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 (4.1 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

Hypoglycemic effects of edible fungus polysaccharides: A mini review

Yan-Long Cui1,2,3Bo Li3( )
Postdoctoral Innovation Practice Base, Henan Institute of Science and Technology, Xinxiang 453000, China
Postdoctoral Research Center, Henan Bainong Seed Industry Co., Ltd. Xinxiang 453000, China
College of Food Science, Henan Institute of Science and Technology, Xinxiang 453000, China
Show Author Information

Highlights

(1) We summarize the hypoglycemic mechanisms of edible fungus polysaccharides (EFP).

(2) The structure-hypoglycemic activity relationship of EFP was fully discussed.

(3) We point out the probable future research directions and trends of EFP involved hypoglycemic effects.

Graphical Abstract

Edible fungus are an important resource of food and medicine. Edible fungus Polysaccharides are one of the bioactive substances and own multiple biological activities, such as immunomodulatory, antioxidant, anti-inflammatory and hypoglycemic effect etc. The possible hypoglycemic mechanisms of edible fungus (EFP) were fully summarized.

Abstract

Diabetes mellitus (DM) is a chronic metabolic disorder with the feature of hyperglycemia, which has severely endangered human health. Edible fungus polysaccharides (EFP), which has great development potential, have attracted enormous attention since their natural and safe characteristics as well as obvious benefits against diabetes. This review summarized the hypoglycemic effect of EFP and the underlying mechanism against diabetes by inhibiting digestive enzyme activity, increasing insulin sensitivity and resistance, improving pancreatic function and lipid metabolism, alleviating oxidative stress and inflammation, regulating gut microbiota and signal transduction. In addition, the relationships between structure and hypoglycemic activity as well as future prospect of EFP were also discussed. This current review provides valuable insights for readers and healthcare professionals developing preventive and therapeutic of diabetes and development of functional products or foods of edible fungi in the future.

References

[1]

Pruhova, S., Dusatkova, P. Monogenic diabetes mellitus hidden in autoantibody-negative diabetes mellitus. Nature Reviews Endocrinology, 2023, 19: 132–133. https://doi.org/10.1038/s41574-022-00800-5

[2]

Ogurtsova, K., Guariguata, L., Barengo, N. C. W., et al. IDF diabetes atlas: global estimates of undiagnosed diabetes in adults. Diabetes Research and Clinical Practice, 2022, 183: 109118. https://doi.org/10.1016/j.diabres.2021.109118

[3]

Jones, V. C., Dietze, E. C., Jovanovic Talisman, T., et al. Metformin and chemoprevention: Potential for heart-healthy targeting of biologically aggressive breast cancer. Frontiers in Public Health, 2020, 8: 509714. https://doi.org/10.3389/fpubh.2020.509714

[4]

Dhameja, M., Gupta, P. Synthetic heterocyclic candidates as promising α-glucosidase inhibitors: An overview. European Journal of Medicinal Chemistry, 2019, 176: 343–377. https://doi.org/10.1016/ j.ejmech.2019.04.025

[5]

Arnold, S. V., Inzucchi, S. E., Echouffo-Tcheugui, J. B., et al. Understanding contemporary use of thiazolidinediones an analysis from the diabetes collaborative registry. Circulation-Heart Failure, 2019, 12: e005855. https://doi.org/10.1161/circheartfailure.118.005855

[6]

Liaw, J., Harhay, M., Setoguchi, S., et al. Trends in prescribing preferences for antidiabetic medications among patients with type 2 diabetes in the UK with and without chronic kidney disease, 2006-2020. Diabetes Care, 2022, 45: 2316–2325. https://doi.org/10.2337/dc22-0224

[7]

Zhu, J., Liu, W., Yu, J., et al. Characterization and hypoglycemic effect of a polysaccharide extracted from the fruit of Lycium barbarum L. Carbohydrate Polymers, 2013, 98: 8–16. https://doi.org/ 10.1016/j.carbpol.2013.04.057

[8]

Ma, W., Xiao, L., Liu, H., et al. Hypoglycemic natural products with in vivo activities and their mechanisms: A review. Food Science and Human Wellness, 2022, 11: 1087–1100. https://doi.org/10.1016/j.fshw.2022.04.001

[9]

Xue, H., Hao, Z., Gao, Y., et al. Research progress on the hypoglycemic activity and mechanisms of natural polysaccharides. International Journal of Biological Macromolecules, 2023, 252: 126199. https://doi.org/10.1016/j.ijbiomac.2023.126199

[10]

Muszynska, B., Grzywacz-Kisielewska, A., Kala, K., et al. Anti-inflammatory properties of edible mushrooms: A review. Food Chemistry, 2018, 243: 373–381. https://doi.org/10.1016/j.foodchem.2017.09.149

[11]

Leong, Y. K., Yang, F. C. and Chang, J. S. Extraction of polysaccharides from edible mushrooms: emerging technologies and recent advances. Carbohydrate Polymers, 2021, 251: 117006. https://doi.org/10.1016/j.carbpol.2020.117006

[12]

Zhang, Y., Wang, D., Chen, Y., et al. Healthy function and high valued utilization of edible fungi. Food Science and Human Wellness, 2021, 10: 408–420. https://doi.org/10.1016/j.fshw.2021.04.003

[13]

Kang, J. Y., Lee, B., Kim, C. H., et al. Enhancing the prebiotic and antioxidant effects of exopolysaccharides derived from Cordyceps militaris by enzyme-digestion. Lwt-Food Science and Technology, 2022, 167: 113830. https://doi.org/10.1016/j.lwt.2022.113830

[14]

Zawadzka, A., Janczewska, A., Kobus-Cisowska, J., et al. The effect of light conditions on the content of selected active ingredients in anatomical parts of the oyster mushroom ( Pleurotus ostreatus L.). Plos One, 2022, 17: 0262279. https://doi.org/10.1371/journal.pone.0262279

[15]

Chen, H. Y., Lei, J. Y., Li, S. L., et al. Progress in biological activities and biosynthesis of edible fungi terpenoids. Critical Reviews In Food Science And Nutrition, 2023, 63: 7288–7310. https://doi.org/10.1080/10408398.2022.2045559

[16]

Magdziak, Z., Siwulski, M. and Mleczek, M. Characteristics of organic acid profiles in 16 species of wild growing edible mushrooms. Journal of Environmental Science and Health Part B-Pesticides Food Contaminants and Agricultural Wastes, 2017, 52: 784–789. https://doi.org/10.1080/03601234.2017.1356676

[17]

Yin, Z. H., Liang, Z. H., Li, C. Q., et al. Immunomodulatory effects of polysaccharides from edible fungus: A review. Food Science and Human Wellness, 2021, 10: 393–400. https://doi.org/10.1016/j.fshw.2021.04.001

[18]

Guo, Q., Liang, S., Ge, C., et al. Research progress on extraction technology and biological activity of polysaccharides from edible Fungi: A review. Food Reviews International, 2023, 39: 4909–4940. https://doi.org/10.1080/87559129.2022.2039182

[19]

Hwang, H. S., Yun, J. W. Hypoglycemic effect of polysaccharides produced by submerged mycelial culture of Laetiporus sulphureus on streptozotocin-induced diabetic rats. Biotechnology and Bioprocess Engineering, 2010, 15: 173–181. https://doi.org/10.1007/s12257-009-0160-6

[20]

Li, Y. M., Zhong, R. F., Chen, J., et al. Structural characterization, anticancer, hypoglycemia and immune activities of polysaccharides from Russula virescens. International Journal of Biological Macromolecules, 2021, 184: 380–392. https://doi.org/10.1016/j.ijbiomac.2021.06.026

[21]

Yang, S., Qu, Y., Zhang, H., et al. Hypoglycemic effects of polysaccharides from Gomphidiaceae rutilus fruiting bodies and their mechanisms. Food & Function, 2020, 11: 424–434. https://doi.org/10.1039/c9fo02283j

[22]

Feng, X. B., Wang, P., Lu, Y. X., et al. A novel polysaccharide from Heimioporus retisporus displays hypoglycemic Activity in a diabetic mouse model. Frontiers in Nutrition, 2022, 9: 964948. https://doi.org/10.3389/fnut.2022.964948

[23]

Kou, L., Du, M., Liu, P., et al. Anti-diabetic and anti-nephritic activities of Grifola frondosa mycelium polysaccharides in diet-streptozotocin-induced diabetic rats via modulation on oxidative stress. Applied Biochemistry and Biotechnology, 2018, 184: 310–322. https://doi.org/10.1007/s12010-018-2803-6

[24]

Chen, Y., Liu, D., Wang, D., et al. Hypoglycemic activity and gut microbiota regulation of a novel polysaccharide from Grifola frondosa in type 2 diabetic mice. Food and Chemical Toxicology, 2019, 126: 295–302. https://doi.org/10.1016/j.fct.2019.02.034

[25]

Liu, N., Chen, X., Song, J., et al. Hypoglycemic effects of Auricularia auricula polysaccharides on high fat diet and streptozotocin-induced diabetic mice using metabolomics analysis. Food & Function, 2021, 12: 9994–10007. https://doi.org/10.1039/d1fo02022f

[26]

Cao, H., Ma, S., Guo, H., et al. Comparative study on the monosaccharide compositions, antioxidant and hypoglycemic activities in vitro of intracellular and extracellular polysaccharides of liquid fermented Coprinus comatus. International Journal of Biological Macromolecules, 2019, 139: 543–549. https://doi.org/10.1016/j.ijbiomac.2019.08.017

[27]

Song, Q., Teng, A. G., Zhu, Z. Chemical structure and inhibition on α-glucosidase of a novel polysaccharide from Hypsizygus marmoreus. Journal of Molecular Structure, 2020, 1211: 128110. https://doi.org/10.1016/j.molstruc.2020.128110

[28]

Gong, P., Long, H., Guo, Y., et al. Isolation, structural characterization, and hypoglycemic activities in vitro of polysaccharides from Pleurotus eryngii. Molecules, 2022, 27: 7140. https://doi.org/10.3390/molecules27207140

[29]

Yin, C., Li, C., Ma, K., et al. The physicochemical, antioxidant, hypoglycemic and prebiotic properties of γ-irradiated polysaccharides extracted from Lentinula edodes. Food Science and Biotechnology, 2023, 32: 987–996. https://doi.org/10.1007/s10068-022-01234-5

[30]

Sun, H., Yu, X., Li, T., et al. Structure and hypoglycemic activity of a novel exopolysaccharide of Cordyceps militaris. International Journal of Biological Macromolecules, 2020, 166: 496–508. https://doi.org/10.1016/j.ijbiomac.2020.10.207

[31]

Tian, R., Zhang, Y. Z., Cheng, X., et al. Structural characterization, and in vitro hypoglycemic activity of a polysaccharide from the mushroom Cantharellus yunnanensis. International Journal of Biological Macromolecules, 2023, 235: 127200. https://doi.org/10.1016/j.ijbiomac.2023.127200

[32]

Yang, H. R., Chen, L. H., Zeng, Y. J. Structure, antioxidant activity and in vitro hypoglycemic activity of a polysaccharide purified from Tricholoma matsutake. Foods, 2021, 10: 2184. https://doi.org/10.3390/foods10092184

[33]

Yang, W., Jiang, W., Liao, W., et al. An estrogen receptor α-derived peptide improves glucose homeostasis during obesity. Nature Communications, 2024, 15: 3410. https://doi.org/10.1038/s41467-024-47687-6

[34]

Yang, S., Yan, J., Yang, L., et al. Alkali-soluble polysaccharides from mushroom fruiting bodies improve insulin resistance. International Journal of Biological Macromolecules, 2018, 126: 466–474. https://doi.org/10.1016/j.ijbiomac.2018.12.251

[35]
Xiao, C., Jiao, C., Xie, Y., et al. Grifola frondosa GF5000 improves insulin resistance by modulation the composition of gut microbiota in diabetic rats. Journal of Functional Foods, 2021 , 77: 104313. https://doi.org/10.1016/j.jff.2020.104313
[36]

Jiao, J., Yong, T., Huang, L., et al. A Ganoderma lucidum polysaccharide F31 alleviates hyperglycemia through kidney protection and adipocyte apoptosis. International Journal of Biological Macromolecules, 2022, 226: 1178–1191. https://doi.org/10.1016/j.ijbiomac.2022.11.231

[37]

Ma, X., Zhou, F., Chen, Y., et al. A polysaccharide from Grifola frondosa relieves insulin resistance of HepG2 cell by Akt-GSK-3 pathway. Glycoconjugate Journal, 2014, 31: 355–363. https://doi.org/ 10.1007/s10719-014-9526-x

[38]

Guo, W. L., Deng, J. C., Pan, Y. Y., et al. Hypoglycemic and hypolipidemic activities of Grifola frondosa polysaccharides and their relationships with the modulation of intestinal microflora in diabetic mice induced by high-fat diet and streptozotocin. International Journal of Biological Macromolecules, 2019, 153: 1231–1240. https://doi.org/10.1016/j.ijbiomac.2019.10.253

[39]

Xiao, C., Wu, Q. P., Cai, W., et al. Hypoglycemic effects of Ganoderma lucidum polysaccharides in type 2 diabetic mice. Archives of Pharmacal Research, 2012, 35: 1793–1801. https://doi.org/10.1007/s12272-012-1012-z

[40]

Wang, J., Hu, W., Li, L., et al. Antidiabetic activities of polysaccharides separated from Inonotus obliquus via the modulation of oxidative stress in mice with streptozotocin-induced diabetes. Plos One, 2017, 12: 0180476. https://doi.org/10.1371/journal.pone.0180476

[41]

Xiang, H., Sun-Waterhouse, D. , Cui, C. Hypoglycemic polysaccharides from Auricularia auricula and Auricularia polytricha inhibit oxidative stress, NF-κB signaling and proinflammatory cytokine production in streptozotocin-induced diabetic mice. Food Science and Human Wellness, 2021, 10: 87–93. https://doi.org/10.1016/j.fshw.2020.06.001

[42]

Liu, Y., Chen, D., You, Y., et al. Structural characterization and antidiabetic activity of a glucopyranose-rich heteropolysaccharide from Catathelasma ventricosum. Carbohydrate Polymers, 2016, 149: 399–407. https://doi.org/10.1016/j.carbpol.2016.04.106

[43]

Jiang, T., Wang, L., Ma, A., et al. The hypoglycemic and renal protective effects of Grifola frondosa polysaccharides in early diabetic nephropathy. Journal of Food Biochemistry, 2020, 44: 13515. https://doi.org/10.1111/jfbc.13515

[44]

Zhao, H., Lai, Q., Zhang, J., et al. Antioxidant and hypoglycemic effects of acidic-extractable polysaccharides from Cordyceps militaris on type 2 diabetes mice. Oxidative Medicine and Cellular Longevity, 2018, 2018: 9150807. https://doi.org/10.1155/2018/9150807

[45]

Sun, W., Zhang, Y. and Jia, L. Polysaccharides from Agrocybe cylindracea residue alleviate type 2-diabetes-induced liver and colon injuries by p38 MAPK signaling pathway. Food Bioscience, 2022, 47: 101690. https://doi.org/10.1016/j.fbio.2022.101690

[46]
Ashcroft, F. M., Rohm, M., Clark, A., et al. Is type 2 diabetes a glycogen storage disease of pancreatic β cells? Cell Metabolism, 2017 , 26: 17-23. https://doi.org/10.1016/j.cmet.2017.05.014
[47]
Zheng, J., Yang, B., Yu, Y., et al. Ganoderma lucidum polysaccharides exert anti-hyperglycemic effect on streptozotocin-induced diabetic rats through affecting β-cells. Combinatorial Chemistry & High Throughput Screening, 2012 , 15: 542–550. https://doi.org/10.2174/138620712801619168
[48]
Zhao, H., Li, M., Liu, L., et al. Cordyceps militaris polysaccharide alleviates diabetic symptoms by regulating gut microbiota against TLR4/NF-κB pathway. International Journal of Biological Macromolecules, 2023 , 230: 123241. https://doi.org/10.1016/j.ijbiomac.2023.123241
[49]

Anders, H.-J., Huber, T. B., Isermann, B., et al. CKD in diabetes: diabetic kidney disease versus nondiabetic kidney disease. Nature Reviews Nephrology, 2018, 14: 361–377. https://doi.org/10.1038/s41581-018-0001-y

[50]
Shang, X. L., Pan, L. C., Tang, Y., et al. 1H NMR-based metabonomics of the hypoglycemic effect of polysaccharides from Cordyceps militaris on streptozotocin-induced diabetes in mice. Natural Product Research, 2018 , 34: 1366–1372. https://doi.org/10.1080/14786419.2018.1516216
[51]

Xiao, C., Wu, Q., Xie, Y., et al. Hypoglycemic mechanisms of Ganoderma lucidum polysaccharides F31 in db/db mice via RNA-seq and iTRAQ. Food & Function, 2018, 9: 6496–6508. https://doi.org/10.1039/c8fo01656a

[52]

Capece, D., Verzella, D., Flati, I., et al. NF-κB: blending metabolism, immunity, and inflammation. Trends in Immunology, 2022, 43: 757–775. https://doi.org/10.1016/j.it.2022.07.004

[53]

Yang, C., Feng, Q., Liao, H., et al. Anti-diabetic nephropathy activities of polysaccharides obtained from Termitornyces albuminosus via regulation of NF-κB signaling in db/db Mice. International Journal of Molecular Sciences, 2019, 20: 5205. https://doi.org/10.3390/ijms20205205

[54]

Zhang, Y., Lu, J., Li, H., et al. Advances in dietary polysaccharides as hypoglycemic agents: Mechanisms, structural characteristics, and innovative applications. Critical Reviews In Food Science And Nutrition, 2023, 2293254: 1–21. https://doi.org/10.1080/10408398.2023.2293254

[55]

Cao, X. Y., Liu, D., Bi, R. C., et al. The protective effects of a novel polysaccharide from Lentinus edodes mycelia on islet β (INS-1) cells damaged by glucose and its transportation mechanism with human serum albumin. International Journal of Biological Macromolecules, 2019, 134: 344–353. https://doi.org/10.1016/j.ijbiomac.2019.05.033

[56]

Xiao, C., Wu, Q. P., Zhang, J. M., et al. Antidiabetic activity of Ganoderma lucidum polysaccharides F31 down-regulated hepatic glucose regulatory enzymes in diabetic mice. Journal of Ethnopharmacology, 2017, 196: 47–57. https://doi.org/10.1016/j.jep.2016.11.044

[57]

Niu, B., Feng, S., Xuan, S., et al. Moisture and caking resistant Tremella fuciformis polysaccharides microcapsules with hypoglycemic activity. Food Research International, 2021, 146: 110420. https://doi.org/10.1016/j.foodres.2021.110420

[58]

Liu, Y., Li, Y., Zhang, W., et al. Hypoglycemic effect of inulin combined with Ganoderma lucidum polysaccharides in T2DM rats. Journal of Functional Foods, 2019, 55: 381–390. https://doi.org/10.1016/j.jff.2019.02.036

[59]

Chen, Y., Liu, Y., Sarker, M. M. R., et al. Structural characterization and antidiabetic potential of a novel heteropolysaccharide from Grifola frondosa via IRS1/PI3K-JNK signaling pathways. Carbohydrate Polymers, 2018, 198: 452–461. https://doi.org/10.1016/j.carbpol.2018.06.077

[60]

Xia, F., Cao S. L., Wang, M. W., et al. Optimizing extraction, structural characterization, and in vitro hypoglycemic activity of a novel polysaccharide component from Lentinus edodes. Food Bioscience, 2023, 55: 103007 . https://doi.org/10.1016/j.fbio.2023.103007

[61]

Cai, W. D., Ding, Z. C., Wang, Y. Y., et al. Hypoglycemic benefit and potential mechanism of a polysaccharide from Hericium erinaceus in streptozotoxin-induced diabetic rats. Process Biochemistry, 2020, 88: 180–188. https://doi.org/10.1016/j.procbio.2019.09.035

[62]

Zuo, C., Cao, H., Song, Y., et al. Nrf2: An all-rounder in depression. Redox Biology, 2022, 58: 102522. https://doi.org/10.1016/j.redox.2022.102522

[63]

Rotimi, D. E., Ojo, O. A., Olaolu, T. D., et al. Exploring Nrf2 as a therapeutic target in testicular dysfunction. Cell and Tissue Research, 2022, 390: 23–33. https://doi.org/10.1007/s00441-022-03664-3

[64]

Gong, P., Wang, X., Liu, M., et al. Hypoglycemic effect of a novel polysaccharide from Lentinus edodes on STZ-induced diabetic mice via metabolomics study and Nrf2/HO-1 pathway. Food & Function, 2022, 13: 3036–3049. https://doi.org/10.1039/d1fo03487a

[65]

Hu, W., Wang, J., Guo, W., et al. Studies on characteristics and anti-diabetic and -nephritic effects of polysaccharides isolated from Paecilomyces hepiali fermentation mycelium in db/db mice. Carbohydrate Polymers, 2020, 232: 115766. https://doi.org/10.1016/j.carbpol.2019.115766

[66]

Defour, M., van Weeghel, M., Hermans, J., et al. Hepatic ADTRP overexpression does not influence lipid and glucose metabolism. American Journal of Physiology-Cell Physiology, 2021, 321: C585–C595. https://doi.org/10.1152/ajpcell.00185.2021

[67]

Gross, B., Pawlak, M., Lefebvre, P., et al. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nature Reviews Endocrinology, 2017, 13: 36–49. https://doi.org/10.1038/nrendo.2016.135

[68]

Huang, H. Y., Korivi, M., Yang, H. T., et al. Effect of Pleurotus tuber-regium polysaccharides supplementation on the progression of diabetes complications in obese-diabetic rats. The Chinese Journal of Physiology, 2014, 57: 198–208. https://doi.org/10.4077/cjp.2014.bac245

[69]

Hu, T., Lin, Q., Guo, T., et al. Polysaccharide isolated from Phellinus linteus mycelia exerts anti-inflammatory effects via MAPK and PPAR signaling pathways. Carbohydrate Polymers, 2018, 200: 487–497. https://doi.org/10.1016/j.carbpol.2018.08.021

[70]

Cho, E. J., Hwang, H. J., Kim, S. W., et al. Hypoglycemic effects of exopolysaccharides produced by mycelial cultures of two different mushrooms Tremella fuciformis and Phellinus baumii in ob/ob mice. Applied Microbiology and Biotechnology, 2007, 75: 1257–65. https://doi.org/10.1007/s00253-007-0972-2

[71]

Dong, Y. H., Wang, Z. X., Chen, C., et al. A review on the hypoglycemic effect, mechanism and application development of natural dietary polysaccharides. International Journal of Biological Macromolecules, 2023, 253: 127267. https://doi.org/10.1016/j.ijbiomac.2023.127267

[72]
Liu, Y., Wang, C., Li, J., et al. Phellinus linteus polysaccharide extract improves insulin resistance by regulating gut microbiota composition. The FASEB Journal, 2019 , 34: 1065–1078. https://doi.org/10.1096/fj.201901943rr
[73]
He, G., Chen, T., Huang, L., et al. Tremella fuciformis polysaccharide reduces obesity in high-fat diet-fed mice by modulation of gut microbiota. Frontiers in Microbiology, 2022 , 13: 1073350. https://doi.org/10.3389/fmicb.2022.1073350
[74]

Shao, W., Xiao, C., Yong, T., et al. A polysaccharide isolated from Ganoderma lucidum ameliorates hyperglycemia through modulating gut microbiota in type 2 diabetic mice. International Journal of Biological Macromolecules, 2021, 197: 23–38. https://doi.org/10.1016/j.ijbiomac.2021.12.034

[75]

Zhan, K., Ji, X., Luo, L. Recent progress in research on Momordica charantia polysaccharides: extraction, purification, structural characteristics and bioactivities. Chemical and Biological Technologies in Agriculture, 2023, 10: 58. https://doi.org/10.1186/s40538-023-00433-4

[76]

Yang, W., Wu, J., Liu, W., et al. Structural characterization, antioxidant and hypolipidemic activity of Grifola frondosa polysaccharides in novel submerged cultivation. Food Bioscience, 2021, 42: 101187. https://doi.org/10.1016/j.fbio.2021.101187

[77]

Gong, L. L., Meng, F. J., Hou, Y. C., et al. Purification, characterization, and bioactivity of two new polysaccharide fractions from Thelephora ganbajun mushroom. Journal of Food Biochemistry, 2020, 44: 13092. https://doi.org/10.1111/jfbc.13092

[78]

Chen, J., Zhang, X., Huo, D., et al. Preliminary characterization, antioxidant and α-glucosidase inhibitory activities of polysaccharides from Mallotus furetianus. Carbohydrate Polymers, 2019, 215: 307–315. https://doi.org/10.1016/j.carbpol.2019.03.099

[79]

Dubey, S. K., Chaturvedi, V. K., Mishra, D., et al. Role of edible mushroom as a potent therapeutics for the diabetes and obesity. 3 Biotech, 2019, 9: 450. https://doi.org/10.1007/s13205-019-1982-3

[80]

Hao, Y., Sun, H., Zhang, X., et al. A novel polysaccharide from Pleurotus citrinopileatus mycelia: Structural characterization, hypoglycemic activity and mechanism. Food Bioscience, 2020, 37: 100735. https://doi.org/10.1016/j.fbio.2020.100735

[81]

Ru, Y., Chen, X., Wang, J., et al. Structural characterization, hypoglycemic effects and mechanism of a novel polysaccharide from Tetrastigma hemsleyanum Diels et Gilg. International Journal of Biological Macromolecules, 2019, 123: 775–783. https://doi.org/10.1016/j.ijbiomac.2018.11.085

[82]

Liu, Q., Wu, J., Wang, P., et al. Neutral polysaccharides from Hohenbuehelia serotina with hypoglycemic effects in a type 2 diabetic mouse model. Frontiers in Pharmacology, 2022, 13: 883653. https://doi.org/10.3389/fphar.2022.883653

[83]

Cao, C., Huang, Q., Zhang, B., et al. Physicochemical characterization and in vitro hypoglycemic activities of polysaccharides from Sargassum pallidum by microwave-assisted aqueous two-phase extraction. International Journal of Biological Macromolecules, 2018, 109: 357–368. https://doi.org/10.1016/j.ijbiomac.2017.12.096

[84]

Feng, S., Luan, D., Ning, K., et al. Ultrafiltration isolation, hypoglycemic activity analysis and structural characterization of polysaccharides from Brasenia schreberi. International Journal of Biological Macromolecules, 2019, 135: 141–151. https://doi.org/10.1016/j.ijbiomac.2019.05.129

[85]

Chen, Z., Yin, C., Fan, X., et al. Characterization of physicochemical and biological properties of Schizophyllum commune polysaccharide extracted with different methods. International Journal of Biological Macromolecules, 2019, 156: 1425–1434. https://doi.org/10.1016/j.ijbiomac.2019.11.183

[86]

Li, Y., He, Y., Zhang, H., et al. Effects of ultrasonic-enzymatic-assisted ethanol precipitation method on the physicochemical characteristics, antioxidant and hypoglycemic activities of Tremella fuciformis polysaccharides. Ultrasonics Sonochemistry, 2023, 101: 106682. https://doi.org/10.1016/j.ultsonch.2023.106682

Food & Medicine Homology
Article number: 9420046
Cite this article:
Cui Y-L, Li B. Hypoglycemic effects of edible fungus polysaccharides: A mini review. Food & Medicine Homology, 2025, 2(1): 9420046. https://doi.org/10.26599/FMH.2025.9420046

297

Views

104

Downloads

0

Crossref

Altmetrics

Received: 28 June 2024
Revised: 10 July 2024
Accepted: 10 July 2024
Published: 28 September 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