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 (1.6 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

The food and medicinal homological resources benefiting patients with hyperlipidemia: categories, functional components, and mechanisms

Xu-Ting Hao1,Rui Peng5,Miao Guan1,4Huan-Jie Zhang1,4Yuhan Guo4Nashwa M. Shalapy2,3Xu-Qiang Liu1,3,4( )Chang-Yang Ma1,3,4( )
National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
Microbial Chemistry Department, Biotechnology Research Institute, National Research Centre, Cairo 12622, Egypt
Joint International Research Laboratory of Food & Medicine Resource Function, Henan, Kaifeng 475004, China
College of Agriculture, Henan University, Kaifeng 475004, China
School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China

These authors contributed equally to this work.

Show Author Information

Highlights

(1) This review focuses on the active ingredients and their mechanisms of action of food & medicine homology resources with lipid-regulating effects.

(2) The effects of 55 active ingredients of 53 food & medicine homology resources on regulating blood lipids were reviewed.

(3) Triterpenes and their glycosides, flavonoids, alkaloids and polysaccharides are the main active ingredients of food & medicine homology resources in regulating blood lipids.

(4) The active ingredients of food & medicine homology resources can regulate blood lipids mainly by regulating AMPK pathway, cholesterol metabolism pathway, PPARγ signaling pathway and gut microbiota.

Graphical Abstract

This article reviews the types, frequency of use, active ingredients, and mechanisms of lipid-lowering drugs and homologous resources in food. A total of 53 drug-food analogues with lipid-regulating effects were found, and the main active ingredients were triterpenoids and their glycosides, flavonoids, alkaloids, and polysaccharides. They mainly regulate blood lipids by controlling AMPK, PPARγ, cholesterol metabolism, and modulating the gut microbiota.

Abstract

Hyperlipidemia is a kind of lipid metabolism disease, whose pathogenesis is complex and diverse, mainly related to abnormal glucose and lipid metabolism, as well as insulin resistance and other characteristics. Because of their low toxic and side effects along with clear medicinal effects, the food and medicinal homological resources are widely used in the regulation of blood lipids in recent years. The State Administration for Market Regulation website and various databases have been searched for the use of food and medicinal homological materials in functional food products, the categories and frequency in food and medicinal homological resources, the lipid-lowering active ingredients and their mechanisms. The results showed that 53 kinds of food and medicinal homological resources were used to regulate blood lipids, of which Crataegus pinnatifida Bge., Cassia obtusifolia L., Nelumbo nucifera Gaertn., Morus alba L. and Pueraria lobata (Willd.) Ohwi were used most frequently. The main active ingredients are triterpenes and their glycosides, flavonoids, alkaloids and polysaccharides, etc., they regulated blood lipid levels and cholesterol metabolism by activating AMPK and PPARγ.

References

[1]

Navar-Boggan, A. M., Peterson, E. D., D'Agostino, R. B., et al. Hyperlipidemia in early adulthood increases long-term risk of coronary heart disease. Circulation, 2015, 131: 451–458. https://doi.org/10.1161/CIRCULATIONAHA.114.012477

[2]

Wilemon, K. A., Patel, J., Aguilar-Salinas, C., et al. Reducing the clinical and public health burden of familial hypercholesterolemia: A global call to action. JAMA Cardiol, 2020, 5: 217–229. https://doi.org/10.1001/jamacardio.2019.5173

[3]

Lemp, J. M., Nuthanapati, M. P., Bärnighausen, T. W., et al. Use of lifestyle interventions in primary care for individuals with newly diagnosed hypertension, hyperlipidaemia or obesity: A retrospective cohort study. Journal of the Royal Society of Medicine, 2022, 115: 289–299. https://doi.org/10.1177/01410768221077381

[4]

Jung, U. J., Choi, M. S. Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. International Journal of Molecular Sciences, 2014, 15: 6184–6223. https://doi.org/10.3390/ijms15046184

[5]

Choi, H. D., Shin, W. G. Safety and efficacy of statin treatment alone and in combination with fibrates in patients with dyslipidemia: A meta-analysis. Current Medical Research and Opinion, 2014, 30: 1–10. https://doi.org/10.1185/03007995.2013.842165

[6]

Mancini, G. B., Baker, S., Bergeron, J., et al. Diagnosis, prevention, and management of statin adverse effects and intolerance: Canadian consensus working group update. The Canadian Journal of Cardiology, 2016, 32: S35–S65. https://doi.org/10.1016/j.cjca.2016.01.003

[7]

Liu, X. Q., Wang, S. Y., Cui, L. L., et al. Flowers: Precious food and medicine resources. Food Science and Human Wellness, 2023, 12: 1020–1052. https://doi.org/10.1016/j.fshw.2022.10.022

[8]

Zhen, H. M., Yan, Q. J., Liu, Y. H., et al. Chitin oligosaccharides alleviate atherosclerosis progress in ApoE-/- mice by regulating lipid metabolism and inhibiting inflammation. Food Science and Human Wellness, 2022, 11: 999–1009. https://doi.org/10.1016/j.fshw.2022.03.027

[9]

Hou, Y., Jiang, J. G. Origin and concept of medicine food homology and its application in modern functional foods. Food & Function, 2013, 4: 1727–1741. https://doi.org/10.1039/c3fo60295h

[10]

Song, D. X., Jiang, J. G. Hypolipidemic components from medicine food homology species used in China: Pharmacological and health effects. Archives of Medical Research, 2017, 48: 569–581. https://doi.org/10.1016/j.arcmed.2018.01.004

[11]

Ma, A., Zou, F. M., Zhang, R. W., et al. The effects and underlying mechanisms of medicine and food homologous flowers on the prevention and treatment of related diseases. Journal of Food Biochemistry, 2022, 46: e14430. https://doi.org/10.1111/jfbc.14430

[12]

Fan, W. X., Huang, Y. L., Zheng, H., et al. Ginsenosides for the treatment of metabolic syndrome and cardiovascular diseases: Pharmacology and mechanisms. Biomedecine & Pharmacotherapie, 2020, 132: 110915. https://doi.org/10.1016/j.biopha.2020.110915

[13]

Sun, Y. Y., Liu, Y., Chen, K. J. Roles and mechanisms of ginsenoside in cardiovascular diseases: Progress and perspectives. Science China Life Sciences, 2016, 59: 292–298. https://doi.org/10.1007/s11427-016-5007-8

[14]

Shuai, M. Y., Yang, Y., Bai, F. Q., et al. Geographical origin of American ginseng ( Panax quinquefolius L.) based on chemical composition combined with chemometric. Journal of Chromatography A, 2022, 1676: 463284. https://doi.org/10.1016/j.chroma.2022.463284

[15]

Lu, C., Zhao, S. J., Wei, G. N., et al. Functional regulation of ginsenoside biosynthesis by RNA interferences of a UDP-glycosyltransferase gene in Panax ginseng and Panax quinquefolius. Plant Physiology and Biochemistry, 2017, 111: 67–76. https://doi.org/10.1016/j.plaphy.2016.11.017

[16]

Hunter, P. M., Hegele, R. A. Functional foods and dietary supplements for the management of dyslipidaemia. Nature Reviews Endocrinology, 2017, 13: 278–288. https://doi.org/10.1038/nrendo.2016.210

[17]

Wu, M., Liu, L. T., Xing, Y. W., et al. Roles and mechanisms of hawthorn and its extracts on atherosclerosis: A review. Frontiers in Pharmacology, 2020, 11: 118. https://doi.org/10.3389/fphar.2020.00118

[18]

Shinozaki, F., Kamei, A., Watanabe, Y., et al. Propagule powder of Japanese yam ( dioscorea japonica) reduces high-fat diet-induced metabolic stress in mice through the regulation of hepatic gene expression. Molecular Nutrition & Food Research, 2020, 64: e2000284. https://doi.org/10.1002/mnfr.202000284

[19]

Cao, H., Chen, X. Q., Jassbi, A. R., et al. Microbial biotransformation of bioactive flavonoids. Biotechnology Advances, 2015, 33: 214–223. https://doi.org/10.1016/j.biotechadv.2014.10.012

[20]

Pei, R. S., Liu, X. C., Bolling, B. Flavonoids and gut health. Current Opinion in Biotechnology, 2020, 61: 153–159. https://doi.org/10.1016/j.copbio.2019.12.018

[21]

Maleki, S. J., Crespo, J. F., Cabanillas, B. Anti-inflammatory effects of flavonoids. Food Chemistry, 2019, 299: 125124. https://doi.org/10.1016/j.foodchem.2019.125124

[22]

Zhou, Y.X., Zhang, H., Peng, C. Puerarin: a review of pharmacological effects. Phytotherapy Research, 2014, 28: 961–975. https://doi.org/10.1002/ptr.5083

[23]

Xu, D. X., Guo, X. X., Zeng, Z., et al. Puerarin improves hepatic glucose and lipid homeostasis in vitro and in vivo by regulating the AMPK pathway. Food & Function, 2021, 12: 2726–2740. https://doi.org/10.1039/d0fo02761h

[24]

Zhang, W., Liu, C. Q., Wang, P. W., et al. Puerarin improves insulin resistance and modulates adipokine expression in rats fed a high-fat diet. European Journal of Pharmacology, 2010, 649: 398–402. https://doi.org/10.1016/j.ejphar.2010.09.054

[25]

Wang, Z. F., Liu, J., Yang, Y. A., et al. A review: The anti-inflammatory, anticancer and antibacterial properties of four kinds of licorice flavonoids isolated from licorice. Current Medicinal Chemistry, 2020, 27: 1997–2011. https://doi.org/10.2174/0929867325666181001104550

[26]

Jafari, F., Jafari, M., Moghadam, A. T., et al. A review of glycyrrhiza glabra (licorice) effects on metabolic syndrome. Advances in Experimental Medicine and Biology, 2021, 1328: 385–400. https://doi.org/10.1007/978-3-030-73234-9_25

[27]

Abookleesh, F. L., Al-Anzi, B. S., Ullah, A. Potential antiviral action of alkaloids. Molecules, 2022, 27: 903. https://doi.org/10.3390/molecules27030903

[28]

Zhou, C., Huang, R., Cai, Y., et al. Research progress on mechanism of hypolipidemic effect of alkaloid components in natural products. Chinese Traditional and Herbal Drugs, 2024, 55: 1717–1727. https://doi.org/10.7501/j.issn.0253-2670.2024.05.029

[29]

Zheng, H. X., Han, L. T., Shi, W. F., et al. Research advances in lotus leaf as Chinese dietary herbal medicine. The American Journal of Chinese Medicine, 2022, 50: 1423–1445. https://doi.org/10.1142/S0192415X22500616

[30]

Zhu, X. Y., Si, F., Hao, R. L., et al. Nuciferine protects against obesity-induced nephrotoxicity through its hypolipidemic, anti-inflammatory, and antioxidant effects. Journal of Agricultural and Food Chemistry, 2023, 71: 18769–18779. https://doi.org/10.1021/acs.jafc.3c05735

[31]

Xu, H. Y., Wang, L. J., Yan, K. M., et al. Nuciferine inhibited the differentiation and lipid accumulation of 3T3-L1 preadipocytes by regulating the expression of lipogenic genes and adipokines. Frontiers in Pharmacology, 2021, 12: 632236. https://doi.org/10.3389/fphar.2021.632236

[32]

Zhou, L., Wang, Q. Y., Zhang, H., et al. YAP inhibition by nuciferine via AMPK-mediated downregulation of HMGCR sensitizes pancreatic cancer cells to gemcitabine. Biomolecules, 2019, 9: 620. https://doi.org/10.3390/biom9100620

[33]

Li, W. F., Yu, J. J., Zhao, J. M., et al. Poria Cocos polysaccharides reduces high-fat diet-induced arteriosclerosis in ApoE (-/-) mice by inhibiting inflammation. Phytotherapy Research, 2021, 35: 2220–2229. https://doi.org/10.1002/ptr.6980

[34]

Liang, Z., Yuan, Z. H., Li, G. Y., et al. Hypolipidemic, antioxidant, and antiapoptotic effects of polysaccharides extracted from reishi mushroom, ganoderma lucidum (leysser: Fr) Karst, in mice fed a high-fat diet. Journal of Medicinal Food, 2018, 21: 1218–1227. https://doi.org/10.1089/jmf.2018.4182

[35]

Park, I. S., Kim, B., Han, Y., et al. Decursin and decursinol angelate suppress adipogenesis through activation of β-catenin signaling pathway in human visceral adipose-derived stem cells. Nutrients, 2019, 12: 13. https://doi.org/10.3390/nu12010013

[36]

Zhou, F., Ding, M. N., Gu, Y. Q., et al. Aurantio-obtusin attenuates non-alcoholic fatty liver disease through AMPK-mediated autophagy and fatty acid oxidation pathways. Frontiers in Pharmacology, 2021, 12: 826628. https://doi.org/10.3389/fphar.2021.826628

[37]

Liu, X. Y., Yang, Z. H., Li, H. X., et al. Chrysophanol alleviates metabolic syndrome by activating the SIRT6/AMPK signaling pathway in brown adipocytes. Oxidative Medicine and Cellular Longevity, 2020, 2020: 7374086. https://doi.org/10.1155/2020/7374086

[38]

Yarley, O. P. N., Kojo, A. B., Zhou, C. S., et al. Reviews on mechanisms of in vitro antioxidant, antibacterial and anticancer activities of water-soluble plant polysaccharides. International Journal of Biological Macromolecules, 2021, 183: 2262–2271. https://doi.org/10.1016/j.ijbiomac.2021.05.181

[39]

Dong, X. L., Zhou, M. Z., Li, Y. H., et al. Cardiovascular protective effects of plant polysaccharides: A review. Frontiers in Pharmacology, 2021, 12: 783641. https://doi.org/10.3389/fphar.2021.783641

[40]

Duan, Y. T., Huang, J. J., Sun, M. J., et al. Poria Cocos polysaccharide improves intestinal barrier function and maintains intestinal homeostasis in mice. International Journal of Biological Macromolecules, 2023, 249: 125953. https://doi.org/10.1016/j.ijbiomac.2023.125953

[41]

Lu, J. H., He, R. J., Sun, P. L., et al. Molecular mechanisms of bioactive polysaccharides from Ganoderma lucidum (Lingzhi), a review. International Journal of Biological Macromolecules, 2020, 150: 765–774. https://doi.org/10.1016/j.ijbiomac.2020.02.035

[42]

Jia, J., Bissa, B., Brecht, L., et al. AMPK, a regulator of metabolism and autophagy, is activated by lysosomal damage via a novel galectin-directed ubiquitin signal transduction system. Molecular Cell, 2020, 77: 951–969.e9. https://doi.org/10.1016/j.molcel.2019.12.028

[43]

Li, T., Xu, L.H., Yan, Q.J., et al. Sucrose-free hawthorn leathers formulated with fructooligosaccharides and xylooligosaccharides ameliorate high-fat diet induced inflammation, glucose and lipid metabolism in liver of mice. Food Science and Human Wellness, 2022, 11: 1064–1075. https://doi.org/10.1016/j.fshw.2022.03.033

[44]

Steinberg, G. R., Carling, D. AMP-activated protein kinase: The Current landscape for drug development. Nature Reviews Drug Discovery, 2019, 18: 527–551. https://doi.org/10.1038/s41573-019-0019-2

[45]

Ma, C. J., Li, G., He, Y. F., et al. Pronuciferine and nuciferine inhibit lipogenesis in 3T3-L1 adipocytes by activating the AMPK signaling pathway. Life Sciences, 2015, 136: 120–125. https://doi.org/10.1016/j.lfs.2015.07.001

[46]

Ryu, J., Kim, M. J., Kim, T. O., et al. Piperlongumine as a potential activator of AMP-activated protein kinase in HepG2 cells. Natural Product Research, 2014, 28: 2040–2043. https://doi.org/10.1080/14786419.2014.919283

[47]

Quan, H. Y., Yuan, H. D., Jung, M. S., et al. Ginsenoside Re lowers blood glucose and lipid levels via activation of AMP-activated protein kinase in HepG2 cells and high-fat diet fed mice. International Journal of Molecular Medicine, 2012, 29: 73–80. https://doi.org/10.3892/ijmm.2011.805

[48]

Du, Q., Zhang, S. H., Li, A. Y., et al. Astragaloside IV inhibits adipose lipolysis and reduces hepatic glucose production via Akt dependent PDE3B expression in HFD-fed mice. Frontiers in Physiology, 2018, 9: 15. https://doi.org/10.3389/fphys.2018.00015

[49]

Yu, X. Z., Ye, L. F., Zhang, H., et al. Ginsenoside Rb1 ameliorates liver fat accumulation by upregulating perilipin expression in adipose tissue of db/db obese mice. Journal of Ginseng Research, 2015, 39: 199–205. https://doi.org/10.1016/j.jgr.2014.11.004

[50]

Liu, H. M., Liu, M. H., Jin, Z. B., et al. Ginsenoside Rg2 inhibits adipogenesis in 3T3-L1 preadipocytes and suppresses obesity in high-fat-diet-induced obese mice through the AMPK pathway. Food & Function, 2019, 10: 3603–3614. https://doi.org/10.1039/c9fo00027e

[51]

Hwang, Y. P., Choi, J. H., Kim, H. G., et al. Saponins, especially platycodin D, from Platycodon grandiflorum modulate hepatic lipogenesis in high-fat diet-fed rats and high glucose-exposed HepG2 cells. Toxicology and Applied Pharmacology, 2013, 267: 174–183. https://doi.org/10.1016/j.taap.2013.01.001

[52]

Kojima, K., Shimada, T., Nagareda, Y., et al. Preventive effect of geniposide on metabolic disease status in spontaneously obese type 2 diabetic mice and free fatty acid-treated HepG2 cells. Biological & Pharmaceutical Bulletin, 2011, 34: 1613–1618. https://doi.org/10.1248/bpb.34.1613

[53]

Zhong, H., Chen, K., Feng, M. Y., et al. Genipin alleviates high-fat diet-induced hyperlipidemia and hepatic lipid accumulation in mice via miR-142a-5p/SREBP-1c axis. The FEBS Journal, 2018, 285: 501–517. https://doi.org/10.1111/febs.14349

[54]

Jeong, Y. U., Park, Y. J. Ergosterol peroxide from the medicinal mushroom ganoderma lucidum inhibits differentiation and lipid accumulation of 3T3-L1 adipocytes. International Journal of Molecular Sciences, 2020, 21: 460. https://doi.org/10.3390/ijms21020460

[55]

Variya, B. C., Bakrania, A. K., Chen, Y. L., et al. Suppression of abdominal fat and anti-hyperlipidemic potential of Emblica officinalis: Upregulation of PPARs and identification of active moiety. Biomedicine & Pharmacotherapy, 2018, 108: 1274–1281. https://doi.org/10.1016/j.biopha.2018.09.158

[56]

Sun, J. H., Wang, Z. D., Chen, L., et al. Hypolipidemic effects and preliminary mechanism of chrysanthemum flavonoids, its main components luteolin and luteoloside in hyperlipidemia rats. Antioxidants, 2021, 10: 1309. https://doi.org/10.3390/antiox1008 1309

[57]

Guo, C. Y., Liao, W. T., Qiu, R. J., et al. Aurantio-obtusin improves obesity and insulin resistance induced by high-fat diet in obese mice. Phytotherapy Research, 2021, 35: 346–360. https://doi.org/10.1002/ptr.6805

[58]

Kang, O. H., Kim, S. B., Mun, S. H., et al. Puerarin ameliorates hepatic steatosis by activating the PPARα and AMPK signaling pathways in hepatocytes. International Journal of Molecular Medicine, 2015, 35: 803–809. https://doi.org/10.3892/ijmm.2015.2074

[59]

Kong, X., Liu, J. J., Li, H., Chen, Z. B. Effect of polysaccharides from Polygonatum sibiricum on lipid-metabolism related mRNA and protein expression in hyperlipidemic mice. China Journal of Chinese Materia Medica, 2018, 43: 3740–3747. https://doi.org/10.19540/j.cnki.cjcmm.20180502.001

[60]

Wang, K. P., Cao, P., Wang, H. X., et al. Chronic administration of Angelica sinensis polysaccharide effectively improves fatty liver and glucose homeostasis in high-fat diet-fed mice. Scientific Reports, 2016, 6: 26229. https://doi.org/10.1038/srep26229

[61]

Liou, C. J., Lee, Y. K., Ting, N. C., et al. Protective effects of licochalcone A ameliorates obesity and non-alcoholic fatty liver disease via promotion of the sirt-1/AMPK pathway in mice fed a high-fat diet. Cells, 2019, 8: 447. https://doi.org/10.3390/cells8050447

[62]

Tian, G., Li, J., Zhou, L. N. Ginsenoside Rg1 regulates autophagy and endoplasmic reticulum stress via the AMPK/mTOR and PERK/ATF4/CHOP pathways to alleviate alcohol-induced myocardial injury. International Journal of Molecular Medicine, 2023, 52: 56. https://doi.org/10.3892/ijmm.2023.5259

[63]

Yu, M. H., Hung, T. W., Wang, C. C., et al. Neochlorogenic acid attenuates hepatic lipid accumulation and inflammation via regulating miR-34a in vitro. International Journal of Molecular Sciences, 2021, 22: 13163. https://doi.org/10.3390/ijms222313163

[64]

Luo, Y., Lu, S., Ai, Q. D., et al. SIRT1/AMPK and Akt/eNOS signaling pathways are involved in endothelial protection of total aralosides of Aralia elata (Miq) Seem against high-fat diet-induced atherosclerosis in ApoE-/ - mice. Phytotherapy Research, 2019, 33: 768–778. https://doi.org/10.1002/ptr.6269

[65]

Zheng, Z. G., Zhou, Y. P., Zhang, X., et al. Anhydroicaritin improves diet-induced obesity and hyperlipidemia and alleviates insulin resistance by suppressing SREBPs activation. Biochemical Pharmacology, 2016, 122: 42–61. https://doi.org/10.1016/j.bcp.2016.10.016

[66]

Moslehi, A., Hamidi-Zad, Z. Role of SREBPs in liver diseases: A mini-review. Journal of Clinical and Translational Hepatology, 2018, 6: 332–338. https://doi.org/10.14218/JCTH.2017.00061

[67]

Lin, Y. K., Yeh, C. T., Kuo, K. T., et al. Pterostilbene increases LDL metabolism in HL-1 cardiomyocytes by modulating the PCSK9/HNF1α/SREBP2/LDLR signaling cascade, upregulating epigenetic hsa-miR-335 and hsa-miR-6825, and LDL receptor expression. Antioxidants, 2021, 10: 1280. https://doi.org/10.3390/antiox10081280

[68]

Feng, Y. C., Gao, S., Zhu, T., et al. Hawthorn fruit acid consumption attenuates hyperlipidemia-associated oxidative damage in rats. Frontiers in Nutrition, 2022, 9: 936229. https://doi.org/10.3389/fnut.2022.936229

[69]

Zhang, Y. Y., Zhang, L., Geng, Y., et al. Hawthorn fruit attenuates atherosclerosis by improving the hypolipidemic and antioxidant activities in apolipoprotein e-deficient mice. Journal of Atherosclerosis and Thrombosis, 2014, 21: 119–128. https://doi.org/10.5551/jat.19174

[70]

Chen, F., Zhou, Y., Yang, K. Y., et al. NPY stimulates cholesterol synthesis acutely by activating the SREBP2-HMGCR pathway through the Y1 and Y5 receptors in murine hepatocytes. Life Sciences, 2020, 262: 118478. https://doi.org/10.1016/j.lfs.2020.118478

[71]

Zou, J., Zhang, S. S., Li, P. Y., et al. Supplementation with curcumin inhibits intestinal cholesterol absorption and prevents atherosclerosis in high-fat diet-fed apolipoprotein E knockout mice. Nutrition Research, 2018, 56: 32–40. https://doi.org/10.1016/j.nutres.2018.04.017

[72]

Feng, D., Zou, J., Zhang, S. S., et al. Hypocholesterolemic activity of curcumin is mediated by down-regulating the expression of niemann-pick C1-like 1 in hamsters. Journal of Agricultural and Food Chemistry, 2017, 65: 276–280. https://doi.org/10.1021/acs.jafc.6b04102

[73]

Dou, X. B., Fan, C. L., Wo, L. K., et al. Curcumin up-regulates LDL receptor expression via the sterol regulatory element pathway in HepG2 cells. Planta Medica, 2008, 74: 1374–1379. https://doi.org/10.1055/s-2008-1081316

[74]

Cheng, Y. J., Tang, K., Wu, S. H., et al. Astragalus polysaccharides lowers plasma cholesterol through mechanisms distinct from statins. PLoS One, 2011, 6: e27437. https://doi.org/10.1371/journal.pone.0027437

[75]

Liu, J. X., Li, Y., Sun, C., et al. Geniposide reduces cholesterol accumulation and increases its excretion by regulating the FXR-mediated liver-gut crosstalk of bile acids. Pharmacological Research, 2020, 152: 104631. https://doi.org/10.1016/j.phrs.2020.104631

[76]

Rao, Y. F., Wen, Q., Liu, R. H., et al. PL-S2, a homogeneous polysaccharide from Radix Puerariae lobatae, attenuates hyperlipidemia via farnesoid X receptor (FXR) pathway-modulated bile acid metabolism. International Journal of Biological Macromolecules, 2020, 165: 1694–1705. https://doi.org/10.1016/j.ijbiomac.2020.10.029

[77]

Zhu, R. G., Sun, Y. D., Li, T. P., et al. Comparative effects of hawthorn ( Crataegus pinnatifida Bunge) pectin and pectin hydrolyzates on the cholesterol homeostasis of hamsters fed high-cholesterol diets. Chemico-Biological Interactions, 2015, 238: 42–47. https://doi.org/10.1016/j.cbi.2015.06.006

[78]

Cho, I. J., Lee, C., Ha, T. Y. Hypolipidemic effect of soluble fiber isolated from seeds of Cassia tora Linn. in rats fed a high-cholesterol diet. Journal of Agricultural and Food Chemistry, 2007, 55: 1592–1596. https://doi.org/10.1021/jf0622127

[79]

Ma, J. Q., Ding, J., Zhao, H., et al. Puerarin attenuates carbon tetrachloride-induced liver oxidative stress and hyperlipidaemia in mouse by JNK/c-Jun/CYP7A1 pathway. Basic & Clinical Pharmacology & Toxicology, 2014, 115: 389–395. https://doi.org/10.1111/bcpt.12245

[80]

Zhang, Z. G., Li, X. Y., Lv, W. S., et al. Ginsenoside Re reduces insulin resistance through inhibition of c-Jun NH2-terminal kinase and nuclear factor-kappaB. Molecular Endocrinology, 2008, 22: 186–195. https://doi.org/10.1210/me.2007-0119

[81]

Kwon, H. C., Kim, T. Y., Lee, C. M., et al. Active compound chrysophanol of Cassia tora seeds suppresses heat-induced lipogenesis via inactivation of JNK/p38 MAPK signaling in human sebocytes. Lipids in Health and Disease, 2019, 18: 135. https://doi.org/10.1186/s12944-019-1072-x

[82]

Li, P., Fan, W., Xu, J., et al. Adipocyte NCoR knockout decreases PPARγ phosphorylation and enhances PPARγ activity and insulin sensitivity. Cell, 2011, 147: 815–826. https://doi.org/10.1016/j.cell.2011.09.050

[83]

Lefterova, M. I., Haakonsson, A. K., Lazar, M. A., Mandrup, S. PPARγ and the global map of adipogenesis and beyond. Trends in Endocrinology and Metabolism, 2014, 25: 293–302. https://doi.org/10.1016/j.tem.2014.04.001

[84]

Kim, J., Lee, Y. J., Kim, J. M., et al. PPARγ agonists induce adipocyte differentiation by modulating the expression of Lipin-1, which acts as a PPARγ phosphatase. The International Journal of Biochemistry & Cell Biology, 2016, 81: 57–66. https://doi.org/10.1016/j.biocel.2016.10.018

[85]

Gu, W., Kim, K. A., Kim, D. H. Ginsenoside Rh1 ameliorates high fat diet-induced obesity in mice by inhibiting adipocyte differentiation. Biological & Pharmaceutical Bulletin, 2013, 36: 102–7. https://doi.org/10.1248/bpb.b12-00558

[86]

Bu, S., Zheng, H. H., Yuan, C. Y., et al. Lingzhi or reishi medicinal mushroom, ganoderma lucidum (agaricomycetes), polysaccharides suppressed adipogenesis and stimulated lipolysis in HPA-v and 3T3-L1 adipocytes. International Journal of Medicinal Mushrooms, 2020, 22: 897–908. https://doi.org/10.1615/IntJMedMushrooms.2020035861

[87]

Xu, X. C., Chen, W. J., Yu, S. K., et al. Inhibition of preadipocyte differentiation by Lycium barbarum polysaccharide treatment in 3T3-L1 cultures. Electronic Journal of Biotechnology, 2021, 50: 53–58. https://doi.org/10.1016/j.ejbt.2021.01.003

[88]

Yang, J. W., Kim, S. S. Ginsenoside Rc promotes anti-adipogenic activity on 3T3-L1 adipocytes by down-regulating C/EBPα and PPARγ. Molecules, 2015, 20: 1293–1303. https://doi.org/10.3390/molecules20011293

[89]

Xue, H.K., Hao, Z.T., Gao, Y.C., et al. Research progress on hypoglycemic effect and mechanism of saponins. Natural Product Research and Development, 2023, 251: 126199. https://doi.org/10.1016/j.ijbiomac.2023.126199

[90]

Variya, B. C., Bakrania, A. K., Patel, S. S. Antidiabetic potential of Gallic acid from Emblica officinalis: Improved glucose transporters and insulin sensitivity through PPAR-γ and Akt signaling. Phytomedicine, 2020, 73: 152906. https://doi.org/10.1016/j.phymed.2019.152906

[91]

Guru, B., Tamrakar, A. K., Mandal, S. P., et al. A novel partial PPARγ agonist has weaker lipogenic effect in adipocytes and stimulates GLUT4 translocation in skeletal muscle cells via AMPK-dependent signaling. Pharmacology, 2022, 107: 90–101. https://doi.org/10.1159/000519331

[92]

Mu, Q. Q., Zuo, J. C., Zhao, D. D., et al. Ginsenoside Rg3 reduces body weight by regulating fat content and browning in obese mice. Journal of Traditional Chinese Medical Sciences, 2021, 8: 65–71. https://doi.org/10.1016/j.jtcms.2021.01.009

[93]

Nishina, A., Sato, D., Yamamoto, J., et al. Antidiabetic-like effects of naringenin-7- O-glucoside from edible chrysanthemum ‘kotobuki’ and naringenin by activation of the PI3K/akt pathway and PPARγ. Chemistry & Biodiversity, 2019, 16: e1800434. https://doi.org/10.1002/cbdv.201800434

[94]

Li, J. S., Ji, T., Su, S. L., et al. Mulberry leaves ameliorate diabetes via regulating metabolic profiling and AGEs/RAGE and p38 MAPK/NF-κB pathway. Journal of Ethnopharmacology, 2022, 283: 114713. https://doi.org/10.1016/j.jep.2021.114713

[95]
Zhang, T. S., Sawada, K., Yamamoto, N., Ashida, H. 4-Hydroxyderricin and xanthoangelol from Ashitaba (Angelica keiskei) suppress differentiation of preadiopocytes to adipocytes via AMPK and MAPK pathways. Molecular Nutrition & Food Research, 2013 , 57: 1729–1740. https://doi.org/10.1002/mnfr.201300020
[96]

Zhang, W. Y., Ho, C. T., Lu, M. W. Piperine improves lipid dysregulation by modulating circadian genes Bmal1 and Clock in HepG2 cells. International Journal of Molecular Sciences, 2022, 23: 5611. https://doi.org/10.3390/ijms23105611

[97]

Ahmadian, M., Suh, J. M., Hah, N., et al. PPARγ signaling and metabolism: The good, the bad and the future. Nature Medicine, 2013, 19: 557–566. https://doi.org/10.1038/nm.3159

[98]

Xian, X., Ding, Y., Dieckmann, M., et al. LRP1 integrates murine macrophage cholesterol homeostasis and inflammatory responses in atherosclerosis. Elife, 2017, 6: e29292. https://doi.org/10.7554/eLife.29292

[99]
Luo, D., Dong, X. K., Huang, J., et al. Pueraria lobata root polysaccharide alleviates glucose and lipid metabolic dysfunction in diabetic db/db mice. Pharmaceutical Biology, 2021 , 59: 380–388. https://doi.org/10.1080/13880209.2021.1898648
[100]

Yang, Y., Li, W., Li, Y., et al. Dietary Lycium barbarum polysaccharide induces Nrf2/ARE pathway and ameliorates insulin resistance induced by high-fat via activation of PI3K/AKT signaling. Oxidative Medicine and Cellular Longevity, 2014, 2014: 145641. https://doi.org/10.1155/2014/145641

[101]

Zhang, Q., Liu, J., Duan, H., et al. Activation of Nrf2/HO-1 signaling: An important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. Journal of Advanced Research, 2021, 34: 43–63. https://doi.org/10.1016/j.jare.2021.06.023

[102]

Schoeler, M., Caesar, R. Dietary lipids, gut microbiota and lipid metabolism. Reviews in Endocrine and Metabolic Disorders, 2019, 20: 461–472. https://doi.org/10.1007/s11154-019-09512-0

[103]

Lei, L. R., Zhao, N., Zhang, L., et al. Gut microbiota is a potential goalkeeper of dyslipidemia. Frontiers in Endocrinology, 2022, 13: 950826. https://doi.org/10.3389/fendo.2022.950826

[104]

Avery, E. G., Bartolomaeus, H., Maifeld, A., et al. The gut microbiome in hypertension: Recent advances and future perspectives. Circulation Research, 2021, 128: 934–950. https://doi.org/10.1161/CIRCRESAHA.121.318065

[105]

Wang, L., Wu, Y. Z., Zhuang, L. J., et al. Puerarin prevents high-fat diet-induced obesity by enriching Akkermansia muciniphila in the gut microbiota of mice. PLoS One, 2019, 14: e0218490. https://doi.org/10.1371/journal.pone.0218490

[106]

Yang, X. Y., Dong, B. J., An, L. J., et al. Ginsenoside Rb1 ameliorates glycemic disorder in mice with high fat diet-induced obesity via regulating gut microbiota and amino acid metabolism. Frontiers in Pharmacology, 2021, 12: 756491. https://doi.org/10.3389/fphar.2021.756491

[107]
Zheng, J. P., Zhu, L., Hu, B. F., et al. 1-Deoxynojirimycin improves high fat diet-induced nonalcoholic steatohepatitis by restoring gut dysbiosis. The Journal of Nutritional Biochemistry, 2019 , 71: 16–26. https://doi.org/10.1016/j.jnutbio.2019.05.013
[108]

Peng, M., Wang, L. M., Su, H., et al. Ginsenoside Rg1 improved diabetes through regulating the intestinal microbiota in high-fat diet and streptozotocin-induced type 2 diabetes rats. Journal of Food Biochemistry, 2022, 46: e14321. https://doi.org/10.1111/jfbc.14321

[109]

Xie, B., Zu, X. P., Wang, Z. C., et al. Ginsenoside Rc ameliorated atherosclerosis via regulating gut microbiota and fecal metabolites. Frontiers in Pharmacology, 2022, 13: 990476. https://doi.org/10.3389/fphar.2022.990476

[110]

Wang, Y., Yao, W. F., Li, B., et al. Nuciferine modulates the gut microbiota and prevents obesity in high-fat diet-fed rats. Experimental & Molecular Medicine, 2020, 52: 1959–1975. https://doi.org/10.1038/s12276-020-00534-2

[111]

Gu, W., Wang, Y. F., Zeng, L. X., et al. Polysaccharides from Polygonatum kingianum improve glucose and lipid metabolism in rats fed a high fat diet. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie, 2020, 125: 109910. https://doi.org/10.1016/j.biopha.2020.109910

[112]

Zhu, L., Ye, C., Hu, B. F., et al. Regulation of gut microbiota and intestinal metabolites by Poria Cocos oligosaccharides improves glycolipid metabolism disturbance in high-fat diet-fed mice. The Journal of Nutritional Biochemistry, 2022, 107: 109019. https://doi.org/10.1016/j.jnutbio.2022.109019

[113]

Sun, S. S., Wang, K., Ma, K., et al. An insoluble polysaccharide from the sclerotium of Poria Cocos improves hyperglycemia, hyperlipidemia and hepatic steatosis in ob/ob mice via modulation of gut microbiota. Chinese Journal of Natural Medicines, 2019, 17: 3–14. https://doi.org/10.1016/S1875-5364(19)30003-2

[114]
Yang, Y., Li, M., Wang, Q., et al. Pueraria lobata starch regulates gut microbiota and alleviates high-fat high-cholesterol diet induced non-alcoholic fatty liver disease in mice. Food Research International , 2022 , 157: 111401. https://doi.org/10.1016/j.foodres.2022.111401
[115]

Lee, Y., Kwon, E. Y., Choi, M. S. Dietary isoliquiritigenin at a low dose ameliorates insulin resistance and NAFLD in diet-induced obesity in C57BL/6J mice. International Journal of Molecular Sciences, 2018, 19: 3281. https://doi.org/10.3390/ijms19103281

[116]

Xu, J., Liu, H. R., Su, G. Y., et al. Purification of ginseng rare sapogenins 25-OH-PPT and its hypoglycemic, antiinflammatory and lipid-lowering mechanisms. Journal of Ginseng Research, 2021, 45: 86–97. https://doi.org/10.1016/j.jgr.2019.11.002

[117]

Huang, Y. C., Chang, W. L., Huang, S. F., et al. Pachymic acid stimulates glucose uptake through enhanced GLUT4 expression and translocation. European Journal of Pharmacology, 2010, 648: 39–49. https://doi.org/10.1016/j.ejphar.2010.08.021

[118]

Cai, H., Cheng, Y. J., Zhu, Q. N., et al. Identification of triterpene acids in Poria cocos extract as bile acid uptake transporter inhibitors. Drug Metabolism and Disposition, 2021, 49: 353–360. https://doi.org/10.1124/dmd.120.000308

[119]

Zhang, W., Li, T., Zhang, X. J., et al. Hypoglycemic effect of glycyrrhizic acid, a natural non-carbohydrate sweetener, on streptozotocin-induced diabetic mice. Food & Function, 2020, 11: 4160–4170. https://doi.org/10.1039/c9fo02114k

[120]

Wang, T. J., Choi, R. C. Y., Li, J., et al. Trillin, a steroidal saponin isolated from the rhizomes of Dioscorea nipponica, exerts protective effects against hyperlipidemia and oxidative stress. Journal of Ethnopharmacology, 2012, 139: 214–220. https://doi.org/10.1016/j.jep.2011.11.001

[121]

Ma, W. L., Ding, H., Gong, X. H., et al. Methyl protodioscin increases ABCA1 expression and cholesterol efflux while inhibiting gene expressions for synthesis of cholesterol and triglycerides by suppressing SREBP transcription and microRNA 33a/b levels. Atherosclerosis, 2015, 239: 566–570. https://doi.org/10.1016/j.atherosclerosis.2015.02.034

[122]

Kusano, Y., Tsujihara, N., Masui, H., et al. Diosgenin supplementation prevents lipid accumulation and induces skeletal muscle-fiber hypertrophy in rats. Journal of Nutritional Science and Vitaminology, 2019, 65: 421–429. https://doi.org/10.3177/jnsv.65.421

[123]

Guo, W. L., Guo, J. B., Liu, B. Y., et al. Ganoderic acid A from Ganoderma lucidum ameliorates lipid metabolism and alters gut microbiota composition in hyperlipidemic mice fed a high-fat diet. Food & Function, 2020, 11: 6818–6833. https://doi.org/10.1039/d0fo00436g

[124]

Liu, F. L., Shi, K. J., Dong, J. J., et al. Ganoderic acid A attenuates high-fat-diet-induced liver injury in rats by regulating the lipid oxidation and liver inflammation. Archives of Pharmacal Research, 2020, 43: 744–754. https://doi.org/10.1007/s12272-020-01256-9

[125]

Li, C. J., Chen, Y. E., Yuan, X., et al. Vitexin ameliorates chronic stress plub high fat diet-induced nonalcoholic fatty liver disease by inhibiting inflammation. European Journal of Pharmacology, 2020, 882: 173264. https://doi.org/10.1016/j.ejphar.2020.173264

[126]

Enkhmaa, B., Shiwaku, K., Katsube, T., et al. Mulberry ( Morus alba L.) Leaves and Their Major Flavonol Quercetin 3-(6-Malonylglucoside) Attenuate Atherosclerotic Lesion Development in LDL Receptor-Deficient Mice. The Journal of Nutrition, 2005, 135: 729–734. https://doi.org/10.1093/jn/135.4.729

[127]
Mbikay, M., Mayne, J., Sirois, F., et al. Mice fed a high-cholesterol diet supplemented with quercetin-3-glucoside show attenuated hyperlipidemia and hyperinsulinemia associated with differential regulation of PCSK9 and LDLR in their liver and pancreas. Molecular Nutrition & Food Research, 2018 , 62 e1700729. https://doi.org/10.1002/mnfr.201700729
[128]

Luo, Y., Sun, G. B., Dong, X., et al. Isorhamnetin attenuates atherosclerosis by inhibiting macrophage apoptosis via PI3K/AKT activation and HO-1 induction. PLoS One, 2015, 10: e0120259. https://doi.org/10.1371/journal.pone.0120259

[129]

Farias-Pereira, R., Savarese, J., Yue, Y. R., et al. Fat-lowering effects of isorhamnetin are via NHR-49-dependent pathway in Caenorhabditis elegans. Current Research in Food Science, 2020, 2: 70–76. https://doi.org/10.1016/j.crfs.2019.11.002

[130]

Sun, C., Wang, L., Sun, J., et al. Hypoglycemic and hypolipidemic effects of rutin on hyperglycemic rats. Journal of Traditional Chinese Medicine, 2020, 40: 640–645. https://doi.org/10.19852/j.cnki.jtcm.2020.04.012

[131]

Cheng, L., Shi, L., He, C. H., et al. Rutin-activated adipose tissue thermogenesis is correlated with increased intestinal short-chain fatty acid levels. Phytotherapy Research, 2022, 36: 2495–2510. https://doi.org/10.1002/ptr.7462

[132]

Zhao, Y., Yang, X. B., Ren, D. Y., et al. Preventive effects of jujube polysaccharides on fructose-induced insulin resistance and dyslipidemia in mice. Food & Function, 2014, 5: 1771–1778. https://doi.org/10.1039/c3fo60707k

[133]

Kim, K. J., Lee, O. H., Han, C. K., et al. Acidic polysaccharide extracts from Gastrodia Rhizomes suppress the atherosclerosis risk index through inhibition of the serum cholesterol composition in Sprague Dawley rats fed a high-fat diet. International Journal of Molecular Sciences, 2012, 13: 1620–1631. https://doi.org/10.3390/ijms13021620

[134]

Li, R. L., Xue, Z. H., Jia, Y. N., et al. Polysaccharides from mulberry ( Morus alba L.) leaf prevents obesity by inhibiting pancreatic lipase in high-fat diet induced mice. International Journal of Biological Macromolecules, 2021, 192: 452–460. https://doi.org/10.1016/j.ijbiomac.2021.10.010

[135]

Qu, L. L., Yu, B., Li, Z., et al. Gastrodin ameliorates oxidative stress and proinflammatory response in nonalcoholic fatty liver disease through the AMPK/Nrf2 pathway. Phytotherapy Research, 2016, 30: 402–411. https://doi.org/10.1002/ptr.5541

[136]

Wan, J., Zhang, Y. Y., Yang, D. Q., et al. Gastrodin improves nonalcoholic fatty liver disease through activation of the adenosine monophosphate-activated protein kinase signaling pathway. Hepatology, 2021, 74: 3074–3090. https://doi.org/10.1002/hep.32068

[137]

Li, W. F., Zhi, W. B., Zhao, J. M., et al. Cinnamaldehyde attenuates atherosclerosis via targeting the IκB/NF-κB signaling pathway in high fat diet-induced ApoE-/- mice. Food & Function, 2019, 10: 4001–4009. https://doi.org/10.1039/c9fo00396g

Food & Medicine Homology
Article number: 9420003
Cite this article:
Hao X-T, Peng R, Guan M, et al. The food and medicinal homological resources benefiting patients with hyperlipidemia: categories, functional components, and mechanisms. Food & Medicine Homology, 2024, 1(2): 9420003. https://doi.org/10.26599/FMH.2024.9420003

1712

Views

595

Downloads

0

Crossref

Altmetrics

Received: 01 April 2024
Revised: 30 April 2024
Accepted: 02 May 2024
Published: 03 June 2024
© National R & D Center for Edible Fungus Processing Technology 2024. Published by Tsinghua University Press.

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

Return