Journal Home > Volume 12 , Issue 6

Alcohol abuse has recently become a serious health concern worldwide, and the incidence of alcoholic liver disease (ALD) is rapidly increasing with high morbidity and mortality. Ferroptosis is a newly recognized form of regulated cell death caused by the iron-dependent accumulation of lipid peroxidation. Here we showed that the circadian clock protein BMAL1 in hepatocytes is both necessary and sufficient to protect against ALD by mitigating ferroptosis. Upon exposure to alcohol (5 % Lieber-DeCarli liquid alcohol diet for 10 days before binged alcohol with 5 g/kg body weight in vivo, 300 mmol/L for 12 h in vitro, respectively), the content of iron, reactive oxygen species (ROS) and malondialdehyde (MDA) was boosted significantly while glutathione (GSH) was decreased that mainly based on the downregulated protein expression of ferritin heavy chain (FTH), ferroportin (FPN), heme oxygenase1(HO-1) and anti-cystine/glutamate antiporter (SLC7A11), while these changes could be abolished by ferroptosis inhibitor Ferrostatin-1[Fer-1 (5 mg/kg body weight for 10 days in vivo, 10 μmol/L for 2 h in vitro, respectively)]. Further study indicated that the alcohol could activate the protein expression of brain and muscle arnt-like protein-1 (BMAL1) which exerts a protective effect against ferroptosis through promoting nuclear factor erythroid 2-related factor 2 (Nrf2) translocation into nuclear and subsequently stimulating its downstream proteins FTH, FPN, glutathione peroxidase 4 activity (GPX4), HO-1, SLC7A11, while knockdown of BMAL1 and Nrf2 by RNA interference further downregulated the expression of these protein and thus promoting ferroptosis in response to alcohol. Collectively, our results unveiled that the protective action of BMAL1 during alcohol challenge depends on its ability to activate Nrf2-ARE antiferroptosis pathway and targeting hepatic BMAL1 to dampen hepatic ferroptosis signaling may have therapeutic potential for ALD.


menu
Abstract
Full text
Outline
About this article

Protective effect of brain and muscle arnt-like protein-1 against ethanol-induced ferroptosis by activating Nrf2 in mice liver and HepG2 cells

Show Author's information Yanan ZhaoaRanran ZhangaZiheng ChenaZiyi WangaShuang Guana,b( )Jing Lua,b( )
College of Food Science and Engineering, Jilin University, Changchun 130062, China
Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

Alcohol abuse has recently become a serious health concern worldwide, and the incidence of alcoholic liver disease (ALD) is rapidly increasing with high morbidity and mortality. Ferroptosis is a newly recognized form of regulated cell death caused by the iron-dependent accumulation of lipid peroxidation. Here we showed that the circadian clock protein BMAL1 in hepatocytes is both necessary and sufficient to protect against ALD by mitigating ferroptosis. Upon exposure to alcohol (5 % Lieber-DeCarli liquid alcohol diet for 10 days before binged alcohol with 5 g/kg body weight in vivo, 300 mmol/L for 12 h in vitro, respectively), the content of iron, reactive oxygen species (ROS) and malondialdehyde (MDA) was boosted significantly while glutathione (GSH) was decreased that mainly based on the downregulated protein expression of ferritin heavy chain (FTH), ferroportin (FPN), heme oxygenase1(HO-1) and anti-cystine/glutamate antiporter (SLC7A11), while these changes could be abolished by ferroptosis inhibitor Ferrostatin-1[Fer-1 (5 mg/kg body weight for 10 days in vivo, 10 μmol/L for 2 h in vitro, respectively)]. Further study indicated that the alcohol could activate the protein expression of brain and muscle arnt-like protein-1 (BMAL1) which exerts a protective effect against ferroptosis through promoting nuclear factor erythroid 2-related factor 2 (Nrf2) translocation into nuclear and subsequently stimulating its downstream proteins FTH, FPN, glutathione peroxidase 4 activity (GPX4), HO-1, SLC7A11, while knockdown of BMAL1 and Nrf2 by RNA interference further downregulated the expression of these protein and thus promoting ferroptosis in response to alcohol. Collectively, our results unveiled that the protective action of BMAL1 during alcohol challenge depends on its ability to activate Nrf2-ARE antiferroptosis pathway and targeting hepatic BMAL1 to dampen hepatic ferroptosis signaling may have therapeutic potential for ALD.

Keywords: Ferroptosis, Alcohol, HepG2 cells, BMAL1, Nrf2, Mice liver

References(60)

[1]

A.K. Singal, R. Bataller, J. Ahn, et al., ACG clinical guideline: alcoholic liver disease, Am. J. Gastroenterol. 113 (2018) 175-194. https://doi.org/10.1038/ajg.2017.469.

[2]

O. Campollo, Alcohol and the liver: The return of the prodigal son, Ann. Hepatol. 18 (2019) 6-10. https://doi.org/10.5604/01.3001.0012.7854.

[3]

S. Hong, Y. Kim, J. Sung, et al., Jujube (Ziziphus jujuba Mill.) Protects hepatocytes against alcohol-induced damage through Nrf2 activation, evidence-based complement. Altern. Med. 2020 (2020) 6684331. https://doi.org/10.1155/2020/6684331.

[4]

P. Lei, T. Bai, Y. Sun, Mechanisms of ferroptosis and relations with regulated cell death: A review, Front. Physiol. 10 (2019) 139. https://doi.org/10.3389/fphys.2019.00139.

[5]

K. Shimada, R. Skouta, A. Kaplan, et al., Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis, Nat. Chem. Biol. 12(7) (2016) 497-503. https://doi.org/10.1038/nchembio.2079.

[6]

Y.T. Bai, R. Chang, H. Wang, et al., ENPP2 protects cardiomyocytes from erastin-induced ferroptosis, Biochem. Biophys. Res. Commun. 499 (2018) 44-51. https://doi.org/10.1016/j.bbrc.2018.03.113.

[7]
M. Abdalkader, R. Lampinen, K.M. Kanninen, et al., Targeting Nrf2 to suppress ferroptosis and mitochondrial dysfunction in neurodegeneration, 12 (2018) 1-9. https://doi.org/10.3389/fnins.2018.00466.
DOI
[8]

Y. Su, B. Zhao, L. Zhou, et al., Ferroptosis, a novel pharmacological mechanism of anti-cancer drugs, Cancer Lett. 483 (2020) 127-136. https://doi.org/10.1016/j.canlet.2020.02.015.

[9]

S. Wei, T. Qiu, X. Yao, et al., Arsenic induces pancreatic dysfunction and ferroptosis via mitochondrial ROS-autophagy-lysosomal pathway, J. Hazard. Mater. 384 (2020) 121390. https://doi.org/10.1016/j.jhazmat.2019.121390.

[10]

H. Zhou, F. Li, J.Y. Niu, et al., Ferroptosis was involved in the oleic acid-induced acute lung injury in mice, Sheng Li Xue Bao. 71 (2019) 689-697. https://doi.org/10.13294/j.aps.2019.0070.

[11]

Y. Zhao, J. Lu, A. Mao, et al., Autophagy inhibition plays a protective role in ferroptosis induced by alcohol via the p62-Keap1-Nrf2 pathway, J. Agric. Food Chem. 69 (2021) 9671-9683. https://doi.org/10.1021/acs.jafc.1c03751.

[12]

Q. Sun, C. Zeng, L. Du, et al., Mechanism of circadian regulation of the NRF2/ARE pathway in renal ischemia‑reperfusion, Exp. Ther. Med. 21 (2021) 1-9. https://doi.org/10.3892/etm.2021.9622.

[13]

M. Benedusi, E. Frigato, M. Beltramello, et al., Circadian clock as possible protective mechanism to pollution induced keratinocytes damage, Mech. Ageing Dev. 172 (2018) 13-20. https://doi.org/10.1016/j.mad.2017.08.017.

[14]

D. Zhang, X. Tong, B.B. Nelson, et al., The hepatic BMAL1/AKT/lipogenesis axis protects against alcoholic liver disease in mice via promoting PPARα pathway, Hepatology. 68 (2018) 883-896. https://doi.org/10.1002/hep.29878.

[15]

E.S. Musiek, M.M. Lim, G. Yang, et al., Circadian clock proteins regulate neuronal redox homeostasis and neurodegeneration, J. Clin. Invest. 123 (2013) 5389-5400. https://doi.org/10.1172/JCI70317.

[16]

M. Hastings, J.S. O’Neill, E.S. Maywood, Circadian clocks: regulators of endocrine and metabolic rhythms, J. Endocrinol. 195 (2007) 187-198. https://doi.org/10.1677/JOE-07-0378.

[17]

G. Qi, W. Wu, Y. Mi, et al., Tea polyphenols direct Bmal1-driven ameliorating of the redox imbalance and mitochondrial dysfunction in hepatocytes, Food Chem. Toxicol. 122 (2018) 181-193. https://doi.org/10.1016/j.fct.2018.10.031.

[18]

F. Okazaki, N. Matsunaga, H. Okazaki, et al., Circadian clock in a mouse colon tumor regulates intracellular iron levels to promote tumor progression, J. Biol. Chem. 291 (2016) 7017-7028. https://doi.org/10.1074/jbc.M115.713412.

[19]

J.O. Early, A.M. Curtis, Immunometabolism: Is it under the eye of the clock?, Semin. Immunol. 28 (2016) 478-490. https://doi.org/10.1016/j.smim.2016.10.006.

[20]

O. Barca-Mayo, M. Pons-Espinal, P. Follert, et al., Astrocyte deletion of Bmal1 alters daily locomotor activity and cognitive functions via GABA signalling., Nat. Commun. 8 (2017) 14336. https://doi.org/10.1038/ncomms14336.

[21]

X. Pan, S. Mota, B. Zhang, Circadian clock regulation on lipid metabolism and metabolic diseases, Adv. Exp. Med. Biol. 1276 (2020) 53-66. https://doi.org/10.1007/978-981-15-6082-8_5.

[22]

A. Angelousi, N. Nasiri-Ansari, E. Spilioti, et al., Altered expression of circadian clock genes in polyglandular autoimmune syndrome type III, Endocrine. 59 (2018) 109-119. https://doi.org/10.1007/s12020-017-1407-1.

[23]

R.K. Alexander, Y.H. Liou, N.H. Knudsen, et al., Bmal1 integrates mitochondrial metabolism and macrophage activation, Elife. 9 (2020). https://doi.org/10.7554/eLife.54090.

[24]

F.C. Kelleher, A. Rao, A. Maguire, Circadian molecular clocks and cancer., Cancer Lett. 342 (2014) 9-18. https://doi.org/10.1016/j.canlet.2013.09.040.

[25]

Q. Liu, L. Xu, M. Wu, et al., Rev-erbα exacerbates hepatic steatosis in alcoholic liver diseases through regulating autophagy, Cell Biosci. 11 (2021) 1-15. https://doi.org/10.1186/s13578-021-00622-4.

[26]

U.S. Udoh, J.A. Valcin, T.M. Swain, et al., Genetic deletion of the circadian clock transcription factor BMAL1 and chronic alcohol consumption differentially alter hepatic glycogen in mice., Am. J. Physiol. Gastrointest. Liver Physiol. 314 (2018) G431-G447. https://doi.org/10.1152/ajpgi.00281.2017.

[27]

M. Abdalkader, R. Lampinen, K.M. Kanninen, et al., Targeting Nrf2 to suppress ferroptosis and mitochondrial dysfunction in neurodegeneration, Front. Neurosci. 12 (2018) 466. https://doi.org/10.3389/fnins.2018.00466.

[28]

R.K. Thimmulappa, K.H. Mai, S. Srisuma, et al., Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray, Cancer Res. 62(18) (2002) 5196-5203.

[29]

A. Anandhan, M. Dodson, C.J. Schmidlin, et al., Breakdown of an ironclad defense system: the critical role of NRF2 in mediating ferroptosis, Cell Chem. Biol. 27(4) (2020) 436-447. https://doi.org/10.1016/j.chembiol.2020.03.011.

[30]

M. Yang, P. Chen, J. Liu, et al., Clockophagy is a novel selective autophagy process favoring ferroptosis, Sci. Adv. 5(7) (2019) eaaw2238. https://doi.org/10.1126/sciadv.aaw2238.

[31]

Y. Liu, Y. Wang, J. Liu, et al., The circadian clock protects against ferroptosis-induced sterile inflammation, Biochem. Biophys. Res. Commun. 525 (2020) 620-625. https://doi.org/10.1016/j.bbrc.2020.02.142.

[32]

A. Bertola, S. Mathews, S.H. Ki, et al., Mouse model of chronic and binge ethanol feeding (the NIAAA model), Nat. Protoc. 8 (2013) 627-637. https://doi.org/10.1038/nprot.2013.032.

[33]

Y.B. Qiu, B.B. Wan, G. Liu, et al., Nrf2 protects against seawater drowning-induced acute lung injury via inhibiting ferroptosis, Respir. Res. 21 (2020) 1-16. https://doi.org/10.1186/s12931-020-01500-2.

[34]

Y.Y. Zhang, Z.J. Ni, E. Elam, et al., Juglone, a novel activator of ferroptosis, induces cell death in endometrial carcinoma Ishikawa cells, Food Funct. 12(11) (2021) 4947-4959. https://doi.org/10.1039/d1fo00790d.

[35]

Y. Zhang, S. Zhao, Y. Fu, et al., Computational repositioning of dimethyl fumarate for treating alcoholic liver disease, Cell Death Dis. 11 (2020). https://doi.org/10.1038/s41419-020-02890-3.

[36]

H. Zhang, L. Xue, B. Li, et al., Vitamin D protects against alcohol-induced liver cell injury within an NRF2-ALDH2 feedback loop, Mol. Nutr. Food Res. 63 (2019) 1-9. https://doi.org/10.1002/mnfr.201801014.

[37]

M. Yao, H. Teng, Q. Lv, et al., Anti-hyperglycemic effects of dihydromyricetin in streptozotocin-induced diabetic rats, Food Sci. Hum. Wellness. 10(2) (2021) 155-162.

[38]

L. Chen, H. Teng, H. Cao, Chlorogenic acid and caffeic acid from Sonchus oleraceus Linn synergistically attenuate insulin resistance and modulate glucose uptake in HepG2 cells, Food Chem. Toxicol. 127 (2019) 182-187. https://doi.org/10.1016/j.fct.2019.03.038.

[39]

C. Lackner, D. Tiniakos, Fibrosis and alcohol-related liver disease, J. Hepatol. 70 (2019) 294-304. https://doi.org/10.1016/j.jhep.2018.12.003.

[40]

Z. Zhou, T.J. Ye, E. DeCaro, et al., Intestinal sIRT1 deficiency protects mice from ethanol-induced liver injury by mitigating ferroptosis, Am. J. Pathol. 190 (2020) 82-92. https://doi.org/10.1016/j.ajpath.2019.09.012.

[41]

J. Liu, H. He, J. Wang, et al., Oxidative stress-dependent frataxin inhibition mediated alcoholic hepatocytotoxicity through ferroptosis, Toxicology. 445 (2020) 152584. https://doi.org/10.1016/j.tox.2020.152584.

[42]

Z. Zhou, T.J. Ye, G. Bonavita, et al., Adipose‐specific lipin‐1 overexpression renders hepatic ferroptosis and exacerbates alcoholic steatohepatitis in mice, Hepatol. Commun. 3(5) (2019) 656-669. https://doi.org/10.1002/hep4.1333.

[43]
D. Zhang, X. Tong, B.B. Nelson, et al., Promoting PPAR a pathway, 68 (2018) 883-896. https://doi.org/10.1002/hep.29878.
DOI
[44]

Q. Wang, T. Gu, L. Ma, et al., Efficient iron utilization compensates for loss of extracellular matrix of ovarian cancer spheroids, Free Radic. Biol. Med. 164 (2021) 369-380. https://doi.org/10.1016/j.freeradbiomed.2021.01.001.

[45]

E. Nemeth, M.S. Tuttle, J. Powelson, et al., Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization, Science 306 (2004) 2090-2093. https://doi.org/10.1126/science.1104742.

[46]

S. Recalcati, M. Correnti, E. Gammella, et al., Iron metabolism in liver cancer stem cells, Front. Oncol. 9 (2019) 1-9. https://doi.org/10.3389/fonc.2019.00149.

[47]

K. Pantopoulos, Iron metabolism and the IRE/IRP regulatory system: An update, Ann. N. Y. Acad. Sci. 1012 (2004) 1-13. https://doi.org/10.1196/annals.1306.001.

[48]

Y. Kohgo, T. Ohtake, K. Ikuta, et al., Iron accumulation in alcoholic liver diseases., Alcohol. Clin. Exp. Res. 29 (2005) 189S-193S. https://doi.org/10.1097/01.alc.0000189274.00479.62.

[49]

J. Varghese, J.V. James, S. Sagi, et al., Decreased hepatic iron in response to alcohol may contribute to alcohol-induced suppression of hepcidin, Br. J. Nutr. 115 (2016) 1978-1986. https://doi.org/10.1017/S0007114516001197.

[50]

W. Dong, Y. Tan, Q. Qin, et al., Polybrominated diphenyl ethers quinone induces NCOA4-mediated ferritinophagy through selectively autophagic degradation of ferritin, Chem. Res. Toxicol. 32 (2019) 2509-2516. https://doi.org/10.1021/acs.chemrestox.9b00350.

[51]

A. Kohsaka, A.D. Laposky, K.M. Ramsey, et al., High-fat diet disrupts behavioral and molecular circadian rhythms in mice, Cell Metab. 6 (2007) 414-421. https://doi.org/10.1016/j.cmet.2007.09.006.

[52]

G. Asher, D. Gatfield, M. Stratmann, et al., SIRT1 regulates circadian clock gene expression through PER2 deacetylation, Cell. 134 (2008) 317-328. https://doi.org/10.1016/j.cell.2008.06.050.

[53]

Y.U. Doruk, D. Yarparvar, Y.K. Akyel, et al., A CLOCK-binding small molecule disrupts the interaction between clock and BMAL1 and enhances circadian rhythm amplitude. J. Biol. Chem. 295(11) (2020) 3518 -3531.

[54]

J.O. Early, D. Menon, C.A. Wyse, et al., Circadian clock protein BMAL1 regulates IL-1β in macrophages via NRF2, Proc. Natl. Acad. Sci. U. S. A. 115 (2018) E8460-E8468. https://doi.org/10.1073/pnas.1800431115.

[55]

X. Tan, L. Li, J. Wang, et al., Resveratrol prevents acrylamide-induced mitochondrial dysfunction and inflammatory responses via targeting circadian regulator bmal1 and Cry1 in hepatocytes, J. Agric. Food Chem. 67 (2019) 8510-8519. https://doi.org/10.1021/acs.jafc.9b03368.

[56]

F. Okazaki, N. Matsunaga, H. Okazaki, et al., Circadian rhythm of transferrin receptor 1 gene expression controlled by c-Myc in colon cancer-bearing mice, Cancer Res. 70 (2010) 6238-6246. https://doi.org/10.1158/0008-5472.CAN-10-0184.

[57]

R. Ben-Shlomo, R.A. Akhtar, B.H. Collins, et al., Light pulse-induced heme and iron-associated transcripts in mouse brain: A microarray analysis, Chronobiol. Int. 22 (2005) 455-471. https://doi.org/10.1081/CBI-200062353.

[58]

J. Wang, Y. Bai, S. Yin, et al., Circadian clock gene BMAL1 reduces urinary calcium oxalate stones formation by regulating NRF2/HO-1 pathway, Life Sci. 265 (2021) 118853. https://doi.org/10.1016/j.lfs.2020.118853.

[59]

Y. Yao, A. Zuo, Q. Deng, et al., Physcion protects against ethanol-induced liver injury by reprogramming of circadian clock, Front. Pharmacol. 11 (2020) 1-14. https://doi.org/10.3389/fphar.2020.573074.

[60]

Y.U. Doruk, D. Yarparvar, Y.K. Akyel, et al., A CLOCK-binding small molecule disrupts the interaction between CLOCK and BMAL1 and enhances circadian rhythm amplitude, J. Biol. Chem. 295 (2020) 3518-3531. https://doi.org/10.1074/jbc.RA119.011332.

Publication history
Copyright
Rights and permissions

Publication history

Received: 20 December 2021
Revised: 19 January 2022
Accepted: 24 February 2022
Published: 04 April 2023
Issue date: November 2023

Copyright

© 2023 Beijing Academy of Food Sciences.

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return