Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2019, Vol. 13 Issue (1) : 104-111    https://doi.org/10.1007/s11684-017-0568-x
RESEARCH ARTICLE
Rdh13 deficiency weakens carbon tetrachloride-induced liver injury by regulating Spot14 and Cyp2e1 expression levels
Xiaofang Cui1,2, Benting Ma1, Yan Wang3, Yan Chen1, Chunling Shen1, Ying Kuang2, Jian Fei2, Lungen Lu3(), Zhugang Wang1,2()
1. State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2. Shanghai Research Center for Model Organisms, Shanghai 201203, China
3. Department of Gastroenterology, Shanghai First People’s Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
 Download: PDF(4753 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Mitochondrion-localized retinol dehydrogenase 13 (Rdh13) is a short-chain dehydrogenase/reductase involved in vitamin A metabolism in both humans and mice. We previously generated Rdh13 knockout mice and showed that Rdh13 deficiency causes severe acute retinal light damage. In this study, considering that Rdh13 is highly expressed in mouse liver, we further evaluated the potential effect of Rdh13 on liver injury induced by carbon tetrachloride (CCl4). Although Rdh13 deficiency showed no significant effect on liver histology and physiological functions under regular culture, the Rdh13−/− mice displayed an attenuated response to CCl4-induced liver injury. Their livers also exhibited less histological changes and contained lower levels of liver-related metabolism enzymes compared with the livers of wild-type (WT) mice. Furthermore, the Rdh13−/− mice had Rdh13 deficiency and thus their liver cells were protected from apoptosis, and the quantity of their proliferative cells became lower than that in WT after CCl4 exposure. The ablation of Rdh13 gene decreased the expression levels of thyroid hormone-inducible nuclear protein 14 (Spot14) and cytochrome P450 (Cyp2e1) in the liver, especially after CCl4 treatment for 48 h. These data suggested that the alleviated liver damage induced by CCl4 in Rdh13−/− mice was caused by Cyp2e1 enzymes, which promoted reductive CCl4 metabolism by altering the status of thyroxine metabolism. This result further implicated Rdh13 as a potential drug target in preventing chemically induced liver injury.

Keywords retinol dehydrogenase 13      carbon tetrachloride      acute liver injury      Cyp2e1      Spot14     
Corresponding Author(s): Lungen Lu,Zhugang Wang   
Just Accepted Date: 25 December 2017   Online First Date: 16 April 2018    Issue Date: 12 March 2019
 Cite this article:   
Xiaofang Cui,Benting Ma,Yan Wang, et al. Rdh13 deficiency weakens carbon tetrachloride-induced liver injury by regulating Spot14 and Cyp2e1 expression levels[J]. Front. Med., 2019, 13(1): 104-111.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-017-0568-x
https://academic.hep.com.cn/fmd/EN/Y2019/V13/I1/104
Fig.1  Liver functions and histology remain normal in Rdh13−/− mice. (A) Serum levels of alanine aminotransferase, aspartate aminotransferase, and albumin are shown as mean values±S.E.M. from sex- and age-matched wild type and Rdh13−/− mice (n = 6, P>0.05). (B) Histological analyses of liver sections stained with H&E are shown at original magnifications of 100× and 400×, correspondingly.
Fig.2  Acute liver injury induced by CCl4 is alleviated in Rdh13−/− mice. (A) Serum ALT levels in WT and Rdh13−/− mice at the indicated times post-CCl4 exposure are expressed as mean±S.E.M. (n = 4, *P<0.01, **P<0.05). (B) Representative H&E-stained liver sections of WT and Rdh13−/− mice at the indicated times post-CCl4 exposure are shown at an original magnification of 100×.
Fig.3  Apoptosis induced by CCl4 treatment is reduced in Rdh13−/− livers. (A) In situ fluorescence TUNEL assay on liver frozen sections shows hepatocyte nuclei (blue, 4′,6-diamidino-2-phenylindole) and apoptotic cells (green, fluorescein isothiocyanate). (B) Quantitative analysis on apoptotic cells in the liver upon CCl4 treatment for the indicated time points (*P<0.05, **P<0.01). (C) Caspase-3 expression by immunohistochemistry in the liver upon CCl4 treatment for the indicated time points. (D) Western blot analyses for the indicated proteins in the livers of mice exposed to CCl4 for the indicated time points. Glyceraldehyde 3-phosphate dehydrogenase is shown as loading control.
Fig.4  Rdh13 deficiency reduced hepatocyte compensatory proliferation after CCl4-induced liver damage in mice. (A) In vivo Brdu incorporation assayed by immunohistochemistry in the liver upon CCl4 treatment for the indicated time points. (B) Percentages of Brdu-positive cells in the livers are shown as mean±S.E.M. (***P<0.001). (C) Cyclin D1 mRNA expression at the indicated time points in the livers by real-time RT-PCR is shown as mean±S.E.M. (*P<0.05, ** P<0.01). Protein levels of cyclin D1 in the livers after CCl4 treatment are shown by immunohistochemistry (D) and Western blot (E). (F) Western blot for p-ERK1/2 and total ERK1/2 in the livers of WT and Rdh13−/− mice after CCl4 treatment is shown.
Fig.5  Decreased expression levels of Cyp2e1 and Spot14 in the livers of Rdh13−/− mice after CCl4 treatment. (A) Expression levels of Cyp2e1 mRNA and protein by real-time PCR (mean±S.E.M., ** P<0.01) and Western blot (B) show reduced Cyp2e1 expression levels in Rdh13−/− livers 24 h after CCl4 exposure. The expression levels of Spot14 in the livers after CCl4 treatment are detected by quantitative real-time PCR (C, mean±S.E.M., * P<0.05, ** P<0.01) and Western blot (D).
1 OVBelyaeva, OV Korkina, AVStetsenko, NYKedishvili. Human retinol dehydrogenase 13 (RDH13) is a mitochondrial short-chain dehydrogenase/reductase with a retinaldehyde reductase activity. FEBS J 2008; 275(1): 138–147
https://doi.org/10.1111/j.1742-4658.2007.06184.x pmid: 18039331
2 KLKavanagh, H Jörnvall, BPersson, UOppermann. Medium- and short-chain dehydrogenase/reductase gene and protein families: the SDR superfamily: functional and structural diversity within a family of metabolic and regulatory enzymes. Cell Mol Life Sci 2008; 65(24): 3895–3906
https://doi.org/10.1007/s00018-008-8588-y pmid: 19011750
3 UOppermann, C Filling, MHult, NShafqat, XWu, M Lindh, JShafqat, ENordling, YKallberg, BPersson, HJörnvall. Short-chain dehydrogenases/reductases (SDR): the 2002 update. Chem Biol Interact 2003; 143-144: 247–253
https://doi.org/10.1016/S0009-2797(02)00164-3 pmid: 12604210
4 HJörnvall, B Persson, MKrook, SAtrian, RGonzàlez-Duarte, JJeffery, DGhosh. Short-chain dehydrogenases/reductases (SDR). Biochemistry 1995; 34(18): 6003–6013
https://doi.org/10.1021/bi00018a001 pmid: 7742302
5 AKramm, M Kisiela, RSchulz, EMaser. Short-chain dehydrogenases/reductases in cyanobacteria. FEBS J 2012; 279(6): 1030–1043
https://doi.org/10.1111/j.1742-4658.2012.08494.x pmid: 22251568
6 ASimon, U Hellman, CWernstedt, UEriksson. The retinal pigment epithelial-specific 11-cis retinol dehydrogenase belongs to the family of short chain alcohol dehydrogenases. J Biol Chem 1995; 270(3): 1107–1112
https://doi.org/10.1074/jbc.270.3.1107 pmid: 7836368
7 CADriessen, BP Janssen, HJWinkens, AHvan Vugt, TLde Leeuw, JJJanssen. Cloning and expression of a cDNA encoding bovine retinal pigment epithelial 11-cis retinol dehydrogenase. Invest Ophthalmol Vis Sci 1995; 36(10): 1988–1996
pmid: 7544779
8 FHaeseleer, GF Jang, YImanishi, CAGGDriessen, MMatsumura, PSNelson, KPalczewski. Dual-substrate specificity short chain retinol dehydrogenases from the vertebrate retina. J Biol Chem 2002; 277(47): 45537–45546
https://doi.org/10.1074/jbc.M208882200 pmid: 12226107
9 HWang, X Cui, QGu, YChen, J Zhou, YKuang, ZWang, X Xu. Retinol dehydrogenase 13 protects the mouse retina from acute light damage. Mol Vis 2012; 18: 1021–1030
pmid: 22605914
10 KKuniyoshi, H Sakuramoto, KYoshitake, KAbe, K Ikeo, MFuruno, KTsunoda, SKusaka, YShimomura, TIwata. Longitudinal clinical course of three Japanese patients with Leber congenital amaurosis/early-onset retinal dystrophy with RDH12 mutation. Doc Ophthalmol 2014; 128(3): 219–228
https://doi.org/10.1007/s10633-014-9436-z pmid: 24752437
11 KKovalovich, RA DeAngelis, WLi, EEFurth, GCiliberto, RTaub. Increased toxin-induced liver injury and fibrosis in interleukin-6-deficient mice. Hepatology 2000; 31(1): 149–159
https://doi.org/10.1002/hep.510310123 pmid: 10613740
12 TOtsuka, H Takagi, NHoriguchi, MToyoda, KSato, H Takayama, MMori. CCl4-induced acute liver injury in mice is inhibited by hepatocyte growth factor overexpression but stimulated by NK2 overexpression. FEBS Lett 2002; 532(3): 391–395
https://doi.org/10.1016/S0014-5793(02)03714-6 pmid: 12482598
13 CYu, F Wang, CJin, XWu, WK Chan, WLMcKeehan. Increased carbon tetrachloride-induced liver injury and fibrosis in FGFR4-deficient mice. Am J Pathol 2002; 161(6): 2003–2010
https://doi.org/10.1016/S0002-9440(10)64478-1 pmid: 12466116
14 CGHuh, VM Factor, ASánchez, KUchida, EAConner, SSThorgeirsson. Hepatocyte growth factor/c-met signaling pathway is required for efficient liver regeneration and repair. Proc Natl Acad Sci USA 2004; 101(13): 4477–4482
https://doi.org/10.1073/pnas.0306068101 pmid: 15070743
15 YYamada, N Fausto. Deficient liver regeneration after carbon tetrachloride injury in mice lacking type 1 but not type 2 tumor necrosis factor receptor. Am J Pathol 1998; 152(6): 1577–1589
pmid: 9626061
16 SBisht, MA Khan, MBekhit, HBai, T Cornish, MMizuma, MARudek, MZhao, A Maitra, BRay, DLahiri, AMaitra, RAAnders. A polymeric nanoparticle formulation of curcumin (NanoCurc™) ameliorates CCl4-induced hepatic injury and fibrosis through reduction of pro-inflammatory cytokines and stellate cell activation. Lab Invest 2011; 91(9): 1383–1395
https://doi.org/10.1038/labinvest.2011.86 pmid: 21691262
17 FZhang, R Shu, XWu, XZhao, D Feng, LWang, SLu, Q Liu, YXiang, JFei, L Huang, ZWang. Delayed liver injury and impaired hepatocyte proliferation after carbon tetrachloride exposure in BPOZ2-deficient mice. Toxicol Lett 2009; 188(3): 201–207
https://doi.org/10.1016/j.toxlet.2009.04.009 pmid: 19393728
18 MNatsume, H Tsuji, AHarada, MAkiyama, TYano, H Ishikura, INakanishi, KMatsushima, SKaneko, NMukaida. Attenuated liver fibrosis and depressed serum albumin levels in carbon tetrachloride-treated IL-6-deficient mice. J Leukoc Biol 1999, 66: 601–608
pmid: 10534116
19 MBBansal, K Kovalovich, RGupta, WLi, A Agarwal, BRadbill, CEAlvarez, RSafadi, MIFiel, SLFriedman, RATaub. Interleukin-6 protects hepatocytes from CCl4-mediated necrosis and apoptosis in mice by reducing MMP-2 expression. J Hepatol 2005; 42(4): 548–556
https://doi.org/10.1016/j.jhep.2004.11.043 pmid: 15763341
20 EPGomez-Sanchez, V Ganjam, YJChen, YLiu, SA Clark, CEGomez-Sanchez. The 11β hydroxysteroid dehydrogenase 2 exists as an inactive dimer. Steroids 2001; 66(11): 845–848
https://doi.org/10.1016/S0039-128X(01)00119-2 pmid: 11576624
21 OVBelyaeva, NY Kedishvili. Human pancreas protein 2 (PAN2) has a retinal reductase activity and is ubiquitously expressed in human tissues. FEBS Lett 2002; 531(3): 489–493
https://doi.org/10.1016/S0014-5793(02)03588-3 pmid: 12435598
22 TSKim, A Maeda, TMaeda, CHeinlein, NKedishvili, KPalczewski, PSNelson. Delayed dark adaptation in 11-cis-retinol dehydrogenase-deficient mice: a role of RDH11 in visual processes in vivo. J Biol Chem 2005; 280(10): 8694–8704
https://doi.org/10.1074/jbc.M413172200 pmid: 15634683
23 AIannaccone, SA Tedesco, KTGallaher, HYamamoto, SCharles, TPDryja. Fundus albipunctatus in a 6-year old girl due to compound heterozygous mutations in the RDH5 gene. Doc Ophthalmol 2007; 115(2): 111–116
https://doi.org/10.1007/s10633-007-9054-0 pmid: 17476461
24 MAjmal, MI Khan, KNeveling, YMKhan, SHAli, W Ahmed, MSIqbal, MAzam, AI den Hollander, RWCollin, RQamar, FPCremers. Novel mutations in RDH5 cause fundus albipunctatus in two consanguineous Pakistani families. Mol Vis 2012; 18: 1558–1571
pmid: 22736946
25 JHAlbrecht, LK Hansen. Cyclin D1 promotes mitogen-independent cell cycle progression in hepatocytes. Cell Growth Differ 1999; 10: 397–404
26 FVégran, R Boidot, ESolary, SLizard-Nacol. A short caspase-3 isoform inhibits chemotherapy-induced apoptosis by blocking apoptosome assembly. PLoS One 2011; 6(12): e29058
https://doi.org/10.1371/journal.pone.0029058 pmid: 22216167
27 RWJohnstone, AA Ruefli, SWLowe. Apoptosis: a link between cancer genetics and chemotherapy. Cell 2002; 108(2): 153–164
https://doi.org/10.1016/S0092-8674(02)00625-6 pmid: 11832206
28 PJDlugosz, LP Billen, MGAnnis, WZhu, Z Zhang, JLin, BLeber, DWAndrews. Bcl-2 changes conformation to inhibit Bax oligomerization. EMBO J 2006; 25(11): 2287–2296
https://doi.org/10.1038/sj.emboj.7601126 pmid: 16642033
29 JHAlbrecht, LK Hansen. Cyclin D1 promotes mitogen-independent cell cycle progression in hepatocytes. Cell Growth Differ 1999; 10(6): 397–404
pmid: 10392901
30 NFausto. Liver regeneration. J Hepatol 2000; 32(1 Suppl): 19–31
https://doi.org/10.1016/S0168-8278(00)80412-2 pmid: 10728791
31 OCoqueret. Linking cyclins to transcriptional control. Gene 2002; 299(1-2): 35–55
https://doi.org/10.1016/S0378-1119(02)01055-7 pmid: 12459251
32 NFausto, JS Campbell, KJRiehle. Liver regeneration. Hepatology 2006; 43(Suppl 1): S45–S53
https://doi.org/10.1002/hep.20969 pmid: 16447274
33 YXie, H Hao, HWang, CGuo, A Kang, GWang. Reversing effects of lignans on CCl4-induced hepatic CYP450 down regulation by attenuating oxidative stress. J Ethnopharmacol 2014; 155(1): 213–221
https://doi.org/10.1016/j.jep.2014.05.016
34 DWRosenberg, GS Drummond, TJSmith. Depletion of cytochrome P-450 by thyroid hormone and cobalt-protoporphyrin IX in rat liver: evidence that susceptibility varies among forms of the heme protein. Pharmacology 1995; 51(4): 254–262
https://doi.org/10.1159/000139367 pmid: 8577819
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed