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 (3.3 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Open Access | Online First

Critical Review for active iridoids in Gardenia jasminoides J.Ellis as a plant of food and medicine homology

Ya-Dan Zhang1Meng-Han Wang1,2Miao Guan1,2Fathy Mohamed Saber Ali Mehaya3Xiao-Yu Chen1,2( )Xu-Qiang Liu1,2,4( )
National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
College of Agriculture, Henan University, Kaifeng 475004, China
Food Technology Department, National Research Centre, Dokki 12622, Egypt
Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Kaifeng 475004, China
Show Author Information

Highlights

(1) Iridoids from Gardenia jasminoides contain six categories according to the structure of iridoids.

(2) Iridoids from Gardenia jasminoides possess anti-inflammatory, antioxidant, anti-tumor, and neuroprotection biological activities.

(3) The potential mechanisms of anti-inflammatory, antioxidant, anti-tumor and neuroprotective effects of iridoids from Gardenia jasminoides were summarized.

Graphical Abstract

There are 94 iridoids isolated from G. jasminoides, and their structures are diversified. The iridoids in G. jasminoides are divided into six categories according to the structure. Main biological activities of iridoids from Gardenia jasminoides (include anti-inflammatory, neuroprotection, and anti-oxidative) and their potential mechanism.

Abstract

Iridoids compounds are one of the main active components of Gardenia jasminoides, which were divided into two categories: iridoid glycosides and non-glucosylated iridoids according to their chemical structures. Relevant studies have shown that components such as geniposide, deacetyl asperulosidic acid methyl ester, and geniposidic acid play an important role in anti-inflammatory, hypoglycemia, and the protection of cardiomyocytes. While the pharmacological effects of most iridoid glycosides are still not clear. This paper’s systematic summary was created by searching for relevant iridoids material on websites such as Google Scholar, PubMed, SciFinder Scholar, Science Direct, and others. Up to now, a total of 94 iridoids had been identified in G. jasminoides, among which 40 iridoids had extensive biological activities and the mechanisms involved in MAPK, NF-κB, JNK signal pathways. This paper reviews the literature on types and pharmacological effects of iridoid compounds in G. jasminoides, in order to provide a reference for its development and application.

References

[1]

Sampaio-Santos, M. I., Kaplan, M. A. C. Biosynthesis significance of iridoids in chemosystematics. Journal of the Brazilian Chemical Society, 2010, 12: 144–153. https://doi.org/10.1590/S0103-50532001000200004

[2]

Franzyk, H. Synthetic aspects of iridoid chemistry. Organic Chemistry Frontiers, 2000, 79: 1–114. https://doi.org/10.1007/978-3-7091-6341-21

[3]

Kong, Y. F. Research progress on structures and structure-activity relationships of iridoids. Natural Product Research, 2021, 33: 1236–1250. https://doi.org/10.7501/j.issn.0253-2670.2013.19.025

[4]

Çelikel, F. G., Reid, M. S., Jiang, C. Z. Postharvest physiology of cut Gardenia jasminoides flowers. Scientia Horticulturae, 2020, 261: 108983. https://doi.org/10.1016/j.scienta.2019.108983

[5]
Chen, L. P., Tian, X. Y., Li, X. L., et al. Gardenia jasminoides Ellis: ethnopharmacology, phytochemistry, and pharmacological and industrial applications of an important traditional Chinese medicine. Journal of Ethnopharmacology, 2020 , 257: 112829. https://doi.org/10.1016/j.jep.2020.112829
[6]

Feng, J., He X., Zhou, S., et al. Preparative separation of crocins and geniposide simultaneously from gardenia fruits using macroporous resin and reversed-phase chromatography. Journal of Separation Science, 2014, 37: 314–322. https://doi.org/10.1002/jssc.201300601

[7]
Li, N., Wu, H. M., Zhou. H. Antioxidative property and molecular mechanisms underlying geniposide-mediated therapeutic effects in diabetes mellitus and cardiovascular disease. Oxidative Medicine and Cellular Longevity, 2019 , 2019: 20. https://doi.org/10.1155/2019/7480512
[8]

Hobbs, C. A., Koyanagi. M., Swartz, C., et al. Genotoxicity evaluation of the naturally-derived food colorant, gardenia blue, and its precursor, genipin. Food and Chemical Toxicology, 2018, 118: 695–708. https://doi.org/10.1016/j.fct.2018.06.001

[9]

Zhang, Q. H. Homology of medicine and food and Chinese herb taken as food. Journal of Liaoning University of Traditional Chinese Medicine, 2009, 11: 54–55. https://doi.org/10.13194/j.jlunivtcm.2009.07.56.zhangqh.096

[10]
Jina, C. Y., Zongoa, A. W., Dub, H. J., et al. Gardenia (Gardenia jasminoides Ellis) fruit: a critical review of its functionalnutrients, processing methods, health-promoting effects, comprehensiveapplication and future tendencies. Critical Reviews in Food Science and Nutrition, 2023 : 1–28. https://doi.org/10.1080/10408398.2023.2270530
[11]
Wang, C., Gong, X., Bo, A., et al. Iridoids: research advances in their phytochemistry. Biological Activities and Pharmacokinetics Molecules, 2020 , 25: 287. https://doi.org/10.3390/molecules25020287
[12]
Ghule, B. V., Kotagale, N. R., Patil, K. S., Inhibition of the pro-inflammatory mediators in rat neutrophils by shanzhiside methyl ester and its acetyl derivative isolated from Barleria prionitis. Journal of Ethnopharmacology, 2020 : 112374. https://doi.org/10.1016/j.jep.2019.112374
[13]

Li, H. B., Ma, J. F., Mei, Y. D., et al. Two new iridoid glycosides from the fruit of Gardenia jasminoides. Natural Product Research, 2022, 36: 186–192. https://doi.org/10.1080/14786419.2020.1775227

[14]

Chen, Y. F., Zhang, J. Y., Zhao, M., et al. The analgesic activity and possible mechanisms of deacetyl asperulosidic acid methyl ester from Ji Shi Teng in mice. Pharmacology Biochemistry and Behavior, 2012, 102: 585–592. https://doi.org/10.1016/j.pbb.2012.07.005

[15]

Xia, T., Zhao, R., He, S., et al. Gardenoside ameliorates inflammation and inhibits ECM degradation in IL-1β-treated rat chondrocytes via suppressing NF-κB signaling pathways. Biochemical and Biophysical Research Communications, 2023, 640: 164–172. https://doi.org/10.1016/j.bbrc.2022.12.016

[16]

Liu, J., Zhao, N., Shi, G., et al. Geniposide ameliorated sepsis-induced acute kidney injury by activating PPARγ. Neurobiology of Aging, 2020, 12: 22744–22758. https://doi.org/10.18632/aging.103902

[17]

Yu, Y., Bian, Y., Shi, J. X., et al. Geniposide promotes splenic Treg differentiation to alleviate colonic inflammation and intestinal barrier injury in ulcerative colitis mice. Bioengineered, 2022, 13: 14616–14631. https://doi.org/10.1080/21655979.2022.2092678

[18]

Chen, J. Y., Li, H. H., Hu, S. L., et al. Anti-inflammatory effects and pharmacokinetics study of geniposide on rats with adjuvant arthritis. International Immunopharmacology, 2015, 24: 102–109. https://doi.org/10.1016/j.intimp.2014. 11.017

[19]

Yu, B., Shen, Y., Qiao, J., et al. Geniposide attenuates Staphylococcus aureus-induced pneumonia in mice by inhibiting NF-κB activation. Microbial Pathogenesis, 2017, 112: 117–121. https://doi.org/10.1016/j.micpath.2017.09.050

[20]

Song, X., Zhang, W., Wang, T., et al. Geniposide plays an anti-inflammatory role via regulating TLR4 and downstream signaling pathways in lipopolysaccharide-induced mastitis in mice. Inflammopharmacology, 2014, 37: 1588–1598. https://doi.org/10.1007/s10753-014-9885-2

[21]

Chang, C. H., Wu, J. B., Yang, J. S., et al. The suppressive effects of geniposide and genipin on Helicobacter pylori infections in vitro and in vivo. Journal of Food Science, 2017, 82: 3021–3028. https://doi.org/10.1111/1750-3841.13955

[22]

Li, C., Mu, Y. R. Penta-acetyl geniposide suppresses migration, invasion, and inflammation of TNF-α-stimulated rheumatoid arthritis fibroblast-like synoviocytes involving Wnt/β-catenin signaling pathway. Inflammopharmacology, 2021, 44: 2232–2245. https://doi.org/10.1007/s10753-021-01495-y

[23]
Song, M., Chen, Z., Qiu, R., et al. Inhibition of NLRP3-mediated crosstalk between hepatocytes and liver macrophages by geniposidic acid alleviates cholestatic liver inflammatory injury. Redox Biology, 2022, 55: 102404. https://doi.org/10.1016/j.redox.2022.102404
[24]

Tang, J. J., Zhao, N., Gao, Y. Q., et al. Phytosterol profiles and iridoids of the edible Eucommia ulmoides Oliver seeds and their anti-inflammatory potential. Food Bioscience, 2021, 43: 2212–4292. https://doi.org/10.1016/j.fbio.2021.101295

[25]

Sohn, Y. A., Hwang, I. Y., Lee, S. Y., Cho, et al. Protective Effects of Genipin on Gastrointestinal Disorders. Biological and Pharmaceutical Bulletin, 2017, 40: 151–154. https://doi.org/10.1248/ bpb.b16-00545

[26]

Kim, T. H., Yoon, S. J., Lee, S. M. Genipin attenuates sepsis by inhibiting Toll-like receptor signaling. Molecular Medicine, 2012, 18: 455–465. https://doi.org/10.2119/molmed.2011.00308

[27]

Li, Z., Ma, T., Zhang, W., et al. Genipin attenuates dextran sulfate sodium-induced colitis via suppressing inflammatory and oxidative responses. Inflammopharmacology, 2020, 28: 333–339. https://doi.org/10.1007/s10787-019-00639-9

[28]

Huertas-Bell, M., Cuéllar-Sáenz, J. A., Rodriguez, C. N., et al. A pilot study to evaluate genipin in Staphylococcus aureus and Pseudomonas aeruginosa keratitis models: modulation of pro-inflammatory cytokines and matrix metalloproteinases. International Journal of Molecular Sciences, 2023, 24: 6904. https://doi.org/10.3390/ijms24086904

[29]

Wang, J., Chen, L. Genipin inhibits LPS-induced inflammatory response in BV2 microglial cells. Neurochemical Research, 2017, 42: 2769–2776. https://doi.org/10.1007/s11064-017-2289-6

[30]

Shindo, S., Hosokawa, Y., Hosokawa, I., et al. Genipin inhibits MMP-1 and MMP-3 release from TNF-a-stimulated human periodontal ligament cells. Biochimie, 2014, 107: 391–395. https://doi.org/10.1016/j.biochi.2014.10.008

[31]

Jeon, W. K., Hong, H. Y., Kim, B. C. Genipin up-regulates heme oxygenase-1 via PI3-kinase-JNK1/2-Nrf2 signaling pathway to enhance the anti-inflammatory capacity in RAW264.7 macrophages. Archives of Biochemistry and Biophysics, 2011, 512: 119–125. https://doi.org/10.1016/j.abb.2011.05.016

[32]

Luo, X., Lin, B., Gao, Y., et al. Genipin attenuates mitochondrial-dependent apoptosis, endoplasmic reticulum stress, and inflammation via the PI3K/AKT pathway in acute lung injury. International Immunopharmacology, 2019, 76: 105842. https://doi.org/10.1016/j.intimp

[33]

Zhang, X., Wang, L., Zheng, Z., et al. Online microdialysis-ultra performance liquid chromatography–mass spectrometry method for comparative pharmacokinetic investigation on iridoids from Gardenia jasminoides Ellis in rats with different progressions of type 2 diabetic complications. Journal of Pharmaceutical and Biomedical Analysis, 2017, 140: 146–154. https://doi.org/10.1016/j.jpba.2017.03.040

[34]

Zeng, X., Jiang, J., Liu, S., et al. Bidirectional effects of geniposide in liver injury: preclinical evidence construction based on meta-analysis. Journal of ethnopharmacology, 2023, 319: 117061. https://doi.org/10.1016/j.jep.2023.117061

[35]

He, J., Lu, X., Wei, T., et al. Asperuloside and asperulosidic acid exert an anti-inflammatory effect via suppression of the NF-κB and MAPK signaling pathways in LPS-Induced RAW 264.7 macrophages. International Journal of Molecular Sciences, 2018, 19: 2027. https://doi.org/10.3390/ijms19072027

[36]

Chen, Y. E., Xu, S. J., Lu, Y. Y., et al. Asperuloside suppressing oxidative stress and inflammation in DSS-induced chronic colitis and RAW 264.7 macrophages via Nrf2/HO-1 and NF-κB pathways. Chemico-Biological Interactions, 2021, 344: 109512. https://doi.org/10.1016/j.cbi.2021.109512

[37]

Zhang, H., Feng, N., Xu, Y. T., et al. Chemical constituents from the flowers of wild Gardenia jasminoides J. Ellis. Chemistry & Biodiversity, 2017, 14: 1600437. https://doi.org/10.1002/cbdv.201600437

[38]

Ijaz, F., Haq, A. U., Ahmad, I., et al. Antioxidative iridoid glycosides from the sky flower ( Duranta repens Linn). Journal of Enzyme Inhibition and Medicinal Chemistry, 2011, 26: 88–92. https://doi.org/ 10.3109/14756361003724778

[39]

Wei, D., Liu, B. L., Shi, Y., et al. Genipin inhibited on oxidative stress injury of ARPE-19 cells by regulating Nrf2/HO-1 pathway. International Journal of Molecular Medicine, 2022, 51: 83–86. https://doi.org/10.3892/ijmm.2018.4027

[40]

Cheng, S., Jia, H., Zhang, Y., et al. Geniposidic acid from Eucommia ulmoides oliver staminate flower tea mitigates cellular oxidative stress via activating AKT/NRF2 signaling. Molecules, 2022, 27: 8568. https://doi.org/10.3390/molecules27238568

[41]

Li, J., Ge, H., Xu, Y., et al. Geniposide alleviates oxidative damage in hepatocytes through regulating miR-27b-3p/Nrf2 axis. Journal of Agricultural and Food Chemistry, 2022, 70: 11544–11553. https://doi.org/10.1021/acs.jafc.2c03856

[42]
Lakshmana Raju, B., Lin, S. J., Hou, W. C., et al. Antioxidant iridoid glucosides from wendlandia formosana. Natural Product Research, 2004 , 18: 357–364. https://doi.org/10.1080/14786410310001622013
[43]

Wu, Q., Gai, S., Zhang, H. J. Asperulosidic acid, a bioactive iridoid, alleviates placental oxidative stress and inflammatory responses in gestational diabetes mellitus by suppressing NF-κB and MAPK signaling pathways. Pharmacology, 2022, 107: 197–205. https://doi.org/10.1159/000521080

[44]

Lin, L., Cheng, X. L., Li, M. Z., et al. Antitumor effects of iridomyrmecin in HeLa cervical cancer cells are mediated via apoptosis induction, loss of mitochondrial membrane potential, cell cycle arrest and down-regulation of PI3K/Akt and up-regulation of lncRNA CCAT2 expression. Bangladesh Journal of Pharmacology, 2016, 11: 856. https://doi.org/10.3329/bjp.v11i4.27539

[45]

Zhang, C., Wang, N., Tan, H. Y., et al. Direct inhibition of the TLR4/MyD88 pathway by geniposide suppresses HIF-1α-independent VEGF expression and angiogenesis in hepatocellular carcinoma. British Journal of Pharmacology, 2020, 14: 3240–3257. https://doi.org/10.1111/bph.15046

[46]

Alamri, M. A., Alawam, A. S., Alshahrani, M. M., et al. Establishing the role of iridoids as potential Kirsten rat sarcoma viral oncogene homolog G12C inhibitors using molecular docking; molecular docking simulation; molecular mechanics poisson–boltzmann surface area; frontier molecular orbital theory; molecular electrostatic potential; and absorption, distribution, metabolism, excretion, and toxicity analysis. Molecules, 2023, 28: 5050. https://doi.org/10.3390/molecules28135050

[47]

Moon, A. Genipin, a constituent of Gardenia jasminoides Ellis, induces apoptosis and inhibits invasion in MDA-MB-231 breast cancer cells. Oncology Reports, 2011, 27: 567–572. https://doi.org/10.3892/or.2011.1508

[48]

Li, Z., Zhang, T. B., Jia, D. H., et al. Genipin inhibits the growth of human bladder cancer cells via inactivation of PI3K/Akt signaling. Oncology Letters, 2018, 15: 2619–2624. https://doi.org/10.3892/ol.2017.7588

[49]

Jo, M. J., Jeong, S., Yun, H. K., et al. Genipin induces mitochondrial dysfunction and apoptosis via downregulation of Stat3/mcl-1 pathway in gastric cancer. BMC Cancer, 2019, 19: 739. https://doi.org/10.1186/s12885-019-5957-x

[50]

Cao, H., Feng, Q., Xu, W., et al. Genipin induced apoptosis associated with activation of the c-Jun NH2-terminal kinase and p53 protein in HeLa cells. Biological & pharmaceutical bulletin, 2010, 33: 1343–1348. https://doi.org/10.1248/bpb

[51]

Yang, X., Yao, J., Luo, Y., et al. P38 MAP kinase mediates apoptosis after genipin treatment in non-small-cell lung cancer H1299 cells via a mitochondrial apoptotic cascade. Journal of Pharmacological Sciences, 2013, 121: 272–281. https://doi.org/10.1254/jphs.12234fp

[52]
Feng, Q., Cao, H., Xu, W., et al. Apoptosis induced by genipin in human leukemia K562 cells: involvement of c-Jun N-terminal kinase in G2/M arrest. Acta Pharmacologica Sinica, 2011, 32: 519–527. https://doi.org/10.1038/aps.2010.158. Epub 2011 Mar 14.
[53]

Peng, C., Huang, C., Wang, C. The anti-tumor effect and mechanisms of action of penta-acetyl geniposide. Current Cancer Drug Targets, 2005, 5: 299–305. https://doi.org/10.2174/1568009054064633

[54]

Qi, Z. M., Wang, X., Liu, X., et al. Asperuloside promotes apoptosis of cervical cancer cells through endoplasmic reticulum stress-mitochondrial pathway. Chinese Journal of Integrative Medicine, 2024, 30: 34–41. https://doi.org/10.1007/s11655-023-3695-z

[55]

Zhang, H., Sun, Y., Yau, S., et al. Synergistic effects of two natural compounds of iridoids on rapid antidepressant action by up-regulating hippocampal PACAP signaling. British Journal of Pharmacology, 2022, 179: 4078–4091. https://doi.org/10.1111/bph.15847

[56]

Sun, Z. W., Zhan, H. G.,Wang, C., et al. Shanzhiside methylester protects against depression by inhibiting inflammation via the miRNA-155-5p/SOCS1 axis. psychopharmacology, 2022, 239: 2201–2213. https://doi.org/10.1007/s00213-022-06107-7

[57]

Luo, X. F., Li, Y., Hu, J. Regulatory effect of gardenoside on sleep disorder in rats with Parkinson’s disease and itsmechanism. Journal of Jilin University Medicine Edition, 2020, 46: 1177–1181. https://doi.org/10.13481/j.1671-587x.20200611

[58]

Ma, W. W., Tao, Y., Wang, Y. Y., et al. Effects of Gardenia jasminoides extracts on cognition and innate immune response in an adult Drosophila model of Alzheimer's disease. Chinese Journal of Natural Medicines, 2017, 15: 899–904. https://doi.org/10.1016/S1875-5364 (18)30005-0

[59]

Chen, G. H., Chen, Q. Role of gardenoside and its aglycone genipine in treatment of depression. Chinese Journal of Chemical Physics, 2022, 38: 1877–1882. https://doi.org/10.3969/j.issn.1000-4718.2022.10.018

[60]

Uczay, M., Pflüger, P., Picada, J. N., et al. Geniposide and asperuloside alter the COX-2 and GluN2B receptor expression after pilocarpine-induced seizures in mice. Naunyn-schmiedebergs Archives of Pharmacology, 2023, 396: 951–962. https://doi.org/10.1007/s00210-022-02367-4

[61]

Wei, H., Duan, G., He, J., et al. Geniposide attenuates epilepsy symptoms in a mouse model through the PI3K/Akt/GSK-3β signaling pathway. Experimental and Therapeutic Medicine, 2017, 1: 1136–1142. https://doi.org/10.3892/etm.2017.5512

[62]

Wang, M., Yang, L., Chen, Z., et al. Geniposide ameliorates chronic unpredictable mild stress induced depression-like behavior through inhibition of ceramide-PP2A signaling via the PI3K/Akt/GSK3β axis. Psychopharmacology, 2021, 238: 2789–2800. https://doi.org/10.1007/s00213-021-05895-8

[63]

Chen, Q. Y., Yin, Y., Li, L., et al. Geniposidic acid confers neuroprotective effects in a mouse model of Alzheimer's disease through activation of a PI3K/AKT/GAP43 regulatory axis. Journal of Prevention of Alzheimers Disease, 2022, 9: 158–171. https://doi.org/10.14283/jpad.2021.60

[64]

Gao, F. Y., Chen, X. F., Cui, L. X., et al. Gut microbiota mediates the pharmacokinetics of Zhi-Zi-Chi decoction for the personalized treatment of depression. Journal of Ethnopharmacology, 2023, 302: 115934. https://doi.org/10.1016/j.jep

[65]

Wang, Q. S., Tian, J. S., Cui, Y. L., et al. Genipin is active via modulating monoaminergic transmission and levels of brain-derived neurotrophic factor (BDNF) in rat model of depression. Neuroscience, 2014, 275: 365–373. https://doi.org/10.1016/j.neuroscience.2014.06.032

[66]

Li, Y., Li, L., Hölscher, C. Therapeutic potential of genipin in central neurodegenerative diseases. CNS Drugs, 2016, 30: 889–897. https://doi.org/10.1007/s40263-016-0369-9

[67]

Ramos, R. R., Gonçalo, M. P., Daniela, M. Genipin prevents alpha-synuclein aggrega tion and toxicity by affecting endocytosis, metabolism and lipid storage. Nature Communictions, 2023, 14: 1918. https://doi.org/10.1038/s41467-023-37561-2

[68]

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

[69]

Chang, W. L., Wang, H. Y., Shi, L. S., et al. Immunosuppressive iridoids from the fruits of gardeniajasminoides. Journal of Natural Products, 2005, 68: 1683–1685. https://doi.org/10.1021/np0580816

[70]

Zhong, A. J., Li, G. Screening of active ingredients in Rehmannia glutinosa and its mechanism of action in the treatment of liver diseases. Indian Journal of Pharmaceutical Sciences, 2023, 85: 445–455. https://doi.org/10.36468/pharmaceutical-sciences.1110

[71]

Fan, H., Li, T. F., Gong, N., et al. Shanzhiside methylester, the principle effective iridoid glycoside from the analgesic herb Lamiophlomis rotata, reduces neuropathic pain by stimulating spinal microglial β-endorphin expression. Neuropharmacology, 2016, 101: 98–109. https://doi.org/10.1016/j.neuropharm.2015.09.010

[72]

Yenigun, S., Basar, Y., Ipek, Y., et al. Comprehensive evaluation of Ixoroside: An iridoid glycoside from Nepeta aristata and N. baytopii, assessing antioxidant, antimicrobial, enzyme inhibitory, DNA protective properties, with computational and pharmacokinetic analyses. Journal of Biologically Active Products from Nature, 2024, 14: 286–315. https://doi.org/10.1080/22311866.2024.2358785

[73]
Ortiz de Urbina, A., Martín, M., Fernández, B., et al. In vitro antispasmodic activity of peracetylated penstemonoside, aucubin and catalpol. Planta Medica, 1994 , 60: 512–515. https://doi.org/10.1055/s-2006-959561
[74]

Zhou, J., Li, S. P., Feng, K. X., et al. Impact of gardenoside on insulin receptor and nuclear factor kappa B of insulin resistant in HepG2 cells. The Chinese Journal of Clinical Pharmacology, 2015, 31: 362–365. https://doi.org/10.13699/j.cnki.1001-6821.2015.05.013

[75]

Song, P., Shen, D. F., Meng, Y. Y., et al. Geniposide protects against sepsis-induced myocardial dysfunction through AMPK α-dependent pathway. Free Radical Biology and Medicine, 2020, 152: 186–196. https://doi.org/10.1016/j.freeradbiomed.2020.02.011

[76]

Fu, Y., Yuan, P., Cao, Y., et al. Geniposide in Gardenia jasminoides var. radicans Makino modulates blood pressure via inhibiting WNK pathway mediated by the estrogen receptors. Journal of Pharmacy and Pharmacology, 2020, 72: 1956–1969. https://doi.org/10.1111/jphp.13361

[77]

Zhang, Y., Ding, Y., Zhong, X., et al. Geniposide acutely stimulates insulin secretion in pancreatic β-cells by regulating GLP-1 receptor/cAMP signaling and ion channels. Molecular and Cellular Endocrinology, 2016, 430: 89–96. https://doi.org/10.1016/j.mce.2016.04.020

[78]

Kwak, J. H., Lee, D. U. Structure–antiamnesic activity relationship of iridoid glycosides from gardenia fruits. Chemistry Letters, 2015, 44: 837–839. https://doi.org/10.1246/cl.150172

[79]
Huang, A. G., Chen, C., Liu, T. Q., et al. scFv antibody-mediated targeted drug delivery system improves the antiviral activity of geniposidic acid against WSSV. Aquaculture, 2022 , 560: 738496. https://doi.org/10.1016/j.aquaculture
[80]

Nakamura, K., Hosoo, S., Yamaguchi, S., et al. Geniposidic acid upregulates atrial natriuretic peptide secretion and lowers blood pressure in spontaneously hypertensive rats. Journal of Functional Foods, 2018, 40: 634–638. https://doi.org/10.1016/j.jff.2017.10.037

[81]

Huang, S. M., Lin, S. Y., Chen, M. K., et al. Effects of geniposide and geniposidic acid on fluoxetine-induced muscle atrophy in C2C12 cells. Processes, 2021, 9: 1649. https://doi.org/10.3390/pr9091649

[82]
Sekandi, P., Namukobe, J., Byamukama, R., et al., Antimicrobial, antioxidant, and sun protection potential of the isolated compounds from Spermacoce princeae (K. Schum). BMC Complementary Medicine and Therapies, 2023 , 23. https://doi.org/10.1186/s12906-023-04026-4
[83]

L. Li, J. Zou, Q. Xia, et al. Anti-TMV and insecticidal potential of four iridoid glycosides from Gardenia jasminoides fruit. Chemical Research in Chinese Universities, 2018, 34: 697–699. https://doi.org/10.1007/s40242-018-8197-8

[84]

Kim, D. H., Lee, H. J., Kim, Y. J., et al. Iridoid glycosides isolated from Oldenlandia diffusa inhibit LDL-oxidation. Archives of Pharmacal Research, 2005, 28: 1156–1160. https://doi.org/10.1007/BF02972979

[85]

Akihisa, T., Watanabe, K., Yamamoto, A., et al. Melanogenesis inhibitory activity of monoterpene glycosides from Gardeniae Fructus. Chemistry & Biodiversity, 2012, 9: 1490–1499. https://doi.org/10.1002/cbdv.201200030

[86]

Hirata, T., Kobayashi, T., Wada, A., et al. Anti-obesity compounds in green leaves of Eucommia ulmoides. Bioorganic & Medicinal Chemistry Letters, 2011, 21: 1786–1791. https://doi.org/10.1016/j.bmcl.2011.01.060

[87]
Nakamura, A., Yokoyama, Y., Tanaka, K., et al. Asperuloside improves obesity and type 2 diabetes through modulation of gut microbiota and metabolic signaling. Science, 2020 , 23: 101522. https://doi.org/10.1016/j.isci.2020.101522
[88]

Li, L., Zou, J., You, S., et al. Natural product cerbinal and its analogues Cyclopenta pyridines: synthesis and discovery as novel pest control agents. Journal of Agricultural and Food Chemistry, 2019, 18: 10498–10504. https://doi.org/10.1021/acs.jafc.9b03699

Food & Medicine Homology
Cite this article:
Zhang Y-D, Wang M-H, Guan M, et al. Critical Review for active iridoids in Gardenia jasminoides J.Ellis as a plant of food and medicine homology. Food & Medicine Homology, 2025, https://doi.org/10.26599/FMH.2025.9420030

269

Views

66

Downloads

0

Crossref

Altmetrics

Received: 25 April 2024
Revised: 01 June 2024
Accepted: 29 June 2024
Published: 09 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