Review

Cassia auriculata L.–A mini review of phytochemical compounds and their antidiabetic mechanisms

  • Received: 31 December 2023 Revised: 21 February 2024 Accepted: 27 February 2024 Published: 20 March 2024
  • Cassia auriculata is an important medicinal herb traditionally used for the treatment and management of diabetes. Scientific research has reported some bioactivities related to traditional roles that include antihyperglycemic and antihyperlipidemic, which could inhibit onset of diabetes. Our aim was twofold: To review the presence of phytochemical compounds in plant extracts and to perform an in-papyro evaluation of their antidiabetic potential. A detailed literature survey was carried out for evaluating metabolic syndrome-related medicinal bioactivities and antidiabetic activity from specific compounds of C. auriculata. We uncovered a wide range of medicinal uses of C. auriculata in Ayurveda and Sri Lankan medicinal traditions and cultures. Many of the compounds in C. auriculata extracts have already been reported for their specific antidiabetic, hypoglycemic, and hypolipidemic activities, which exhibited positive effects on neuro, renal, and liver support. In conclusion, our findings suggested that the phytocomposition of C. auriculata could be attributed to the presence of antidiabetic activity through various mechanisms.

    Citation: Zipora Tietel, Devanesan Arul Ananth, Thilagar Sivasudha, Liron Klipcan. Cassia auriculata L.–A mini review of phytochemical compounds and their antidiabetic mechanisms[J]. AIMS Agriculture and Food, 2024, 9(1): 374-392. doi: 10.3934/agrfood.2024022

    Related Papers:

  • Cassia auriculata is an important medicinal herb traditionally used for the treatment and management of diabetes. Scientific research has reported some bioactivities related to traditional roles that include antihyperglycemic and antihyperlipidemic, which could inhibit onset of diabetes. Our aim was twofold: To review the presence of phytochemical compounds in plant extracts and to perform an in-papyro evaluation of their antidiabetic potential. A detailed literature survey was carried out for evaluating metabolic syndrome-related medicinal bioactivities and antidiabetic activity from specific compounds of C. auriculata. We uncovered a wide range of medicinal uses of C. auriculata in Ayurveda and Sri Lankan medicinal traditions and cultures. Many of the compounds in C. auriculata extracts have already been reported for their specific antidiabetic, hypoglycemic, and hypolipidemic activities, which exhibited positive effects on neuro, renal, and liver support. In conclusion, our findings suggested that the phytocomposition of C. auriculata could be attributed to the presence of antidiabetic activity through various mechanisms.



    加载中


    [1] Ananth DA, Mahalakshmi V, Sivasudha T, et al. (2021) Identification and quantification of polyphenols from Cassia auriculata L. leaf, flower and flower bud using UPLC-QqQ-MS/MS. Isr J Plant Sci 68: 133–141. https://doi.org/10.1163/22238980-bja10027 doi: 10.1163/22238980-bja10027
    [2] Tietel Z, Ananth DA, Sivasudha T, et al. (2021) Metabolomics of Cassia auriculata plant parts (leaf, flower, bud) and their antidiabetic medicinal potentials. OMICS: J Integr Biol 25: 294–301. https://doi.org/10.1089/omi.2021.0010 doi: 10.1089/omi.2021.0010
    [3] Anitha R, Subashini R, Kumar PS (2020) In silico and in vitro approaches to evaluate the bioactivity of Cassia auriculata L extracts. IET Nanobiotechnol 14: 210–216. https://doi.org/10.1049/iet-nbt.2019.0364 doi: 10.1049/iet-nbt.2019.0364
    [4] Girme A, Saste G, Ghule C, et al. (2019) Phytoanalytical profiling of Cassia auriculata by LC-PDA-ESI-MS/MS and HPTLC supporting its metabolic claims. Planta Med 85: 1439. https://doi.org/10.1055/s-0039-3399767 doi: 10.1055/s-0039-3399767
    [5] Aye MM, Aung HT, Sein MM, et al. (2019) A review on the phytochemistry, medicinal properties and pharmacological activities of 15 selected Myanmar medicinal plants. Molecules 24: 293. https://doi.org/10.3390/molecules24020293 doi: 10.3390/molecules24020293
    [6] Anandan A, Eswaran R, Doss A, et al. (2011) Chemical compounds investigation of Cassia auriculata leaves–a potential folklore medicinal plant. Bull Environ, Pharm Life Sci 1: 20–23.
    [7] Meena V, Baruah H, Parveen R (2019) Cassia auriculata: A healing herb for all remedy. J Pharm Phytochem 8: 4093–4097.
    [8] Nille GC, Reddy KR (2015) A phytopharmacological review of plant–Cassia auriculata. Int J Pharm Biol Arch 6: 1–9.
    [9] Khurm M, Wang X, Zhang H, et al. (2020) The genus Cassia L.: Ethnopharmacological and phytochemical overview. Phytother Res 35: 2336–2385. https://doi.org/10.1002/ptr.6954 doi: 10.1002/ptr.6954
    [10] Shafeeq RS, Shekshavali T, Ahamed SSS (2018) A Review on Cassia auriculata. Res J Pharmacol Pharmacodyn 10: 141–145. https://doi.org/10.5958/2321-5836.2018.00026.5 doi: 10.5958/2321-5836.2018.00026.5
    [11] Wasana KGP, Attanayake AP, Arawwawala LDAM (2022) Ethnobotanical survey on medicinal plants used for the treatment of diabetes mellitus by Ayurveda and traditional medicine practitioners in Galle district of Sri Lanka. Eur J Integr Med 55: 102177. https://doi.org/10.1016/j.eujim.2022.102177 doi: 10.1016/j.eujim.2022.102177
    [12] Latha M, Pari L (2003) Antihyperglycaemic effect of Cassia auriculata in experimental diabetes and its effects on key metabolic enzymes involved in carbohydrate metabolism. Clin Exp Pharmacol Physiol 30: 38–43. https://doi.org/10.1046/j.1440-1681.2003.03785.x doi: 10.1046/j.1440-1681.2003.03785.x
    [13] Nambirajan G, Karunanidhi K, Ganesan A, et al. (2018) Evaluation of antidiabetic activity of bud and flower of Avaram Senna (Cassia auriculata L.) In high fat diet and streptozotocin induced diabetic rats. Biomed Pharmacother 108: 1495–1506. https://doi.org/10.1016/j.biopha.2018.10.007 doi: 10.1016/j.biopha.2018.10.007
    [14] Sathasivampillai SV, Rajamanoharan PR, Munday M, et al. (2017) Plants used to treat diabetes in Sri Lankan Siddha Medicine–An ethnopharmacological review of historical and modern sources. J Ethnopharmacol 198: 531–599. https://doi.org/10.1016/j.jep.2016.07.053 doi: 10.1016/j.jep.2016.07.053
    [15] Rajagopal SK, Manickam P, Periyasamy V, et al. (2003) Activity of Cassia auriculata leaf extract in rats with alcoholic liver injury. J Nutr Biochem 14: 452–458. https://doi.org/10.1016/S0955-2863(03)00053-6 doi: 10.1016/S0955-2863(03)00053-6
    [16] Nakamura S, Xu F, Ninomiya K, et al. (2014) Chemical structures and hepatoprotective effects of constituents from Cassia auriculata leaves. Chem Pharm Bull 62: 1026–1031. https://doi.org/10.1248/cpb.c14-00420 doi: 10.1248/cpb.c14-00420
    [17] Sharmila G, Nikitha V, Ilaiyarasi S, et al. (2016) Ultrasound assisted extraction of total phenolics from Cassia auriculata leaves and evaluation of its antioxidant activities. Ind Crops Prod 84: 13–21. https://doi.org/10.1016/j.indcrop.2016.01.010 doi: 10.1016/j.indcrop.2016.01.010
    [18] Gunathilake K, Ranaweera K, Rupasinghe H (2018) Analysis of rutin, β‐carotene, and lutein content and evaluation of antioxidant activities of six edible leaves on free radicals and reactive oxygen species. J Food Biochem 42: e12579. https://doi.org/10.1111/jfbc.12579 doi: 10.1111/jfbc.12579
    [19] Prasathkumar M, Raja K, Vasanth K, et al. (2021) Phytochemical screening and in vitro antibacterial, antioxidant, anti-inflammatory, anti-diabetic, and wound healing attributes of Senna auriculata (L.) Roxb. leaves. Arab J Chem 14: 103345. https://doi.org/10.1016/j.arabjc.2021.103345 doi: 10.1016/j.arabjc.2021.103345
    [20] Bandawane D, Beautikumari S, Gate S, et al. (2014) Evaluation of anti-arthritic activity of ethyl acetate fraction of Cassia auriculata Linn. leaves. Biomed Aging Pathol 4: 105–115. https://doi.org/10.1016/j.biomag.2013.10.009 doi: 10.1016/j.biomag.2013.10.009
    [21] Sutar S, Korpale S, Nadaf S, et al. (2023) Anti-arthritic activity of Senna auriculata leaves extract on formaldehyde-induced arthritic rats. J Res Pharm 27: 1402. https://doi.org/10.29228/jrp.427 doi: 10.29228/jrp.427
    [22] Gupta S, Sharma SB, Bansal SK, et al. (2009) Antihyperglycemic and hypolipidemic activity of aqueous extract of Cassia auriculata L. leaves in experimental diabetes. J Ethnopharmacol 123: 499–503. https://doi.org/10.1016/j.jep.2009.02.019 doi: 10.1016/j.jep.2009.02.019
    [23] Shanmugam H, Venkatesan RS (2022) Determination of antihyperglycemic activity of ethanolic crude leaf extract of Cassia auriculata in the streptozocin induced male wistar albino rats. Int J Health Sci 6: 5617–5630. https://doi.org/10.53730/ijhs.v6nS3.7190 doi: 10.53730/ijhs.v6nS3.7190
    [24] Khader SZA, Ahmed SSZ, Balasubramanian SK, et al. (2017) Modulatory effect of dianthrone rich alcoholic flower extract of Cassia auriculata L. on experimental diabetes. Integr Med Res 6: 131–140. https://doi.org/10.1016/j.imr.2017.01.007 doi: 10.1016/j.imr.2017.01.007
    [25] Fauzi FM, John CM, Karunanidhi A, et al. (2017) Understanding the mode-of-action of Cassia auriculata via in silico and in vivo studies towards validating it as a long term therapy for type Ⅱ diabetes. J Ethnopharmacol 197: 61–72. https://doi.org/10.1016/j.jep.2016.07.058 doi: 10.1016/j.jep.2016.07.058
    [26] Pari L, Latha M (2002) Effect of Cassia auriculata flowers on blood sugar levels, serum and tissue lipids in streptozotocin diabetic rats. Singapore Med J 43: 617–621.
    [27] Abesundara KJ, Matsui T, Matsumoto K (2004) α-Glucosidase inhibitory activity of some Sri Lanka plant extracts, one of which, Cassia auriculata, exerts a strong antihyperglycemic effect in rats comparable to the therapeutic drug acarbose. J Agric Food Chem 52: 2541–2545. https://doi.org/10.1021/jf035330s doi: 10.1021/jf035330s
    [28] Grace B, Viswanathan M, Wilson DD (2022) A new silver nano-formulation of Cassia auriculata flower extract and its anti-diabetic effects. Recent Pat Nanotechnol 16: 160–169. https://doi.org/10.2174/1872210515666210329160523 doi: 10.2174/1872210515666210329160523
    [29] Vijayakumar R, Nachiappan V (2017) Cassia auriculata flower extract attenuates hyperlipidemia in male Wistar rats by regulating the hepatic cholesterol metabolism. Biomed Pharmacother 95: 394–401. https://doi.org/10.1016/j.biopha.2017.08.075 doi: 10.1016/j.biopha.2017.08.075
    [30] Vijayaraj P, Muthukumar K, Sabarirajan J, et al. (2013) Antihyperlipidemic activity of Cassia auriculata flowers in triton WR 1339 induced hyperlipidemic rats. Exp Toxicol Pathol 65: 135–141. https://doi.org/10.1016/j.etp.2011.07.001 doi: 10.1016/j.etp.2011.07.001
    [31] Lingaiah, Mamidala E, Rao PN (2017) Modulatory effect of Cassia auriculata plant extraction on glucose metabolism in alloxan induced diabetic wistar rats. Amer J Sci Med Res 3: 8–11.
    [32] Murugan P, Sakthivel V (2021) Effect of cassia auriculata on lipid profiles in streptozotocin–nicotinamide induced type 2 diabetes mellitus. J Popul Ther Clin Pharmacol 28: 73–79. https://doi.org/10.53555/jptcp.v28i01.2440 doi: 10.53555/jptcp.v28i01.2440
    [33] Kumaran A, Karunakaran RJ (2007) Antioxidant activity of Cassia auriculata flowers. Fitoterapia 78: 46–47. https://doi.org/10.1016/j.fitote.2006.09.031 doi: 10.1016/j.fitote.2006.09.031
    [34] Latha M, Pari L (2003) Preventive effects of Cassia auriculata L. flowers on brain lipid peroxidation in rats treated with streptozotocin. Mol Cell Biochem 243: 23–28. https://doi.org/10.1023/A:1021697311150 doi: 10.1023/A:1021697311150
    [35] Kolar FR, Gogi CL, Khudavand MM, et al. (2018) Phytochemical and antioxidant properties of some Cassia species. Nat Prod Res 32: 1324–1328. https://doi.org/10.1080/14786419.2017.1342085 doi: 10.1080/14786419.2017.1342085
    [36] Kumar JSP, Tharaheswari M, Subhashree S, et al. (2014) Cassia auriculata flower extract articulate its antidiabetic effects by regulating antioxidant levels in plasma, liver and pancreas in T2DM rats. AJPCT 2: 705–722.
    [37] John CM, Sandrasaigaran P, Tong CK, et al. (2011) Immunomodulatory activity of polyphenols derived from Cassia auriculata flowers in aged rats. Cell Immunol 271: 474–479. https://doi.org/10.1016/j.cellimm.2011.08.017 doi: 10.1016/j.cellimm.2011.08.017
    [38] Jancy VJJ, Kalaichelvan V, Balakrishnan N (2020) Phytochemical analysis and anti-oxidant activity of various extracts of plant Cassia auriculata. Res J Pharm Technol 13: 6150–6155. https://doi.org/10.5958/0974-360X.2020.01073.2 doi: 10.5958/0974-360X.2020.01073.2
    [39] Juan-Badaturuge M, Habtemariam S, Thomas MJ (2011) Antioxidant compounds from a South Asian beverage and medicinal plant, Cassia auriculata. Food Chem 125: 221–225. https://doi.org/10.1016/j.foodchem.2010.08.065 doi: 10.1016/j.foodchem.2010.08.065
    [40] Habtemariam S (2013) Antihyperlipidemic components of Cassia auriculata aerial parts: Identification through in vitro studies. Phytother Res 27: 152–155. https://doi.org/10.1002/ptr.4711 doi: 10.1002/ptr.4711
    [41] Annie S, Rajagopal P, Malini S (2005) Effect of Cassia auriculata Linn. root extract on cisplatin and gentamicin-induced renal injury. Phytomedicine 12: 555–560. https://doi.org/10.1016/j.phymed.2003.11.010 doi: 10.1016/j.phymed.2003.11.010
    [42] Jaydeokar AV, Bandawane DD, Bibave KH, et al. (2014) Hepatoprotective potential of Cassia auriculata roots on ethanol and antitubercular drug-induced hepatotoxicity in experimental models. Pharm Biol 52: 344–355. https://doi.org/10.3109/13880209.2013.837075 doi: 10.3109/13880209.2013.837075
    [43] Deshpande S, Kewatkar SM, Paithankar VV (2013) In-vitro antioxidant activity of different fraction of roots of Cassia auriculata Linn. Drug Invent Today 5: 164–168. https://doi.org/10.1016/j.dit.2013.05.006 doi: 10.1016/j.dit.2013.05.006
    [44] Salma B, Janhavi P, Muthaiah S, et al. (2020) Ameliorative efficacy of the Cassia auriculata root against high-fat-diet+ STZ-induced Type-2 diabetes in C57BL/6 mice. ACS Omega 6: 492–504. https://doi.org/10.1021/acsomega.0c04940 doi: 10.1021/acsomega.0c04940
    [45] Rao GN, Kumar PM, Dhandapani V, et al. (2000) Constituents of Cassia auriculata. Fitoterapia 71: 82–83. https://doi.org/10.4103/0250-474X.113546 doi: 10.4103/0250-474X.113546
    [46] Varshney S, Rizvi S, Gupta P (1973) Chemical and spectral studies of novel keto–alcohols from the leaves of Cassia auriculata. Planta Med 23: 363–369. https://doi.org/10.1055/s-0028-1099456 doi: 10.1055/s-0028-1099456
    [47] Murugan T, Wins JA, Murugan M (2013) Antimicrobial activity and phytochemical constituents of leaf extracts of Cassia auriculata. Indian J Pharm Sci 75: 122. https://doi.org/10.4103/0250-474X.113546 doi: 10.4103/0250-474X.113546
    [48] Gunathilake K, Ranaweera K, Rupasinghe H (2020) Optimization of polyphenols and carotenoids extraction from leaves of Cassia auriculata for natural health products. Asian Plant Res J 6: 14–25. https://doi.org/10.9734/APRJ/2020/v6i130118 doi: 10.9734/APRJ/2020/v6i130118
    [49] Abdulwaliyu I, Arekemase SO, Adudu JA, et al. (2019) Investigation of the medicinal significance of phytic acid as an indispensable anti-nutrient in diseases. Clin Nutr Exp 28: 42–61. https://doi.org/10.1016/j.yclnex.2019.10.002 doi: 10.1016/j.yclnex.2019.10.002
    [50] Bartolome AP, Villaseñor IM, Yang WC (2013) Bidens pilosa L.(Asteraceae): Botanical properties, traditional uses, phytochemistry, and pharmacology. Evidence-based Complementary Altern Med 2013: 340215. https://doi.org/10.1155/2013/340215 doi: 10.1155/2013/340215
    [51] Amsalu N, Asfaw Z (2020) Review of the antioxidant properties of wild edible plants in Ethiopia. Afr J Med Health Scis 19: 84–102. https://doi.org/10.5897/AJMHS2019.0082 doi: 10.5897/AJMHS2019.0082
    [52] Nagarani G, Abirami A, Siddhuraju P (2014) Food prospects and nutraceutical attributes of Momordica species: A potential tropical bioresources–A review. Food Sci Human Wellness 3: 117–126. https://doi.org/10.1016/j.fshw.2014.07.001 doi: 10.1016/j.fshw.2014.07.001
    [53] Ramakrishnan P, Kalakandan S, Pakkirisamy M (2018) Studies on positive and negative ionization mode of ESI-LC-MS/MS for screening of Phytochemicals on Cassia auriculata (Aavaram Poo). Pharm J 10: 457–462. https://doi.org/10.5530/pj.2018.3.75 doi: 10.5530/pj.2018.3.75
    [54] Rajkumar P, Selvaraj S, Suganya R, et al. (2016) GC-MS characterization of the anti-diabetic compounds from the flowers of Cassia auriculata (AVARAM): A structure based molecular docking studies. Int J Innov Res Sci Eng Technol 1: 85–93.
    [55] Bargah RK, Kushwaha A, Tirkey A, et al. (2020) In vitro antioxidant and antibacterial screening of flowers extract from Cassia auriculata Linn. Res J Pharm Technol 13: 2624–2628. https://doi.org/10.5958/0974-360X.2020.00466.7 doi: 10.5958/0974-360X.2020.00466.7
    [56] Sahoo J, Kamalaja T, Devi SS, et al. (2020) Nutritional composition of Cassia auriculata flower powder. J Pharmacogn Phytochem9: 867–870.
    [57] Girme A, Saste G, Chinchansure A, et al. (2020) Simultaneous determination of anthraquinone, flavonoids, and phenolic antidiabetic compounds from Cassia auriculata seeds by validated UHPLC based MS/MS method. Mass Spectrom Lett 11: 82–89.
    [58] Zhang Y, Nakamura S, Nakashima S, et al. (2015) Chemical structures of constituents from the seeds of Cassia auriculata. Tetrahedron 71: 6727–6732. https://doi.org/10.1016/j.tet.2015.07.045 doi: 10.1016/j.tet.2015.07.045
    [59] Raj JY, Peter MPJ, Joy V (2012) Chemical compounds investigation of Cassia auriculata seeds: A potential folklore medicinal plant. Asian J Plant Sci Res 2: 187–192.
    [60] Dave H, Ledwani L (2012) A review on anthraquinones isolated from Cassia species and their applications. IJNPR 3: 291–319.
    [61] Sivakumar V, Ilanhtiraiyan S, Ilayaraja K, et al. (2014) Influence of ultrasound on Avaram bark (Cassia auriculata) tannin extraction and tanning. Chem Eng Res Des 92: 1827–1833. https://doi.org/10.1016/j.cherd.2014.04.007 doi: 10.1016/j.cherd.2014.04.007
    [62] Chatterjee S, Khunti K, Davies MJ (2017) Type 2 diabetes. Lancet 389: 2239–2251. https://doi.org/10.1016/S0140-6736(17)30058-2 doi: 10.1016/S0140-6736(17)30058-2
    [63] Yoshinari O, Igarashi K (2011) Anti-diabetic effect of pyroglutamic acid in type 2 diabetic Goto-Kakizaki rats and KK-A y mice. Br J Nutr 106: 995–1004. https://doi.org/10.1017/S0007114511001279 doi: 10.1017/S0007114511001279
    [64] Chou J, Liu R, Yu J, et al. (2018) Fasting serum α‑hydroxybutyrate and pyroglutamic acid as important metabolites for detecting isolated post-challenge diabetes based on organic acid profiles. J Chromatogr B 1100: 6–16. https://doi.org/10.1016/j.jchromb.2018.09.004 doi: 10.1016/j.jchromb.2018.09.004
    [65] Grioli S, Lomeo C, Quattropani M, et al. (1990) Pyroglutamic acid improves the age associated memory impairment. Fund Clin Pharmacol 4: 169–173. https://doi.org/10.1111/j.1472-8206.1990.tb00485.x doi: 10.1111/j.1472-8206.1990.tb00485.x
    [66] Foster AC, Kemp JA (2006) Glutamate-and GABA-based CNS therapeutics. Curr Opin Pharmacol 6: 7–17. https://doi.org/10.1016/j.coph.2005.11.005 doi: 10.1016/j.coph.2005.11.005
    [67] Gao K, Mu C-l, Farzi A, et al. (2020) Tryptophan metabolism: A link between the gut microbiota and brain. Adv Nutr 11: 709–723. https://doi.org/10.1093/advances/nmz127 doi: 10.1093/advances/nmz127
    [68] Li P, Yin Y-L, Li D, et al. (2007) Amino acids and immune function. Br J Nutr 98: 237–252. https://doi.org/10.1017/S000711450769936X doi: 10.1017/S000711450769936X
    [69] Nimalaratne C, Lopes-Lutz D, Schieber A, et al. (2011) Free aromatic amino acids in egg yolk show antioxidant properties. Food Chem 129: 155–161. https://doi.org/10.1016/j.foodchem.2011.04.058 doi: 10.1016/j.foodchem.2011.04.058
    [70] Ming X-F, Rajapakse AG, Carvas JM, et al. (2009) Inhibition of S6K1 accounts partially for the anti-inflammatory effects of the arginase inhibitor L-norvaline. BMC Cardiovasc Disor 9: 12. https://doi.org/10.1186/1471-2261-9-12 doi: 10.1186/1471-2261-9-12
    [71] Karak S, Nag G, De B (2017) Metabolic profile and β-glucuronidase inhibitory property of three species of Swertia. Rev Bras Farmacogn 27: 105–111. https://doi.org/10.1016/j.bjp.2016.07.007 doi: 10.1016/j.bjp.2016.07.007
    [72] Wakuda T, Azuma K, Saimoto H, et al. (2013) Protective effects of galacturonic acid-rich vinegar brewed from Japanese pear in a dextran sodium sulfate-induced acute colitis model. J Funct Foods 5: 516–523. https://doi.org/10.1016/j.jff.2012.10.010 doi: 10.1016/j.jff.2012.10.010
    [73] Suzuki M, Kajuu T (1983) Suppression of hepatic lipogenesis by pectin and galacturonic acid orally-fed at the separate timing from digestion-absorption of nutrients in rat. J Nutr Sci Vitaminol 29: 553–562. https://doi.org/10.3177/jnsv.29.553 doi: 10.3177/jnsv.29.553
    [74] Nguyen NK, Nguyen PB, Nguyen HT, et al. (2015) Screening the optimal ratio of symbiosis between isolated yeast and acetic acid bacteria strain from traditional kombucha for high-level production of glucuronic acid. LWT-Food Sci Technol 64: 1149–1155. https://doi.org/10.1016/j.lwt.2015.07.018 doi: 10.1016/j.lwt.2015.07.018
    [75] Biagi G, Piva A, Moschini M, et al. (2006) Effect of gluconic acid on piglet growth performance, intestinal microflora, and intestinal wall morphology. J Anim Sci 84: 370–378. https://doi.org/10.2527/2006.842370x doi: 10.2527/2006.842370x
    [76] Surman C, Vaudreuil C, Boland H, et al. (2020) L-threonic acid magnesium salt supplementation in ADHD: An open-label pilot study. J Diet Suppl 18: 119–131. https://doi.org/10.1080/19390211.2020.1731044 doi: 10.1080/19390211.2020.1731044
    [77] Banerjee S, Bhattacharjee P, Kar A, et al. (2019) LC–MS/MS analysis and network pharmacology of Trigonella foenum-graecum–A plant from Ayurveda against hyperlipidemia and hyperglycemia with combination synergy. Phytomedicine 60: 152944. https://doi.org/10.1016/j.phymed.2019.152944 doi: 10.1016/j.phymed.2019.152944
    [78] Vogt JA, Ishii-Schrade KB, Pencharz PB, et al. (2006) L-rhamnose and lactulose decrease serum triacylglycerols and their rates of synthesis, but do not affect serum cholesterol concentrations in men. J Nutr 136: 2160–2166.
    [79] Nagata Y, Mizuta N, Kanasaki A, et al. (2018) Rare sugars, d‐allulose, d‐tagatose and d‐sorbose, differently modulate lipid metabolism in rats. J Sci Food Agric 98: 2020–2026. https://doi.org/10.1002/jsfa.8687 doi: 10.1002/jsfa.8687
    [80] Oku T, Murata-Takenoshita Y, Yamazaki Y, et al. (2014) D-sorbose inhibits disaccharidase activity and demonstrates suppressive action on postprandial blood levels of glucose and insulin in the rat. Nutr Res 34: 961–967. https://doi.org/10.1016/j.nutres.2014.09.009 doi: 10.1016/j.nutres.2014.09.009
    [81] Yamada T, Hayashi N, Iida T, et al. (2014) Dietary D-sorbose decreases serum insulin levels in growing Sprague-Dawley rats. J Nutr Sci Vitaminol 60: 297–299. https://doi.org/10.3177/jnsv.60.297 doi: 10.3177/jnsv.60.297
    [82] Seri K, Sanai K, Matsuo N, et al. (1996) L-arabinose selectively inhibits intestinal sucrase in an uncompetitive manner and suppresses glycemic response after sucrose ingestion in animals. Metabolism 45: 1368–1374. https://doi.org/10.1016/S0026-0495(96)90117-1 doi: 10.1016/S0026-0495(96)90117-1
    [83] Li Y, Pan H, Liu JX, et al. (2019) L-Arabinose inhibits colitis by modulating gut microbiota in mice. J Agric Food Chem 67: 13299–13306. https://doi.org/10.1021/acs.jafc.9b05829 doi: 10.1021/acs.jafc.9b05829
    [84] Roy S, Chikkerur J, Roy SC, et al. (2018) Tagatose as a potential nutraceutical: Production, properties, biological roles, and applications. J Food Sci 83: 2699–2709. https://doi.org/10.1111/1750-3841.14358 doi: 10.1111/1750-3841.14358
    [85] Chen Z, Chen J, Zhang W, et al. (2018) Recent research on the physiological functions, applications, and biotechnological production of D-allose. Appl Microbiol Biotechnol 102: 4269–4278. https://doi.org/10.1007/s00253-018-8916-6 doi: 10.1007/s00253-018-8916-6
    [86] Salkovic-Petrisic M, Osmanovic-Barilar J, Knezovic A, et al. (2014) Long-term oral galactose treatment prevents cognitive deficits in male Wistar rats treated intracerebroventricularly with streptozotocin. Neuropharmacology 77: 68–80. https://doi.org/10.1016/j.neuropharm.2013.09.002 doi: 10.1016/j.neuropharm.2013.09.002
    [87] Park M-O, Lee B-H, Lim E, et al. (2016) Enzymatic process for high-yield turanose production and its potential property as an adipogenesis regulator. J Agric Food Chem 64: 4758–4764. https://doi.org/10.1021/acs.jafc.5b05849 doi: 10.1021/acs.jafc.5b05849
    [88] Chung J-Y, Kim Y-S, Kim Y, et al. (2017) Regulation of inflammation by sucrose isomer, Turanose, in raw 264.7 cells. J Cancer Prev 22: 195. https://doi.org/10.15430/JCP.2017.22.3.195 doi: 10.15430/JCP.2017.22.3.195
    [89] Kim E, Bae J, Lee J, et al. (2019) Purification and characterization of turanose, a sucrose isomer and its anti-inflammatory effects in dextran sulfate sodium (DSS)-induced colitis model. J Funct Foods 63: 103570. https://doi.org/10.1016/j.jff.2019.103570 doi: 10.1016/j.jff.2019.103570
    [90] Mizunoe Y, Kobayashi M, Sudo Y, et al. (2018) Trehalose protects against oxidative stress by regulating the Keap1–Nrf2 and autophagy pathways. Redox Biol 15: 115–124. https://doi.org/10.1016/j.redox.2017.09.007 doi: 10.1016/j.redox.2017.09.007
    [91] Laihia J, Kaarniranta K (2020) Trehalose for ocular surface health. Biomolecules 10: 809. https://doi.org/10.3390/biom10050809 doi: 10.3390/biom10050809
    [92] Thompson J, Neutel J, Homer K, et al. (2014) Evaluation of D‐ribose pharmacokinetics, dose proportionality, food effect, and pharmacodynamics after oral solution administration in healthy male and female subjects. J Clin Pharmacol 54: 546–554. https://doi.org/10.1002/jcph.241 doi: 10.1002/jcph.241
    [93] Addis P, Shecterle LM, Cyr JASt (2012) Cellular protection during oxidative stress: A potential role for D-ribose and antioxidants. J Diet Suppl 9: 178–182. https://doi.org/10.3109/19390211.2012.708715 doi: 10.3109/19390211.2012.708715
    [94] Croze ML, Soulage CO (2013) Potential role and therapeutic interests of myo-inositol in metabolic diseases. Biochimie 95: 1811–1827. https://doi.org/10.1016/j.biochi.2013.05.011 doi: 10.1016/j.biochi.2013.05.011
    [95] Corrado F, D'Anna R, Di Vieste G, et al. (2011) The effect of myoinositol supplementation on insulin resistance in patients with gestational diabetes. Diabetic Med 28: 972–975. https://doi.org/10.1111/j.1464-5491.2011.03284.x doi: 10.1111/j.1464-5491.2011.03284.x
    [96] Pintaudi B, Di Vieste G, Bonomo M (2016) The effectiveness of myo-inositol and D-chiro inositol treatment in type 2 diabetes. Int J Endocrinol 2016: 9132052. https://doi.org/10.1155/2016/9132052 doi: 10.1155/2016/9132052
    [97] Unfer V, Facchinetti F, Orrù B, et al. (2017) Myo-inositol effects in women with PCOS: A meta-analysis of randomized controlled trials. Endocr Connect 6: 647–658. https://doi.org/10.1530/EC-17-0243 doi: 10.1530/EC-17-0243
    [98] Benahmed AG, Gasmi A, Arshad M, et al. (2020) Health benefits of xylitol. Appl Microbiol Biotechnol 104: 109495. https://doi.org/10.1016/j.enzmictec.2019.109495 doi: 10.1016/j.enzmictec.2019.109495
    [99] Ur-Rehman S, Mushtaq Z, Zahoor T, et al. (2015) Xylitol: A review on bioproduction, application, health benefits, and related safety issues. Crit Rev Food Sci Nutr 55: 1514–1528. https://doi.org/10.1080/10408398.2012.702288 doi: 10.1080/10408398.2012.702288
    [100] Salli K, Lehtinen MJ, Tiihonen K, et al. (2019) Xylitol's health benefits beyond dental health: A comprehensive review. Nutrients 11: 1813. https://doi.org/10.3390/nu11081813 doi: 10.3390/nu11081813
    [101] Ching T-L, Haenen GR, Bast A (1993) Cimetidine and other H2 receptor antagonists as powerful hydroxyl radical scavengers. Chem-Biol Interact 86: 119–127. https://doi.org/10.1016/0009-2797(93)90116-G doi: 10.1016/0009-2797(93)90116-G
    [102] Saha BC, Racine FM (2011) Biotechnological production of mannitol and its applications. Appl Microbiol Biotechnol 89: 879–891. https://doi.org/10.1007/s00253-010-2979-3 doi: 10.1007/s00253-010-2979-3
    [103] den Hartog GJ, Boots AW, Adam-Perrot A, et al. (2010) Erythritol is a sweet antioxidant. Nutrition 26: 449–458. https://doi.org/10.1016/j.nut.2009.05.004 doi: 10.1016/j.nut.2009.05.004
    [104] Flint N, Hamburg NM, Holbrook M, et al. (2014) Effects of erythritol on endothelial function in patients with type 2 diabetes mellitus: A pilot study. Acta Diabetolo 51: 513–516. https://doi.org/10.1007/s00592-013-0534-2 doi: 10.1007/s00592-013-0534-2
    [105] Wen H, Tang B, Stewart AJ, et al. (2018) Erythritol attenuates postprandial blood glucose by inhibiting α-glucosidase. J Agric Food Chem 66: 1401–1407. https://doi.org/10.1021/acs.jafc.7b05033 doi: 10.1021/acs.jafc.7b05033
    [106] Wölnerhanssen BK, Meyer-Gerspach AC, Beglinger C, et al. (2020) Metabolic effects of the natural sweeteners xylitol and erythritol: A comprehensive review. Crit Rev Food Sci Nutr 60: 1986–1998. https://doi.org/10.1080/10408398.2019.1623757 doi: 10.1080/10408398.2019.1623757
    [107] Chauhan PS, Gupta KK, Bani S (2011) The immunosuppressive effects of Agyrolobium roseum and pinitol in experimental animals. Int Immunopharmacol 11: 286–291. https://doi.org/10.1016/j.intimp.2010.11.028 doi: 10.1016/j.intimp.2010.11.028
    [108] Kim HJ, Park KS, Lee SK, et al. (2012) Effects of pinitol on glycemic control, insulin resistance and adipocytokine levels in patients with type 2 diabetes mellitus. Ann Nutr Metab 60: 1–5. https://doi.org/10.1159/000334834 doi: 10.1159/000334834
    [109] Dang NT, Mukai R, Yoshida K-i, et al. (2010) D-pinitol and myo-inositol stimulate translocation of glucose transporter 4 in skeletal muscle of C57BL/6 mice. Biosci, Biotechnol, Biochem 74: 1062–1067. https://doi.org/10.1271/bbb.90963 doi: 10.1271/bbb.90963
    [110] Hernández-Mijares A, Bañuls C, Peris JE, et al. (2013) A single acute dose of pinitol from a naturally-occurring food ingredient decreases hyperglycaemia and circulating insulin levels in healthy subjects. Food Chem 141: 1267–1272. https://doi.org/10.1016/j.foodchem.2013.04.042 doi: 10.1016/j.foodchem.2013.04.042
    [111] Gao Y, Zhang M, Wu T, et al. (2015) Effects of D-pinitol on insulin resistance through the PI3K/Akt signaling pathway in type 2 diabetes mellitus rats. J Agric Food Chem 63: 6019–6026. https://doi.org/10.1021/acs.jafc.5b01238 doi: 10.1021/acs.jafc.5b01238
    [112] Geethan PA, Prince PSM (2008) Antihyperlipidemic effect of D‐pinitol on streptozotocin‐induced diabetic wistar rats. J Biochem Mol Toxicol 22: 220–224. https://doi.org/10.1002/jbt.20218 doi: 10.1002/jbt.20218
    [113] Choi MS, Lee MK, Jung UJ, et al. (2009) Metabolic response of soy pinitol on lipid‐lowering, antioxidant and hepatoprotective action in hamsters fed‐high fat and high cholesterol diet. Mol Nutr Food Res 53: 751–759. https://doi.org/10.1002/mnfr.200800241 doi: 10.1002/mnfr.200800241
    [114] Sousa LGF, de Souza Cortez LUA, Evangelista JSAM, et al. (2020) Renal protective effect of pinitol in experimental diabetes. Eur J Pharmacol 880: 173130. https://doi.org/10.1016/j.ejphar.2020.173130 doi: 10.1016/j.ejphar.2020.173130
    [115] Lee E, Lim Y, Kwon SW, et al. (2019) Pinitol consumption improves liver health status by reducing oxidative stress and fatty acid accumulation in subjects with non-alcoholic fatty liver disease: A randomized, double-blind, placebo-controlled trial. J Nutr Biochem 68: 33–41. https://doi.org/10.1016/j.jnutbio.2019.03.006 doi: 10.1016/j.jnutbio.2019.03.006
    [116] Vasaikar N, Mahajan U, Patil KR, et al. (2018) D-pinitol attenuates cisplatin-induced nephrotoxicity in rats: Impact on pro-inflammatory cytokines. Chem-Biol Interact 290: 6–11. https://doi.org/10.1016/j.cbi.2018.05.003 doi: 10.1016/j.cbi.2018.05.003
    [117] Sivakumar S, Palsamy P, Subramanian SP (2010) Attenuation of oxidative stress and alteration of hepatic tissue ultrastructure by D-pinitol in streptozotocin-induced diabetic rats. Free Radical Res 44: 668–678. https://doi.org/10.3109/10715761003733901 doi: 10.3109/10715761003733901
    [118] López-Domènech S, Bañuls C, de Marañón AM, et al. (2018) Pinitol alleviates systemic inflammatory cytokines in human obesity by a mechanism involving unfolded protein response and sirtuin 1. Clin Nutr 37: 2036–2044. https://doi.org/10.1016/j.clnu.2017.09.015 doi: 10.1016/j.clnu.2017.09.015
    [119] Kim JC, Shin JY, Shin DH, et al. (2005) Synergistic antiinflammatory effects of pinitol and glucosamine in rats. Phytother Res 19: 1048–1051. https://doi.org/10.1002/ptr.1788 doi: 10.1002/ptr.1788
    [120] Su H, Liu R, Chang M, et al. (2018) Effect of dietary alpha-linolenic acid on blood inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Eur J Nutr 57: 877–891. https://doi.org/10.1007/s00394-017-1386-2 doi: 10.1007/s00394-017-1386-2
    [121] Yue H, Qiu B, Jia M, et al. (2020) Effects of α-linolenic acid intake on blood lipid profiles: A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 61: 2894–2910. https://doi.org/10.1080/10408398.2020.1790496 doi: 10.1080/10408398.2020.1790496
    [122] Yoshida Y, Niki E (2003) Antioxidant effects of phytosterol and its components. J Nutr Sci Vitaminol 49: 277–280. https://doi.org/10.3177/jnsv.49.277 doi: 10.3177/jnsv.49.277
    [123] Fatahi S, Kord-Varkaneh H, Talaei S, et al. (2019) Impact of phytosterol supplementation on plasma lipoprotein (a) and free fatty acid (FFA) concentrations: A systematic review and meta-analysis of randomized controlled trials. Nutr, Metab Cardiovas Dis 29: 1168–1175. https://doi.org/10.1016/j.numecd.2019.07.011 doi: 10.1016/j.numecd.2019.07.011
    [124] Umeno A, Horie M, Murotomi K, et al. (2016) Antioxidative and antidiabetic effects of natural polyphenols and isoflavones. Molecules 21: 708. https://doi.org/10.3390/molecules21060708 doi: 10.3390/molecules21060708
    [125] Habtemariam S, Varghese GK (2014) The antidiabetic therapeutic potential of dietary polyphenols. Curr Pharm Biotechnol 15: 391–400.
    [126] Vinayagam R, Jayachandran M, Xu B (2016) Antidiabetic effects of simple phenolic acids: A comprehensive review. Phytother Res 30: 184–199. https://doi.org/10.1002/ptr.5528 doi: 10.1002/ptr.5528
    [127] Dludla PV, Nkambule BB, Jack B, et al. (2019) Inflammation and oxidative stress in an obese state and the protective effects of gallic acid. Nutrients 11: 23. https://doi.org/10.3390/nu11010023 doi: 10.3390/nu11010023
    [128] Kahkeshani N, Farzaei F, Fotouhi M, et al. (2019) Pharmacological effects of gallic acid in health and diseases: A mechanistic review. Iran J Basic Med Sci 22: 225. https://doi.org/10.22038/ijbms.2019.32806.7897 doi: 10.22038/ijbms.2019.32806.7897
    [129] Badhani B, Sharma N, Kakkar R (2015) Gallic acid: A versatile antioxidant with promising therapeutic and industrial applications. RSC Adv 5: 27540–27557. https://doi.org/10.1039/C5RA01911G doi: 10.1039/C5RA01911G
    [130] Serra A, Macià A, Romero M-P, et al. (2012) Metabolic pathways of the colonic metabolism of flavonoids (flavonols, flavones and flavanones) and phenolic acids. Food Chem 130: 383–393. https://doi.org/10.1016/j.foodchem.2011.07.055 doi: 10.1016/j.foodchem.2011.07.055
    [131] Kumar N, Goel N (2019) Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnol Rep 24: e00370. https://doi.org/10.1016/j.btre.2019.e00370 doi: 10.1016/j.btre.2019.e00370
    [132] Abd El-Aziz TA, Mohamed RH, Pasha HF, et al. (2012) Catechin protects against oxidative stress and inflammatory-mediated cardiotoxicity in adriamycin-treated rats. Clin Exper Med 12: 233–240. https://doi.org/10.1007/s10238-011-0165-2 doi: 10.1007/s10238-011-0165-2
    [133] Shafabakhsh R, Milajerdi A, Reiner Ž, et al. (2020) The effects of catechin on endothelial function: A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 60: 2369–2378. https://doi.org/10.1080/10408398.2019.1639037 doi: 10.1080/10408398.2019.1639037
    [134] Pedro AC, Maciel GM, Rampazzo Ribeiro V, et al. (2020) Fundamental and applied aspects of catechins from different sources: A review. Int J Food Sci Tech 55: 429–442. https://doi.org/10.1111/ijfs.14371 doi: 10.1111/ijfs.14371
    [135] Guo T, Song D, Cheng L, et al. (2019) Interactions of tea catechins with intestinal microbiota and their implication for human health. Food Sci Biotechnol 28: 1617–1625. https://doi.org/10.1007/s10068-019-00656-y doi: 10.1007/s10068-019-00656-y
    [136] Márquez Campos E, Jakobs L, Simon M-C (2020) Antidiabetic effects of flavan-3-ols and their microbial metabolites. Nutrients 12: 1592. https://doi.org/10.3390/nu12061592 doi: 10.3390/nu12061592
    [137] Bai L, Li X, He L, et al. (2019) Antidiabetic potential of flavonoids from traditional Chinese medicine: a review. Amer J Chin Med 47: 933–957. https://doi.org/10.1142/S0192415X19500496 doi: 10.1142/S0192415X19500496
    [138] Takahashi M, Miyashita M, Suzuki K, et al. (2014) Acute ingestion of catechin-rich green tea improves postprandial glucose status and increases serum thioredoxin concentrations in postmenopausal women. Br J Nutr 112: 1542–1550. https://doi.org/10.1017/S0007114514002530 doi: 10.1017/S0007114514002530
    [139] Eid HM, Ouchfoun M, Saleem A, et al. (2016) A combination of (+)-catechin and (−)-epicatechin underlies the in vitro adipogenic action of Labrador tea (Rhododendron groenlandicum), an antidiabetic medicinal plant of the Eastern James Bay Cree pharmacopeia. J Ethnopharmacol 178: 251–257. https://doi.org/10.1016/j.jep.2015.12.021 doi: 10.1016/j.jep.2015.12.021
    [140] Mrabti HN, Jaradat N, Fichtali I, et al. (2018) Separation, identification, and antidiabetic activity of catechin isolated from Arbutus unedo L. plant roots. Plants 7: 31. https://doi.org/10.3390/plants7020031 doi: 10.3390/plants7020031
    [141] Wang W, Sun C, Mao L, et al. (2016) The biological activities, chemical stability, metabolism and delivery systems of quercetin: A review. Trends Food Sci Technol 56: 21–38. https://doi.org/10.1016/j.tifs.2016.07.004 doi: 10.1016/j.tifs.2016.07.004
    [142] D'Andrea G (2015) Quercetin: A flavonol with multifaceted therapeutic applications? Fitoterapia 106: 256–271. https://doi.org/10.1016/j.fitote.2015.09.018 doi: 10.1016/j.fitote.2015.09.018
    [143] Lesjak M, Beara I, Simin N, et al. (2018) Antioxidant and anti-inflammatory activities of quercetin and its derivatives. J Funct Foods 40: 68–75. https://doi.org/10.1016/j.jff.2017.10.047 doi: 10.1016/j.jff.2017.10.047
    [144] Xu D, Hu M-J, Wang Y-Q, et al. (2019) Antioxidant activities of quercetin and its complexes for medicinal application. Molecules 24: 1123. https://doi.org/10.3390/molecules24061123 doi: 10.3390/molecules24061123
    [145] Zaplatic E, Bule M, Shah SZA, et al. (2019) Molecular mechanisms underlying protective role of quercetin in attenuating Alzheimer's disease. Life Sci 224: 109–119. https://doi.org/10.1016/j.lfs.2019.03.055 doi: 10.1016/j.lfs.2019.03.055
    [146] Patel RV, Mistry BM, Shinde SK, et al. (2018) Therapeutic potential of quercetin as a cardiovascular agent. Eur J Med Chem 155: 889–904. https://doi.org/10.1016/j.ejmech.2018.06.053 doi: 10.1016/j.ejmech.2018.06.053
    [147] Dabeek WM, Marra MV (2019) Dietary quercetin and kaempferol: Bioavailability and potential cardiovascular-related bioactivity in humans. Nutrients 11: 2288. https://doi.org/10.3390/nu11102288 doi: 10.3390/nu11102288
    [148] Bule M, Abdurahman A, Nikfar S, et al. (2019) Antidiabetic effect of quercetin: A systematic review and meta-analysis of animal studies. Food Chem Toxicol 125: 494–502. https://doi.org/10.1016/j.fct.2019.01.037 doi: 10.1016/j.fct.2019.01.037
    [149] Ahn J, Lee H, Kim S, et al. (2008) The anti-obesity effect of quercetin is mediated by the AMPK and MAPK signaling pathways. Biochem Biophys Res Commun 373: 545–549. https://doi.org/10.1016/j.bbrc.2008.06.077 doi: 10.1016/j.bbrc.2008.06.077
    [150] Nabavi SF, Russo GL, Daglia M, et al. (2015) Role of quercetin as an alternative for obesity treatment: you are what you eat! Food Chem 179: 305–310. https://doi.org/10.1016/j.foodchem.2015.02.006 doi: 10.1016/j.foodchem.2015.02.006
    [151] Mohammadi A, Kazemi S, Hosseini M, et al. (2019) Chrysin effect in prevention of acetaminophen-induced hepatotoxicity in rat. Chem Res Toxicol 32: 2329–2337. https://doi.org/10.1021/acs.chemrestox.9b00332 doi: 10.1021/acs.chemrestox.9b00332
    [152] Zhang Z, Li G, Szeto SS, et al. (2015) Examining the neuroprotective effects of protocatechuic acid and chrysin on in vitro and in vivo models of Parkinson disease. Free Radical Biol Med 84: 331–343. https://doi.org/10.1016/j.freeradbiomed.2015.02.030 doi: 10.1016/j.freeradbiomed.2015.02.030
    [153] Ramírez-Espinosa JJ, Saldaña-Ríos J, García-Jiménez S, et al. (2018) Chrysin induces antidiabetic, antidyslipidemic and anti-inflammatory effects in athymic nude diabetic mice. Molecules 23: 67. https://doi.org/10.3390/molecules23010067 doi: 10.3390/molecules23010067
    [154] Taslimi P, Kandemir FM, Demir Y, et al. (2019) The antidiabetic and anticholinergic effects of chrysin on cyclophosphamide‐induced multiple organ toxicity in rats: Pharmacological evaluation of some metabolic enzyme activities. J Biochem Mol Toxicol 33: e22313. https://doi.org/10.1002/jbt.22313 doi: 10.1002/jbt.22313
    [155] Malik EM, Müller CE (2016) Anthraquinones as pharmacological tools and drugs. Med Res Rev 36: 705–748. https://doi.org/10.1002/med.21391 doi: 10.1002/med.21391
    [156] Duval J, Pecher V, Poujol M, et al. (2016) Research advances for the extraction, analysis and uses of anthraquinones: A review. Ind Crop Prod 94: 812–833. https://doi.org/10.1016/j.indcrop.2016.09.056 doi: 10.1016/j.indcrop.2016.09.056
    [157] Fouillaud M, Caro Y, Venkatachalam M, et al. (2018) Anthraquinones, In: Nollet L, Gutierrez-Uribe J (Eds.), Phenolic compounds in food, Boca Raton: CRC Press, 131–172.
    [158] Li Y, Jiang J-G (2018) Health functions and structure–activity relationships of natural anthraquinones from plants. Food Funct 9: 6063–6080. https://doi.org/10.1039/C8FO01569D doi: 10.1039/C8FO01569D
    [159] Yen G-C, Duh P-D, Chuang D-Y (2000) Antioxidant activity of anthraquinones and anthrone. Food Chem 70: 437–441. https://doi.org/10.1016/S0308-8146(00)00108-4 doi: 10.1016/S0308-8146(00)00108-4
    [160] Li X, Chu S, Liu Y, et al. (2019) Neuroprotective effects of anthraquinones from rhubarb in central nervous system diseases. Evidence-Based Complementary Altern Med 2019: 3790728. https://doi.org/10.1155/2019/3790728 doi: 10.1155/2019/3790728
    [161] Chen Db, Gao Hw, Peng C, et al. (2020) Quinones as preventive agents in Alzheimer's diseases: Focus on NLRP3 inflammasomes. J Pharm Pharmacol 72: 1481–1490. https://doi.org/10.1111/jphp.13332 doi: 10.1111/jphp.13332
    [162] Chien S-C, Wu Y-C, Chen Z-W, et al. (2015) Naturally occurring anthraquinones: Chemistry and therapeutic potential in autoimmune diabetes. Evidence-Based Complementary Altern Med 2015: 357357. https://doi.org/10.1155/2015/357357 doi: 10.1155/2015/357357
    [163] Zhao XY, Qiao GF, Li BX, et al. (2009) Hypoglycaemic and hypolipidaemic effects of emodin and its effect on L-type calcium channnels in dyslipidaemic-diabetic rats. Clin Exp Pharmacol Physiol 36: 29–34. https://doi.org/10.1111/j.1440-1681.2008.05051.x doi: 10.1111/j.1440-1681.2008.05051.x
    [164] Mishra SK, Tiwari S, Shrivastava A, et al. (2014) Antidyslipidemic effect and antioxidant activity of anthraquinone derivatives from Rheum emodi rhizomes in dyslipidemic rats. J Nat Med 68: 363–371. https://doi.org/10.1007/s11418-013-0810-z doi: 10.1007/s11418-013-0810-z
    [165] Li P, Lu Q, Jiang W, et al. (2017) Pharmacokinetics and pharmacodynamics of rhubarb anthraquinones extract in normal and disease rats. Biomed Pharmacother 91: 425–435. https://doi.org/10.1016/j.biopha.2017.04.109 doi: 10.1016/j.biopha.2017.04.109
    [166] Dalirfardouei R, Karimi G, Jamialahmadi K (2016) Molecular mechanisms and biomedical applications of glucosamine as a potential multifunctional therapeutic agent. Life Sci 152: 21–29. https://doi.org/10.1016/j.lfs.2016.03.028 doi: 10.1016/j.lfs.2016.03.028
    [167] Shintani T, Yamazaki F, Katoh T, et al. (2010) Glucosamine induces autophagy via an mTOR-independent pathway. Biochem Biophys Res Commun 391: 1775–1779. https://doi.org/10.1016/j.bbrc.2009.12.154 doi: 10.1016/j.bbrc.2009.12.154
    [168] Lee JH, Jia Y, Thach TT, et al. (2017) Hexacosanol reduces plasma and hepatic cholesterol by activation of AMP-activated protein kinase and suppression of sterol regulatory element-binding protein-2 in HepG2 and C57BL/6J mice. Nutr Res 43: 89–99. https://doi.org/10.1016/j.nutres.2017.05.013 doi: 10.1016/j.nutres.2017.05.013
    [169] Hsu C, Shih H, Chang Y, et al. (2015) The beneficial effects of tetracosanol on insulin-resistance by insulin receptor kinase sensibilisation. J Funct Foods 14: 174–182. https://doi.org/10.1016/j.jff.2015.01.033 doi: 10.1016/j.jff.2015.01.033
    [170] Ninh The S (2017) A review on the medicinal plant Dalbergia odorifera species: Phytochemistry and biological activity. Evidence-Based Complementary Altern Med 2017: 7142370. https://doi.org/10.1155/2017/7142370 doi: 10.1155/2017/7142370
    [171] Lee D-S, Jeong G-S (2014) Arylbenzofuran isolated from Dalbergia odorifera suppresses lipopolysaccharide-induced mouse BV2 microglial cell activation, which protects mouse hippocampal HT22 cells death from neuroinflammation-mediated toxicity. Eur J Pharmacol 728: 1–8. https://doi.org/10.1016/j.ejphar.2013.12.041 doi: 10.1016/j.ejphar.2013.12.041
  • Reader Comments
  • © 2024 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(540) PDF downloads(86) Cited by(0)

Article outline

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog