-10 are released after I/R injury [11]. Several reports indicate that Kupffer cells are activated during ischemia/reperfusion injury increasing the production of cytokines and ROS [40, 41]. In the present study, a significant decrease of serum levels of cytokines in the TD treated animals was observed (Fig 4A, 4B and 4C). This may reflect a reduction of hepatic lesion after the ischemia/ reperfusion injury. However, other inflammatory triggers, such as damage-associated molecular patterns (DAMPs), that are released from necrotic cells, may also contribute to inflammation associated with hepatic ischemia-reperfusion injury [42,43]. Excessive ROS signaling leads to cell death after I/R and, similarly to the ischemic accumulation of succinate, calcium also participates in the reactions responsible for an extensive ROS generation [44, 45].

An anti-inflammatory effect of the low molecular weight heparin enoxaparin was described on carbon tetrachloride induced hepatic necrosis and apoptosis in the liver of rats [46]. In the present study, TD pretreatment also suppressed the increases of TNF-α, IL-6, and IL-10 serum levels that occur after hepatic I/R, suggesting that modulation of the inflammatory response could be another mechanism of TD liver protection. This anti-inflammatory effect appears to be a local and immediate effect of TD since no distant benefit, as measured by pulmonary vascular permeability, was obtained after the four-hour observation period in this study. The decrease of IL-10 levels might be explained by the diminished cell damage determined by the TD pretreatment.

Lipid peroxidation, as evaluated by MDA, is a mechanism of cell injury mediated by reactive oxygen species (ROS). Both excessive ROS signaling and calcium overload lead to cell death after I/R as calcium participates in the reactions that generate ROS [44, 45]. Our findings showed that MDA levels in the ischemic liver lobe of TD pretreated animals were lower than saline treated rats also suggesting a liver protective effect of TD. This observation is consistent with the histologic observations that showed much less necrosis after I/R in the TD group. The sustained increase in cytosolic calcium is commonly accompanied by mitochondrial calcium uptake. Such increased mitochondrial calcium uptake results mitochondrial membrane permeabilization leading to caspase activation and disruption of multiple cellular functions [47]. Our assessment of mitochondrial function revealed the maintenance of oxygen consumption rates in TD animal group comparable to the control group and higher than the saline group. This mitochondrial protective effect of TD, also suggested by the histological findings, is possibly related to calcium extrusion via NCX.

Animal models are frequently used to study possible pharmacological agents to attenuate I/R in liver transplantation. A recent systematic search of the PubMed database for literature in which pharmacological agents against I/R injury have been studied using rat liver transplant models showed, by the results of a meta-analysis, that pharmacological treatment is effective to reduce I/R liver damage in hepatic transplantation [48]. Based on the experimental data that are available today, the authors propose that identified agents should be further evaluated in human liver transplantation. Considering the findings of the present study TD represents a promising agent that might be considered for evaluation in human liver protection. Although this rat I/R model may not be completely translatable to human clinical characteristics of hepatic I/R [49], it was purposely used in the present study with the intend of testing the eventual protective effect of TD under extreme liver damage conditions. In conclusion, the results of this study demonstrate that trisulfated disaccharide pretreatment in a rat model of liver I/R injury achieves hepatic protection, probably via modulation of intracellular calcium concentrations mediated by Na-Ca exchanger. This hypothesis was demonstrated by the acceleration of calcium extrusion and decrease of calcium levels in liver cell cultures pretreated with DT. More data are necessary to further elucidate the mechanisms involved before its application in the clinical setting is justified.

Author Contributions

Conceived and designed the experiments: ERV JEMC. Performed the experiments: ERV AMMC SNS RAP EEA TR. Analyzed the data: ERV JEMC AMMC RAP TR CMGG. Contributed reagents/materials/analysis tools: HBN MAL EC. Wrote the paper: ERV JEMC EC ILST LACD.

References

  1. 1. Toledo-Pereyra LH, Simmons RL, Najarian JS. Protection of the ischemic liver by donor pre-treatment before transplantation. Am J Surg 1975; 129:513–517. pmid:1093421
  2. 2. Moussa ME, Sarraf CE, Uemoto S, Sawada H, Habib NA. Effect of total hepatic vascular exclusion during liver resection on hepatic ultrastructure. Liver TransplSurg 1996; 2:461–467.
  3. 3. Crisostomo PR, Wairiuko GM, Wang M, Tsai BM, Morrell ED, Meldrum DR. Preconditioning versus post conditioning: mechanisms and therapeutic potentials. J Am CollSurg 2006; 202:797–812.
  4. 4. Coelho AM, Machado MC, Takahashi HK, Sampietre SN, Stefano JT, Leite AZ, et al. Ischemic preconditioning-like effect of polyunsaturated fatty acid-rich diet on hepatic ischemia/reperfusion injury. J GastrointestSurg 2011; 15:1679–1688.
  5. 5. Teixeira AR, Molan NT, Kubrusly MS, Bellodi-Privato M, Coelho AM, Leite KR, et al. Post conditioning ameliorates lipid peroxidation in liver ischemia-reperfusion injury in rats. Acta Cir Bras 2009; 24:52–56. pmid:19169543
  6. 6. Seabra AL, Savassi-Rocha PR, Vasconcelos AC, Lima AS, Rodrigues KC, Almeida HM. Ischemia/Reperfusion injury after continuous or intermittent hepatic pedicle clamping in rabbits. Arq Bras Cir Dig 2012; Apr-Jun; 25(2):105–9. pmid:23381753
  7. 7. Zhang WX, Zhou LF, Zhang L., Bao L, Wang CC, Meng HY, et al. Protective effects of glutamine preconditioning on ischemia-reperfusion injury in rats. Hepatobiliary Pancreat Dis Int 2011; 10: 78–82. pmid:21269939
  8. 8. Schwartz J, Holmuhamedov E, Zhang X, Lovelace GL, Smith CD, Lemasters JJ. Minocycline and doxycycline, but not other tetracycline-derived compounds, protect liver cells from chemical hypoxia and ischemia/reperfusion injury by inhibition of the mitochondrial calcium uniporter. Toxicol Appl Pharmacol 2013; 273: 172–179. pmid:24012766
  9. 9. Cámara-Lemarroy CR, Guzmán-de la Garza FJ, Alarcón-Galván G, Cordero-Pérez P, Muñoz-Espinosa L, Torres-González L, et al. Hepatic Ischemia/Reperfusion Injury Is Diminished by Atorvastatin in Wistar Rats. Arch Med Res. 2014; 45:210–216. pmid:24726586
  10. 10. Amberger A, Weiss H, Haller T, Köck G, Hermann M, Widschwendter M, et al. A subpopulation of mitochondria prevents cytosolic calcium overload in endothelial cells after cold ischemia/reperfusion. Transplantation 2001; 71:1821–1827. pmid:11455264
  11. 11. Chang WJ, Chehab M, Kink S, Toledo-Pereyra LH. Intracellular calcium signaling pathways during liver ischemia and reperfusion. J Invest Surg 2010; 23:228–238. pmid:20690849
  12. 12. Xing WJ, Kong FJ, Li GW, Qiao K, Zhang WH, Zhang L, et al. Calcium-sensing receptors induce apoptosis during simulated ischemia-reperfusion in Buffalo rat liver cells. Clin Exp Pharmacol Physiol 2011; 38: 605–612. pmid:21692826
  13. 13. Nicoud IB, Knox CD, Jones CM, Anderson CD, Pierce JM, Belous AE, et al. 2-APB protects against liver ischemia reperfusion injury by reducing cellular and mitochondrial calcium uptake. Am J PhysiolGastrointest Liver Physiol 2007; 293: G623–G230.
  14. 14. Chattopadhyay P, Chaudhury P, Wahi AK. Ca2+ concentrations are key determinants of ischemia-reperfusion-induced apoptosis: significance for the molecular mechanism of Bcl-2 action. Appl Biochem Biotechnol. 2010; 160: 1968–1977. pmid:19763897
  15. 15. Maack C1, Ganesan A, Sidor A, O’Rourke B. Cardiac sodium-calcium exchanger is regulated by allosteric calcium and exchanger inhibitory peptide at distinct sites. Circ Res. 2005 Jan 7; 96(1):91–9. pmid:15550690
  16. 16. Carini R, Bellomo G, Dianzani MU. The operation of Na+/Ca2+exchanger prevents intracellular Ca2 + overload and hepatocyte killing following iron-induced lipid peroxidation. Biochem Biophys Res Commun 1995; 208: 813–818. pmid:7695640
  17. 17. Shinjo SK, Tersariol IL, Oliveira V, Nakaie CR, Oshiro ME, Ferreira AT, et al. Heparin and heparan sulfate disaccharides bind to the exchanger inhibitor peptide region of Na+/Ca2+exchanger and reduce the cytosolic calcium of smooth muscle cell lines. Requirement of C4-C5 unsaturation and 1—> 4 glycosidic linkage for activity. J Biol Chem. 2002;277(50):48227–33. pmid:12374809
  18. 18. Figueira ER, Bacchella T, Coelho AM, Sampietre SN, Leitão RM, Machado MC. Timing-dependent protection of hypertonic saline solution administration in experimental liver ischemia/reperfusion injury. Surgery, 2010; 147: 415–423. pmid:20004454
  19. 19. Canedo LF, Lima GV, Machado MC. Pulmonary effects after partial liver ischemia and reperfusion—experimental model. Rev. Col. Bras. Cir. 2012; 39(3): 216–221. pmid:22836571
  20. 20. Nogueira MA, Coelho AM, Sampietre SN, Patzina RA, Pinheiro da Silva F, D'Albuquerque LA, et al. Beneficial effects of adenosine triphosphate-sensitive K+ channel opener on liver ischemia/reperfusion injury. World J Gastroenterol. 2014 Nov 7;20(41):15319–26. pmid:25386080
  21. 21. Coelho AM, Machado MC, Sampietre SN, Leite KR, Oliveira VL, Pinotti HW. Hepatic damage during acute pancreatitis in the rat. Braz J Med Biol Res 1997; 30: 947–53. pmid:9361723
  22. 22. Estabrook RW. Mitochondrial respiratory control and the polarographic measurement of ADP/O ratios. Methods in Enzymology. New York: Academic Press, 1967, 41–47.
  23. 23. Chance B, Williams GR. A simple and rapid assay of oxidative phosphorylation. Nature 1955; 175(4469): 1120–1121. pmid:14394122
  24. 24. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J BiolChem 1951; 193: 265–275.
  25. 25. Soriano FG, Liaudet L, Szabrr AL, Szabó E, Virág L, Mabley JG, et al. Resistance to acute septic peritonitis in poly (ADP-ribose) polymerase-1-deficient mice. Shock 2002; 17: 286–292. pmid:11954828
  26. 26. Jancar S, De Giaccobi G, Mariano M, Mencia-Huerta JM, Sirois P, Braquet P. Immune complex induced pancreatitis: effect of BN 52021, a selective antagonist of platelet-activating factor. Prostaglandins 1988; 35: 757–770. pmid:2969600
  27. 27. Quireze C, Montero EF, Leitao RM, Juliano Y, Fagundes DJ, Poli-de-Figueiredo LF. Ischemic preconditioning prevents apoptotic cell death and necrosis in early and intermediate phases of liver ischemia-reperfusion injury in rats. J Invest Surg 2006; 19: 229–236. pmid:16835137
  28. 28. Dietrich Carl P.. Isolation and characterizarion of novel disaccharides and oligosaccharides produced from heparin by bacterial degradation.Biochem. J. (1968) 108, 647.
  29. 29. Sakamoto K, Owada Y, Shikama Y, Wada I, Waguri S, Iwamoto T, et al. Involvement of Na+/Ca2+ exchanger in migration and contraction of rat cultured tendon fibroblasts. J Physiol. 2009 Nov 15; 587(Pt 22):5345–59. pmid:19770194
  30. 30. Clavien PA, Emond J, Vauthey JN, Belghiti J, Chari RS, Strasberg SM. Protection of the liver during hepatic surgery. J GastrointestSurg 8:313–327, 2004.
  31. 31. Kupiec-Weglinski JW, Busuttil RW. Ischemia and reperfusion injury in liver transplantation. Transplant Proc 37(4):1653–6. pmid:15919422
  32. 32. Serracino-Inglott F, Habib NA, Mathie RT. Hepatic ischemia- reperfusion injury. Am J Surg 181: 160–166, 2001. pmid:11425059
  33. 33. Li J, Li RJ, Lv GY, Liu HQ. The mechanisms and strategies to protect from hepatic ischemia-reperfusion injury. Eur Rev Med PharmacolSci 2015; 19(11): 2036–2047.
  34. 34. Yener AÜ, Çiçek MC, Genç SB, Özkan T, Doğan E, Bilgin BC, et al. Hepatic ischemia- reperfusion in protective role of heparin in the injury of the liver and kidney on the experimental model of ischemia/reperfusion. J Cardiothorac Surg. 2014 Feb17; 9:35.
  35. 35. Taha MO, Gonçalves PF, Vidigal RO, Sacchi GP, Pinheiro FG, Brandão LS. Protective effects of heparin on hepatic ischemia and reperfusion lesions in rabbits. Transplant Proc. 2009 Apr; 41(3):812–5. pmid:19376359
  36. 36. Harada N, Okajima K, Uchiba M. Dalteparin, a low molecular weight heparin, attenuates inflammatory responses and reduces ischemia-reperfusion-induced liver injury in rats. Crit Care Med. 2006 Jul; 34(7):1883–91. pmid:16641616
  37. 37. Zhou T, Chen JL, Song W, Wang F, Zhang MJ, Ni PH, et al. Effect of N-desulfated heparin on hepatic/renal ischemia reperfusion injury in rats. J Gastroenterol. 2002 Oct; 8(5):897–900.
  38. 38. Abdel-Salam OM, Baiuomy AR, Ameen A, Hassan NS. A study of unfractionated and low molecular weight heparins in a model of cholestatic liver injury in the rat. Pharmacol Res. 2005; 51(1):59–67. pmid:15519536
  39. 39. Kim JA, Kang YS, Lee SH, Lee YS. Inhibitors of Na+/Ca2+ exchanger prevent oxidant-induced intracellular Ca2+ increase and apoptosis in a human hepatoma cell line. Free Radic Res. 2000; 33 (3):267–77. pmid:10993480
  40. 40. Bilzer M, Jaeschke H, Vollmar AM, Paumgartner G, Gerbes AL. Prevention of Kupffer cell-induced oxidant injury in rat liver by atrial natriuretic peptide. Am J Physiol 1999; 276: G1137–1144. pmid:10330004
  41. 41. Wheeler M.D. Endotoxin and Kupffer cell activation in alcoholic liver disease. Alcohol Res Health 2003; 27:300–306. pmid:15540801
  42. 42. Luedde T, Kaplowitz N, Schwabe RF Cell death and cell death responses in liver disease: mechanisms and clinical relevance. Gastroenterology. 2014 Oct; 147(4):765–783 pmid:25046161
  43. 43. Tsung A, Sahai R, Tanaka H, et al. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med 2005; 201:1135–1143. pmid:15795240
  44. 44. Yan Yuan, Wei Chao-liang, Zhang Wan-rui, Cheng He-ping, Liu Jie. Cross talk between calcium and reactive oxygen species signaling. ActaPharmacologicaSinica (2006) 27, 821–826.
  45. 45. Chouchani ET, Pell VR, Gaude E, Aksentijević D, Sundier SY, Robb EL, et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 2014 Nov 20; 515(7527):431–5. pmid:25383517
  46. 46. Kukner A, Tore F, Firat T, Terzi EH, Oner H, Balaban YH. The preventive effect of low molecular weight heparin on CCL4-induced necrosis and apoptosis in rat liver. Ann Hepatol. 2010; 9(4):445–54. pmid:21057164
  47. 47. Chan CC, Lee KC, Huang YH, Chou CK, Lin HC, Lee FY. Regulation by resveratrol of the cellular factors mediating liver damage and regeneration after acute toxic liver injury. J Gastroenterol Hepatol. 2014 Mar; 29(3):603–13. pmid:23981054
  48. 48. Yamanaka K, Houben P, Bruns H, Schultze D, Hatano E, Schemmer P. A systematic review of pharmacological treatment options used to reduce ischemia reperfusion injury in rat liver transplantation PLoS One. 2015 Apr 28; 10(4):e0122214. pmid:25919110
  49. 49. Van Golen RF, Reiniers MJ, Heger M, Verheij J. Solutions to the discrepancies in the extent of liver damage following ischemia/reperfusion in standard mouse models. J Hepatol. 2015 Apr;62(4):975–7. pmid:25529620