<<

Available online at www.sciencedirect.com

Chinese Journal of Natural Medicines 2020, 18(7): 526-535 doi: 10.1016/S1875-5364(20)30063-7

•Review•

Advances on hormone-like activity of and ginsenosides

TIAN Mei, LI Lin-Nan, ZHENG Rui-Rong, YANG Li, WANG Zheng-Tao*

The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China Available online 20 Jul., 2020

[ABSTRACT] Traditional Chinese medicine (TCM) has been paid much attentions due to the prevention and treatment of hor- mone disorders. Ginseng, the root of C. A. Meyer (), is one of the most valuable herbs in complementary and alternative medicines around the world. A series of triterpenoid , also known as phytosteroids, were reported as the primary ingredients of Ginseng, and indicated broad spectral pharmacological actions, including anti-cancer, anti-inflammation and anti-fatigue. The skeletons of the dammarane triterpenoid aglycone are structurally similar to the steroid hormones. Both in vitro and in vivo studies showed that Ginseng and its active ingredients have beneficial hormone-like role in hormonal disorders. This review thus summarizes the structural similarities between hormones and dammarane ginsenosides and integrates the analogous effect of Ginseng and ginsenosides on prevention and treatment of hormonal disorders published in recent twenty years (1998–2018). The review may provide convenience for anticipate structure-function relationship between saponins structure and hormone-like effect. [KEY WORDS] Panax ginseng; steroid hormones; triterpenoid saponins; hormone-like activity; molecular signaling [CLC Number] R965 [Document code] A [Article ID] 2095-6975(2020)07-0526-10

Introduction verse effects, a large number of researchers are concentrating on using available and alternative traditional Chinese medi- Steroid hormones play an indispensable role in regulat- cine (TCM). Especially, -derived natural products which ing physical and emotional growth, sex differentiation, repro- are similar to synthetic hormones are applied to prevent endo- duction, immune functions, protein synthesis and metabo- [9-13] crine disorder and treat disease . [1] lism . Generally, the steroid hormones can be grouped into Panax ginseng C. A. Meyer, is a perennial plant belongs four main classes by their structural characteristics: progesto- to the genus Panax of Araliaceae family. It is widely used for [2] gens, corticosteroids, androgens and estrogens (Fig. 1A) . curing miscellaneous diseases all over the world. As early as The clinical practice showed that the imbalance of hormone 4000 years ago, P. ginseng was acquainted as “The king of level would directly or indirectly lead to the occurrence of the grass bouquet” by means of nourishing weakness, replen- [3, 4] many physiological and psychological diseases . With the ishing energy, supporting body balance and consolidating ro- increasing environmental pollution and social pressure, the bustness [14, 15]. Nowadays, there are plenty of herbal prepara- disorder of endocrine system is becoming more complicated tions including tablets, capsules, granules that containing gin- [5, 6] and serious . Many diseases caused by hormone imbal- senosides [16, 17]. Triterpenoid saponins are the principal com- ance were treated with synthetic hormones, nevertheless, pro- ponents in Panax species, which refer to a range of dam- longed intake of these artificial drugs might lead to severe ad- marane- or oleanane-type triterpenoid ginsenosides [18-21]. verse effects [7, 8]. Therefore, due to lower cost and minor ad- Ginsenoside Ro belongs to oleanane-type (OA) saponins which has a five-link trans ring [22]. Dammarane-type ginsen- [Received on] 20-Dec.-2019 osides share a four-ring hydrophobic steroid-like structure [Research funding] This work was supported by the National Natur- skeleton, and can be divided into two functional groups based al Science Foundation of China (Nos. 81530096, 81920108033, on their preferred glycosylation sites at C6 position: proto- 81573581) and the Shanghai Sailing Program (No. 19YF 1448800). * panaxadiol (PPD), such as ginsenoside Rb1, Rg3; protopan- [ Corresponding author] Tel: 021-51322507, E-mail: ztwang@shu- [18, 23] tcm.edu.cn axatriol (PPT) such as ginsenoside Rg1, Rh1 (Fig. 2) .

These authors have no conflict of interest to declare. Triterpenoid saponins are considered to be the main pharma-

– 526 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535

A O O OH O O SCC 3β-HSD1/2 CYP21/CYP11β2 HO H H H H H HH H H H H H HO HO O Cholesterol Progesterone O Aldosterone

OH OH CYP19 H H

H H H H HO O Testosterone Estradiol

B 21 HO 20 22 OH 24 20 17 23 25 27 17 2324 11 25 26 6 3 HO 3 OH Steroid scaffold Dammarane ginsenoside skeleton Fig. 1 (A) The main steroidal structure of the four categories of hormones from cholesterol. The steroid hormones are divided into the progestogens, corticosteroids, androgens and estrogens are shown in purple, green, blue and red respectively. (B) Car- bon numbering of a typical steroid scaffold and common functionalized positions (C3, 11, 17, 20, and 21); Carbon numbering of dammarane ginsenoside skeleton, the common functionalized positions at C17, 23, 24 and 25, the typical glycosylation group sites at C3, 6, 20 for these aglycone are labeled in green

R2 OH 20

R2 28

3 6 R1

R1 3 R3

Dammarane ginsenosides Oleanane ginsenosides

Fig. 2 Classification and chemical structure of dammarane ginsenosides and oleanane ginsenosides. PPD, Protopanaxadiol; PPT, Protopanaxatriol; Glc, β-D-glucopyranosyl; GlcA, β-D-glucuropyronosyl; OA, Oleanane acid

cological bioactive ingredients. They can boost immunity idant, cardiovascular system protection, anti-inflammatory, from diseases and enhance physical functions, such as antiox- and anti-tumor [24-28]. More to the point, the study also showed

– 527 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535 that ginsenosides act as an estrogen analogue by activating C17, 23, 24 and 25), and glycosyl (Glc, Ara, etc.) group at estrogen receptor (ER) [29, 30]. C3, 6, 20 (Fig. 1B) [46]. In terms of the molecular structure, In the last decades, in vitro and in vivo results have re- ginsenosides is analogous to steroid hormones, and it would vealed that P. ginseng and its active compounds are equipped provide a solid structural basis for the hormone-like effect of with hormone analogous effects. Many studies have demon- dammarane ginsenosides. strated that dammarane ginsenosides possess greater poten- Hormone-like Activity of P. ginseng and Ginsen- tial in hormone-like effects [31, 32]. Therefore, to gather all of osides the literature relevant to hormone-like activities of P. gin- seng and ginsenosides is of great importance. The objective In vitro study on the hormone-like activity of Ginseng and of this review is to provide valuable research results on the ginsenosides roles and mechanisms of the hormone-like activities of P. A few studies have demonstrated that the hormone-like ginseng and ginsenosides between 1998 and 2018 from the effects of P. ginseng and ginsenosides in hormone-associated Web of Science, ScienceDirect and GeenMedical databases. cells. In oestrogen receptor-positive cells, such as human And it may provide insight into opportunity for anticipating breast cancer MCF-7 cells, often used as the preferred cell structure-function relationship of saponins in the activity of line for estrogen assay, exhibited cells proliferation distinctly hormone-like. after treatment with P. ginseng water extract and ginsenoside Rg1 [47]. In glucocorticoid receptor-related cells, like murine Structure of Endogenous Steroid Hormones and macrophagic RAW264.7 cells, ginsenoside Rg1 could en- Dammarane Ginsenosides hance the effect of anti-inflammatory by up-regulating gluco- The structure characterization of endocrine steroid hor- corticoid receptor (GR) expression [48]. These may indicate mones and triterpenoid saponins lead to a better understand- that P. ginseng and ginsenosides regulate the production, con- ing of their semblable biological activities [33]. Eighteen ster- version and of hormones in different organs and oid hormones are derived from cleavage of the cholesterol cells. side chain by cytochrome P450 cholesterol side-chain cleav- In vivo study on the hormone-like activity of ginseng and gin- age enzyme (SCC) [34, 35]. Cholesterol is converted into pro- senosides gestogens in mammals by SCC, corticosteroids and andro- Except extracorporeal exploration, internal experiments gens are bioconverted from progestogens by enzyme CYP21 also verified the analogous steroid hormone effects of P. gin- and CYP17, respectively. And then enzyme CYP19 turns the seng and ginsenosides in many explorations. In vivo studies, androgen into estrogen (Fig. 1A) [36, 37]. These enzymes play a surgical removal of sexual organs, drinks or drug-induced crucial role in transformation of one hormone into another in hormonal disorders and ageing rats were commonly used as line with indicating arrow [38, 39]. experiment model systems. In a general way, castrated rats or Visually from the structure of ginsenosides and hor- ovariectomized (OVX) rats have been adopted for simulating mones: 1) all share the same basic 17 carbon back- model of testosterone deficiency or estrogen decline in con- bone which are known as the cyclopentanophenanthrene ring. siderable literatures [49, 50]. Chronic consumption may Three six-carbon cyclohexane rings are designated A, B, and bring about the problem of nervous system and male infertil- C rings and the five-carbon cyclopentane ring is denoted as ity [51]. Cyclosporine A (CsA), which has been reported that the D ring. All of the A ring are lipid rings, except for oestro- the concentration in serum were negatively correlated with gens——the C18 steroids estranges, which A ring is aromat- vitality and motility of sperm [52]. Hence, Pan et al. [53] have ic [40]. The other three main groups of steroids of interest in adopted mice model of cyclosporine-induced damage of sper- clinical endocrinology consist of 21, 19 or 21 carbon atoms, matogenic apoptosis and testosterone synthesis to investigate representing the progestogen, androgen and corticosteroid the potential protective effects of decoction, which contain- skeleton [41, 42]. Similarly, the parent ring of PPD and PPT ing P. ginseng. In addition, using ageing-induced testoster- type ginsenosides have a steroid-like four-link trans ring, and one deficiency or estrogen decline rat model in a natural state the C17 side chains of ginsenosides are similar in structure to is also a good alternative method to perform an exploration, cholesterol [43, 44]. 2) In addition, most of steroids have angu- which could limit the risk of damage to experimental animals lar methyl group in C10 or C13. Similarly, the structure of a and in line with the provisions of the experimental animal comprises a sapogenin moiety and one or two glyc- protection laws [54, 55]. osyl units and/or chains, and the sapogenin has the same an- This is the fact that accumulating evidence have demon- gular methyl group in C10 or C13 as the parent nucleus of the strated P. ginseng or its saponins have a potent steroid hor- hormone. 3) Generally, steroid has shown to possess at least monal activity by evaluating physiological change compared one hydroxy (−OH) or ketone (=O) or methyl (-CH3) group at with placebo control. The regain of estrus cycle, improve- various carbon positions (typically C3, 11, 17, 20, and 21) of ment of target tissue function, reversal of the atrophy of the their scaffold [45]. Besides, there also have structures of gin- vagina and uterus could be regarded as the effective reflec- seng saponins whose genin has a hydroxyl (−OH) or methyl tions of P. ginseng and ginsenosides estrogen-like activity in

(−CH3) group or double bond at some carbon sites (generally OVX rats. Moreover, the balance of steroid hormone levels in

– 528 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535 serum or plasma is also the expression of effectiveness of dative stress kinase are also common index in functional or- ginsenosides, which were detected by radioimmunoassay gans [64-66]. According to the enzyme-immunoassay (EIA), (RIA) kit or ultra-performance liquid chromatography hy- ginsenoside Rg1 could protect tissue and cell from oxidative phenated with mass spectrometry (UPLC-MS/MS) after treat- stress damage by increasing the activities of superoxide dis- ments [56-58]. In the testosterone decline male rat model, the mutase (SOD) and catalase (CAT) and decreasing the level of number and vitality of fully mature sperm cells at the center malondialdehyde (MDA) [67, 68]. Additionally, observing of the tubules was limited; the number of cells lining the tu- change of rat sex organs weight, like epididymis, testis and bular membrane was decreased; and the tissue around the tu- seminal vesicles, were often considered as a simple and wide- bule was degraded, these symptoms all indicated that cells spread assessment [51, 69]. Table 1 displays the hormone-like was late maturation arrest or premature death in reproduc- activity of Ginseng and ginsenosides in detail. tive organ [59, 60]. Hence, the improvement of symptoms can Mechanism research on the hormone-like activity of Ginseng be analyzed to evaluate the androgen-like effect of P. gin- and ginsenosides seng and ginsenosides [61-63]. The level and activity of oxi- Many studies have shown that the molecular target path-

Table 1 Preclinical studies of Ginseng and ginsenosides for hormone-like activities compared with placebo control

Active substance Experimental Treatment Steroidal Dose (mg·kg−1) Treatment activities Ref. material model duration (weeks) activity P. ginseng water Nile tilapia Improved: testes and spleen somatic indexes; Oral, 400 12 Androgen [69] extract fingerlings plasma T level Korean red Ageing -induced Improved: body and testis weights; serum T ginseng water testicular Oral, 200 24 Androgen level; cell numbers; the process of [54] extract dysfunction rat spermatogenesis Korean red Improved: apoptosis of prostate epithelial T-inducedprostate ginseng water IP, 100 or 200 4 Androgen cells.Decreased: epithelial layer thickness; [117] hyperplasia rat extract prostate cells proliferation Improved: estrous stage; uterine weight index; P. ginseng water OVX rat Oral, 6000 8 Estrogen serum E2 level.Decreased: body weight; serum [50] extract LH level P. ginseng water Immature and Improved: serum E2 level; uterus and vagina Oral, 24000 4 Estrogen [118] extract OVX mice weights.Decreased: LH and FSH level P. ginseng water MCF-7 cells 0.1–100 μg·mL−1 - Estrogen Improved: cells Proliferation [118] extract Ginsenoside Rc Human sperm 1 and 10 μg·mL−1 - Androgen Improved: sperm motility; sperm progression [119] Improved: 5-HT concentration; HTP Ginsenoside Rb1 OVX mice IP, 10 1 Estrogen [120] accumulations.Decreased: MAO activity Improved: apoptosis of ectopic endometrial Allotransplantatio Ginsenoside Rg3 Oral, 10 4 Estrogen cells.Decreased: serum E2 level; volume and [120] n- induced EM rat height of ectopic endometrial lesions Improved: epididymis, testis and seminal vesicles weights; the count, viability, and Ethanol-induced Ginsenoside Rg1 Oral, 20 and 40 4 Androgen motility of testicular sperm; serum T level; [51] infertility rat SOD and CAT activities.Decreased: MDA levels Gonadectomized Improved: sleep disruption; AR protein rat level.Decreased: serum CORT level Androgen; Ginsenoside Rg1 Chronic Oral, 20 4 [122] corticosteroid Improved: serum T level; GR protein unpredictable level.Decreased: serum CORT level stress rat ALD-induced Improved: T-SOD activity. Decreased: Ginsenoside Rg1 MPC5 mouse 0.08 μg·mL−1 - Corticosteroid oxidative stress; autophagy; MDA level; ROS [123] podocyte cells production Protopanaxatriol ACTH-stimulated Decreased: corticosteroid generation; the saponins and bovine adrenal 9.53 μg·mL−1 - Corticosteroid [113] conversion from cholesterol to pregnenolone metabolites fasciculata cells T, testosterone; IP, Intraperitoneal; E2, estradiol; LH, luteinizing hormone; LHR, luteinizing hormone receptor; FSH, follicle-stimulating hormone; CORT, corticosterone; HPA axis, The hypothalamic-pituitary-adrenal axis; HPG axis, The hypothalamus- pituitary-gonadal axis; VEGF, Vascular endothelial growth factor; 5-HT, 5-hydroxytryptamine; HTP, tryptophan hydroxylase; MAO, monoamine oxidase; EM, endometriosis; ALD, Aldosterone; ROS, reactive oxygen species

– 529 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535 way of P. ginseng and ginsenosides effect on steroid hor- proving the activities of related hormone enzymes [51, 84-86]. mones by binding or activating intracellular nuclear, Simultaneously, minor ginsenosides has been shown to re- or cell membrane hormone receptors, such as ER, androgen lieve a high does synthetic hormones-induced sexual dys- receptor (AR), GR, either directly or indirectly, and sub- function in rats through classical signaling pathways [87]. sequently regulating the associated hormone levels [70-74]. Female steroid hormones consist of estrogens secreted Saponins regulate intracellular hormone balance by non-ge- from the ovaries in the body. The cellular response to estro- netic pathways of intracellular signaling molecules [75, 76]. On gen is mediated by the ERα and ERβ isoforms, estrogen re- the other hand, ginsenosides also could active the expression ceptors α and β were differentially exhibited and expressed in of related invertases like endogenous antioxidant enzymes by different tissue distribution patterns. ERβ is mainly ex- regulating intracellular target proteins or genes [77-80]. In many pressed in the ovaries, whereas, ERα is voiced in many or- cases, ginsenosides can cross-talk with multiple signaling gans, like breast, kidney, bone, uterus, ovaries (where ERα pathway at the same time, from enzyme level, protein level to upregulation was stronger than that of ERβ) [67, 88-90]. There is gene level, which directly or indirectly play an important role evidence that ginsenosides Rg1 exert estrogen-like effect via in preventing and treating diseases caused by hormone dis- the activation of ERα, whereas ginsenoside Rb1 have the orders [81]. ability to active ERα and ERβ [81, 91-93]. Ginsenosides not only Androgens play their biological roles predominately in have sex hormone-like activity, but also bind GR to produce binding to the AR whether directly or indirectly [82, 83]. By corticosteroid-like effects. Ginsenosides has been observed to binding to receptors, P. ginseng and ginsenosides have abil- promote the upregulation of cytoplasmic and nuclear GR pro- ity to deeply regulate genes level of hormone metabolism in tein level in the rat liver and lipopolysaccharide (LPS)-stimu- the nucleus [80]. And on the membranes of mitochondria, P. lated RAW264.7 macrophage cells [94]. Table 2 displays the ginseng and ginsenosides play an advantageous role in im- possible pathway of the regulation mechanisms of action of

Table 2 Pathway of the regulation mechanisms of hormone-like effect of Ginseng and ginsenosides Signaling molecules Active substance material Observations Steroidal activity Experimental model Ref. monitored Korean red ginseng Steroids metabolism Androgen; Estrogen Aged male rat testes CYP11a1 [80] water extract improvement and Corticosteroid Korean red ginseng AR transcription suppression Androgen LNCaP cells Kallikrein-3 Mrna, AR [117] water extract Korean red ginseng Spermatogenesis Androgen Aged male rat Inhibin-α, nectin-2, CREB-1 [124] water extract Ginsenoside Ro 5α-reductase inhibitory Androgen Androgenetic alopecia rat 5α reductase [125] Hydrocortisone-induced Ginsenosides Rh4 and Rg5 Endothelial NO production Androgen NO/cGMP/PKGI [87] male mice 20(S)-protopanaxadiol AR protein level diminution Androgen C4-2 xenograft tumors AR [126]

P. ginseng water extract Estrogen-like activity Estrogen OVX mice ERα, ERβ [118] P. ginseng water extract Estrogen-like activity Estrogen Female immature mice ERα, ERβ [127] Ginsenoside Rb1 Estrogen-like activity Estrogen MCF-7 cells ER [128] Endometriosis-associated Allotransplantation- induced Ginsenoside Rg3 Estrogen PI3K-Akt-mTOR [121] angiogenesis rat Ginsenoside Rh1 Estrogen-like activity Estrogen MCF-7 cells ER [129] Ginsenoside Rg1 Estrogen-like activity Estrogen MCF-7 cells ERE-luciferase [47] Ginsenoside Rg1 Estrogen-like activity Estrogen MCF-7 cells pS2 gene expression, ERα [81] Ginsenosides Glucocorticoid-like activity Corticosteroid Hydrocortisone-induced rat GR, TAT, mRNA [94] GR translocation Ginsenoside Rg6 Corticosteroid Human cancer cells GR [130] suppression GR transcription Ginsenoside Rh1 Corticosteroid CIA mice NF-Κb, DUSP1 [131] improvement Ginsenoside Rg1 Glucocorticoid-like activity Corticosteroid RAW 264.7 cells eNOS, TNF-α, GR [48] Human umbilical vein Ginsenoside Rg1 GR activation Corticosteroid GR [132] endothelial cells PI3K/Akt/mTOR, phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin; ERE, estrogen response element; CIA, collagen-induced arthritis; NF-κB, nuclear factor kappa B; DUSP1, dual specificity protein phosphatase 1; eNOS, endothelial nitric oxide synthase; NO, Nitric oxide

– 530 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535

P. ginseng and ginsenosides. The summarized pathway of the metabolism of ginsenosides revealed that ginsenosides were regulation mechanisms of representative ginsenosides effect transformed via multiple pathways including deglycosylation, on the steroid hormones are shown in Fig. 3. dehydration, oxygenation and hydration both in vitro and in vivo, these transformed ginsenosides further contribute to the Conclusion and Future Perspectives chemical diversity of minor ginsenosides [99]. For example, TCM have attracted considerable concern from the pub- under the conditions of artificial gastric juice, the deglyc- lic, medical community, and governmental agencies, the osylated and dehydrated ginsenosides possessed slightly in- search for lower side effects with a similar endogenous hor- creased than primary ginsenosides [100]. And in vivo studies, mones effect of steroidal constituents like phytohormones in- there have reported that the moieties could de- stead of synthetic hormones has been in full swing [95-97]. In grade ginsenosides via cleavage of the sugar [101-104]. The de- this review, from the perspective of structure-activity rela- graded minor ginsenosides have been proven to traverse tionship, dammarane ginsenosides possess cholesterol-like membrane more efficient than the intact ginsenosides, and four connected trans-ring steroid skeleton with various free or further yield higher bioavailability in cells [104, 105]. Therefore, sugar-bound hydroxyl groups. Meanwhile, there are similarit- lower polar ginsenosides might have great potential to be ab- ies between ligand of saponins and hormones, all the more so sorbed into circulation system [106-109]. For instance, ginsenos- the degraded minor ginsenosides, such as ginsenoside Rg5, ide Rg3, Rg5, Rh1 has been demonstrated to exhibit remark- 25-OH-PPD [98]. Internal studies have manifested that the gin- able anticancer and antioxidant pharmacological activities seng water extract, or individual ginsenosides have activity of than polar ginsenosides [110-112]. Thus, we would be correct to analogous endogenous hormone in mammals. Different from assume that the rare minor ginsenosides may play a more ef- fresh P. ginseng, the main ingredients of Korean Red gin- fective role in keeping regular of hormonal level after treat- seng are the minor ginsenosides. And according to the results, ment with P. ginseng saponins. the minor ginsenosides may have stronger activities than the Current studies speculate that sugar linkage at C-6 was native ginsenosides [87]. Further, ginsenosides could bind and more active than linkage at C-3, total number of sugar mo- activate nuclear hormone receptors to maintain a healthy lecules within a ginsenoside indicated the negative correla- level of physiology in target tissues or cells. In addition, the tion with steroid hormones effect, elimination of glycosyl moderating effect of dammarane saponins on the expression bonds may be consistent with their steroid hormone-like of some invertase proteins also promotes the balanced trans- effects, the steamed red ginseng better than untreated gin- formation and metabolism of hormones. seng [113, 114]. However, whether stereoselectivity, C17 side- To expand structure–activity relationship between gin- chain variation or location of hydroxyl group have a signific- senosides and hormones, thus attention also should be paid to ant impact on the biological activity of ginsenosides still need the structure of ginsenosides after metabolism. Studies on to be further investigated, whether oleanane type saponins

Rb1 PPD Rg1 Rg3

Rg1 Membrane Rb1

P13K Rg3 Rg1 ERα P13K P ERα Akt P Rb1 PPD eNOS ERβ Akt Rg1 AR GR L-Arginine NO MEK mTOR

Regulation Regulation of Nucleus of gene transcription

Fig. 3 A summarized pathway of the regulation mechanisms of representative ginsenosides effect on the steroid hormones via genes and transcriptions pathways. PI3K, phosphatidylinositol 3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; eNOS, endothelial nitric oxide synthase; NO, Nitric oxide; MEK, mitogen-activated protein kinase kinase

– 531 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535 like ginsenoside Ro are more active than dammarane type and chemical composition [J]. J Ginseng Res, 2015, 39(4): still need to be further explored. In addition, many diseases 384-391. are linked to endogenous steroid hormones, such as depres- [15] Park H, Kim D, Park S, et al. Ginseng in traditional herbal prescriptions [J]. J Ginseng Res, 2012, 36(3): 225-241. sion and menopause [115, 116]. The possible related mechan- [16] Shi X, Yang W, Qiu S, et al. An in-source multiple collision- isms by which ginsenosides act may differ, the mechanism of neutral loss filtering based nontargeted metabolomics ap- different types of ginseng corresponding to its activity also proach for the comprehensive analysis of malonyl-ginsenos- deserves further research. ides from Panax ginseng, P. quinquefolius, and P. notogin- seng [J]. Anal Chim Acta, 2017, 952: 59-70.

References [17] Jia L, Zhao Y. Current evaluation of the millennium phytomedicine--ginseng (I): etymology, pharmacognosy,

[1] Caron P, Turcotte V, Guillemette C. A chromatography/tan- phytochemistry, market and regulations [J]. Curr Med Chem, dem mass spectrometry method for the simultaneous profiling 2009, 16(19): 2475-2484. of ten endogenous steroids, including progesterone, adrenal

[18] Liu J, Wang Y, Qiu L, et al. Saponins of Panax notoginseng: precursors, androgens and estrogens, using low serum chemistry, cellular targets and therapeutic opportunities in car- volume [J]. Steroids, 2015, 104: 16-24. diovascular diseases [J]. Expert Opin Inv Drug, 2014, 23(4):

[2] Abdel-Khalik J, Björklund E, Hansen M. Simultaneous de- 523-539. termination of endogenous steroid hormones in human and an-

[19] Dean M, Murphy BT, Burdette JE. Phytosteroids beyond es- imal plasma and serum by liquid or gas chromatography trogens: regulators of reproductive and endocrine function in coupled to tandem mass spectrometry [J]. J Chromatogr B, natural products [J]. Mol Cell Endocrinol, 2017, 442: 98-105. 2013, 928: 58-77.

[20] Lee J, Yoon S, Lee S, et al. Ginsenoside Rg3 ameliorated

[3] Rossi C, Calton L, Hammond G, et al. Serum steroid profil- HFD-induced hepatic steatosis through downregulation of ing for Congenital Adrenal Hyperplasia using liquid chroma- STAT5-PPARγ [J]. J Endocrinol, 2017, 235(3): 223-235. tography–tandem mass spectrometry [J]. Clin Chim Acta,

[21] Zhang Y, Han L, Sakah K, et al. Bioactive Protopanaxatriol 2010, 411(3−4): 222-228. type saponins isolated from the roots of Panax Notoginseng

[4] Stolze BR, Gounden V, Gu J, et al. An improved micro-meth- (Burk.) F. H. Chen [J]. Molecules, 2013, 18(9): 10352-10366. od for the measurement of steroid profiles by APPI-

[22] Cheng Y, Liang Q, Hu P, et al. Combination of normal-phase LC–MS/MS and its use in assessing diurnal effects on steroid medium-pressure liquid chromatography and high-perform- concentrations and optimizing the diagnosis and treatment of adrenal insufficiency and CAH [J]. J Steroid Biochem, 2016, ance counter-current chromatography for preparation of gin- 162: 110-116. senoside-Ro from Panax ginseng with high recovery and effi- ciency [J]. Sep Purif Technol, 2010, 73(3): 397-402.

[5] Meng Q, Ren A, Zhang L, et al. Incidence of infertility and risk factors of impaired fecundity among newly married [23] Yang W, Hu Y, Wu W, et al. Saponins in the genus Panax L. couples in a Chinese population [J]. Reprod Biomed Online, (Araliaceae): a systematic review of their chemical 2015, 30(1): 92-100. diversity [J]. Phytochemistry, 2014, 106: 7-24.

[24] Xin X, Zhong J, Wei D, et al. Protection effect of 20(S)-gin- [6] Flanagan SD, DuPont WH, Caldwell LK, et al. The effects of a korean ginseng, GINST15, on Hypo-Pituitary-Adrenal and senoside Rg2 extracted from cultured Panax notoginseng cells oxidative activity induced by intense work stress [J]. J Med on hydrogen peroxide-induced cytotoxity of human umbilical Food, 2018, 21(1): 104-112. cord vein endothelial cells in vitro [J]. Process Biochem, 2005, 40(10): 3202-3205. [7] Akomolafe SF, Oboh G, Akindahunsi AA, et al. Ethanol-in-

duced male infertility: effects of aqueous leaf extract of Tetra- [25] Kim JH, Yi Y, Kim M, et al. Role of ginsenosides, the main carpidium conophorum [J]. Andrologia, 2017, 49(10): active components of Panax ginseng, in inflammatory re- e12759. sponses and diseases [J]. J Ginseng Res, 2017, 41(4): 435- 443. [8] Borst SE, Yarrow JF. Injection of testosterone may be safer

and more effective than transdermal administration for com- [26] Lee CH, Kim J. A review on the medicinal potentials of gin- bating loss of muscle and bone in older men [J]. Am J Physiol- seng and ginsenosides on cardiovascular diseases [J]. J Gin- Endoc M, 2015, 308(12): E1035-E1042. seng Res, 2014, 38(3): 161-166.

[27] Wang XD, Sun YY, Zhao C, et al. 12-Chloracetyl-PPD, a [9] Chan S. Panax ginseng, Rhodiola rosea and Schisandra chinensis [J]. Int J Food Sci Nutr, 2012, 63(sup1): 75-81. novel dammarane derivative, shows anti-cancer activity via delay the progression of cell cycle G2/M phase and reactive [10] Jiang D, Coscione A, Li L, et al. Effect of Chinese herbal medicine on male infertility [J]. Int Rev Neurobiol, 2017, 135: oxygen species-mediate cell apoptosis [J]. Eur J Pharmacol, 297-311. 2017, 798: 49-56.

[11] Panossian A, Wikman G, Sarris J. Rosenroot (Rhodiola [28] Park JD, Rhee DK, Lee YH. Biological activities and chem- rosea): traditional use, chemical composition, istry of saponins from Panax ginseng C. A. Meyer [J]. Phyto- and clinical efficacy [J]. Phytomedicine, 2010, 17(7): 481- chem Rev, 2005, 4(2−3): 159-175.

493. [29] Chan RY, Chen WF, Dong A, et al. Estrogen-like activity of

[12] Schloms L, Storbeck K, Swart P, et al. The influence of As- ginsenoside Rg1 derived from Panax notoginseng [J]. J Clin palathus linearis (Rooibos) and dihydrochalcones on adrenal Endocr & Metab, 2002, 87(8): 3691-5.

steroidogenesis: quantification of steroid intermediates and [30] Amato P, Christophe S, Mellon PL. Estrogenic activity of end products in H295R cells [J]. J Steroid Biochem, 2012, herbs commonly used as remedies for menopausal 128(3−5): 128-138. symptoms [J]. Menopause, 2002, 9(2): 145-150.

[13] Yao DF, Mills JN. Male infertility: lifestyle factors and holist- [31] Jang D, Lee MS, Shin B, et al. Red ginseng for treating ic, complementary, and alternative therapies [J]. Asian J An- erectile dysfunction: a systematic review [J]. Brit J Clin Phar- drol, 2016, 18(3): 410-418. maco, 2008, 66(4): 444-450.

[14] Lee SM, Bae B, Park H, et al. Characterization of Korean Red [32] Sun S, Wang C, Tong R, et al. Effects of steaming the root of Ginseng (Panax ginseng Meyer): history, preparation method, Panax notoginseng on chemical composition and anticancer

– 532 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535

activities [J]. Food Chem, 2010, 118(2): 307-314. [53] Pan X, Wang X, Wang X, et al. Protective effects of new

[33] Shahidi NT. A review of the chemistry, biological action, and Wenshen Shengjing Decoction on cyclosporine-induced clinical applications of anabolic-androgenic steroids [J]. Clin impairment of testosterone synthesis and spermatogenic apop- Ther, 2001, 23(9): 1355-1390. tosis [J]. Exp Ther Med, 2017, 15: 813-821.

[34] Mellon SH, Griffin LD. Neurosteroids: Biochemistry and [54] Kopalli SR, Cha K, Ryu J, et al. Specific activity of Korean clinical significance [C]. Elsevier Ltd., 2002: 35−43. red ginseng saponin and non-saponin fractions in ageing-in-

[35] Schumacher M, Guennoun R, Mattern C, et al. Analytical duced rat testicular dysfunction [J]. J Funct Foods, 2017, 29: challenges for measuring steroid responses to stress, neurode- 226-237.

generation and injury in the [J]. Ster- [55] Blanchard TL, Varner DD, Love CC, et al. Management op- oids, 2015, 103: 42-57. tions for the aged breeding stallion with declining testicular

[36] Gracia T, Jones PD, Higley EB, et al. Modulation of steroido- function [J]. J Equine Vet Sci, 2012, 32(8): 430-435.

genesis by coastal waters and sewage effluents of Hong Kong, [56] Irwin S, Kenny AP, O'Halloran J, et al. Adaptation and valid- China, using the H295R assay [J]. Environ Scid Poll R, 2008, ation of a radioimmunoassay kit for measuring plasma cortisol 15(4): 332-343. in turbot [J]. Comp Biochem Physiol C , 1999, 124(1): 27-31.

[37] Krone CKSW. Steroid biochemistry [J]. Technological Ad- [57] Skenandore CS, Pineda A, Bahr JM, et al. Evaluation of a vances, 2014(27): 41-53. commercially available radioimmunoassay and enzyme im-

[38] Munkboel CH, Baake MLK, Styrishave B. Atorvastatin de- munoassay for the analysis of progesterone and estradiol and creases steroid production in H295R cells and in major endo- the comparison of two extraction efficiency methods [J]. crine tissues of male rats [J]. Arch Toxicol, 2018, 92(5): 1703- Domest Anim Endocrin, 2017, 60: 61-66.

1715. [58] Ke Y, Gonthier R, Simard J, et al. A validated LC–MS/MS

[39] Winther CS, Nielsen FK, Hansen M, et al. Corticosteroid pro- method for the sensitive quantitation of serum 7alpha hy- duction in H295R cells during exposure to 3 endocrine dis- droxy-, 7beta hydroxy- and 7keto-dehydroepiandrosterone us- rupters analyzed with LC-MS/MS [J]. Int J Toxicol, 2013, ing a novel derivatization reagent [J]. Steroids, 2016, 108: 32(3): 219-227. 112-117.

[40] Wudy SA, Schuler G, Sánchez-Guijo A, et al. The art of [59] Xiao CY, Wang YQ, Li JH, et al. Transformation, migration measuring steroids [J]. J Steroid Biochem, 2018, 179: 88-103. and outcome of residual bodies in the seminiferous tubules of

[41] Greaves RF, Jevalikar G, Hewitt JK, et al. A guide to under- the rat testis [J]. Andrologia, 2017, 49(10): e12786.

standing the steroid pathway: new insights and diagnostic im- [60] Sarıözkan S, Türk G, Çıkla Süzgün P, et al. Effect of etodolac plications [J]. Clin Biochem, 2014, 47(15): 5-15. hydrazone, a new compound synthesised from etodolac, on

[42] Miller WL, Auchus RJ. The molecular biology, biochemistry, spermatozoon quality, testicular lipid peroxidation, apoptosis and physiology of human steroidogenesis and its disorders [J]. and spermatozoon DNA integrity [J]. Andrologia, 2016, Endocr Rev, 2011, 32(1): 81-151. 48(2): 177-188.

[43] Cho C, Kim Y, Rhee YK, et al. Chemical composition charac- [61] Gray SL, Lackey BR, Boone WR. Effects of Panax ginseng, teristics of Korean straight ginseng products [J]. J Ethnic zearalenol, and estradiol on sperm function [J]. J Ginseng Res, Foods, 2014, 1(1): 24-28. 2016, 40(3): 251-259.

[44] Liu Q, Lv J, Xu M, et al. Dammarane-type saponins from [62] Won Y, Kim B, Shin Y, et al. Pectinase-treated Panax gin- steamed leaves of Panax notoginseng [J]. Natur Prod seng extract (GINST) rescues testicular dysfunction in aged Bioprosp, 2011, 1(3): 124-128. rats via redox-modulating proteins [J]. Exp Gerontol, 2014,

[45] Maltais R, Tremblay MR, Ciobanu LC, et al. Steroids and 53: 57-66.

combinatorial chemistry [J]. J Comb Chem, 2004, 6(4): 443- [63] Xia C, Chu S, Zhang S, et al. Ginsenoside Rg1 alleviates cor- 456. ticosterone-induced dysfunction of gap junctions in

[46] Shin B, Kwon SW, Park JH. Chemical diversity of ginseng astrocytes [J]. J Ethnopharmacol, 2017, 208: 207-213.

saponins from Panax ginseng [J]. J Ginseng Res, 2015, 39(4): [64] Turner TT, Lysiak JJ. Oxidative stress: a common factor in 287-298. testicular dysfunction [J]. J Andrology, 2008, 29(5): 488-498.

[47] Zhang X, Shi Y, Tang T, et al. Osteogenic differentiation of [65] Samie A, Sedaghat R, Baluchnejadmojarad T, et al. Hesper- mesenchymal stem cells under continuous cyclic mechanical etin, a citrus flavonoid, attenuates testicular damage in diabet- tension stimulation [J]. Bone, 2010, 47: S439. ic rats via inhibition of oxidative stress, inflammation, and ap-

[48] Song Y, Zhao F, Zhang L, et al. Ginsenoside Rg1 exerts syn- optosis [J]. Life Sci, 2018, 210: 132-139.

ergistic anti-inflammatory effects with low doses of glucocor- [66] Park S, Kim J, Lee DG, et al. Peroxiredoxin 2 deficiency ac- ticoids in vitro [J]. Fitoterapia, 2013, 91: 173-179. celerates age-related ovarian failure through the reactive oxy-

[49] Shin SS, Yoon M. Korean red ginseng (Panax ginseng) inhib- gen species-mediated JNK pathway in mice [J]. Free Radic its obesity and improves lipid metabolism in high fat diet-fed Biol Med, 2018, 123: 96-106.

castrated mice [J]. J Ethnopharmacol, 2018, 210: 80-87. [67] Haffner-Luntzer M, Kovtun A, Lackner I, et al. Estrogen re-

[50] He Lin ZLZP. Urinary metabolomic study of antagonistic ef- ceptor α- (ERα), but not ERβ-signaling, is crucially involved fect of Panax ginseng in rat with estrogen decline using ultra in mechanostimulation of bone fracture healing by whole- performance liquid chromatography coupled with quadrupole body vibration [J]. Bone, 2018, 110: 11-20.

time-of-flight mass spectrometry [J]. Food Funct, 2018, 3(9): [68] Liu X, Gu X, Yu M, et al. Effects of ginsenoside Rb1 on oxid- 1444-1453. ative stress injury in rat spinal cords by regulating the

[51] Zhenzhen Jin MZAY, Wang X. Protective Effect of ginsenos- eNOS/Nrf2/HO-1 signaling pathway [J]. Exp Ther Med, 2018, ide Rg1 against ethanol-induced male infertility in sprague- 16(2): 1079-1086.

dawley rats [J]. Int J Pharmacol, 2018(14): 513-521. [69] Mansour AT, Omar EA, Srour TM, et al. Effect of three nat-

[52] Zahra A, Gholamreza N, Farokhi F, et al. Attenuation of Cyc- ural phytochemicals supplementation on growth performance, losporine-Induced sperm impairment and embryotoxicity by testosterone level and feed utilization of Nile tilapia (Oreo- crataegus monogyna fruit aqueous extract [J]. Cell J, 2013, chromis niloticus) [J]. Aquacult Nutr, 2018, 24(1): 408-415.

15(3): 198-203. [70] Chung E, Lee KY, Lee YJ, et al. Ginsenoside Rg1 down-regu-

– 533 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535

lates glucocorticoid receptor and displays synergistic effects regulated distinct breast cancer cell progression through bind- with Camp [J]. Steroids, 1998, 63(7−8): 421-424. ing with estrogen receptor alpha or beta isoforms [J]. Environ

[71] Nah S. Ginseng ginsenoside pharmacology in the nervous sys- Pollut, 2018, 239: 300-307.

tem: involvement in the regulation of ion channels and recept- [89] Park J, Bui PTC, Song H, et al. Ginseng on nuclear hormone ors [J]. Front Physiol, 2014, 5: 98. receptors [J]. Am J Chin Med, 2017, 45(06): 1147-1156.

[72] Shen K, Leung SWS, Ji L, et al. Notoginsenoside Ft1 activ- [90] Zhong L, Zhou XL, Liu YS, et al. Estrogen receptor alpha ates both glucocorticoid and estrogen receptors to induce en- mediates the effects of notoginsenoside R1 on endotoxin-in- dothelium-dependent, nitric oxide-mediated relaxations in rat duced inflammatory and apoptotic responses in H9c2 cardi- mesenteric arteries [J]. Biochem Pharmacol, 2014, 88(1): 66- omyocytes [J]. Mol Med Rep, 2015, 12(1): 119-26.

74. [91] Cho J, Park W, Lee S, et al. Ginsenoside-Rb1 from Panax

[73] Wu J, Pan Z, Wang Z, et al. Ginsenoside Rg1 protection ginseng C.A. Meyer activates estrogen receptor-α and -β, in- against β-amyloid peptide-induced neuronal apoptosis via es- dependent of ligand binding [J]. J Clin Endocr Metab, 2004, trogen receptor α and glucocorticoid receptor-dependent anti- 89(7): 3510-3515.

protein nitration pathway [J]. Neuropharmacology, 2012, [92] Lau W, Chan RY, Guo D, et al. Ginsenoside Rg1 exerts estro- 63(3): 349-361. gen-like activities via ligand-independent activation of ERα

[74] Raheja S, Girdhar A, Lather V, et al. Biochanin a: a phytoes- pathway [J]. J Steroid Biochem, 2008, 108(1-2): 64-71.

trogen with therapeutic potential [J]. Trends Food Sci Tech, [93] Gao Q, Zhou L, Lee VH, et al. Ginsenoside Rg1 activates lig- 2018, 79: 55-66. and-independent estrogenic effects via rapid estrogen recept-

[75] Tamaoki J, Nakata J, Kawatani K, et al. Ginsenoside‐in- or signaling pathway [J]. J Ginseng Res, 2019, 43(4): 527- duced relaxation of human bronchial smooth muscle via re- 538.

lease of nitric oxide [J]. Brit J Pharmacol, 2000, 130(8): [94] Binbin C, Yinglu F, Juan D, et al. Upregulation effect of gin- 1859-1864. senosides on glucocorticoid receptor in rat liver [J]. Horm

[76] Zhang H, Zhou Q, Li X, et al. Ginsenoside Re promotes hu- Metab Res, 2009, 41(7): 531-6.

man sperm capacitation through nitric oxide-dependent path- [95] Patel S. Phytochemicals for taming agitated immune-endo- way [J]. Mol Reprod Dev, 2007, 74(4): 497-501. crine-neural axis [J]. Biomed Pharmacother, 2017, 91: 767-

[77] Li Q, Xiang Y, Chen Y, et al. Ginsenoside rg1 protects cardi- 775.

omyocytes against Hypoxia/Reoxygenation injury via activa- [96] Ma WG, Jia JM. The effects and prospects of the integration tion of Nrf2/HO-1 signaling and inhibition of JNK [J]. Cell of traditional Chinese medicine and Western medicine on an- Physiol Biochem, 2018, 44(1): 21-37. drology in China [J]. Asian J Androl, 2011, 13(4): 592-595.

[78] Li J, Gao Y, Chu S, et al. Nrf2 pathway activation contributes [97] Fung FY, Linn YC. Steroids in traditional Chinese medicine: to anti-fibrosis effects of ginsenoside Rg1 in a rat model of al- what is the evidence? [J]. Singap Med J, 2017, 58(3): 115- cohol-and CCI4-induced hepatic fibrosis [J]. Acta Pharmacol 120.

Sin, 2014, 35(8): 1031-1044. [98] Yuan W, Guo J, Wang X, et al. Non-protein amino acid deriv-

[79] Fan J, Liu D, He C, et al. Inhibiting adhesion events by Panax atives of 25-methoxylprotopanaxadiol/25-hydroxyprotopanax- notoginseng saponins and Ginsenoside Rb1 protecting arter- adioland their anti-tumour activity evaluation [J]. Steroids, ies via activation of Nrf2 and suppression of p38 – VCAM-1 2018, 129: 1-8.

signal pathway [J]. J Ethnopharmacol, 2016, 192: 423-430. [99] Mohanan P, Subramaniyam S, Mathiyalagan R, et al. Molecu-

[80] Kim I, Kim S, Kim E, et al. Korean red ginseng up-regulates lar signaling of ginsenosides Rb1, Rg1, and Rg3 and their C21-Steroid hormone metabolism via cyp11a1 gene in senes- mode of actions [J]. J Ginseng Res, 2018, 42(2): 123-132.

cent rat testes [J]. J Ginseng Res, 2011, 35(3): 272-282. [100] Wang J, Yau LF, Zhang R, et al. Transformation of ginsenos-

[81] Gao Q, Chan H, Man CW, et al. Differential ERα-mediated ides from notoginseng by artificial gastric juice can increase rapid estrogenic actions of ginsenoside Rg1 and estren in hu- cytotoxicity toward cancer cells [J]. J Agr Food Chem, 2014, man breast cancer MCF-7 cells [J]. J Steroid Biochem, 2014, 62(12): 2558-2573.

141: 104-112. [101] Kim D. Gut microbiota-mediated pharmacokinetics of gin-

[82] Santi D, Spaggiari G, Gilioli L, et al. Molecular basis of an- seng saponins [J]. J Ginseng Res, 2018, 42(3): 255-263.

drogen action on human sexual desire [J]. Mol Cell Endocrin- [102] Liu H, Yang J, Du F, et al. Absorption and disposition of gin- ol, 2018, 467: 31-41. senosides after oral administration of Panax notoginseng ex-

[83] Lee DK, Chang C. Molecular communication between andro- tract to rats [J]. Drug Metab Dispos, 2009, 37(12): 2290-2298.

gen receptor and general transcription machinery [J]. J Ster- [103] Dong W, Zhao J, Zhong F, et al. Biotransformation of Panax oid Biochem, 2003, 84(1): 41-49. ginseng extract by rat intestinal microflora: identification and

[84] Saadeldin IM, Hussein MA, Suleiman AH, et al. Ameliorat- quantification of metabolites using liquid chromatography- ive effect of ginseng extract on phthalate and bisphenol a re- tandem mass spectrometry [J]. J Ginseng Res, 2017, 41(4): protoxicity during pregnancy in rats [J]. Environ Sci Pollut R, 540-547.

2018, 25(21): 21205-21215. [104] Pan W, Xue B, Yang C, et al. Biopharmaceutical characters

[85] Chatuphonprasert W, Jarukamjorn K, Ellinger I. Physiology and bioavailability improving strategies of ginsenosides [J]. and pathophysiology of steroid biosynthesis, transport and Fitoterapia, 2018, 129: 272-282.

metabolism in the human placenta [J]. Front Pharmacol, [105] Won HJ, Kim HI, Park T, et al. Non-clinical pharmacokinetic 2018, 9: 102. behavior of ginsenosides [J]. J Ginseng Res, 2018, 43(3): 354-

[86] Payne AH, Hales DB. Overview of steroidogenic enzymes in 360.

the pathway from cholesterol to active steroid hormones [J]. [106] Zheng P, Chen Y, Fu Y, et al. Influence of B-complex vitam- Endocr Rev, 2004, 25(6): 947-970. ins on the pharmacokinetics of ginsenosides rg(1), rb-1, and ro

[87] Ying A, Yu Q, Guo L, et al. Structural–activity relationship of after oral administration [J]. J Med Food, 2017, 20(11): 1127- ginsenosides from steamed ginseng in the treatment of erectile 1132.

dysfunction [J]. Am J Chinese Med, 2018, 46(01): 137-155. [107] Bae E, Choo M, Park E, et al. Metabolism of ginsenoside rc

[88] Song W, Liu QS, Sun Z, et al. Polyfluorinated iodine alkanes by human intestinal bacteria and its related antiallergic activ-

– 534 – TIAN Mei, et al. / Chin J Nat Med, 2020, 18(7): 526-535

ity [J]. Biol Pharm Bull, 2002, 25(6): 743-747. of ginsenoside Rb1, an active component of Panax ginseng,

[108] Tawab MA, Bahr U, Karas M, et al. Degradation of ginsenos- on neural 5-HT disposition and behavioral tasks in ovariec- ides in humans after oral administration [J]. Drug Metab Dis- tomized mice [J]. Eur J Pharmacol, 2011, 659(1): 15-25.

pos, 2003, 31(8): 1065-1071. [121] Cao Y, Ye Q, Zhuang M, et al. Ginsenoside Rg3 inhibits an-

[109] Yu H, Wang Y, Liu C, et al. Conversion of ginsenoside rb1 giogenesis in a rat model of endometriosis through the VEG- into six types of highly bioactive ginsenoside rg3 and its de- FR-2-mediated PI3K/Akt/mTOR signaling pathway [J]. PLoS rivatives by FeCl3 catalysis [J]. Chem Pharm Bull, 2018, ONE, 2017, 12(11): e0186520.

66(9): 901-906. [122] Mou Z, Huang Q, Chu S, et al. Antidepressive effects of gin-

[110] Hwang IG, Kim HY, Joung EM, et al. Changes in ginsenos- senoside Rg1 via regulation of HPA and HPG axis [J]. Bio- ides and antioxidant activity of Korean ginseng (Panax gin- med Pharmacother, 2017, 92: 962-971. seng C. A. Meyer) with heating temperature and pressure [J]. [123] Mao N, Cheng Y, Shi XL, et al. Ginsenoside Rg1 protects Food Sci Biotechnol, 2010, 19(4): 941-949. mouse podocytes from aldosterone-induced injury in vitro [J].

[111] Xie H, Wang G, Lv H, et al. Development of a HPLC-MS as- Acta Pharmacol Sin, 2014, 35(4): 513-22. say for ginsenoside Rh2, a new anti-tumor substance from nat- [124] Kopalli SR, Cha K, Ryu J, et al. Korean red ginseng im- ural product and its pharacokinetic study in dogs [J]. Eur J proves testicular ineffectiveness in aging rats by modulating Drug Metab Pharmacokinet, 2005, 30(1-2): 63. spermatogenesis-related molecules [J]. Exp Gerontol, 2017, [112] Kim WY, Kim JM, Han SB, et al. Steaming of ginseng at high 90: 26-33. temperature enhances biological activity [J]. J Nat Prod,

[125] Murata K, Takeshita F, Samukawa K, et al. Effects of gin- 2000, 63(12): 1702-1704. seng rhizome and ginsenoside ro on testosterone 5α-Re-

[113] Hasegawa E, Nakagawa S, Miyate Y, et al. Inhibitory effect ductase and hair re-growth in testosterone-treated mice [J]. of protopanaxatriol ginseng metabolite M4 on the production Phytother Res, 2012, 26(1): 48-53. of corticosteroids in ACTH-stimulated bovine adrenal fascicu-

[126] Ben-Eltriki M, Deb S, Hassona M, et al. 20(S)-protopanaxadi- lata cells [J]. Life Sci, 2013, 92(12): 687-693. ol regio-selectively targets androgen receptor: anticancer ef-

[114] Dong S, Kiyama R. Characterisation of oestrogenic activity of fects in castration-resistant prostate tumors [J]. Oncotarget, ginsenosides in MCF-7 cells using a customised DNA mi- 2018, 9(30): 20965-20978. croarray [J]. Food Chem, 2009, 113(2): 672-678.

[127] Ding Jie XYMX. Estrogenic effect of the extract of Renshen

[115] Ke Y, Bertin J, Gonthier R, et al. A sensitive, simple and ro- (Radix Ginseng) on reproductive tissues in immature mice [J]. bust LC–MS/MS method for the simultaneous quantification of seven androgen- and estrogen-related steroids in postmeno- J Tradit Chin Med, 2015, 35(4): 460-467. pausal serum [J]. J Steroid Biochem, 2014, 144: 523-534. [128] Lee YJ, Jin YR, Lim WC, et al. Ginsenoside-Rb1 acts as a weak phytoestrogen in MCF-7 human breast cancer cells [J]. [116] Zhai X, Chen F, Zhu C, et al. A simple LC–MS/MS method for the determination of cortisol, cortisone and tetrahydro- Arch Pharm Res, 2003, 26(1): 58-63.

metabolites in human urine: assay development, validation [129] Lee Y, Jin Y, Lim W, et al. A ginsenoside-Rh1, a component and application in depression patients [J]. J Pharmaceut Bio- of ginseng saponin, activates estrogen receptor in human med, 2015, 107: 450-455. breast carcinoma MCF-7 cells [J]. J Steroid Biochem, 2003, 84(4): 463-468. [117] Bae J, Park H, Park J, et al. Red ginseng and 20(S)-Rg3 con-

trol testosterone-induced prostate hyperplasia by deregulating [130] Cho J, Chun H, Lee JS, et al. Prevention effect of rare ginsen- androgen receptor signaling [J]. J Nat Med-Tokyo, 2012, osides against stress-hormone induced MTOC 66(3): 476-485. amplification [J]. Oncotarget, 2016, 7(23): 35144.

[118] Xu Y, Ding J, An J, et al. Effect of the interaction of [131] Li J, Du J, Liu D, et al. Ginsenoside Rh1 potentiates dexa- nigrum with Panax ginseng on estrogenic activity in vivo and methasone's anti-inflammatory effects for chronic inflammat- in vitro [J]. Sci Rep-Uk, 2016, 6(1): 7827-7840. ory disease by reversing dexamethasone-induced

[119] Chen JC, Chen LD, Tsauer W, et al. Effects of Ginsenoside resistance [J]. Arthritis Res Ther, 2014, 16(3): R106.

Rb2 and Rc on inferior human sperm motility in vitro [J]. Am [132] Leung KW, Cheng Y, Mak NK, et al. Signaling pathway of J Chin Med, 2001, 29(1): 155-160. ginsenoside-Rg1 leading to nitric oxide production in en-

[120] Hao K, Gong P, Sun S, et al. Beneficial estrogen-like effects dothelial cells [J]. FEBS Lett, 2006, 580(13): 3211-3216.

Cite this article as: TIAN Mei, LI Lin-Nan, ZHENG Rui-Rong, YANG Li, WANG Zheng-Tao. Advances on hormone-like activity of Panax ginseng and ginsenosides [J]. Chin J Nat Med, 2020, 18(7): 526-535.

– 535 –