Skip to main content
Journal of Ginseng Research logoLink to Journal of Ginseng Research
. 2016 Aug 23;41(2):215–221. doi: 10.1016/j.jgr.2016.08.005

Effects of ginseng on two main sex steroid hormone receptors: estrogen and androgen receptors

Joonwoo Park 1, Heewon Song 1, Si-Kwan Kim 2, Myeong Soo Lee 3, Dong-Kwon Rhee 4, YoungJoo Lee 1,
PMCID: PMC5386121  PMID: 28413327

Abstract

Ginseng has been used in China for at least two millennia and is now popular in over 35 countries. It is one of the world's popular herbs for complementary and alternative medicine and has been shown to have helpful effects on cognition and blood circulation, as well as anti-aging, anti-cancer, and anti-diabetic effects, among many others. The pharmacological activities of ginseng are dependent mainly on ginsenosides. Ginsenosides have a cholesterol-like four trans-ring steroid skeleton with a variety of sugar moieties. Nuclear receptors are one of the most important molecular targets of ginseng, and reports have shown that members of the nuclear receptor superfamily are regulated by a variety of ginsenosides. Here, we review the published literature on the effects of ginseng and its constituents on two main sex steroid hormone receptors: estrogen and androgen receptors. Furthermore, we discuss applications for sex steroid hormone receptor modulation and their therapeutic efficacy.

Keywords: androgen receptor, estrogen receptor, ginseng, nuclear receptor, steroid hormone receptor

1. Introduction

Ginseng has been used for over 2,000 y as a medicine in East Asia. It is a popular herb in the world, and is used in more than 35 countries as a food, health supplement, and natural remedy [1]. Ginseng has been demonstrated to have an extensive range of pharmacological effects on the reproductive, cardiovascular, endocrine, and immune systems. Its ability to diminish fatigue, enhance blood circulation, aid menopausal symptoms, boost immune function, and enhance concentration has been verified in certain countries [2]. Many components of ginseng, such as ginsenosides, polysaccharides, peptides, polyacetylenic alcohols, and fatty acids, have been isolated and characterized [3], [4]. However, further research is necessary to understand how these components contribute to ginseng's pharmacological properties. Among these constituents, dammarane-type ginsenosides, comprising a rigid steroid skeleton consisting of four transrings with a modified side chain at C–20, have received considerable attention because of their biological activity [5], [6]. To date, more than 100 different ginsenosides with various pharmacological effects have been isolated and identified from the root of Panax ginseng, with ginsenosides Rb1, Rb2, Rg1, and Re being most abundant. In addition, steaming and drying during processing of ginseng can affect its pharmacological activity by altering the characteristics of the constituent ginsenosides [7], [8], [9], [10], [11], [12]. Red ginseng, produced by steaming fresh ginseng, possesses unique ginsenosides (Rg3, Rg5, Rh2, Rh3, Rh4, Rs3, and F4) [13], [14], [15].

The molecular target of ginsenosides may be located either in the cellular membrane or inside the cell, depending on the hydrophobicity of the ginsenoside [16]. Most ginsenosides are lipophilic in nature; with their steroidal backbone they can traverse cell membranes by simple diffusion, and regulate cellular functions by binding to specific intracellular target proteins in the cytoplasm and nucleus [16]. The nongenomic pathway of ginsenoside activity involves binding to membrane-associated receptors that initiate the activation of the phosphorylation cascade and generation of second messengers. Ginsenosides activate the genomic pathway by initially binding to intracellular nuclear hormone receptors, such as the glucocorticoid receptor, progesterone receptor, androgen receptor (AR), mineralocorticoid receptor, estrogen receptor (ER), peroxisome proliferator-activated receptor, and liver X receptor [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29]. Many studies have shown that ginsenosides act through nuclear hormone receptors. For example, one study found that administration of ginseng extract for about 1.5 wk inhibited the activity of peroxisome proliferator-activated receptor-α and the expression of several genes associated with lipid and lipoprotein metabolism [30]; presumably, ginsenosides were the active constituents. Ginsenosides enhanced the expression levels of glucocorticoid receptor in the brain cytosol of heat-injured rats [31]. In another study, the interaction of ginsenoside-activated glucocorticoid receptors triggered the activation of the phosphatidylinositol-3 kinase (PI3K)/Akt pathway and increased nitric oxide production in human umbilical vein endothelial cells [32]. Ginsenosides act through not only steroid hormone receptors, but also ion channels. One study showed that ginsenosides inhibit voltage-dependent calcium, potassium, and sodium channel activities in a stereospecific manner, and block subtypes of nicotinic acetylcholine and 5-hydroxytryptamine type 3 receptors [33].

Here, we review the ginsenosides that affect estrogen and androgen activities, including those that directly or indirectly modulate ERs and ARs. We also discuss clinical studies of ginseng on female and male reproductive functions.

2. Effects on ERα and ERβ

2.1. Genomic and nongenomic action

Estrogens are female steroid hormones that are produced mainly by the ovaries through the conversion of cholesterol. Estrogens can also be produced locally in the placenta, adrenal glands, adipose tissue, and brain, where they act in a paracrine fashion [34]. Estrogens are essential in the development and maintenance of the female reproductive system, immune system, cardiovascular system, and central and peripheral nervous systems; they also regulate bone metabolism [35]. As a consequence of their wide range of physiological roles, estrogens are also involved in many pathological states, including cancer, metabolic and cardiovascular diseases, neurodegeneration, and osteoporosis [36].

Phytoestrogens are compounds of plant origin that can exert estrogenic properties, through either directly binding to ER or indirectly activating ERs [36], [37], [38]. Phytoestrogens, such as genistein and daidzein, have shown protective effects on conditions related to decreased estrogen, including menopause, osteoporosis, and cognitive disorders [39], [40]. The interest in the use of phytoestrogens stems from epidemiologic studies suggesting a decreased incidence of breast cancer, and lower occurrence and complaints of menopausal symptoms and osteoporosis in women from countries with high consumption of phytoestrogens, mainly found in soy products [41], [42], [43], [44].

Cellular effects of estrogens are mediated by ERs, which are transcription factors activated by ligands. Two distinct isoforms exist: ERα and ERβ. The expression patterns of these two receptors differ and display distinct characteristics. ERα is expressed mainly in reproductive tissues such as the uterus and ovary, breast, bone, white adipose tissue, and liver. ERβ is expressed mainly in the ovary, central nervous system, cardiovascular system, lung, prostate, colon, and the immune system [36], [45], [46].

The classical estrogen action model suggests that nuclear ER changes its conformation upon ligand binding, binds to the cognate estrogen-responsive element, and modulates the transcription of target genes [47], [48]. Hundreds of coactivator/corepressor regulatory proteins affect the ER-mediated transcriptional response. ERs are mainly found in the nucleus and also in the cytoplasmic membrane. Estrogen binds to the membrane receptors and stimulates signaling proteins. The nongenomic pathway triggers G protein-coupled receptors, generates calcium flux, stimulates cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) production, and activates the PI3K and extracellular-signal-regulated kinase (ERK) pathways [49], [50], [51]. Recent researches indicate that ERs were also identified in mitochondria and endosomes, and that the act of signaling from all these sites must be integrated with nuclear ER action to produce the final functions of the steroid [52], [53]. Studies using a xenograft model of MCF-7 human breast cancer cells injected into nude mice have shown that engagement of only the membrane receptor by an estrogenic compound failed to stimulate tumor proliferation [54]. This indicates that nongenomic and genomic pathways of ER communicate, which is likely to play a role in promoting human breast tumor growth. Cellular effects of ginsenosides can be influenced by many factors, including concentration, receptor status, amount of endogenous estrogens, and target tissue [55]. Some phytoestrogens display different affinities to ER isoforms [56]. No study has shown antiestrogenic effects of ginsenosides, but both estrogenic and antiestrogenic activities have been reported for genistein [57], [58].

Treatment with P. ginseng upregulated both ERα and ERβ in the reproductive organs (e.g., uterus and vagina) of both normal and ovariectomized mice, demonstrating that P. ginseng had a potent estrogenic activity [25], [26]. However, P. ginseng upregulated the expression of ERα to a greater extent than that of ERβ in the uterus and vagina, suggesting that P. ginseng selectively binds to ERα in these reproductive tissues [25], [26]. However, ERβ antagonizes ERα-mediated effects in reproductive tissues, such as the breast, ovary, prostate, and uterus [59], [60]. In contrast to the effects seen in reproductive organs, treatment of immobilized mice brains and in vitro treatment of neuroblastoma SK-N-SH cells with hydrogen peroxide depressed ERβ, but not ERα [24]. Korean Red Ginseng (KRG) upregulates ERβ and subsequently mitigates stress-induced gene expression [24]. Consistently, in vitro studies also demonstrated that KRG represses apoptosis by potentiating PI3K/Akt signaling via ERβ upregulation [61], [62]. For instance, KRG appears to repress stress-induced tumor necrosis factor-α converting enzyme and nuclear factor-κB in immobilized mice brains and H2O2-induced neuroblastoma SK-N-SH cells, thus preventing the production of reactive oxygen species and protecting brain cells from apoptosis [24], [61], [62]. Thus, in the brain, ERβ seems to inhibit apoptosis and antagonize oxidative stress.

Regarding the mechanisms of action of KRG components, the major ginsenoside Rb1 dose dependently activated both ERα and ERβ in COS monkey kidney cells, but it did not bind to the ER receptors in MCF-7 breast cancer cells [63]. In contrast, the antiangiogenic effects of the ginsenoside Rb1 have been ascribed to its interaction with ERβ in vitro. Rb1 specifically increased the expression of a potent antiangiogenic protein pigment epithelium-derived factor by binding to ERβ, but not to ERα, resulting in the inhibition of endothelial tube formation [64]. These results suggest that KRG might affect ERβ expression in a tissue- and organ-specific manner.

Reproduction is controlled mainly by estradiol in an ERα-dependent manner. Furthermore, brain functions are profoundly mediated by ERα, but not by ERβ [65], [66], and most research on ERβ has focused on brain function and behavior [67]. However, in the brain, ERα and ERβ are usually tightly controlled in an interrelated and estradiol-dependent manner for a particular brain function [67]. ERβ modulation is also common; for example, hypernatremia stress and immobilization stress downregulate ERβ expression [24], [68]. In contrast, P. ginseng may upregulate ERβ in the brain, and ERβ may counteract stress via antiapoptotic and antioxidative activities [24]. Although the underlying mechanism for this effect requires further study, one possible explanation is that KRG upregulates the steroidogenic enzyme P450 (Cyp11a1) [69]. Cyp11a1 catalyzes the initial and rate-limiting step in steroid hormone synthesis, and cleaves the cholesterol side chain into pregnenolone with a steroidogenic potential [70]. However, an interaction between ERα and ERβ cannot be ruled out; further research on the interaction between these two proteins in the brain is therefore required to clarify the underlying mechanism.

Fig. 1 and Table 1 summarizes the modulating effects of ginsenosides upon the ER pathway.

Fig. 1.

Fig. 1

Ginsenosides modulating estrogen receptor pathway. A summarized diagram of ginsenosides affecting the estrogen receptor pathway via genomic and nongenomic pathways. COX-2, cyclooxygenase-2; eNOS, endothelial nitric oxide synthase; ER, estrogen receptor; iNOS, inducible nitric oxide synthase; PI3K, phosphatidylinositol-3 kinase; NO, nitric oxide; MAPK, p38 mitogen-activated protein kinase; AMPK, AMP-activated protein kinase.

Table 1.

Ginsenosides modulating estrogen receptor pathway

Ginsenosides/total ginseng Experimental model systems Observations Signaling molecules monitored Reference no.
Total ginseng
 Korean Red Ginseng SK-N-SH human neuroblastoma cells, mate ICR mice model Inhibits oxidative stress and apoptosis PADI4, p53, Bcl-2 COX-2, ERβ [24]
SK-N-SH human neuroblastoma cells Antioxidant and represses apoptosis PI3K/Akt, Bcl-2, p53, Caspase-3 [61]
 Radix Ginseng Immature Kunming mice Estrogen effect on reproductive tissues ERα, ERβ [25]
 P. ginseng Ovariectomized mice Can treat postmenopausal symptoms through action as an estrogen agonist ERα, ERβ [26]
MCF-7 human breast cancer cells Estrogenic activity in vitro but not in vivo ERα [27]
Ginsenosides
 Rb1 MCF-7 human breast cancer cells, COS monkey kidney cells Estrogen-like activity [63]
Human umbilical vein endothelial cells Inhibits capillary morphogenesis [71]
Ovariectomized mice Estrogen-like effect on brain 5-HT levels [72]
PC12 rat pheochromocytoma cells Prevents MPP+-induced apoptosis ERK1/2, SAPK/JNK, p38 MAPK [73]
Human articular chondrocytes Anti-inflammatory and apoptotic properties IL-1β, PGE2, COX-2, iNOS, Caspase-3, NO2–, MMP-3 [74]
 Rg1 MCF-7 human breast cancer cells Proliferation IGF-IR [75]
MCF-7 human breast cancer cells Estrogen-like activity MAPK [76]
Ovariectomized rat model of Alzheimer's disease Neuroprotective effect Caspase-3 [77]
Primary rat cerebrocortical neurons Neuroprotective effect ERK1/2 [19]
HT-22 hippocampal neuronal cells, SH-SY5Y human neuroblastoma cells, Ovariectomized rats Promotes nonamyloidgenic cleavage of beta-amyloid precursor protein PI3K/Akt, MAPK [78]
N2a-APP694 mouse neuronal cells Prevents memory loss PPARγ [28]
SK-N-SH human neuroblastoma cells Apoptosis IGF-IR [79]
Bone marrow stromal cells Proliferation, activates ER-mediated signaling [80]
MCF-7, MDAMB human breast cancer cells Estrogen-like activity [81]
 Rg3 PC-3M prostate cancer cells Inhibits migration AQP1, p38 MAPK [29]
ECV304 human endothelial cells Endothelial NO production PI3K/Akt, AMPK, JNK, p38 [82]
 Re Embryonic rat thoracic aortic smooth muscle cells from DBIX rat (A10 cells) 1. Releases NO via membrane sex steroid receptors
2. Promotes vasodilation
3. Activates potassium channels
PI3K/Akt [83]
Single ventricular myocytes, MCF-7 human breast cancer cells, LNCaP human prostate cancer cells Activates cardiac potassium channels via nongenomic pathway of sex hormone [84]
 Notoginsenoside Ft1 Rat mesenteric arteries model NO-mediated relaxation PI3K/Akt, ERK1/2 [20]
 Notoginsenoside R1 Endotoxemic mice Anti-inflammation, protects the heart from septic shock NF-κB, I-κB, PI3K/Akt [85]
Rat primary osteoblastic cells Stimulates osteoblast differentiation via ER signaling [86]
 P. ginseng ginsenoside Rh1 MCF-7 human breast cancer cells Weak phytoestrogen [87]
Ginsenoside metabolites
 Protopanaxadiol Primary human umbilical vein endothelial cells Functional ligands for both GR and ERβ [88]
 Protopanaxatriol Primary human umbilical vein endothelial cells Functional ligands for both GR and ERβ [88]
AR
 P. ginseng LNCaP human prostate cancer cells Inhibit AR signaling by stimulating the degradation of AR protein
Antiproliferation, apoptosis
AR [21]
C57BL/6 mice model, human hair follicle papilla cells Antiandrogen, promotes hair growth in humans ERK, Akt [23]
 20(s)-25-Methoxyl-dammarane-3beta, 12beta, 20-triol (20-OCH3-PPD) In vitro:
Human LNCaP (P53 wild type, androgen dependent), PC3 (P53 null, androgen independent), IMR90-EEA and IMR90-E1A human primary fibroblast cell lines
In vivo:
Prostate cancer (PC3) xenograft model
Inhibits the expression of androgen receptors, antiproliferation, apoptosis PARP, Bax, Bcl-2, cyclin D [22]

AR, androgen receptor; COX-2, cyclooxygenase-2; ER, estrogen receptor; GR, glucocorticoid receptor; IL, interleukin; iNOS, inducible nitric oxide synthase; MMP-3, matrix metalloproteinase-3; NF-κB, nuclear factor-κB; NO, nitric oxide; PI3K, phosphatidylinositol-3 kinase; PPAR, peroxisome proliferator-activated receptor; 5-HT, 5-hydroxytryptamine

2.2. Clinical studies on menopausal symptoms, erectile dysfunction, and women's sexual function

Ginseng has often been used for management of menopausal symptoms in postmenopausal women, and sexual function in both men and women. One systematic review suggested that the evidence on efficacy of ginseng for managing menopausal symptoms was limited [89]. Three randomized controlled trials (RCTs), which were included in the review, tested the efficacy of KRG compared with placebo, and their results showed superior effects of KRG on sexual arousal and global health [90], [91], [92]. Another trial reported favorable effects of ginseng supplements on well-being and depression as compared with placebo [93].

Regarding the efficacy of KRG for sexual function in men, one systematic review investigated the efficacy of KRG for erectile dysfunction (ED) compared with placebo [94]. This review included seven RCTs, and their results suggested superior effects of KRG on improvement of ED compared with placebo. Recently, two additional RCTs also reported positive effects of KRG on ED [95], [96]. Collectively, the evidence on the efficacy of KRG for treating ED is highly significant.

For the effects of KRG on women's sexual function, three crossover RCTs tested the efficacy of KRG for the sexual function of women compared with placebo control [90], [97], [98]. One RCT showed positive effects of KRG on sexual arousal [90], and the other two RCTs reported improvement of sexual function without statistical significance compared with placebo control [97], [98].

3. Effects on AR

Androgens mediate a wide range of male developmental processes, and are especially important in male sexual differentiation as well as pubertal maturation, maintenance of spermatogenesis, and male gonadotropin regulation [99]. Testosterone and its metabolite 5-α-dihydrotestosterone, two principal androgens, exert their function predominately through AR-mediated pathways [100]. Disorganization of the androgen–AR complex results in androgen insensitivity syndrome, eventually leading to dysfunctions of the male reproductive system, such as subfecundity or infertility. Defects in the AR lead to distinct signs of undervirilization and undermasculinization in males [101]. Disturbance of the androgen–AR complex is triggered mainly by mutations in the AR gene. Mutations in the AR gene are inherited in an X-linked recessive pattern and, therefore, affect males much more frequently than females [102].

Melo et al [103] studied the relationship between male infertility and androgen AR mutations in Brazilian patients, and postulated that patients who do not produce sperm have a higher number of AR mutations than those with merely impaired sperm production.

While the expression level of AR itself, as well as luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR), may also contribute to androgen insensitivity and male sub- and/or infertility, this idea has not received much scientific attention to date. This topic has largely been overlooked in the biochemical and pharmacological communities, as well as in urology. The expression levels of proteins and mRNAs for AR, LHR, and FSHR in testicular tissue from aged and doxorubicin-sensitized Sprague-Dawley rats, were significantly downregulated in these animals as well as in animals stress loaded by intermittent immobilization (2 h/d for 8 wk/6 mo) and heat (32°C, 2 h/d for 8 wk/6 mo). Pretreatment with KRG markedly prevented the downregulation of these receptors [104], [105]. Wang et al [106], at the Center for Disease Prevention and Control in Shenyang Command, Shenyang, China, carried out a similar experiment in cold-stressed rats. They reported that ginseng polysaccharides upregulated AR mRNA expression levels and promoted testosterone. These results indicate that P. ginseng plays an important role in maintaining healthy levels of steroid hormone receptors, including AR, which in turn ensures the proper functioning of androgens.

4. Conclusion

Ginseng has been used for various pathological problems, as well as for preventing and enhancing overall physiological functions. The effects of ginsengs in cancer chemoprevention and therapeutics are one of the most intensively studied areas. Identification of cellular targets responsible for such activity and characterization of active red ginseng compounds still requires continuous research. Nuclear hormone receptors are one of the cellular targets of ginsenosides that are lipophilic in nature. ER is one of the well-studied targets for ginsenosides among the members of nuclear steroid hormone receptors. Modulation of ERs by ginseng components can be beneficial to estrogen-dependent cancer and many physiological states deriving from low levels of estrogen hormones due to aging as well as inevitable ovary-related surgery in younger women. As shown, studies targeting ARs and androgen-associated physiological states are less studied. Even with accumulating data on estrogen-mimicking or supplemental activity of ginseng in humans, additional accumulative data are still required to establish and provide a scientific basis of ginseng use on sex steroid hormone-dependent function.

Acknowledgments

This research was supported by 2015 grant from the Korean Society of Ginseng to YJL.

References

  • 1.Baeg I.H., So S.H. The world ginseng market and the ginseng (Korea) J Ginseng Res. 2013;37:1–7. doi: 10.5142/jgr.2013.37.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Park H.J., Kim D.H., Park S.J., Kim J.M., Ryu J.H. Ginseng in traditional herbal prescriptions. J Ginseng Res. 2012;36:225–241. doi: 10.5142/jgr.2012.36.3.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kim M.K., Lee J.W., Lee K.Y., Yang D.C. Microbial conversion of major ginsenoside rb(1) to pharmaceutically active minor ginsenoside rd. J Microbiol. 2005;43:456–462. [PubMed] [Google Scholar]
  • 4.Kang S., Min H. Ginseng, the ‘immunity boost’: the effects of Panax ginseng on immune system. J Ginseng Res. 2012;36:354–368. doi: 10.5142/jgr.2012.36.4.354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Shin B.K., Kwon S.W., Park J.H. Chemical diversity of ginseng saponins from Panax ginseng. J Ginseng Res. 2015;39:287–298. doi: 10.1016/j.jgr.2014.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Nah S.Y., Park H.J., McCleskey E.W. A trace component of ginseng that inhibits Ca2+ channels through a pertussis toxin-sensitive G protein. Proc Natl Acad Sci U S A. 1995;92:8739–8743. doi: 10.1073/pnas.92.19.8739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kim S.K., Park J.H. Trends in ginseng research in 2010. J Ginseng Res. 2011;35:389–398. doi: 10.5142/jgr.2011.35.4.389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lee D.H., Cho H.J., Kim H.H., Rhee M.H., Ryu J.H., Park H.J. Inhibitory effects of total saponin from Korean Red Ginseng via vasodilator-stimulated phosphoprotein-Ser(157) phosphorylation on thrombin-induced platelet aggregation. J Ginseng Res. 2013;37:176–186. doi: 10.5142/jgr.2013.37.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Siddiqi M.H., Siddiqi M.Z., Ahn S., Kang S., Kim Y.J., Sathishkumar N., Yang D.U., Yang D.C. Ginseng saponins and the treatment of osteoporosis: mini literature review. J Ginseng Res. 2013;37:261–268. doi: 10.5142/jgr.2013.37.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kang K.S., Ham J., Kim Y.J., Park J.H., Cho E.J., Yamabe N. Heat-processed Panax ginseng and diabetic renal damage: active components and action mechanism. J Ginseng Res. 2013;37:379–388. doi: 10.5142/jgr.2013.37.379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lee S., Kim M.G., Ko S.K., Kim H.K., Leem K.H., Kim Y.J. Protective effect of ginsenoside Re on acute gastric mucosal lesion induced by compound 48/80. J Ginseng Res. 2014;38:89–96. doi: 10.1016/j.jgr.2013.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lee C.H., Kim J.H. A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases. J Ginseng Res. 2014;38:161–166. doi: 10.1016/j.jgr.2014.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Fishbein A.B., Wang C.Z., Li X.L., Mehendale S.R., Sun S., Aung H.H., Yuan C.S. Asian ginseng enhances the anti-proliferative effect of 5-fluorouracil on human colorectal cancer: comparison between white and red ginseng. Arch Pharm Res. 2009;32:505–513. doi: 10.1007/s12272-009-1405-9. [DOI] [PubMed] [Google Scholar]
  • 14.Du G.J., Wang C.Z., Zhang Z.Y., Wen X.D., Somogyi J., Calway T., He T.C., Du W., Yuan C.S. Caspase-mediated pro-apoptotic interaction of panaxadiol and irinotecan in human colorectal cancer cells. J Pharm Pharmacol. 2012;64:727–734. doi: 10.1111/j.2042-7158.2012.01463.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Baek S.H., Piao X.L., Lee U.J., Kim H.Y., Park J.H. Reduction of cisplatin-induced nephrotoxicity by ginsenosides isolated from processed ginseng in cultured renal tubular cells. Biol Pharm Bull. 2006;29:2051–2055. doi: 10.1248/bpb.29.2051. [DOI] [PubMed] [Google Scholar]
  • 16.Attele A.S., Wu J.A., Yuan C.S. Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol. 1999;58:1685–1693. doi: 10.1016/s0006-2952(99)00212-9. [DOI] [PubMed] [Google Scholar]
  • 17.Paik N.H., Park M.K., Choi K.J., Cho Y.H. Isolation of ginsenosides Rb1, Rb2, Rc, Rd, Re, Rf and Rg1 from ginseng root by high performance liquid chromatography. Arch Pharm Res. 1982;5:7–12. [Google Scholar]
  • 18.Chung E., Lee K.Y., Lee Y.J., Lee Y.H., Lee S.K. Ginsenoside Rg1 down-regulates glucocorticoid receptor and displays synergistic effects with cAMP. Steroids. 1998;63:421–424. doi: 10.1016/s0039-128x(98)00043-9. [DOI] [PubMed] [Google Scholar]
  • 19.Wu J., Pan Z., Wang Z., Zhu W., Shen Y., Cui R., Lin J., Yu H., Wang Q., Qian J. Ginsenoside Rg1 protection against beta-amyloid peptide-induced neuronal apoptosis via estrogen receptor alpha and glucocorticoid receptor-dependent anti-protein nitration pathway. Neuropharmacology. 2012;63:349–361. doi: 10.1016/j.neuropharm.2012.04.005. [DOI] [PubMed] [Google Scholar]
  • 20.Shen K., Leung S.W., Ji L., Huang Y., Hou M., Xu A., Wang Z., Vanhoutte P.M. Notoginsenoside Ft1 activates both glucocorticoid and estrogen receptors to induce endothelium-dependent, nitric oxide-mediated relaxations in rat mesenteric arteries. Biochem Pharmacol. 2014;88:66–74. doi: 10.1016/j.bcp.2014.01.007. [DOI] [PubMed] [Google Scholar]
  • 21.Bae J.S., Park H.S., Park J.W., Li S.H., Chun Y.S. Red ginseng and 20(S)-Rg3 control testosterone-induced prostate hyperplasia by deregulating androgen receptor signaling. J Nat Med. 2012;66:476–485. doi: 10.1007/s11418-011-0609-8. [DOI] [PubMed] [Google Scholar]
  • 22.Wang W., Wang H., Rayburn E.R., Zhao Y., Hill D.L., Zhang R. 20(S)-25-methoxyl-dammarane-3beta, 12beta, 20-triol, a novel natural product for prostate cancer therapy: activity in vitro and in vivo and mechanisms of action. Br J Cancer. 2008;98:792–802. doi: 10.1038/sj.bjc.6604227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Park G.H., Park K.Y., Cho H.I., Lee S.M., Han J.S., Won C.H., Chang S.E., Lee M.W., Choi J.H., Moon K.C. Red ginseng extract promotes the hair growth in cultured human hair follicles. J Med Food. 2015;18:354–362. doi: 10.1089/jmf.2013.3031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kim E.H., Kim I.H., Lee M.J., Thach Nguyen C., Ha J.A., Lee S.C., Choi S., Choi K.T., Pyo S., Rhee D.K. Anti-oxidative stress effect of red ginseng in the brain is mediated by peptidyl arginine deiminase type IV (PADI4) repression via estrogen receptor (ER) beta up-regulation. J Ethnopharmacol. 2013;148:474–485. doi: 10.1016/j.jep.2013.04.041. [DOI] [PubMed] [Google Scholar]
  • 25.Ding J., Xu Y., Ma X., An J., Yang X., Liu Z., Lin N. Estrogenic effect of the extract of Renshen (Radix Ginseng) on reproductive tissues in immature mice. J Tradit Chin Med. 2015;35:460–467. doi: 10.1016/s0254-6272(15)30125-4. [DOI] [PubMed] [Google Scholar]
  • 26.Xu Y., Ding J., Ma X.P., Ma Y.H., Liu Z.Q., Lin N. Treatment with Panax ginseng antagonizes the estrogen decline in ovariectomized mice. Int J Mol Sci. 2014;15:7827–7840. doi: 10.3390/ijms15057827. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 27.Shim M.K., Lee Y.J. Estrogen receptor is activated by Korean Red Ginseng in vitro but not in vivo. J Ginseng Res. 2012;36:169–175. doi: 10.5142/jgr.2012.36.2.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chen L.M., Lin Z.Y., Zhu Y.G., Lin N., Zhang J., Pan X.D., Chen X.C. Ginsenoside Rg1 attenuates beta-amyloid generation via suppressing PPARgamma-regulated BACE1 activity in N2a-APP695 cells. Eur J Pharmacol. 2012;675:15–21. doi: 10.1016/j.ejphar.2011.11.039. [DOI] [PubMed] [Google Scholar]
  • 29.Pan X.Y., Guo H., Han J., Hao F., An Y., Xu Y., Xiaokaiti Y., Pan Y., Li X.J. Ginsenoside Rg3 attenuates cell migration via inhibition of aquaporin 1 expression in PC-3M prostate cancer cells. Eur J Pharmacol. 2012;683:27–34. doi: 10.1016/j.ejphar.2012.02.040. [DOI] [PubMed] [Google Scholar]
  • 30.Yoon M., Lee H., Jeong S., Kim J.J., Nicol C.J., Nam K.W., Kim M., Cho B.G., Oh G.T. Peroxisome proliferator-activated receptor alpha is involved in the regulation of lipid metabolism by ginseng. Br J Pharmacol. 2003;138:1295–1302. doi: 10.1038/sj.bjp.0705169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Li M., Ling C.Q., Huang X.Q., Shen Z.L. Effects of ginsenosides extracted from ginseng stem and leaves on glucocorticoid receptor in different viscera in heat-damaged rats. Zhong Xi Yi Jie He Xue Bao. 2006;4:156–159. doi: 10.3736/jcim20060210. [DOI] [PubMed] [Google Scholar]
  • 32.Leung K.W., Cheng Y.K., Mak N.K., Chan K.K., Fan T.P., Wong R.N. Signaling pathway of ginsenoside-Rg1 leading to nitric oxide production in endothelial cells. FEBS Lett. 2006;580:3211–3216. doi: 10.1016/j.febslet.2006.04.080. [DOI] [PubMed] [Google Scholar]
  • 33.Nah S.Y. Ginseng ginsenoside pharmacology in the nervous system: involvement in the regulation of ion channels and receptors. Front Physiol. 2014;5:98. doi: 10.3389/fphys.2014.00098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cui J., Shen Y., Li R. Estrogen synthesis and signaling pathways during aging: from periphery to brain. Trends Mol Med. 2013;19:197–209. doi: 10.1016/j.molmed.2012.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mauvais-Jarvis F., Clegg D.J., Hevener A.L. The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev. 2013;34:309–338. doi: 10.1210/er.2012-1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Paterni I., Granchi C., Katzenellenbogen J.A., Minutolo F. Estrogen receptors alpha (ERalpha) and beta (ERbeta): subtype-selective ligands and clinical potential. Steroids. 2014;90:13–29. doi: 10.1016/j.steroids.2014.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Younes M., Honma N. Estrogen receptor beta. Arch Pathol Lab Med. 2011;135:63–66. doi: 10.5858/2010-0448-RAR.1. [DOI] [PubMed] [Google Scholar]
  • 38.Rietjens I.M., Sotoca A.M., Vervoort J., Louisse J. Mechanisms underlying the dualistic mode of action of major soy isoflavones in relation to cell proliferation and cancer risks. Mol Nutr Food Res. 2013;57:100–113. doi: 10.1002/mnfr.201200439. [DOI] [PubMed] [Google Scholar]
  • 39.Sirotkin A.V., Harrath A.H. Phytoestrogens and their effects. Eur J Pharmacol. 2014;741:230–236. doi: 10.1016/j.ejphar.2014.07.057. [DOI] [PubMed] [Google Scholar]
  • 40.Anderson J.J., Garner S.C. Phytoestrogens and bone. Baillieres Clin Endocrinol Metab. 1998;12:543–557. doi: 10.1016/s0950-351x(98)80003-7. [DOI] [PubMed] [Google Scholar]
  • 41.Patisaul H.B., Jefferson W. The pros and cons of phytoestrogens. Front Neuroendocrinol. 2010;31:400–419. doi: 10.1016/j.yfrne.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Glazier M.G., Bowman M.A. A review of the evidence for the use of phytoestrogens as a replacement for traditional estrogen replacement therapy. Arch Inter Med. 2001;161:1161–1172. doi: 10.1001/archinte.161.9.1161. [DOI] [PubMed] [Google Scholar]
  • 43.Horn-Ross P.L., Barnes S., Lee M., Coward L., Mandel J.E., Koo J., John E.M., Smith M. Assessing phytoestrogen exposure in epidemiologic studies: development of a database (United States) Cancer Causes Control. 2000;11:289–298. doi: 10.1023/a:1008995606699. [DOI] [PubMed] [Google Scholar]
  • 44.Chen M.N., Lin C.C., Liu C.F. Efficacy of phytoestrogens for menopausal symptoms: a meta-analysis and systematic review. Climacteric. 2015;18:260–269. doi: 10.3109/13697137.2014.966241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dahlman-Wright K., Cavailles V., Fuqua S.A., Jordan V.C., Katzenellenbogen J.A., Korach K.S., Maggi A., Muramatsu M., Parker M.G., Gustafsson J.A. International Union of Pharmacology. LXIV. Estrogen receptors. Pharmacol Rev. 2006;58:773–781. doi: 10.1124/pr.58.4.8. [DOI] [PubMed] [Google Scholar]
  • 46.Heldring N., Pike A., Andersson S., Matthews J., Cheng G., Hartman J., Tujague M., Strom A., Treuter E., Warner M. Estrogen receptors: how do they signal and what are their targets. Physiol Rev. 2007;87:905–931. doi: 10.1152/physrev.00026.2006. [DOI] [PubMed] [Google Scholar]
  • 47.McKeage K., Curran M.P., Plosker G.L. Fulvestrant: a review of its use in hormone receptor-positive metastatic breast cancer in postmenopausal women with disease progression following antiestrogen therapy. Drugs. 2004;64:633–648. doi: 10.2165/00003495-200464060-00009. [DOI] [PubMed] [Google Scholar]
  • 48.Klinge C.M. Estrogen receptor interaction with co-activators and co-repressors. Steroids. 2000;65:227–251. doi: 10.1016/s0039-128x(99)00107-5. [DOI] [PubMed] [Google Scholar]
  • 49.Prossnitz E.R., Barton M. Estrogen biology: new insights into GPER function and clinical opportunities. Mol Cell Endocrinol. 2014;389:71–83. doi: 10.1016/j.mce.2014.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Filardo E.J., Quinn J.A., Frackelton A.R., Jr., Bland K.I. Estrogen action via the G protein-coupled receptor, GPR30: stimulation of adenylyl cyclase and cAMP-mediated attenuation of the epidermal growth factor receptor-to-MAPK signaling axis. Mol Endocrinol. 2002;16:70–84. doi: 10.1210/mend.16.1.0758. [DOI] [PubMed] [Google Scholar]
  • 51.Filardo E.J., Quinn J.A., Bland K.I., Frackelton A.R., Jr. Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol Endocrinol. 2000;14:1649–1660. doi: 10.1210/mend.14.10.0532. [DOI] [PubMed] [Google Scholar]
  • 52.Levin E.R., Pietras R.J. Estrogen receptors outside the nucleus in breast cancer. Breast Cancer Res Treat. 2008;108:351–361. doi: 10.1007/s10549-007-9618-4. [DOI] [PubMed] [Google Scholar]
  • 53.Levin E.R. Elusive extranuclear estrogen receptors in breast cancer. Clin Cancer Res. 2012;18:6–8. doi: 10.1158/1078-0432.CCR-11-2547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chambliss K.L., Wu Q., Oltmann S., Konaniah E.S., Umetani M., Korach K.S., Thomas G.D., Mineo C., Yuhanna I.S., Kim S.H. Non-nuclear estrogen receptor alpha signaling promotes cardiovascular protection but not uterine or breast cancer growth in mice. J Clin Invest. 2010;120:2319–2330. doi: 10.1172/JCI38291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Setchell K.D. Phytoestrogens: the biochemistry, physiology, and implications for human health of soy isoflavones. Am J Clin Nutr. 1998;68:1333S–1346S. doi: 10.1093/ajcn/68.6.1333S. [DOI] [PubMed] [Google Scholar]
  • 56.Barone M., Tanzi S., Lofano K., Scavo M.P., Guido R., Demarinis L., Principi M.B., Bucci A., Di Leo A. Estrogens, phytoestrogens and colorectal neoproliferative lesions. Genes Nutr. 2008;3:7–13. doi: 10.1007/s12263-008-0081-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Miyazaki K. Novel approach for evaluation of estrogenic and anti-estrogenic activities of genistein and daidzein using B16 melanoma cells and dendricity assay. Pigment Cell Res. 2004;17:407–412. doi: 10.1111/j.1600-0749.2004.00167.x. [DOI] [PubMed] [Google Scholar]
  • 58.Sun Z., Biela L.M., Hamilton K.L., Reardon K.F. Concentration-dependent effects of the soy phytoestrogen genistein on the proteome of cultured cardiomyocytes. J Proteomics. 2012;75:3592–3604. doi: 10.1016/j.jprot.2012.04.001. [DOI] [PubMed] [Google Scholar]
  • 59.Weihua Z., Saji S., Makinen S., Cheng G., Jensen E.V., Warner M., Gustafsson J.A. Estrogen receptor (ER) beta, a modulator of ERalpha in the uterus. Proc Natl Acad Sci U S A. 2000;97:5936–5941. doi: 10.1073/pnas.97.11.5936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lindberg M.K., Moverare S., Skrtic S., Gao H., Dahlman-Wright K., Gustafsson J.A., Ohlsson C. Estrogen receptor (ER)-beta reduces ERalpha-regulated gene transcription, supporting a “ying yang” relationship between ERalpha and ERbeta in mice. Mol Endocrinol. 2003;17:203–208. doi: 10.1210/me.2002-0206. [DOI] [PubMed] [Google Scholar]
  • 61.Nguyen C.T., Luong T.T., Kim G.L., Pyo S., Rhee D.K. Korean Red Ginseng inhibits apoptosis in neuroblastoma cells via estrogen receptor beta-mediated phosphatidylinositol-3 kinase/Akt signaling. J Ginseng Res. 2015;39:69–75. doi: 10.1016/j.jgr.2014.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kim E.H., Kim I.H., Ha J.A., Choi K.T., Pyo S., Rhee D.K. Antistress effect of red ginseng in brain cells is mediated by TACE repression via PADI4. J Ginseng Res. 2013;37:315–323. doi: 10.5142/jgr.2013.37.315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Cho J., Park W., Lee S., Ahn W., Lee Y. Ginsenoside-Rb1 from Panax ginseng C.A. Meyer activates estrogen receptor-alpha and -beta, independent of ligand binding. J Clin Endocrinol Metab. 2004;89:3510–3515. doi: 10.1210/jc.2003-031823. [DOI] [PubMed] [Google Scholar]
  • 64.Leung K.W., Cheung L.W., Pon Y.L., Wong R.N., Mak N.K., Fan T.P., Au S.C., Tombran-Tink J., Wong A.S. Ginsenoside Rb1 inhibits tube-like structure formation of endothelial cells by regulating pigment epithelium-derived factor through the oestrogen beta receptor. Br J Pharmacol. 2007;152:207–215. doi: 10.1038/sj.bjp.0707359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hewitt S.C., Korach K.S. Oestrogen receptor knockout mice: roles for oestrogen receptors alpha and beta in reproductive tissues. Reproduction. 2003;125:143–149. doi: 10.1530/rep.0.1250143. [DOI] [PubMed] [Google Scholar]
  • 66.Nogawa S., Forster C., Zhang F., Nagayama M., Ross M.E., Iadecola C. Interaction between inducible nitric oxide synthase and cyclooxygenase-2 after cerebral ischemia. Proc Natl Acad Sci U S A. 1998;95:10966–10971. doi: 10.1073/pnas.95.18.10966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Handa R.J., Ogawa S., Wang J.M., Herbison A.E. Roles for oestrogen receptor beta in adult brain function. J Neuroendocrinol. 2012;24:160–173. doi: 10.1111/j.1365-2826.2011.02206.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Jensen E.V., Jacobson H.I., Walf A.A., Frye C.A. Estrogen action: a historic perspective on the implications of considering alternative approaches. Physiol Behav. 2010;99:151–162. doi: 10.1016/j.physbeh.2009.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kim I.H., Kim S.K., Kim E.H., Kim S.W., Sohn S.H., Lee S.C., Choi S., Pyo S., Rhee D.K. Korean Red Ginseng up-regulates C21-steroid hormone metabolism via Cyp11a1 gene in senescent rat testes. J Ginseng Res. 2011;35:272–282. doi: 10.5142/jgr.2011.35.3.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lavoie H.A., King S.R. Transcriptional regulation of steroidogenic genes: STARD1, CYP11A1 and HSD3B. Exp Biol Med (Maywood) 2009;234:880–907. doi: 10.3181/0903-MR-97. [DOI] [PubMed] [Google Scholar]
  • 71.Papapetropoulos A. A ginseng-derived oestrogen receptor beta (ERbeta) agonist, Rb1 ginsenoside, attenuates capillary morphogenesis. Br J Pharmacol. 2007;152:172–174. doi: 10.1038/sj.bjp.0707360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hao K., Gong P., Sun S.Q., Hao H.P., Wang G.J., Dai Y., Chen Y.C., Liang Y., Xie L., Li F.Y. Mechanism-based pharmacokinetic-pharmacodynamic modeling of the estrogen-like effect of ginsenoside Rb1 on neural 5-HT in ovariectomized mice. Eur J Pharm Sci. 2011;44:117–126. doi: 10.1016/j.ejps.2011.06.014. [DOI] [PubMed] [Google Scholar]
  • 73.Hashimoto R., Yu J., Koizumi H., Ouchi Y., Okabe T. Ginsenoside Rb1 prevents MPP(+)-induced apoptosis in PC12 cells by stimulating estrogen receptors with consequent activation of ERK1/2, Akt and inhibition of SAPK/JNK, p38 MAPK. Evid Based Complement Alternat Med. 2012;2012:693717. doi: 10.1155/2012/693717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cheng W., Wu D., Zuo Q., Wang Z., Fan W. Ginsenoside Rb1 prevents interleukin-1 beta induced inflammation and apoptosis in human articular chondrocytes. Int Orthop. 2013;37:2065–2070. doi: 10.1007/s00264-013-1990-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen W.F., Lau W.S., Cheung P.Y., Guo D.A., Wong M.S. Activation of insulin-like growth factor I receptor-mediated pathway by ginsenoside Rg1. Br J Pharmacol. 2006;147:542–551. doi: 10.1038/sj.bjp.0706640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gao Q.G., Chan H.Y., Man C.W., Wong M.S. Differential ERalpha-mediated rapid estrogenic actions of ginsenoside Rg1 and estren in human breast cancer MCF-7 cells. J Steroid Biochem Mol Biol. 2014;141:104–112. doi: 10.1016/j.jsbmb.2014.01.014. [DOI] [PubMed] [Google Scholar]
  • 77.Zhang X., Wang J., Xing Y., Gong L., Li H., Wu Z., Li Y., Wang J., Wang Y., Dong L. Effects of ginsenoside Rg1 or 17beta-estradiol on a cognitively impaired, ovariectomized rat model of Alzheimer's disease. Neuroscience. 2012;220:191–200. doi: 10.1016/j.neuroscience.2012.06.027. [DOI] [PubMed] [Google Scholar]
  • 78.Shi C., Zheng D.D., Fang L., Wu F., Kwong W.H., Xu J. Ginsenoside Rg1 promotes nonamyloidgenic cleavage of APP via estrogen receptor signaling to MAPK/ERK and PI3K/Akt. Biochim Biophys Acta. 2012;1820:453–460. doi: 10.1016/j.bbagen.2011.12.005. [DOI] [PubMed] [Google Scholar]
  • 79.Gao Q.G., Chen W.F., Xie J.X., Wong M.S. Ginsenoside Rg1 protects against 6-OHDA-induced neurotoxicity in neuroblastoma SK-N-SH cells via IGF-I receptor and estrogen receptor pathways. J Neurochem. 2009;109:1338–1347. doi: 10.1111/j.1471-4159.2009.06051.x. [DOI] [PubMed] [Google Scholar]
  • 80.Lu X.Z., Wang J.H., Wu X., Zhou L., Wang L., Zhang X.W., Cao K.J., Huang J. Ginsenoside Rg1 promotes bone marrow stromal cells proliferation via the activation of the estrogen receptor-mediated signaling pathway. Acta Pharmacol Sin. 2008;29:1209–1214. doi: 10.1111/j.1745-7254.2008.00874.x. [DOI] [PubMed] [Google Scholar]
  • 81.Chan R.Y., Chen W.F., Dong A., Guo D., Wong M.S. Estrogen-like activity of ginsenoside Rg1 derived from Panax notoginseng. J Clin Endocrinol Metab. 2002;87:3691–3695. doi: 10.1210/jcem.87.8.8717. [DOI] [PubMed] [Google Scholar]
  • 82.Hien T.T., Kim N.D., Pokharel Y.R., Oh S.J., Lee M.Y., Kang K.W. Ginsenoside Rg3 increases nitric oxide production via increases in phosphorylation and expression of endothelial nitric oxide synthase: essential roles of estrogen receptor-dependent PI3-kinase and AMP-activated protein kinase. Toxicol Appl Pharmacol. 2010;246:171–183. doi: 10.1016/j.taap.2010.05.008. [DOI] [PubMed] [Google Scholar]
  • 83.Nakaya Y., Mawatari K., Takahashi A., Harada N., Hata A., Yasui S. The phytoestrogen ginsenoside Re activates potassium channels of vascular smooth muscle cells through PI3K/Akt and nitric oxide pathways. J Med Invest. 2007;54:381–384. doi: 10.2152/jmi.54.381. [DOI] [PubMed] [Google Scholar]
  • 84.Furukawa T., Bai C.X., Kaihara A., Ozaki E., Kawano T., Nakaya Y., Awais M., Sato M., Umezawa Y., Kurokawa J. Ginsenoside Re, a main phytosterol of Panax ginseng, activates cardiac potassium channels via a nongenomic pathway of sex hormones. Mol Pharmacol. 2006;70:1916–1924. doi: 10.1124/mol.106.028134. [DOI] [PubMed] [Google Scholar]
  • 85.Sun B., Xiao J., Sun X.B., Wu Y. Notoginsenoside R1 attenuates cardiac dysfunction in endotoxemic mice: an insight into oestrogen receptor activation and PI3K/Akt signalling. Br J Pharmacol. 2013;168:1758–1770. doi: 10.1111/bph.12063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Wang T., Wan D., Shao L., Dai J., Jiang C. Notoginsenoside R1 stimulates osteogenic function in primary osteoblasts via estrogen receptor signaling. Biochem Biophys Res Commun. 2015;466:232–239. doi: 10.1016/j.bbrc.2015.09.014. [DOI] [PubMed] [Google Scholar]
  • 87.Lee Y., Jin Y., Lim W., Ji S., Choi S., Jang S., Lee S. A ginsenoside-Rh1, a component of ginseng saponin, activates estrogen receptor in human breast carcinoma MCF-7 cells. J Steroid Biochem Mol Biol. 2003;84:463–468. doi: 10.1016/s0960-0760(03)00067-0. [DOI] [PubMed] [Google Scholar]
  • 88.Leung K.W., Leung F.P., Mak N.K., Tombran-Tink J., Huang Y., Wong R.N. Protopanaxadiol and protopanaxatriol bind to glucocorticoid and oestrogen receptors in endothelial cells. Br J Pharmacol. 2009;156:626–637. doi: 10.1111/j.1476-5381.2008.00066.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kim M.S., Lim H.J., Yang H.J., Lee M.S., Shin B.C., Ernst E. Ginseng for managing menopause symptoms: a systematic review of randomized clinical trials. J Ginseng Res. 2013;37:30–36. doi: 10.5142/jgr.2013.37.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Oh K.J., Chae M.J., Lee H.S., Hong H.D., Park K. Effects of Korean Red Ginseng on sexual arousal in menopausal women: placebo-controlled, double-blind crossover clinical study. J Sex Med. 2010;7:1469–1477. doi: 10.1111/j.1743-6109.2009.01700.x. [DOI] [PubMed] [Google Scholar]
  • 91.Kim S.Y., Seo S.K., Choi Y.M., Jeon Y.E., Lim K.J., Cho S., Choi Y.S., Lee B.S. Effects of red ginseng supplementation on menopausal symptoms and cardiovascular risk factors in postmenopausal women: a double-blind randomized controlled trial. Menopause. 2012;19:461–466. doi: 10.1097/gme.0b013e3182325e4b. [DOI] [PubMed] [Google Scholar]
  • 92.Kim H., Yoon Y., Lee J., Lee C., Jang J., Lee K., Cho J. A clinical study on the effect of red ginseng for postmenopausal hot flushes. J Orient Obstet Gynecol. 2009;22:132–139. [Google Scholar]
  • 93.Wiklund I.K., Mattsson L.A., Lindgren R., Limoni C. Effects of a standardized ginseng extract on quality of life and physiological parameters in symptomatic postmenopausal women: a double-blind, placebo-controlled trial. Swedish Alternative Medicine Group. Int J Clin Pharmacol Res. 1999;19:89–99. [PubMed] [Google Scholar]
  • 94.Jang D.J., Lee M.S., Shin B.C., Lee Y.C., Ernst E. Red ginseng for treating erectile dysfunction: a systematic review. Br J Clin Pharmacol. 2008;66:444–450. doi: 10.1111/j.1365-2125.2008.03236.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kim S.W., Paick J.S. Clinical efficacy of Korean Red Ginseng on vasculogenic impotent patients. Korean J Androl. 1999;17:23–28. [Google Scholar]
  • 96.Ham W.S., Kim W.T., Lee J.S., Ju H.J., Kang S.J., Oh J.H., Her Y., Chung J.Y., Park K., Choi Y.D. Efficacy and safety of red ginseng extract powder in patients with erectile dysfunction: multicenter, randomized, double-blind, placebo-controlled study. Korean J Urol. 2009;50:159–164. [Google Scholar]
  • 97.Chung H.S., Hwang I., Oh K.J., Lee M.N., Park K. The effect of Korean Red Ginseng on sexual function in premenopausal women: placebo-controlled, double-blind, crossover clinical trial. Evid Based Complement Alternat Med. 2015;2015:913158. doi: 10.1155/2015/913158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kim D.I., Choi M.S., Alm H.Y. Efficacy and safety of red ginseng on women's health related quality of life and sexual function. J Ginseng Res. 2009;33:115–126. [Google Scholar]
  • 99.Heinlein C.A., Chang C. The roles of androgen receptors and androgen-binding proteins in nongenomic androgen actions. Mol Endocrinol. 2002;16:2181–2187. doi: 10.1210/me.2002-0070. [DOI] [PubMed] [Google Scholar]
  • 100.Lee D.K., Chang C. Molecular communication between androgen receptor and general transcription machinery. J Steroid Biochem Mol Biol. 2003;84:41–49. doi: 10.1016/s0960-0760(03)00005-0. [DOI] [PubMed] [Google Scholar]
  • 101.Hiort O., Holterhus P.M. Androgen insensitivity and male infertility. Int J Androl. 2003;26:16–20. doi: 10.1046/j.1365-2605.2003.00369.x. [DOI] [PubMed] [Google Scholar]
  • 102.McPhaul M.J. Androgen receptor mutations and androgen insensitivity. Mol Cell Endocrinol. 2002;198:61–67. doi: 10.1016/s0303-7207(02)00369-6. [DOI] [PubMed] [Google Scholar]
  • 103.Melo C.O., Danin A.R., Silva D.M., Tacon J.A., Moura K.K., Costa E.O., da Cruz A.D. Association between male infertility and androgen receptor mutations in Brazilian patients. Genet Mol Res. 2010;9:128–133. doi: 10.4238/vol9-1gmr661. [DOI] [PubMed] [Google Scholar]
  • 104.Kopalli S.R., Won Y.-J., Hwang S.-Y., Cha K.-M., Kim S.-Y., Han C.-K., Lee S.-H., Hong J.-Y., Kim S.-K. Korean Red Ginseng protects against doxorubicin-induced testicular damage: an experimental study in rats. J Funct Foods. 2016;20:96–107. [Google Scholar]
  • 105.Kopalli S.R., Cha K.-M., Jeong M.-S., Lee S.-H., Sung J.-H., Seo S.-K., Kim S.-K. Pectinase-treated Panax ginseng ameliorates hydrogen peroxide-induced oxidative stress in GC-2 sperm cells and modulates testicular gene expression in aged rats. J Ginseng Res. 2016;40:185–195. doi: 10.1016/j.jgr.2015.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wang H., Feng L., Chu Z., Yu N., Yang Q., Zhang Z., Wang L., Wu Y. Study on the changes of rat testis androgen receptor mRNA expression and plasma testosterone after cold stress. Lishizhen Med Mater Med Res. 2008;4:929–930. [Google Scholar]

Articles from Journal of Ginseng Research are provided here courtesy of Elsevier

RESOURCES