AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (706.8 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

The triterpenoids-enriched extracts from Antrodia cinnamomea mycelia attenuate alcohol-induced chronic liver injury via suppression lipid accumulation in C57BL/6 mice

Yange Liua,bRonglong ChenaLanzhou LibRuitao DongbHui YincYawen WangaAnhui YangbJianbin WangaChangtian Lid( )Di Wangb,d( )
School of Basic Medical Sciences, Nanchang University, Nanchang 330038, China
School of Life Sciences, Jilin University, Changchun 130012, China
Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China

Peer review under responsibility of KeAi Communications Co., Ltd

Show Author Information

Abstract

The major pathologic hallmark of the alcoholic liver disease (ALD) is the representation of chronic alcohol-induced hepatocyte lipid accumulation. This study aims to investigate the hepatoprotective role of triterpenoids-enriched extracts from Antrodia cinnamomea mycelia (ACT) in chronic alcohol-induced liver injury mice, establishing in C57BL/6 mice through gradient alcohol feeding for 24 weeks. In long-term alcohol consumption mice, the significantly lost body weight, increased organ indexes, hepatic alanine aminotransferase and aspartate aminotransferase levels were all remissed after 6-week ACT orally administration, showing its hepatoprotective property. ACT suppressed the triglyceride, total cholesterol and low-density lipoprotein levels, and enhanced high-density lipoprotein levels in serum or/and liver of chronic alcohol damaged mice. Combining with the pathological observations, ACT displayed an anti-steatosis effects to restrain the progress of ALD. Based on proteomic analysis and enzyme-linked immunosorbent assay, ACT had been confirmed to regulate the levels of lipid biogeneration-related factors and depressed the over-accumulation of hepatic reactive oxygen species. According to further data, ACT prevented alcoholic liver injury may be associated with mediating lipid metabolism-related to PGC-1α and NF-κB signaling. In summary, ACT protected the body against chronic alcohol ingest induced liver injury through its regulation lipid on metabolism.

References

[1]

S. Ramaiah, C. Rivera, G. Arteel, Early-phase alcoholic liver disease: an update on animal models, pathology, and pathogenesis, Int, J. Toxicol. 23 (4) (2004) 217-231. https://doi.org/10.1080/10915810490502069

[2]

P. Sakhuja, Pathology of alcoholic liver disease, can it be differentiated from nonalcoholic steatohepatitis?, World J. Gastroenterol. 20 (44) (2014) 16474-16479. https://doi.org/10.3748/wjg.v20.i44.16474

[3]

A. Mahli, C. Hellerbrand, Alcohol and obesity: a dangerous association for fatty liver disease, Dig. Dis. 34 (Suppl 1) (2016) 32-39. https://doi.org/10.1159/000447279

[4]

C.S. Lieber, R. Schmid, The effect of ethanol on fatty acid metabolism; stimulation of hepatic fatty acid synthesis in vitro, J. Clin. Invest. 40 (2) (1961) 394-399. https://doi.org/10.1172/JCI104266

[5]

A. Galli, D. Price, D. Crabb, High-level expression of rat class I alcohol dehydrogenase is sufficient for ethanol-induced fat accumulation in transduced HeLa cells, Hepatology 29 (4) (1999) 1164-1170. https://doi.org/10.1002/hep.510290420

[6]

C.P. Day, O.F. James, Steatohepatitis: a tale of two "hits"?, Gastroenterology 11 (4) (1998) 842-845. https://doi.org/10.1016/s0016-5085(98)70599-2

[7]

S. Bansal, S. Srinivasan, S. Anandasadagopan, et al., Additive effects of mitochondrion-targeted cytochrome CYP2E1 and alcohol toxicity on cytochrome c oxidase function and stability of respirosome complexes, J. Biol. Chem. 287 (19) (2012) 15284-15297. https://doi.org/10.1074/jbc.M111.314062

[8]

Y.K. Lu, J. Zhuge, X.D. Wang, et al., Cytochrome P450 2E1 contributes to ethanol-induced fatty liver in mice, Hepatology 47 (5) (2008) 1483-1494. https://doi.org/10.1002/hep.22222

[9]

J.M. Cacicedo, N. Yagihashi, J.F. Keaney, et al., AMPK inhibits fatty acid-induced increases in NF-kappaB transactivation in cultured human umbilical vein endothelial cells, Biochem. Biophys. Res. Commun. 324 (4) (2004) 1204-1209. https://doi.org/10.1016/j.bbrc.2004.09.177

[10]

A.A. Nanji, K. Jokelainen, G.L. Tipoe, et al., Curcumin prevents alcohol-induced liver disease in rats by inhibiting the expression of NF-κB-dependent genes, Am. J. Physiol. Gastrointest. Liver Physiol. 284 (2) (2003) 321-327. https://doi.org/10.1152/ajpgi.00230.2002

[11]

M.K. Lu, M.H. Lee, C.H. Chao, et al., Physiochemical changes and mechanisms of anti-inflammation effect of sulfated polysaccharides from ammonium sulfate feeding of Antrodia cinnamomea, Int. J. Biol. Macromol. 148 (2020) 715-721. https://doi.org/10.1016/j.ijbiomac.2020.01.110

[12]

Y.Y. Chang, Y.C. Liu, Y.H. Kuo, et al., Effects of antrosterol from Antrodia camphorata submerged whole broth on lipid homeostasis, antioxidation, alcohol clearance, and anti-inflammation in livers of chronic-alcohol fed mice, J, Ethnopharmacol. 202 (2017) 200-207. https://doi.org/10.1016/j.jep.2017.03.003

[13]

K.J.S. Kumar, F.H. Chu, H.W. Hsieh, et al., Antroquinonol from ethanolic extract of mycelium of Antrodia cinnamomea protects hepatic cells from ethanol-induced oxidative stress through Nrf-2 activation, J. Ethnopharmacol. 136 (1) (2011) 168-177. https://doi.org/10.1016/j.jep.2011.04.030

[14]

Y. Liu, Z.Q. Wang, F.G. Kong, et al., Triterpenoids extracted from Antrodia cinnamomea mycelia attenuate acute alcohol-induced liver injury in C57BL/6 mice via suppression inflammatory response, Front. Microbiol. 11 (2020) 1113. https://doi.org/10.3389/fmicb.2020.01113

[15]

T.W. Ma, Y. Lai, F.C. Yang, Enhanced production of triterpenoid in submerged cultures of Antrodia cinnamomea with the addition of citrus peel extract, Bioprocess Biosyst. Eng. 37 (11) (2014) 2251-2261. https://doi.org/10.1007/s00449-014-1203-8

[16]

Z.Q. Wang, Y.G. Liu, X.Y. Zhao, et al., Aronia melanocarpa prevents alcohol-induced chronic liver injury via regulation of Nrf2 signaling in C57BL/6 mice, Oxid. Med. Cell. Longev. 2020 (2020) 1-13. https://doi.org/10.1155/2020/4054520

[17]

M.L. Ojeda, O. Carreras, P. Sobrino, et al., Biological implications of selenium in adolescent rats exposed to binge drinking: oxidative, immunologic and apoptotic balance, Toxicol, Appl. Pharmacol. 329 (2017) 165-172. https://doi.org/10.1016/j.taap.2017.05.037

[18]

B. Nath, I. Levin, T. Csak, et al., Hepatocyte-specific hypoxia-inducible factor-1α is a determinant of lipid accumulation and liver injury in alcohol-induced steatosis in mice, Hepatology 53 (5) (2011) 1526-1537. https://doi.org/10.1002/hep.24256

[19]

A. Mahli, W.E. Thasler, E. Patsenker, et al., Identification of cytochrome CYP2E1 as critical mediator of alcohol effects on steatotic hepatocytes, Oncogarget. 53 (1) (2015) 40464-40478. https://doi.org/10.18632/oncotarget.6203

[20]

C.F. Cheng, H.C. Ku, H. Lin, PGC-1α as a pivotal factor in lipid and metabolic regulation, Int. J. Mol. Sci. 19 (11) (2018) 3447. https://doi.org/10.3390/ijms19113447

[21]

B. Li, D.S. Lee, Y. Kang, et al., Protective effect of ganodermanondiol isolated from the Lingzhi mushroom against tert-butyl hydroperoxide-induced hepatotoxicity through Nrf2-mediated antioxidant enzymes, Food Chem. Toxicol. 53 (2013) 317-324. https://doi.org/10.1016/j.fct.2012.12.016

[22]

X.R. Peng, J.Q. Liu, X.H. Han, et al., Protective effects of triterpenoids from Ganoderma resinaceum on H2O2-induced toxicity in HepG2 cells, Food Chem. 141 (2) (2013) 920-926. https://doi.org/10.1016/j.foodchem.2013.03.071

[23]

L.P. Breitling, V. Arndt, C. Drath, et al., Liver enzymes: interaction analysis of smoking with alcohol consumption or BMI, comparing AST and ALT to γ-GT, Plos One 6 (11) (2011) e27951. https://doi.org/10.1371/journal.pone.0027951

[24]

A. Mahli, C. Hellerbrand, Alcohol and obesity: a dangerous association for fatty liver disease, Dig. Dis. 34 (Suppl 1) (2016) 32-39. https://doi.org/10.1159/000447279

[25]

Y. Israel, J.M. Khanna, H. Orrego, et al., Studies on metabolic tolerance to alcohol, hepatomegaly and alcoholic liver disease, Drug Alcohol Depend. 4 (1–2) (1979) 107. https://doi.org/10.1016/0376-8716(79)90051-6

[26]

A. Pashaj, X.H. Yi, M.N. Xia, et al., Characterization of genome-wide transcriptional changes in liver and adipose tissues of ZDF (fa/fa) rats fed R-α-lipoic acid by next-generation sequencing, Physiol. Genomics. 45 (23) (2013) 1136-1143. https://doi.org/10.1152/physiolgenomics.00138.2013

[27]

T.R. Altamimi, P.D. Thomas, A.M. Darwesh, et al., Cytosolic carnitine acetyltransferase as a source of cytosolic acetyl-CoA: a possible mechanism for regulation of cardiac energy metabolism, Biochem. J. 475 (5) (2018) 959-976. https://doi.org/10.1042/BCJ20170823

[28]

K. Kienesberger, A.G. Pordes, T.G. Völk, et al., L-carnitine and PPARα-agonist fenofibrate are involved in the regulation of Carnitine Acetyltransferase (CrAT) mRNA levels in murine liver cells, BMC Genomics 15 (1) (2014) 514. https://doi.org/10.1186/1471-2164-15-514

[29]

K. Rasineni, P.G. Thomes, J.L. Kubik, et al., Chronic alcohol exposure alters circulating insulin and ghrelin levels: role of ghrelin in hepatic steatosis, Am. J. Physiol. -Gastr. L. 316 (4) (2019) G453-G461. https://doi.org/10.1152/ajpgi.00334.2018

[30]

E.J. Freyse, T. Giessmann, K.J. Petzke, et al., Effects of fatty acids on hepatic amino acid catabolism and fibrinogen synthesis in young healthy volunteers, Am. J. Physiol. Endocrinol. Metab. 285 (1) (2003) E54-E62. https://doi.org/10.1152/ajpendo.00324.2002

[31]

F.A. Hunt, D.B. Rylatt, R.A.H. Art, et al., Serum crosslinked fibrin (XDP) and fibrinogen/fibrin degradation products (FDP) in disorders associated with activation of the coagulation or fibrinolytic systems, Br. J. Haematol. 60 (4) (1985) 715-722. https://doi.org/10.1111/j.1365-2141.1985.tb07476.x

[32]

D.B. Jeffrey, J.D. Horton, Molecular mediators of hepatic steatosis and liver injury, J. Clin. Invest. 2 (114) (2004) 147-152. https://doi.org/10.1172/JCI22422

[33]

J. Wang, M. Olin, B. Rozell, et al., Differential hepatocellular zonation pattern of cholesterol 7α-hydroxylase (Cyp7a1) and sterol 12α-hydroxylase (Cyp8b1) in the mouse, Histochem. Cell Biol. 127 (3) (2007) 253-261. https://doi.org/10.1007/s00418-006-0239-5

[34]

B. Yerushalmi, R. Dahl, M.W. Devereaux, et al., Bile acid-induced rat hepatocyte apoptosis is inhibited by antioxidants and blockers of the mitochondrial permeability transition, Hepatology 33 (3) (2003) 616-626. https://doi.org/10.1053/jhep.2001.22702

[35]

E. Ip, G.C. Farrell, G. Robertson, et al., Central role of PPARalpha-dependent hepatic lipid turnover in dietary steatohepatitis in mice, Hepatology 38 (1) (2003) 123-132. https://doi.org/10.1053/jhep.2003.50307

[36]

A.K. Madiraju, T. Alves, X.J. Zhao, et al., Argininosuccinate synthetase regulates hepatic AMPK linking protein catabolism and ureagenesis to hepatic lipid metabolism, PNAS 113 (24) (2016) E3423-E3430. https://doi.org/10.1073/pnas.1606022113

[37]

L.L. Li, Y. Yao, J.L. Zhao, et al., Dehydroepiandrosterone protects against hepatic glycolipid metabolic disorder and insulin resistance induced by high fat via activation of AMPK-PGC-1alpha-NRF-1 and IRS1-AKT-GLUT2 signaling pathways, Int. J. Obes (Lond). 44 (5) (2020) 1075-1086. https://doi.org/10.1038/s41366-019-0508-8

[38]

S.C. Fu, J.M. Liu, K.I. Lee, et al., Cr(VI) induces ROS-mediated mitochondrial-dependent apoptosis in neuronal cells via the activation of Akt/ERK/AMPK signaling pathway, Toxicol. In Vitro 65 (2020) 104795. https://doi.org/10.1016/j.tiv.2020.104795

[39]

R. Memmott, J. Gills, P. Dennis, Activation of AMP-activated protein kinase (AMPK) by the lipid-based Akt inhibitors, phosphatidylinositol ether lipid analogues (PIAs), Cancer Res

[40]

T.Y. Huang, D.H. Zheng, J.A. Houmard, et al., Overexpression of PGC-1α increases peroxisomal and mitochondrial fatty acid oxidation in human primary myotubes, Am. J. Physiol. Endocrinol. Metab. 312 (4) (2017) E253-E263. https://doi.org/10.1152/ajpendo.00331.2016

[41]

Q.H. Huang, L.Q. Xu, Y.H. Liu, et al., Polydatin protects rat liver against ethanol-induced injury: involvement of CYP2E1/ROS/Nrf2 and TLR4/NF-κB p65 pathway, Evid. Based Complement Alternat. Med. 2017 (1) (2017) 7953850. https://doi.org/10.1155/2017/7953850

[42]

M. Morgan, Z.G. Liu, Crosstalk of reactive oxygen species and NF-κB signaling, Cell Res. 21 (1) (2011) 103-115. https://doi.org/10.1038/cr.2010.178

[43]

L.V. Madrid, M.W. Mayo, J.Y. Reuther, et al., Akt stimulates the transactivation potential of the RelA/p65 Subunit of NF-kappa B through utilization of the Ikappa B kinase and activation of the mitogen-activated protein kinase p38, J. Biol. Chem. 276 (22) (2001) 18934-18940. https://doi.org/10.1074/jbc.M101103200

[44]

R.M. Hashem, L.A. Rashed, K.M.A. Hassanin, et al., Effect of 6-gingerol on AMPK- NF-kappa B axis in high fat diet fed rats, Biomed. Pharmacother. 88 (2017) 293-301. https://doi.org/10.1016/j.biopha.2017.01.035

[45]

P.S. Eisele, S. Salatino, J. Sobek, et al., The peroxisome proliferator-activated receptor γ coactivator 1α/β (PGC-1) coactivators repress the transcriptional activity of NF-κB in skeletal muscle cells, J. Biol. Chem. 288 (4) (2013) 2246-2260. https://doi.org/10.1074/jbc.M112.375253

[46]

A.A. Nanji, K. Jokelainen, G.L. Tipoe, et al., Curcumin prevents alcohol-induced liver disease in rats by inhibiting the expression of NF-kappa B-dependent genes, Am. J. Physiol. Gastrointest. Liver Physiol. 284 (2) (2003) G321-G327. https://doi.org/10.1152/ajpgi.00230.2002

Food Science and Human Wellness
Pages 497-507
Cite this article:
Liu Y, Chen R, Li L, et al. The triterpenoids-enriched extracts from Antrodia cinnamomea mycelia attenuate alcohol-induced chronic liver injury via suppression lipid accumulation in C57BL/6 mice. Food Science and Human Wellness, 2021, 10(4): 497-507. https://doi.org/10.1016/j.fshw.2021.04.012

550

Views

39

Downloads

7

Crossref

9

Web of Science

8

Scopus

1

CSCD

Altmetrics

Received: 30 August 2020
Revised: 29 September 2020
Accepted: 30 September 2020
Published: 04 June 2021
© 2021 Beijing Academy of Food Sciences. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

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

Return