Laminarin ameliorates alcohol-induced liver damage and its molecular mechanism in mice
Tianyi Guo
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorLingfeng Zhu
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, China
Search for more papers by this authorYaping Zhou
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorShuai Han
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorYunyun Cao
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorZuomin Hu
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorYi Luo
Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha, China
Search for more papers by this authorLiyuan Bao
Department of logistics, Changsha University, Changsha, China
Search for more papers by this authorXiuxiu Wu
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorDandan Qin
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorCorresponding Author
Qinlu Lin
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Correspondence
Qinlu Lin and Feijun Luo, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China.
Email: [email protected] and [email protected]
Search for more papers by this authorCorresponding Author
Feijun Luo
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Correspondence
Qinlu Lin and Feijun Luo, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China.
Email: [email protected] and [email protected]
Search for more papers by this authorTianyi Guo
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorLingfeng Zhu
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, China
Search for more papers by this authorYaping Zhou
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorShuai Han
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorYunyun Cao
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorZuomin Hu
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorYi Luo
Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha, China
Search for more papers by this authorLiyuan Bao
Department of logistics, Changsha University, Changsha, China
Search for more papers by this authorXiuxiu Wu
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorDandan Qin
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Search for more papers by this authorCorresponding Author
Qinlu Lin
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Correspondence
Qinlu Lin and Feijun Luo, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China.
Email: [email protected] and [email protected]
Search for more papers by this authorCorresponding Author
Feijun Luo
Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, China
Correspondence
Qinlu Lin and Feijun Luo, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China.
Email: [email protected] and [email protected]
Search for more papers by this authorAbstract
Alcoholic liver disease (ALD) has become a health issue globally. Laminarin, a low molecular weight marine-derived β-glucan, has been identified with multiple biological activities. In this study, the ameliorative effect on ALD of laminarin isolated from brown algae was investigated. Phenotypic, pathological alterations and biochemical characteristics indicated that laminarin administration (100 mg/kg/day) significantly alleviated liver injury and improved liver function in the alcohol-induced mice. Gene chip results indicated that laminarin treatment caused 52 up-regulated and 13 down-regulated genes in the hepatic tissues of alcohol-induced damage mice, and most of these genes are associated with regulation of oxidative stress (such as CYP450/glutathione-dependent antioxidation), Wnt signaling pathway, retinol metabolism, and cAMP pathway based on GO and KEGG analysis. PPI network analysis indicated that the downstream target genes lied in the hub of the net. Our experiments also confirmed the changed expressions of some target genes. Taken together, these results suggest that laminarin can ameliorate alcohol-induced damage in mice and its molecular mechanism lies in modulating anti-oxidation pathway, WNT pathway, and cAMP pathway, which regulate the expressions of downstream target genes and alleviate alcohol-induced damage. Our study provides new clue to prevent alcohol-induced damage and will be benefit to develop functional foods.
Practical applications
This study verified the positive effect on alcoholic liver disease (ALD) of laminarin, a water-soluble brown algae-derived β-glucan, linked by β-(1,3) glycosidic bonds with β-(1,6) branches. Laminarin significantly alleviated liver injury and improved liver function of ALD mice. Moreover, transcriptomics and bioinformatics analysis further revealed the gene expression patterns, hub targets, and signalings including CYP450/glutathione, Wnt, retinol metabolism, cAMP pathways regulated by laminarin. This research is the first evidence for hepatoprotective effect of laminarin against ALD and its molecular mechanism, which will be advantage to develop functional foods or adjuvant therapy of ALD.
CONFLICT OF INTEREST
The authors confirm that they have no conflicts of interest with respect to the work described in this manuscript.
Open Research
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the corresponding author upon reasonable request.
REFERENCES
- Anders, S., Pyl, P. T., & Huber, W. (2015). HTSeq—A python framework to work with high-throughput sequencing data. Bioinformatics, 31(2), 166–169. https://doi.org/10.1093/bioinformatics/btu638
- Aroor, A. R., Jackson, D. E., & Shukla, S. D. (2012). Dysregulated phosphorylation and nuclear translocation of cyclic AMP response element binding protein (CREB) in rat liver after chronic ethanol binge. European Journal of Pharmacology, 679, 101–108. https://doi.org/10.1016/j.ejphar.2011.12.045
- Avila, D. V., Barker, D. F., Zhang, J., McClain, C. J., Barve, S., & Gobejishvili, L. (2016). Dysregulation of hepatic cAMP levels via altered Pde4b expression plays a critical role in alcohol-induced steatosis. Journal of Pathology, 240(1), 96–107. https://doi.org/10.1002/path.4760
- Baraona, E., & Lieber, C. S. (1979). Effects of ethanol on lipid metabolism. Journal of Lipid Research, 20(3), 289–315. https://doi.org/10.1007/978-3-642-70048-4_4
- Bates, C. J. (1995). Vitamin A. Lancet, 345(8941), 31–35. https://doi.org/10.1016/s0140-6736(95)91157-x
- Bolger, A. M., Lohse, M., & Usadel, B. (2014). Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics, 30(15), 2114–2120. https://doi.org/10.1093/bioinformatics/btu170
- Cederbaum, A. I. (2012). Alcohol metabolism. Clinical Liver Disease, 16(4), 667–685. https://doi.org/10.1016/j.cld.2012.08.002
10.1016/j.cld.2012.08.002 Google Scholar
- Cederbaum, A. I., Lu, Y., & Wu, D. (2009). Role of oxidative stress in alcohol-induced liver injury. Archives of Toxicology, 83(6), 519–548. https://doi.org/10.1007/s00204-009-0432-0
- Chen, J., Schenker, S., & Henderson, G. I. (2002). 4-hydroxynonenal detoxification by mitochondrial glutathione S-transferase is compromised by short-term ethanol consumption in rats. Alcoholism-Clinical and Experimental Research, 26(8), 1252–1258. https://doi.org/10.1097/01.ALC.0000024081.89523.81
- Chen, J., Yang, J., Du, H., Aslam, M., Wang, W., Chen, W., Li, T., Liu, Z., & Liu, X. (2021). Laminarin, a major polysaccharide in stramenopiles. Marine Drugs, 19(10), 576. https://doi.org/10.3390/md19100576
- de Rooij, J., Zwartkruis, F. J., Verheijen, M. H., Cool, R. H., Nijman, S. M., Wittinghofer, A., & Bos, J. L. (1998). Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature, 396(6710), 474–477. https://doi.org/10.1038/24884
- Elnagdy, M., Barve, S., McClain, C., & Gobejishvili, L. (2020). cAMP signaling in pathobiology of alcohol associated liver disease. Biomolecules, 10(10), 1433. https://doi.org/10.3390/biom10101433
- Eriksen, J., Olsen, P. S., & Thomsen, A. C. (1984). Gamma-glutamyltranspeptidase, aspartate aminotransferase, and erythrocyte mean corpuscular volume as indicators of alcohol consumption in liver disease. Scandinavian Journal of Gastroenterology, 19(6), 813–819. https://doi.org/10.1080/00365521.1984.12005813
- Guo, F., Zheng, K., Benedé-Ubieto, R., Cubero, F. J., & Nevzorova, Y. A. (2018). The Lieber-Decarli diet—A flagship model for experimental alcoholic liver disease. Alcoholism-Clinical and Experimental Research, 42(10), 1828–1840. https://doi.org/10.1111/acer.13840
- Guo, T., Akan, O. D., Luo, F., & Lin, Q. (2021). Dietary polysaccharides exert biological functions via epigenetic regulations: Advance and prospectives. Critical Reviews in Food Science and Nutrition, 6, 1–11. https://doi.org/10.1080/10408398.2021.1944974
- Guo, T., Lin, Q., Li, X., Nie, Y., Wang, L., Shi, L., Xu, W., Hu, T., Guo, T., & Luo, F. (2017). Octacosanol attenuates inflammation in both raw264.7 macrophages and a mouse model of colitis. Journal of Agricultural and Food Chemistry, 65(18), 3647–3658. https://doi.org/10.1021/acs.jafc.6b05465
- Haberkorn, V., Heydel, J. M., Mounie, J., Artur, Y., & Goudonnet, H. (2002). Vitamin a modulates the effects of thyroid hormone on UDP-glucuronosyltransferase expression and activity in rat liver. Molecular and Cellular Endocrinology, 190, 167–175. https://doi.org/10.1016/s0303-7207(01)00681-5
- Hoyumpa, A. M. (1986). Mechanisms of vitamin deficiencies in alcoholism. Alcoholism-Clinical and Experimental Research, 10(6), 573–581. https://doi.org/10.1111/j.1530-0277.1986.tb05147.x
- Hugbart, C., Verres, Y., Le Daré, B., Bucher, S., Vène, E., Bodin, A., Lagente, V., Fromenty, B., Bouvet, R., Morel, I., Loyer, P., & Gicquel, T. (2020). Non-oxidative ethanol metabolism in human hepatic cells in vitro: Involvement of uridine diphospho-glucuronosyltransferase 1A9 in ethylglucuronide production. Toxicology in Vitro, 66, 104842. https://doi.org/10.1016/j.tiv.2020.104842
- Kanehisa, M., Araki, M., Goto, S., Hattori, M., Hirakawa, M., Itoh, M., Katayama, T., Kawashima, S., Okuda, S., Tokimatsu, T., & Yamanishi, Y. (2008). KEGG for linking genomes to life and the environment. Nucleic Acids Research, 36, D480–D484. https://doi.org/10.1093/nar/gkm882
- Kim, D., Langmead, B., & Salzberg, S. L. (2015). HISAT: A fast spliced aligner with low memory requirements. Nature Methods, 12(4), 357–360. https://doi.org/10.1038/nmeth.3317
- Leung, T. M., & Lu, Y. (2017). Alcoholic liver disease: From CYP2E1 to CYP2A5. Current Molecular Pharmacology, 10(3), 172–178. https://doi.org/10.2174/1874467208666150817111846
- Li, H. L., Xie, Z. Y., Zhang, Y., Liu, Y., Niu, A. J., Liu, Y. Y., Zhang, L. B., & Guan, L. L. (2021). Rosa rugosa polysaccharide attenuates alcoholic liver disease in mice through the gut-liver axis. Food Bioscience, 44, 101385. https://doi.org/10.1016/j.fbio.2021.101385
- Lieber, C. S. (1991). Alcohol, liver, and nutrition. Journal of the American College of Nutrition, 10(6), 602–632. https://doi.org/10.1080/07315724.1991.10718182
- Liu, Y., Tang, T., Duan, S., Li, C., Lin, Q., Wu, H., Liu, A., Hu, B., Wu, D., Li, S., Shen, L., & Wu, W. (2020). The purification, structural characterization and antidiabetic activity of a polysaccharide from Anoectochilus roxburghii. Food & Function, 11(4), 3730–3740. https://doi.org/10.1039/c9fo00860h
- Louvet, A., & Mathurin, P. (2015). Alcoholic liver disease: Mechanisms of injury and targeted treatment. Nature Reviews Gastroenterology and Hepatology, 12(4), 231–242. https://doi.org/10.1038/nrgastro.2015.35
- Malyarenko, O. S., Usoltseva, R. V., Zvyagintseva, T. N., & Ermakova, S. P. (2019). Laminaran from brown alga Dictyota dichotoma and its sulfated derivative as radioprotectors and radiosensitizers in melanoma therapy. Carbohydrate Polymers, 206, 539–547. https://doi.org/10.1016/j.carbpol.2018.11.008
- Matsushita, H., & Takaki, A. (2019). Alcohol and hepatocellular carcinoma. BMJ Open Gastroenterology, 6(1), e000260. https://doi.org/10.1136/bmjgast-2018-000260
- Mercer, K. E., Hennings, L., & Ronis, M. J. (2015). Alcohol consumption, Wnt/β-catenin signaling, and hepatocarcinogenesis. Advances in Experimental Medicine and Biology, 815, 185–195. https://doi.org/10.1007/978-3-319-09614-8_11
- Neyrinck, A. M., Mouson, A., & Delzenne, N. M. (2007). Dietary supplementation with laminarin, a fermentable marine beta (1-3) glucan, protects against hepatotoxicity induced by LPS in rat by modulating immune response in the hepatic tissue. International Immunopharmacology, 7(12), 1497–1506. https://doi.org/10.1016/j.intimp.2007.06.011
- Nguyen, S. G., Kim, J., Guevarra, R. B., Lee, J. H., Kim, E., Kim, S. I., & Unno, T. (2016). Laminarin favorably modulates gut microbiota in mice fed a high-fat diet. Food & Function, 7(10), 4193–4201. https://doi.org/10.1039/c6fo00929h
- Perugorria, M. J., Olaizola, P., Labiano, I., Esparza-Baquer, A., Marzioni, M., Marin, J. J. G., Bujanda, L., & Banales, J. M. (2019). Wnt-β-catenin signalling in liver development, health and disease. Nature Reviews Gastroenterology & Hepatology, 16(2), 121–136. https://doi.org/10.1038/s41575-018-0075-9
- Petrella, C., Carito, V., Carere, C., Ferraguti, G., Ciafrè, S., Natella, F., Bello, C., Greco, A., Ralli, M., Mancinelli, R., Messina, M. P., Fiore, M., & Ceccanti, M. (2020). Oxidative stress inhibition by resveratrol in alcohol-dependent mice. Nutrition, 79-80, 110783. https://doi.org/10.1016/j.nut.2020.110783
- Rashed, Z. E., Grasselli, E., Khalifeh, H., Canesi, L., & Demori, I. (2022). Brown-algae polysaccharides as active constituents against nonalcoholic fatty liver disease. Planta Medica, 88(1), 9–19. https://doi.org/10.1055/a-1273-3159
- Roberts, A., Trapnell, C., Donaghey, J., Rinn, J. L., & Pachter, L. (2011). Improving RNA-seq expression estimates by correcting for fragment bias. Genome Biology, 12(3), R22. https://doi.org/10.1186/gb-2011-12-3-r22
- Seitz, H. K., Bataller, R., Cortez-Pinto, H., Gao, B., Gual, A., Lackner, C., Mathurin, P., Mueller, S., Szabo, G., & Tsukamoto, H. (2018). Alcoholic liver disease. Nature Reviews Disease Primers, 4(1), 16. https://doi.org/10.1038/s41572-018-0014-7
- Shaywitz, A. J., & Greenberg, M. E. (1999). CREB: A stimulus-induced transcription factor activated by a diverse array of extracellular signals. Annual Review of Biochemistry, 68, 821–861. https://doi.org/10.1146/annurev.biochem.68.1.821
- Trapnell, C., William, B. A., Pertea, G., Mortazavi, A., Kwan, G., van Baren, M. J., Salzberg, S. L., Wold, B. J., & Pachter, L. (2010). Transcript assembly and quantification by RNA-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nature Biotechnolgy, 28(5), 511–515. https://doi.org/10.1038/nbt.1621
- Undi, R. B., Gutti, U., Sahu, I., Sarvothaman, S., Pasupuleti, S. R., Kandi, R., & Gutti, R. K. (2016). Wnt signaling: Role in regulation of haematopoiesis. Indian Journal of Hematology and Blood Transfusion, 32(2), 123–134. https://doi.org/10.1007/s12288-015-0585-3
- Wahlang, B., McClain, C., Barve, S., & Gobejishvili, L. (2018). Role of cAMP and phosphodiesterase signaling in liver health and disease. Cellular Signalling, 49, 105–115. https://doi.org/10.1016/j.cellsig.2018.06.005
- Wang, Q., Dai, X., Yang, W., Wang, H., Zhao, H., Yang, F., Yang, Y., Li, J., & Lv, X. (2015). Caffeine protects against alcohol-induced liver fibrosis by dampening the cAMP/PKA/CREB pathway in rat hepatic stellate cells. International Immunopharmacology, 25(2), 340–352. https://doi.org/10.1016/j.intimp.2015.02.012
- Wei, J., Zhang, L., Liu, J., Pei, D., Wang, N., Wang, H., Di, D., & Liu, Y. (2020). Protective effect of Lycium barbarum polysaccharide on ethanol-induced injury in human hepatocyte and its mechanism. Journal of Food Biochemistry, 44(10), e13412. https://doi.org/10.1111/jfbc.13412
- Wu, D., & Cederbaum, A. I. (2009). Oxidative stress and alcoholic liver disease. Seminars in Liver Disease, 29(2), 141–154. https://doi.org/10.1055/s-0029-1214370
- Yan, X., Liu, X., Wang, Y., Ren, X., Ma, J., Song, R., Wang, X., Dong, Y., Fan, Q., Wei, J., Yu, A., Sui, H., & She, G. (2022). Multi-omics integration reveals the hepatoprotective mechanisms of ursolic acid intake against chronic alcohol consumption. European Journal of Nutrition, 61(1), 115–126. https://doi.org/10.1007/s00394-021-02632-x
- Yang, Y., Ji, J., Di, L., Li, J., Hu, L., Qiao, H., Wang, L., & Feng, Y. (2020). Resource, chemical structure and activity of natural polysaccharides against alcoholic liver damages. Carbohydrate Polymers, 241, 116355. https://doi.org/10.1016/j.carbpol.2020.116355
- Yang, Y., Yang, F., Wu, X., Lv, X., & Li, J. (2016). EPAC activation inhibits acetaldehyde-induced activation and proliferation of hepatic stellate cell via Rap1. Canadian Journal of Physiology and Pharmacology, 94(5), 498–507. https://doi.org/10.1139/cjpp-2015-0437
- Zargarzadeh, M., Amaral, A. J. R., Custódio, C. A., & Mano, J. F. (2020). Biomedical applications of laminarin. Carbohydrate Polymers, 232, 115774. https://doi.org/10.1016/j.carbpol.2019.115774