Remote ischemic preconditioning protects against liver ischemia-reperfusion injury via heme oxygenase-1-induced autophagy

Yun Wang, Jian Shen, Xuanxuan Xiong, Yonghua Xu, Hai Zhang, Changjun Huang, Yuan Tian, Chengyu Jiao, Xuehao Wang, Xiangcheng Li, Yun Wang, Jian Shen, Xuanxuan Xiong, Yonghua Xu, Hai Zhang, Changjun Huang, Yuan Tian, Chengyu Jiao, Xuehao Wang, Xiangcheng Li

Abstract

Background: Growing evidence has linked autophagy to a protective role of preconditioning in liver ischemia/reperfusion (IR). Heme oxygenase-1 (HO-1) is essential in limiting inflammation and preventing the apoptotic response to IR. We previously demonstrated that HO-1 is up-regulated in liver graft after remote ischemic preconditioning (RIPC). The aim of this study was to confirm that RIPC protects against IR via HO-1-mediated autophagy.

Methods: RIPC was performed with regional ischemia of limbs before liver ischemia, and HO-1 activity was inhibited pre-operation. Autophagy was assessed by the expression of light chain 3-II (LC3-II). The HO-1/extracellular signal-related kinase (ERK)/p38/mitogen-activated protein kinase (MAPK) pathway was detected in an autophagy model and mineral oil-induced IR in vitro.

Results: In liver IR, the expression of LC3-II peaked 12-24 h after IR, and the ultrastructure revealed abundant autophagosomes in hepatocytes after IR. Autophagy was inhibited when HO-1 was inactivated, which we believe resulted in the aggravation of liver IR injury (IRI) in vivo. Hemin-induced autophagy also protected rat hepatocytes from IRI in vitro, which was abrogated by HO-1 siRNA. Phosphorylation of p38-MAPK and ERK1/2 was up-regulated in hemin-pretreated liver cells and down-regulated after treatment with HO-1 siRNA.

Conclusions: RIPC may protect the liver from IRI by induction of HO-1/p38-MAPK-dependent autophagy.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Hepatic inflow occlusion and reflow…
Figure 1. Hepatic inflow occlusion and reflow increased hepatic HO-1 and autophagic signaling.
(A) Western blotting for HO-1 and LC3-II in liver lysates from sham-operated or IR-treated mice shows increased HO-1 and LC3-II in the IR group, and the expression of HO-1, LC3-II peaked at 6–12 h and 12–24 h, respectively(*P

Figure 2. Remote ischemic preconditioning induced HO-1…

Figure 2. Remote ischemic preconditioning induced HO-1 to increase autophagy, but the inhibition of HO-1…

Figure 2. Remote ischemic preconditioning induced HO-1 to increase autophagy, but the inhibition of HO-1 by Znpp decreased autophagy.
(A) HO-1 was downregulated in the 12-h Znpp+IR group compared with the sham and 12-h IR groups (*P#P<0.05 compared with the IR group). (B) The data were quantified by counting the number of autophagosomes per cross-sectioned cell (*P<0.05 compared with the IR group; #P<0.05 compared with the RIPC+IR group, n = 15). (C) IR treatment increased autophagy compared to sham as observed by transmission electron microscopy and increased autophagosome formation in the RIPC+IR group, whereas autophagosomes were rare in the Znpp+IR group (black arrows). (D) Increased HO-1 and LC3-II staining was observed in the RIPC+IR group compared with the IR group, and LC3-II staining was decreased after inhibition of HO-1 (*P<0.05 compared with the IR group at the corresponding time point; #P<0.05 compared with the RIPC+IR group at the corresponding time point).

Figure 3. Increased HO-1 and autophagy alleviated…

Figure 3. Increased HO-1 and autophagy alleviated liver damage.

(A) IR treatment increases the histopathologic…

Figure 3. Increased HO-1 and autophagy alleviated liver damage.
(A) IR treatment increases the histopathologic changes of liver. Extensive areas of hepatocyte necrosis (N), vacuolation (V) and sinusoidal congestion were observed in the IR group. The Znpp+IR and Znpp+RIPC+IR group showed markedly higher hepatocyte necrosis accumulation (N. A) and vacuo(V), whereas the RIPC+IR group still showed large areas of normal liver architecture, similar to the sham group mice (original magnification 200×; insets 400×). (B) The mean injury score of the RIPC+IR group was significantly lower than the IR group according to Suzuki's histologic classification. The mean injury scores in the Znpp+IR and Znpp+RIPC+IR groups were significantly higher than in the IR and RIPC+IR groups (*P#P<0.05 compared with the RIPC+IR group). (C) IR treatment increased serum aminotransferase levels in mice compared with controls. Compared with the IR group, serum aminotransferase levels were lower in the RIPC+IR group. The mean serum levels of ALT and AST were highest in the Znpp+IR and Znpp+RIPC+IR groups. There were no significant differences in aminotransferase levels in the Znpp+IR and Znpp+RIPC+IR groups (*P<0.05 compared with the IR group; #P<0.05 compared with the RIPC+IR group).

Figure 4. Induction of autophagy was dependent…

Figure 4. Induction of autophagy was dependent on HO-1 in vivo.

(A) Increased HO-1 and…

Figure 4. Induction of autophagy was dependent on HO-1 in vivo.
(A) Increased HO-1 and LC3-II staining was observed in the RIPC+IR group compared with the Sham, IR, Znpp+IR, and Znpp+RIPC+IR groups, and there were no significant differences in the expression of HO-1 and LC3-II protein between the Znpp+IR and Znpp+RIPC+IR groups (*P#P<0.05 compared with the RIPC+IR group). (B) The data were quantified by counting the number of autophagosomes per cross-sectioned cell (*P<0.05 compared with the IR group; #P<0.05 compared with the RIPC+IR group, n = 15). (C) IR treatment increased autophagy as observed by transmission electron microscopy compared to the sham group and showed increased autophagosome formation in the RIPC+IR group, whereas autophagosomes were rare in the Znpp+IR and Znpp+RIPC+IR groups.

Figure 5. IR simulation in AML12 cells…

Figure 5. IR simulation in AML12 cells increased LC3-II expression; HO-1 siRNA effectively inhibited HO-1…

Figure 5. IR simulation in AML12 cells increased LC3-II expression; HO-1 siRNA effectively inhibited HO-1 expression.
(A) LC3-II staining increased in the IR cell model and peaked at 12–24 h (*P#P<0.05 compared with the SS group). (D) Western blotting showed lower expression of HO-1 protein in the S1 group compared with the SS and S2 groups (*P<0.05 compared with the sham group; #P<0.05 compared with the SS group).

Figure 6. Inhibition of HO-1 increased cell…

Figure 6. Inhibition of HO-1 increased cell death and decreased autophagy in AML12 cells.

(A)…

Figure 6. Inhibition of HO-1 increased cell death and decreased autophagy in AML12 cells.
(A) Immunofluorescence of AML12 cells demonstrated increased LC3-II staining in the HIR group; however, downregulated HO-1 decreased autophagy. (B) Increased photometric values of protein in the HIR group compared with the IR and HSIR groups are shown, and the lowest photometric values of protein were observed in the HSIR group (*P#P<0.05 compared with the IR group; &P<0.05 compared with the HIR group). (C) Induction of HO-1 and autophagy decreased biochemical enzyme levels, whereas inhibition of HO-1 and autophagy increased biochemical enzyme levels (*P<0.01 compared with the IR group; #P<0.05 compared with the HIR group). (D) The expression of HO-1 and autophagy proteins was upregulated by hemin, but downregulated by treatment with HO-1 siRNA (*P<0.01 compared with the sham group; #P<0.05 compared with the IR group; &P<0.05 compared with the HIR group).

Figure 7. HO-1 induced autophagy through activation…

Figure 7. HO-1 induced autophagy through activation of ERK1/2 and p38-MAPK.

(A) Apoptotic rates of…

Figure 7. HO-1 induced autophagy through activation of ERK1/2 and p38-MAPK.
(A) Apoptotic rates of the sham group and three treated groups are shown. Mineral oil treatment increased the rate of early and late apoptosis, and HO-1 siRNA treatment before IR increased the rate of apoptosis compared with the IR group. The HIR group had lower rates of apoptosis, similar to the sham group (*P#P<0.05 compared with the IR group; &P<0.05 compared with the HIR group). (B) Hemin treatment before IR increased phosphorylated ERK1/2 staining, as shown in immunoblotting, and HSIR group cells showed decreased phosphorylated ERK1/2 expression. Western blotting for phosphorylated p38-MAPK demonstrated increased phosphorylated p38-MAPK in the HIR group, but decreased signaling in the HSIR group (*P<0.01 compared with the sham group; #P<0.05 compared with the IR group; &P<0.05 compared with the HIR group). (C) HO-1 expression was decreased in the EIIR group compared with the IR group, and LC3-II expression was decreased in the PIIR group compared with the IR group (*P<0.05 compared with the sham group; #P<0.05 compared with the IR group). (D) HO-1 expression was nearly the same as in the PIIR and IR groups, but LC3-II expression was decreased in the EIIR group compared with the IR group (*P<0.05 compared with the sham group; #P<0.05 compared with the IR group).
All figures (7)
Figure 2. Remote ischemic preconditioning induced HO-1…
Figure 2. Remote ischemic preconditioning induced HO-1 to increase autophagy, but the inhibition of HO-1 by Znpp decreased autophagy.
(A) HO-1 was downregulated in the 12-h Znpp+IR group compared with the sham and 12-h IR groups (*P#P<0.05 compared with the IR group). (B) The data were quantified by counting the number of autophagosomes per cross-sectioned cell (*P<0.05 compared with the IR group; #P<0.05 compared with the RIPC+IR group, n = 15). (C) IR treatment increased autophagy compared to sham as observed by transmission electron microscopy and increased autophagosome formation in the RIPC+IR group, whereas autophagosomes were rare in the Znpp+IR group (black arrows). (D) Increased HO-1 and LC3-II staining was observed in the RIPC+IR group compared with the IR group, and LC3-II staining was decreased after inhibition of HO-1 (*P<0.05 compared with the IR group at the corresponding time point; #P<0.05 compared with the RIPC+IR group at the corresponding time point).
Figure 3. Increased HO-1 and autophagy alleviated…
Figure 3. Increased HO-1 and autophagy alleviated liver damage.
(A) IR treatment increases the histopathologic changes of liver. Extensive areas of hepatocyte necrosis (N), vacuolation (V) and sinusoidal congestion were observed in the IR group. The Znpp+IR and Znpp+RIPC+IR group showed markedly higher hepatocyte necrosis accumulation (N. A) and vacuo(V), whereas the RIPC+IR group still showed large areas of normal liver architecture, similar to the sham group mice (original magnification 200×; insets 400×). (B) The mean injury score of the RIPC+IR group was significantly lower than the IR group according to Suzuki's histologic classification. The mean injury scores in the Znpp+IR and Znpp+RIPC+IR groups were significantly higher than in the IR and RIPC+IR groups (*P#P<0.05 compared with the RIPC+IR group). (C) IR treatment increased serum aminotransferase levels in mice compared with controls. Compared with the IR group, serum aminotransferase levels were lower in the RIPC+IR group. The mean serum levels of ALT and AST were highest in the Znpp+IR and Znpp+RIPC+IR groups. There were no significant differences in aminotransferase levels in the Znpp+IR and Znpp+RIPC+IR groups (*P<0.05 compared with the IR group; #P<0.05 compared with the RIPC+IR group).
Figure 4. Induction of autophagy was dependent…
Figure 4. Induction of autophagy was dependent on HO-1 in vivo.
(A) Increased HO-1 and LC3-II staining was observed in the RIPC+IR group compared with the Sham, IR, Znpp+IR, and Znpp+RIPC+IR groups, and there were no significant differences in the expression of HO-1 and LC3-II protein between the Znpp+IR and Znpp+RIPC+IR groups (*P#P<0.05 compared with the RIPC+IR group). (B) The data were quantified by counting the number of autophagosomes per cross-sectioned cell (*P<0.05 compared with the IR group; #P<0.05 compared with the RIPC+IR group, n = 15). (C) IR treatment increased autophagy as observed by transmission electron microscopy compared to the sham group and showed increased autophagosome formation in the RIPC+IR group, whereas autophagosomes were rare in the Znpp+IR and Znpp+RIPC+IR groups.
Figure 5. IR simulation in AML12 cells…
Figure 5. IR simulation in AML12 cells increased LC3-II expression; HO-1 siRNA effectively inhibited HO-1 expression.
(A) LC3-II staining increased in the IR cell model and peaked at 12–24 h (*P#P<0.05 compared with the SS group). (D) Western blotting showed lower expression of HO-1 protein in the S1 group compared with the SS and S2 groups (*P<0.05 compared with the sham group; #P<0.05 compared with the SS group).
Figure 6. Inhibition of HO-1 increased cell…
Figure 6. Inhibition of HO-1 increased cell death and decreased autophagy in AML12 cells.
(A) Immunofluorescence of AML12 cells demonstrated increased LC3-II staining in the HIR group; however, downregulated HO-1 decreased autophagy. (B) Increased photometric values of protein in the HIR group compared with the IR and HSIR groups are shown, and the lowest photometric values of protein were observed in the HSIR group (*P#P<0.05 compared with the IR group; &P<0.05 compared with the HIR group). (C) Induction of HO-1 and autophagy decreased biochemical enzyme levels, whereas inhibition of HO-1 and autophagy increased biochemical enzyme levels (*P<0.01 compared with the IR group; #P<0.05 compared with the HIR group). (D) The expression of HO-1 and autophagy proteins was upregulated by hemin, but downregulated by treatment with HO-1 siRNA (*P<0.01 compared with the sham group; #P<0.05 compared with the IR group; &P<0.05 compared with the HIR group).
Figure 7. HO-1 induced autophagy through activation…
Figure 7. HO-1 induced autophagy through activation of ERK1/2 and p38-MAPK.
(A) Apoptotic rates of the sham group and three treated groups are shown. Mineral oil treatment increased the rate of early and late apoptosis, and HO-1 siRNA treatment before IR increased the rate of apoptosis compared with the IR group. The HIR group had lower rates of apoptosis, similar to the sham group (*P#P<0.05 compared with the IR group; &P<0.05 compared with the HIR group). (B) Hemin treatment before IR increased phosphorylated ERK1/2 staining, as shown in immunoblotting, and HSIR group cells showed decreased phosphorylated ERK1/2 expression. Western blotting for phosphorylated p38-MAPK demonstrated increased phosphorylated p38-MAPK in the HIR group, but decreased signaling in the HSIR group (*P<0.01 compared with the sham group; #P<0.05 compared with the IR group; &P<0.05 compared with the HIR group). (C) HO-1 expression was decreased in the EIIR group compared with the IR group, and LC3-II expression was decreased in the PIIR group compared with the IR group (*P<0.05 compared with the sham group; #P<0.05 compared with the IR group). (D) HO-1 expression was nearly the same as in the PIIR and IR groups, but LC3-II expression was decreased in the EIIR group compared with the IR group (*P<0.05 compared with the sham group; #P<0.05 compared with the IR group).

References

    1. Teoh NC, Farrell GC (2003) Hepatic ischemia reperfusion injury: Pathogenic mechanisms and basis for hepatoprotection. Journal of Gastroenterology and Hepatology 18: 891–902.
    1. Selzner N, Rudiger H, Graf R, Clavien PA (2003) Protective Strategies Against Ischemic Injury of the Liver. Gastroenterology 125: 917–936.
    1. Przyklenk K, Bauer B, Ovize M, Kloner RA, Whittaker P (1993) Regional ischemic ‘preconditioning’ protects remote virgin myocardium from subsequent sustained coronary occlusion. Circulation 87: 893–9.
    1. Abu-Amara M, Yang SY, Quaglia A, Rowley P, Tapuria N, et al. (2011) Effect of Remote Ischemic Preconditioning on Liver Ischemia/Reperfusion Injury Using a New Mouse Model. Liver transplantation 17: 70–82.
    1. Morse D, Lin L, Choi AM, Ryter SW (2009) Heme Oxygenase-1, a Critical Arbitrator of Cell Death Pathways in Lung Injury and Disease. Free Radic Biol Med 47: 1–12.
    1. David A, Ferenbach David C, Kluth Jeremy Hughes (2010) Hemeoxygenase-1 and Renal Ischemia-Reperfusion Injury. Nephron Exp Nephrol 115: e33–7.
    1. Devey L, Mohr E, Bellamy C, Simpson K, Henderson N, et al. (2009) c-Jun Terminal Kinase-2 Gene Deleted Mice Overexpress Hemeoxygenase-1 and are Protected From Hepatic Ischemia Reperfusion Injury. Transplantation 88: 308–316.
    1. Lai IR, Chang KJ, Chen CF, Tsai HW (2006) Transient limb ischemia induces remote preconditioning in liver among rats: the protective role of heme oxygenase-1. Transplantation 81: 1311–7.
    1. Wang JH, Behrns KE, Leeuwenburgh C, Kim JS (2012) Critical role of autophagy in ischemia/reperfusion injury to aged livers. Autophagy 8: 140–141.
    1. Zeng S, Feirt N, Goldstein M, Guarrera J, Ippagunta N, et al. (2004) Blockade of Receptor for Advanced Glycation End Product (RAGE) Attenuates Ischemia and Reperfusion Injury to the Liver in Mice. Hepatology 39: 422–32.
    1. Zhang W, Zhang X, Lu H, Matsukura M, Zhao J, et al. (2013) Silencing heme oxygenase-1 gene expression in retinal pigment epithelial cells inhibits proliferation, migration and tube formation of cocultured endothelial cells. Biochemical and Biophysical Research Communications 434: 492–7.
    1. Abu-Amara M, Yang SY, Quaglia A, Rowley P, Tapuria N, et al. (2011) Remote Ischemic Preconditioning on Liver Ischemia/Reperfusion Injury Using a New Mouse Model. Liver transplantation 17: 70–82.
    1. Suzuki S, Toledo-Pereyra LH, Rodriguez FJ, Cejalvo D (1993) Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury. Modulating effects of FK506 and cyclosporine. Transplantation 55: 1265–72.
    1. Meldrum KK, Burnett AL, Meng X, Misseri R, Shaw MB, et al. (2003) Liposomal delivery of heat shock protein 72 into renal tubular cells blocks nuclear factor-kappaB activation, tumor necrosis factor-alpha production, and subsequent ischemia-induced apoptosis. Circ Res 92: 293–9.
    1. Choi BM, Pae HO, Kim YM, Chung HT (2003) Nitric Oxide-Mediated Cytoprotection of Hepatocytes From Glucose Deprivation-Induced Cytotoxicity Involvement of Heme Oxygenase-1. Hepatology 37: 810–23.
    1. Settembre C, Di Malta C, Polito VA, Garcia Arencibia M, Vetrini F, et al. (2011) TFEB Links Autophagy to Lysosomal Biogenesis. Science 332: 1429–33.
    1. Wang HQ, Xu YX, Zhu CQ (2012) Upregulation of Heme Oxygenase-1 by Acteoside Through ERK and PI3 K/Akt Pathway Confer Neuroprotection Against Beta-Amyloid-Induced Neurotoxicity. Neurotox Res 21: 368–78.
    1. Carchman EH, Rao J, Loughran PA, Rosengart MR, Zuckerbraun BS (2011) Heme oxygenase-1-mediated autophagy protects against hepatocyte cell death and hepatic injury from infection/sepsis in mice. Hepatology 53: 2053–62.
    1. Tapuria N, Junnarkar SP, Dutt N, Abu-Amara M, Fuller B, et al. (2009) Effect of remote ischemic preconditioning on hepatic microcirculation and function in a rat model of hepatic ischemia reperfusion injury. HPB (Oxford) 11: 108–17.
    1. Yun N, Kim SH, Lee SM (2012) Differential consequences of protein kinase C activation during early and late hepatic ischemic preconditioning. J Physiol Sci 62: 199–209.
    1. Devey L, Ferenbach D, Mohr E, Sangster K, Bellamy CO, et al. (2009) Tissue-resident Macrophages Protect the Liver From Ischemia Reperfusion Injury via a Heme Oxygenase-1-Dependent Mechanism. Molecular Therapy 17: 65–72.
    1. Nari Yun, Hyun-Ae Eum, Sun-Mee Lee (2010) Protective Role of Heme Oxygenase-1 Against Liver Damage Caused by Hepatic Ischemia and Reperfusion in Rats. Antioxid Redox Signal 13: 1503–12.
    1. Wang JH, Ahn IS, Fischer TD, Byeon JI, Dunn WA Jr, et al. (2011) Autophagy suppresses age-dependent ischemia and reperfusion injury in livers of mice. Gastroenterology 141 2188–2199: e6.
    1. Esposti DD, Domart MC, Sebagh M, Harper F, Pierron G, et al. (2010) Autophagy is induced by ischemic preconditioning in human livers formerly treated by chemotherapy to limit necrosis. Autophagy 6: 172–4.
    1. Rautou PE, Mansouri A, Lebrec D, Durand F, Valla D, et al. (2010) Autophagy in liver diseases. Journal of Hepatology 53: 1123–34.
    1. Kim JS, Nitta T, Mohuczy D, O'Malley KA, Moldawer LL, et al. (2008) Impaired autophagy: a mechanism of mitochondrial dysfunction in anoxic rat hepatocytes. Hepatology 47: 1725–1736.
    1. Cardinal J, Pan P, Dhupar R, Ross M, Nakao A, et al. (2009) Cisplatin prevents high mobility group box 1 release and is protective in a murine model of hepatic ischemia/reperfusion injury. Hepatology 50: 565–574.
    1. Kim I, Lemasters JJ (2011) Mitophagy Selectively Degrades Individual Damaged Mitochondria After Photoirradiation. Antioxid Redox Signal 14(10): 1919–28.
    1. Elmore SP, Qian T, Grissom SF, Lemasters JJ (2001) The mitochondrial permeability transition initiates autophagy in rat hepatocytes. The FASEB Journal express article 15: 2286–7.
    1. Shan Y, Pepe J, Lu TH, Elbirt KK, Lambrecht RW, et al. (2000) Induction of the Heme Oxygenase-1 Gene by Metalloporphyrins. Archives of Biochemistry and Biophysics. 380: 219–27.
    1. Wang HQ, Sun XB, Xu YX, Zhao H, Zhu QY, et al. (2010) Astaxanthin up-regulates heme oxygenase-1 expression through ERK1/2 pathway and its protective effect against beta-amyloid-induced cytotoxicity in SH-SY5Y cells. Brain research 1360: 159–67.
    1. Yang YC, Lii CK, Lin AH, Yeh YW, Yao HT, et al. (2011) Induction of glutathione synthesis and heme oxygenase 1 by the flavonoids butein and phloretin is mediated through the ERK/Nrf2 pathway and protects against oxidative stress. Free Radic Biol Med 51: 2073–81.
    1. Yao P, Nussler A, Liu L, Hao L, Song F, et al. (2007) Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. Journal of Hepatology 47: 253–61.
    1. Vardanian AJ, Busuttil RW, Kupiec-Weglinski JW (2008) Molecular Mediators of Liver Ischemia and Reperfusion Injury: A Brief Review. Mol Med 14: 337–45.
    1. Kim HS, Loughran PA, Billiar TR (2008) Carbon monoxide decreases the level of iNOS protein and active dimer in IL-1b-stimulated hepatocytes. Nitric Oxide 18: 256–65.

Source: PubMed

3
Abonnere