j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 5 ) 1 e9

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 Q17 17 Q1 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65

Available online at www.sciencedirect.com

ScienceDirect journal homepage: www.JournalofSurgicalResearch.com

Propofol alleviates liver oxidative stress via activating Nrf2 pathway Mian Ge,1 Weifeng Yao,1 Yanling Wang, Dongdong Yuan, Xinjin Chi, Gangjian Luo,2 and Ziqing Hei*,2 Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China

article info

abstract

Article history:

Background: Nuclear factor-E2erelated factor 2 (Nrf2)emediated antioxidant response is the

Received 21 October 2014

main protective system of graft-liver against ischemiaereperfusion injury after liver

Received in revised form

transplantation. Propofol is considered to confer protective effects on different organs;

9 January 2015

thus, we explored the possibility that whether propofol could attenuate graft-liver injury in

Accepted 11 March 2015

a rat autologous orthotopic liver transplantation (AOLT) model and mechanisms were

Available online xxx

associated with activation of Nrf2 pathway. Methods: SpragueeDawley rats were randomly divided into four groups: sham-operated

Keywords:

group, saline-treated AOLT group, low-dose propofol intervention group, and high-dose

Liver transplantation

propofol intervention group. Liver injury was determined, and concentration of hydroxyl

Oxidative stress

free radical (OH), superoxide anion (O 2 ), and malondialdehyde in the liver tissue were

Propofol

detected. The expression of Keap1, Nrf2, HO-1, and NQO1 were explored by Western

Nuclear factor E2erelated factor 2

blotting, and also the change of Nrf2 and keap1 was assessed by immunofluorescence. Results: Compared with sham group, pathologic damage of graft-livers was in a timedependent manner, accompanied with the increased level of oxidative stress in the AOLT group, and nuclear Nrf2 expression and its downstream antioxidant enzyme, HO-1 and NQO1, were also increased in this group. However, in propofol pretreatment groups especially in the high-dose group, the pathologic score was significantly decreased, accompanied with a lower level of OH, O 2 , and malondialdehyde than that of the AOLT group. The change of oxidative stress might be related to the Nrf2 pathway, evidenced as the elevation of protein expression level of NQO1, HO-1, and nuclear Nrf2. Conclusions: Protective effects of propofol against liver transplantationeinduced graft-liver injury may be related with Keap1-Nrf2 signal pathway activation. ª 2015 Elsevier Inc. All rights reserved.

1.

Introduction

Liver ischemiaereperfusion injury (IRI) is a major cause of graft dysfunction or nonfunction and results in a high mortality after liver transplantation [1]. Various kinds of mechanisms interact with each other and lead to the complex

pathophysiological processes of hepatic IRI. As reported, reactive oxygen species (ROS)emediated oxidative stress plays a central role in the early stage of IRI [2e6], and antioxidant enzyme activation induced by antioxidants has been shown to attenuate liver IRI [7,8], but still, specific strategies are lacking.

* Corresponding author. Department of Anesthesiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. Q2 Tel./fax: þ86 20 85252297. E-mail address: [email protected] (Z. Hei). 1 M.G. and W.Y. contributed equally to this work. 2 Z.H. and G.L. are co-corresponding authors. 0022-4804/$ e see front matter ª 2015 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.jss.2015.03.016

5.2.0 DTD  YJSRE13195_proof  1 April 2015  11:32 pm  ce

66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130

2

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 5 ) 1 e9

Reducing ROS levels or enhancing antioxidant capacity is 131 always considered to be the primary strategy to prevent 132 oxidative damage [9]. Recently, it has been demonstrated that 133 nuclear factor E2erelated factor 2 (Nrf2), a kind of transcrip134 tion factor, could activate different genes encoding various 135 136 antioxidant enzymes and phase II detoxification enzymes, 137 which enabled responses to oxidative stress and ROS scav138 Q3 enging [10,11]. Nrf2 is normally sequestered in the cytoplasm 139 by Kelch-like ECH-associated protein 1 (Keap1), which is 140 considered to be a negative regulator of Nrf2 [12]. Oxidants 141 and other stimuli inactivate Keap1, resulting in the stabiliza142 tion of Nrf2 and its translocation into the nucleus, where Nrf2 143 activates cytoprotective target genes by binding antioxidant 144 response elements [13]. However, whether Nrf2 nuclear 145 translocation could protect against graft-liver injury after liver 146 transplantation has not been reported. 147 148 Nrf2 activation has been confirmed to protect different 149 organs against IRI, including livers [14], brain [15], intestine 150 [16], and hearts [17]; thus, Nrf2 is considered to be a potential 151 strategy for preventing oxidative damage. Propofol, a widely 152 used intravenous anesthetic, has marked antioxidant capac153 ity to attenuate organ IRI effectively [18e22]; however, 154 whether propofol could ameliorate liver IRI after liver trans155 plantation remains unknown, and the interaction between 156 propofol and Nrf2 antioxidant pathway was still unclear. 157 To explore protective effects of propofol on IRI and its relative 158 159 mechanisms, the rat autologous orthotopic liver transplantation 160 (AOLT) model was used to recapitulate liver transplantation 161 in vivo. We focus on the dynamic alterations of Nrf2 expression 162 during and after liver transplantation and confirmed that 163 propofol-ameliorated liver oxidative injury followed AOLT via 164 Keap1 downregulation and Nrf2 upregulation. 165 166 167 2. Materials and methods 168 169 2.1. Animals 170 171 172 Sixty-three healthy male SpragueeDawley rats, aged 8e10 wk 173 and weighing 220e280 g, were provided by the Medical 174 Experimental Animal Center of Guangdong Province. This 175 study was approved by the Institutional Animal Care and Use 176 Committee and Animal Ethics Committee of Sun Yat-sen 177 University in Guangzhou, China, and followed the “Guide for 178 the Care and Use of Laboratory Animals” (National Institutes 179 of Health publication 86-23 revised 1985) guidelines for the 180 treatment of animals. 181 182 183 2.2. AOLT model and experimental groups 184 185 The AOLT model was described in our previous study [23] and 186 consists of primary surgical procedures, such as the dissoci187 ation of vessels and ligaments, the blockade of liver vessels, 188 liver infusion with cold preservation solution, vascular off189 clamping, and hepatic reperfusion. The entire anhepatic 190 phase was controlled within 20  1 min for a high mortality 191 rate of rats when cold ischemia time was longer than 25 min 192 in our preliminary study. The sham group underwent 193 194 abdominal surgery and liver dissection under anesthesia 195 without cold perfusion and reperfusion. The AOLT model rats

were subjected to the typical pathophysiological hepatic IRI processes during liver transplantation.

2.3.

Experimental groups

Initially, 35 male SpragueeDawley rats were randomized into a sham-operated group (sham group, n ¼ 7), a 4 h after AOLT group (4-h group, n ¼ 7), an 8 h after AOLT group (8-h group, n ¼ 7), a 16 h after AOLT group (16-h group, n ¼ 7), and a 24 h after AOLT group (24-h group, n ¼ 7). In the following experiment, 28 rats were divided into a shamoperated group (sham group, n ¼ 7), a saline-treated AOLT group (AOLT group, n ¼ 7), a low-dose propofol intervention AOLT group (L-Pro þ AOLT group, n ¼ 7), and a high-dose propofol intervention AOLT group (H-Pro þ AOLT group, n ¼ 7). The sham and AOLT groups received 3 mL of normal saline via intraperitoneal injections for 3 consecutive days before the experiment, and the LPro þ AOLT and H-Pro þ AOLT groups were pretreated with propofol (Diprivan, 1% propofol, CG411; AstraZeneca, Caponago, Italy) intraperitoneally at a dose or rate of 50 mg/kg (low dose, sedative effect [24]) or 100 mg/kg (high dose, anesthetic effect [25]) daily for 3 d before AOLT.

2.4.

Histologic assessment

Liver tissues were collected immediately after the rats were euthanized. The liver tissues were then fixed with 10% para- Q4 formaldehyde (pH 7.0; Sigma) for 48 h and embedded in paraffin. After hematoxylineeosin staining, liver sections (5 mm) were analyzed microscopically for pathology. Ten randomly selected images were captured in a blinded manner from each slide using a light microscope. The severity of IRI was graded based on Suzuki criteria [26].

2.5.

Biochemical analysis of serum samples

Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities in serum were measured with the test kits from SigmaeAldrich (St. Louis, MO) [27].

2.6. Superoxide anion (O 2 ), hydroxyl radical ($OH) and malondialdehyde assays Spectrophotometer methods were used to determine O 2 and $OH levels. The superoxide anion radical scavenging ability was measured by the method of the active oxygen generation of xanthineexanthine oxidase and detected at 550 nm with the spectrophotometer [28]. Determining the $OH suppression of the propofol was conducted according to the Fenton reaction principle. The procedure was performed using a $OH assay kit, according to the manufacturer’s instructions [29]. Malondialdehyde (MDA) content was detected using the thiobarbituric acid method. All procedures were performed according to the manufacturer’s instructions (Nanjing KeyGen Biotech. Co., Ltd, Nanjing, China).

2.7.

Immunofluorescence staining

Frozen sections of liver tissue were washed three times with phosphate-buffered saline (PBS). After fixing with PBS

5.2.0 DTD  YJSRE13195_proof  1 April 2015  11:32 pm  ce

196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 5 ) 1 e9

261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325

Q5

containing 5% bovine serum albumin and 0.3% Triton X-100 for 1 h, tissue sections were incubated with anti-Nrf2 (1:150) or anti-Keap1 (1:100) antibodies at 4 C overnight. After washing with PBS, the slides were incubated with a fluorescently labeled secondary antibody (1:100) for 1 h at 25 C. The cover slips were then washed again and mounted using a mounting medium (Applygen Technologies, Beijing. No. 01210) and observed using a fluorescent microscope (DM LB2; Leica Microsystems, Germany).

2.8.

Q6 Q7 Q8

Q9

Protein expression of Nrf2, Keap1, HO-1, and NQO1 in the nuclear and cytosolic fractions was detected using Western blotting. Nuclear and cytoplasmic proteins were separated according to the protocol described in the Nuclear and Cytoplasmic Extraction Kit (Thermo Scientific). Protein concentrations were measured using the BCA method (KeyGen BioTech, China). The primary antibodies used were monoclonal rabbit anti-mouse Keap1 (1:1000 dilution; Millipore Company), polyclonal rabbit anti-mouse Nrf2 (1:250 dilution; Santa Cruz Biotechnology), anti-HO-1 (1:250 dilution; Santa Cruz Biotechnology), anti-NQO1 NQO1 (1:250 dilution; Santa Cruz Biotechnology), antieb-actin (1:2000 dilution; Santa Cruz Biotechnology), and anti-H2A (1:1000 dilution; Santa Cruz Biotechnology). The secondary antibody used was goat antirabbit HRP-conjugated IgG (1:2000 dilution; Boster, China). The optical density values of the bands were normalized to those of b-actin or H2A.

2.9.

Q10

Western blotting

Data analyses and statistics

The data were expressed as the mean  standard deviation or as the median (range) where appropriate. Comparisons of more than two groups were performed using a one-way analysis of variance followed by LSD multiple-comparison

3

tests. P < 0.05 was selected to indicate statistically significant differences.

3.

Results

3.1. Dynamic AOLT-induced pathologic changes in the liver and biochemical assessment As depicted in Figure 1, the sinusoids and hepatocytes were normal in the sham-operated group (Fig. 1A). Most severe hepatic injury occurred at 4 and 8 h after reperfusion, which manifested as impaired sinusoids and hepatocytes with edema and congestion, obstructed sinusoids, infiltration of inflammatory cells, and necrosis and vacuolation (Fig. 1B and C). After 16 h of reperfusion, pathologic injuries were alleviated significantly (Fig. 1D). Even after 24 h of reperfusion, only slight congestion and sinusoid obstruction were observed, and a small quantity of hepatocytes showed necrosis and vacuolation (Fig. 1E). These results suggested that hepatic IRI that followed AOLT was obvious at the early stage after reperfusion, and then resolved gradually, which correlated with changes in the Suzuki score (Fig. 1F). Serum AST and ALT levels, two indexes of hepatocellular injury, began to increase significantly at 4 h after reperfusion, peaked at 8 h, and then gradually declined, indicating that the most serious liver damage occurred at 8 h after AOLT (Fig. 1G and H).

3.2. Changes in OH, O 2 , and MDA levels in the liver after AOLT OH and O 2 levels reflect ROS levels, and MDA levels were used to determine the extent of oxidative injury in the liver tissue. As shown in Figure 2, OH and O 2 levels increased significantly at 4, 8, and 16 h after AOLT but returned to near

Fig. 1 e AOLT-induced dynamic pathologic changes in the liver visualized using HE staining (3200) and serum biochemical changes. Images AeE are representative of the sham group (Sham) and the experimental groups 4 h after AOLT (4-h group), 8 h after AOLT (8-h group), 16 h after AOLT (16-h group), and 24 h after AOLT (24-h group), respectively; image F shows Suzuki histologic grading of IRI at each time point after AOLT. Images G and H show the serum biochemical changes of AST O and ALT. *P < 0.05 versus sham group; P < 0.05 versus 24-h group. (Color version of figure available online.) 5.2.0 DTD  YJSRE13195_proof  1 April 2015  11:32 pm  ce

Q15

326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390

4

391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 Q11 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 5 ) 1 e9

Fig. 2 e Changes in liver OH, OL 2 , and MDA levels after AOLT. AeC show changes in hydroxyl radical (OH), superoxide anion (OL 2 ), and MDA levels over time. Sham: sham-operated group; 4 h: 4 h after AOLT; 8 h: 8 h after AOLT; 16 h: 16 h after AOLT; 24 h: 24 h after AOLT. *P < 0.05 versus sham group. the baseline at 24 h after AOLT (Fig. 2A and B). MDA levels showed a similar pattern to OH and O 2 levels (Fig. 2C).

3.3. Dynamic alteration in Nrf2 and HO-1 expression after liver IRI Nrf2 is the key protein regulating downstream antioxidant expression, and its nuclear levels were always used to determine Nrf2 nuclear translocation. Figure 3A shows that Nrf2 expression increased from 8 h after reperfusion (P < 0.05 versus sham group) and peaked at 24 h. A similar tendency was observed for HO-1 expression, which is a downstream Nrf2 antioxidant. HO-1 expression increased gradually from 8e24 h after reperfusion (P < 0.05 versus sham group), increased dramatically at 16 h after AOLT compared with that at 4 h (P < 0.05 versus 4-h group), and peaked at 24 h (P < 0.05 versus 4- and 8-h groups; Fig. 3B).

3.4. Propofol-attenuated liver injury and oxidative stress induced by hepatic IRI in AOLT rats We selected the time point of 8 h after reperfusion for the following intervention studies, for pathologic changes were

the most obvious at that time. Rats were pretreated with propofol intraperitoneally at low and high doses, and particularly in the high-dose group, propofol ameliorated hepatic IRI markedly, evidenced as minimal pathologic changes with less sinusoidal congestion, cytoplasmic vacuolization, and hepatocellular necrosis (Fig. 4AeD), as well as reversed the elevated levels of plasma AST and ALT (Fig. 4C and D). In the low-dose propofol group, there was minor congestion, obstruction, and ballooning, and 200 antioxidants that contributed to the majority of important endogenous antioxidant systems [34]. Keap1, a master regulator of intracellular redox homeostasis, has been shown to interact with Nrf2 [12]. As mentioned previously, Nrf2 is normally sequestered in the cytoplasm by Keap1, which prevents Nrf2 nuclear translocation and induces Nrf2 ubiquitin-mediated degradation. Oxidative or other stimuli enhance Keap1 degradation, and Nrf2 activation results in nuclear translocation and activation of genes encoding various antioxidant enzymes that protect cells from oxidative damage. Thus, the molecular mechanism has been confirmed by in vivo and in vitro experiments using genetic approaches including gene knockout and gene silencing [10]. Tanaka et al. [35] demonstrated that transient middle cerebral artery occlusion induced a progressive decrease in Keap1 expression in ischemic brains relative to Nrf2 and its downstream antioxidants. However, thus far, studies of Nrf2 expression after liver transplantation have been lacking. As shown in our results, during24 h after AOLT, a dynamic process of pathologic and oxidative injury occurs in which the injury is worse during the early stages and that gradually resolves or improves; this process occurs parallel to dynamic changes in nuclear levels of Nrf2. We suggest that Nrf2 and its antioxidant effects play a crucial role in preventing hepatic IRI

induced by AOLT and that it promotes hepatic repair. It is critical to identify effective methods that alleviate hepatic IRI. This study shows that targeting Nrf2 is a feasible strategy for early hepatic protection against IRI. Propofol is a commonly used sedative [18,19] and has been shown to promote the antioxidant capacity of various antioxidants and attenuates heart, brain, liver, and renal IRI [20e22]. However, mechanisms of propofol on its antioxidant activity remain unclear. In this study, we focused on the influence of propofol pretreatment on nuclear Nrf2 expression during hepatic IRI induced by AOLT. Immunofluorescence assays and Western blotting results showed that propofol pretreatment promoted Keap1 degradation and nuclear translocation of Nrf2. Consistent with these results, ROS levels decreased and oxidative damage was ameliorated. HO-1 and NQO1 are regarded as antioxidants that act downstream of Nrf2 [34]. In previous studies, it had been demonstrated that NQO1 expression was mediated by Nrf2 [36,37], and more importantly, HO-1 expression upregulation is always considered to be the evidence for the activation of Nrf2 signaling pathway [38]. NQO1 facilitates scavenging of ROS generated by mitochondria [39e42], and HO-1 exerts multiple regulatory functions during oxidative stress [43e47]. Our study revealed that propofol enhanced antioxidant capacity by upregulating HO-1 and NQO1 through Nrf2 activation. Based on these results and those of other studies, propofol enhances the nuclear translocation of Nrf2 and upregulates antioxidant expression, which alleviates hepatic IRI. In summary, our findings document that propofol ameliorated whole liver IRI in AOLT via downregulation of Keap1 and Nrf2 nuclear translocation. However, the optimal

5.2.0 DTD  YJSRE13195_proof  1 April 2015  11:32 pm  ce

846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910

8

911 912 Q12 913 914 915 916 917 918 919 920 Q13 Q14

921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 5 ) 1 e9

time for propofol administration requires further investigation. For the future studies, we will use more different methods to get the direct and specific evidence for the conclusion that the role of propofol was Nrf2 dependent, such as Nrf2 inhibitors, Nrf2 KO mice or Nrf2-siRNA on cells.

Acknowledgment This work was supported by grants from the National Natural Science Foundation of China (No. 81372090) and the Planned Science and Technology Project of Guangzhou (No. 2013B021800181 and 2013B051000035). The authors acknowledge the language help from Nature Publishing Group Language Editing. Authors’ contribution: M.G. and W.Y. performed the experiment and wrote the draft, Y.W. analyzed the data, D.Y. and X.C. finished the immunofluorescence staining, and G.L. and Z.H. designed and revised the article.

Disclosure The authors have no conflicts of interests, financial, or otherwise related to the publication of this study or its findings.

references

[1] Montalvo-Jave EE, Pin˜a E, Montalvo-Arenas C, et al. Role of ischemic preconditioning in liver surgery and hepatic transplantation. J Gastrointest Surg 2009;13:2074. [2] Zhai Y, Busuttil RW, Kupiec-Weglinski JW. Liver ischemia and reperfusion injury: new insights into mechanisms of innate-adaptive immune-mediated tissue inflammation. Am J Transplant 2011;11:1563. [3] Kalogeris T, Baines CP, Krenz M, et al. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol 2012;298: 229. [4] Klune JR, Tsung A. Molecular biology of liver ischemia/ reperfusion injury: established mechanisms and recent advancements. Surg Clin North Am 2010;90:665. [5] Yang MQ, Ma YY, Tao SF, et al. Mast cell degranulation promotes ischemia-reperfusion injury in rat liver. J Surg Res 2014;186:170. [6] Elias-Miro´ M, Jime´nez-Castro MB, Rode´s J, Peralta C. Current knowledge on oxidative stress in hepatic ischemia/ reperfusion. Free Radic Res 2013;47:555. [7] Jaeschke H, Woolbright BL. Current strategies to minimize hepatic ischemia-reperfusion injury by targeting reactive oxygen species. Transplant Rev 2012;26:103. [8] Czubkowski P, Socha P, Pawlowska J. Oxidative stress in liver transplant recipients. Ann Transplant 2011;16:99. [9] Fukuda K, Asoh S, Ishikawa M, et al. Inhalation of hydrogen gas suppresses hepatic injury caused by ischemia/ reperfusion through reducing oxidative stress. Biochem Biophys Res Commun 2007;361:670. [10] Baird L, Dinkova-Kostova AT. The cytoprotective role of the Keap1-Nrf2 pathway. Arch Toxicol 2011;85:241. [11] Hyeon S, Lee H, Yang Y, Jeong W. Nrf2 deficiency induces oxidative stress and promotes RANKL-induced osteoclast differentiation. Free Radic Biol Med 2013;65:789.

[12] McMahon M, Itoh K, Yamamoto M, Hayes JD. Keap1dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression. J Biol Chem 2003; 278:21592. [13] Chen XL, Dodd G, Thomas S, et al. Activation of Nrf2/ARE pathway protects endothelial cells from oxidant injury and inhibits inflammatory gene expression. Am J Physiol Heart Circ Physiol 2006;290:H1862. [14] Ke B, Shen XD, Zhang Y, et al. KEAP1-NRF2 complex in ischemia-induced hepatocellular damage of mouse liver transplants. J Hepatol 2013;59:1200. [15] Zhai X, Chen X, Shi J, et al. Lactulose ameliorates cerebral ischemia-reperfusion injury in rats by inducing hydrogen by activating Nrf2 expression. Free Radic Biol Med 2013;65:731. [16] Ge M, Chi X, Zhang A, et al. Intestinal NF-E2-related factor-2 expression and antioxidant activity changes in rats undergoing orthotopic liver autotransplantation. Oncol Lett 2013;6:1307. [17] Peake BF, Nicholson CK, Lambert JP, et al. Hydrogen sulfide preconditions the db/db diabetic mouse heart against ischemia-reperfusion injury by activating Nrf2 signaling in an Erk-dependent manner. Am J Physiol Heart Circ Physiol 2013;304:H1215. [18] Kobayashi K, Yoshino F, Takahashi SS, et al. Direct assessments of the antioxidant effects of propofol medium chain triglyceride/long chain triglyceride on the brain of stroke-prone spontaneously hypertensive rats using electron spin resonance spectroscopy. Anesthesiology 2008;109:426. [19] Murphy PG, Myers DS, Davies MJ, Webster NR, Jones JG. The antioxidant potential of propofol (2,6-diisopropylphenol). Br J Anaesth 1992;68:613. [20] King N, Al Shaama M, Suleiman MS. Propofol improves recovery of the isolated working hypertrophic heart from ischaemia-reperfusion. Pflugers Arch 2012;464:513. [21] Zhao G, Ma H, Shen X, et al. Role of glycogen synthase kinase 3b in protective effect of propofol against hepatic ischemiareperfusion injury. J Surg Res 2013;185:388. [22] Yoo YC, Yoo KJ, Lim BJ, et al. Propofol attenuates renal ischemia-reperfusion injury aggravated by hyperglycemia. J Surg Res 2013;183:783. [23] Chi X, Zhang A, Luo G, et al. Knockdown of myeloid differentiation protein-2 reduces acute lung injury following orthotopic autologous liver transplantation in a rat model. Pulm Pharmacol Ther 2013;26:380. [24] Liu KX, Chen SQ, Huang WQ, et al. Propofol pretreatment reduces ceramide production and attenuates intestinal mucosal apoptosis induced by intestinal ischemia/ reperfusion in rats. Anesth Analg 2008;107:1884. [25] Mu X, Wu A, Wu J, et al. Effects of anesthetic propofol on release of amino acids from the spinal cord during visceral pain. Neurosci Lett 2010;484:206. [26] Zhao HF, Zhang GW, Zhou J, et al. Biliary tract injury caused by different relative warm ischemia time in liver transplantation in rats. Hepatobiliary Pancreat Dis Int 2009;8: 247. [27] Lin Z, Wu F, Lin S, et al. Adiponectin protects against acetaminophen-induced mitochondrial dysfunction and acute liver injury by promoting autophagy in mice. J Hepatol 2014 Oct;61:825. [28] Oyetayo VO. Free radical scavenging and antimicrobial properties of extracts of wild mushrooms. Braz J Microbiol 2009 Apr;40:380. [29] Hao Jianxiong, Qiu Shuang, Li Huiying, et al. Roles of hydroxyl radicals in electrolyzed oxidizing water (EOW) for the inactivation of Escherichia coli. Int J Food Microbiol 2012;155:99. [30] Zhang W, Wang M, Xie HY, et al. Role of reactive oxygen species in mediating hepatic ischemia-reperfusion injury

5.2.0 DTD  YJSRE13195_proof  1 April 2015  11:32 pm  ce

975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039

j o u r n a l o f s u r g i c a l r e s e a r c h x x x ( 2 0 1 5 ) 1 e9

1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073

[31]

[32]

[33] Q16

[34]

[35]

[36]

[37]

[38]

[39]

and its therapeutic applications in liver transplantation. Transplant Proc 2007;39:1332. Kadkhodaee M, Mikaeili S, Zahmatkesh M, et al. Alteration of renal functional, oxidative stress and inflammatory indices following hepatic ischemia-reperfusion. Gen Physiol Biophys 2012;31:195. Karabulut AB, Gu¨l M, Karabulut E, et al. Oxidant and antioxidant activity in rabbit livers treated with zoledronic acid. Transplant Proc 2010;42:3820. Jaeschke H. Antioxidant defense mechanisms. In: McQueen CA, editor. Comprehensive toxicology, 9. Oxford: Academic; 2010. p. 319e37. Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem 2009;284:13291. Tanaka N, Ikeda Y, Ohta Y, et al. Expression of Keap1-Nrf2 system and antioxidative proteins in mouse brain after transient middle cerebral artery occlusion. Brain Res 2011; 1370:246. Silva-Gomes S, Santos AG, Caldas C, et al. Transcription factor NRF2 protects mice against dietary iron-induced liver injury by preventing hepatocytic cell death. J Hepatol 2014; 60:354. Tanigawa S, Fujii M, Hou D-X. Action of Nrf2 and Keap1 in ARE-mediated NQO1 expression by quercetin. Free Radic Biol Med 2007;42:1690. Anwar AA, Li FYL, Leake DS, et al. Induction of heme oxygenase 1 by moderately oxidized low-density lipoproteins in human vascular smooth muscle cells: role of mitogen-activated protein kinases and Nrf2. Free Radic Biol Med 2005;39:227. Guerrero-Beltra´n CE, Caldero´n-Oliver M, Martı´nezAbundis E, et al. Protective effect of sulforaphane against cisplatin-induced mitochondrial alterations and

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

9

impairment in the activity of NAD(P)H: quinone oxidoreductase 1 and g glutamyl cysteine ligase: studies in mitochondria isolated from rat kidney and in LLC-PK1 cells. Toxicol Lett 2010;199:80. Gang GT, Kim YH, Noh JR, et al. Protective role of NAD(P) H:quinone oxidoreductase 1 (NQO1) in cisplatin-induced nephrotoxicity. Toxicol Lett 2013;221:165. Lamberti MJ, Vittar NB, da Silva Fde C, Ferreira VF, Rivarola VA. Synergistic enhancement of antitumor effect of b-Lapachone by photodynamic induction of quinone oxidoreductase (NQO1). Phytomedicine 2013;20:1007. Meyer ML, Potts-Kant EN, Ghio AJ, et al. NAD(P)H quinone oxidoreductase 1 regulates neutrophil elastase-induced mucous cell metaplasia. Am J Physiol Lung Cell Mol Physiol 2012;303:L181. Yun N, Cho HI, Lee SM. Impaired autophagy contributes to hepatocellular damage during ischemia/reperfusion: heme oxygenase-1 as a possible regulator. Free Radic Biol Med 2013;68C:168. Katori M, Busuttil RW, Kupiec-Weglinski JW. Heme oxygenase-1 system in organ transplantation. Transplantation 2002;74:905. Gozzelino R, Jeney V, Soares MP. Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol Toxicol 2010;50:323. Zhao Y, Zhang L, Qiao Y, et al. Heme oxygenase-1 prevents cardiac dysfunction in streptozotocin-diabetic mice by reducing inflammation, oxidative stress, apoptosis and enhancing autophagy. PLoS One 2013;8:e75927. Haines DD, Lekli I, Teissier P, Bak I, Tosaki A. Role of haeme oxygenase-1 in resolution of oxidative stressrelated pathologies: focus on cardiovascular, lung, neurological and kidney disorders. Acta Physiol (oxf) 2012; 204:487.

5.2.0 DTD  YJSRE13195_proof  1 April 2015  11:32 pm  ce

1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107

Propofol alleviates liver oxidative stress via activating Nrf2 pathway.

Nuclear factor-E2-related factor 2 (Nrf2)-mediated antioxidant response is the main protective system of graft-liver against ischemia-reperfusion inju...
3MB Sizes 1 Downloads 8 Views