Skip to main content

REVIEW article

Front. Pharmacol., 28 April 2023
Sec. Ethnopharmacology
This article is part of the Research Topic New Insights in the Pharmacological Research, Quality Control, and Pharmaceutical Analysis of Panax Notoginseng View all 3 articles

Saponins of ginseng products: a review of their transformation in processing

Xian-Wen Ye,,Xian-Wen Ye1,2,3Chun-Shuai LiChun-Shuai Li1Hai-Xia ZhangHai-Xia Zhang1Qian LiQian Li1Shui-Qing ChengShui-Qing Cheng1Jia WenJia Wen1Xuan WangXuan Wang1Hong-Min RenHong-Min Ren1Liang-Jing XiaLiang-Jing Xia1Xu-Xing WangXu-Xing Wang1Xin-Fang Xu,,
Xin-Fang Xu1,2,3*Xiang-Ri Li,
Xiang-Ri Li1,2*
  • 1Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
  • 2Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
  • 3Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China

The primary processed product of Panax ginseng C.A. Meyer (P. ginseng) is red ginseng. As technology advances, new products of red ginseng have arisen. Red ginseng products, e.g., traditional red ginseng, sun ginseng, black ginseng, fermented red ginseng, and puffed red ginseng, are commonly used in herbal medicine. Ginsenosides are the major secondary metabolites of P. ginseng. The constituents of P. ginseng are significantly changed during processing, and several pharmacological activities of red ginseng products are dramatically increased compared to white ginseng. In this paper, we aimed to review the ginsenosides and pharmacological activities of various red ginseng products, the transformation law of ginsenosides in processing, and some clinical trials of red ginseng products. This article will help to highlight the diverse pharmacological properties of red ginseng products and aid in the future development of red ginseng industrialization.

1 Introduction

Panax ginseng C.A. Meyer (Panax ginseng) is an ancient Chinese medicinal material used in Asian nations for over 2,000 years (Kennedy and Scholey, 2003). It has been listed as a medicinal herb in Shennong Bencao Jing, a standard Chinese herbal dictionary. P. ginseng, Panax quinquefolius L., and P. notoginseng (Burk) F.H. Chen (P. notoginseng), all from the Araliaceae family, are widely used herbs. Several studies over the past few decades have shown that P. ginseng and P. notoginseng have various pharmacological effects on immunological and neurological system disorders (Liu et al., 2020). The kinds and concentrations of their primary active ingredients, saponins, may be altered throughout the steaming process, and the therapeutic efficacies of raw and steaming P. ginseng and P. notoginseng vary. These variations in saponins are causally significant (Zhang et al., 2019). Meanwhile, according to traditional Chinese medicine (TCM) theory, their uses differ, since P. ginseng strengthens vital energy while P. notoginseng encourages blood circulation (Xiong et al., 2022).

P. ginseng is often processed into white ginseng (WG) and traditional red ginseng (TRG), most well-used in clinical applications for their great pharmacological activity. In TCM, the steaming method of P. ginseng is initially listed in the Complete Manual of Experience in the Treatment of Sores. The character of TRG is detailed and described in The Ming dynasty’s Enlightening Primer of Materia Medica. In steaming processing, the quality of TRG improves with an increase of P. ginseng cultivation age. Typically, TRG is produced by six-year-old ginseng in Korea. In addition, the unique and advanced processing technology makes Korean red ginseng (KRG) predominate the world ginseng market. With the development of steaming, fermenting, and puffing technology, many new red ginseng products are being produced, including sun ginseng (SG), black ginseng (BG), fermented red ginseng (FRG), and puffed red ginseng (PRG). These process conditions directly influence the pharmacological activity of red ginseng.

It is widely known ginsenosides can be classified into three types: the protopanaxadiol type (Rb1, Rc, Rb2, and Rd); the protopanaxatriol type (ginsenoside Rg1 and Re); and the oleanolic acid type (ginsenoside Ro and polyacetylene ginsenoside Ro) according to different aglycones (Xu et al., 2014). The various structures of ginsenosides endow them with rich pharmacological activities, such as antioxidation, anti-inflammation, anti-apoptosis, and so on (Choi et al., 2014; Li et al., 2014). Additionally, ginsenosides can be divided into major and rare ginsenosides according to the different content of ginseng, which all have significant pharmacological activities (Wei et al., 2011).

The major ginsenosides occur in WG, TRG, and other new types of red ginseng. In contrast, rare ginsenosides are present in TRG at a trim level, including Rg2, Rg3, Rh1, and Rh2. However, compared with TRG, the rare ginsenosides are abundant in the new types of red ginseng. The types and contents of ginsenosides in different red ginseng products result in various pharmacological activities, and the relationship between ginsenosides and their bioactivities help in the application of red ginseng products in clinical settings.

We aimed to review the relevant clinical studies of red ginseng products and summarize the discovered ginsenoside components of red ginseng. Furthermore, we discuss the structure–functional relationship of ginsenosides. The transformation law of ginsenosides in red ginseng processing are revealed to illustrate the medicinal composition transformation. The other pharmacological activities of red ginseng, such as TRG, SG, BG, FRG, and PRG, are also discussed. We summarize the process and character of the new type of red ginseng, which provides the basis for further research to facilitate the development of red ginseng industrialization in the future.

2 Red ginseng products

2.1 TRG

Generally, TRG is steamed at 90°C–100°C for 2-3 h and then dried (Chung et al., 2014). The large-scale application of TRG began in the Qing Dynasty. According to TCM theory, WG is used to “supply qi and promote the production of body fluids” and enhance physical fitness and disease resistance. In contrast, TRG is often used to “boost yang” and replenishing vital essence with the “warming effect” (Zhang et al., 2012).

During TRG processing, one ginsenoside can be transformed into another by demalonylating, decarboxylating, deglycosylating, and dehydrating (Figure 1). Compared with WG, the rare ginsenosides are the characteristic compound with significant pharmacological activities. TRG exhibits more potent anticancer activity than WG due to the abundance of rare ginsenosides generated from processing (Li et al., 2011; Kim J. H. et al., 2014), which has been further developed into drugs and health products. The pharmacological activity of TRG focuses on anti-aging (Peng et al., 2021), treating erectile dysfunction (Jang et al., 2008), immune-modulating (Kim et al., 2021), the antidepressant effect (Lee et al., 2020), and anti-inflammation (Min et al., 2022). In addition, TRG can inhibit tau aggregation and promote tau dissociation in vitro, which can be a potential therapeutic agent to treat neurodegenerative diseases (Shin et al., 2020).

FIGURE 1
www.frontiersin.org

FIGURE 1. Various types of red ginseng.

2.2 SG

SG is prepared by steaming fresh ginseng at a temperature of 120°C or higher, which is a higher temperature than during TRG processing (Figure 1). SG contains approximately equal amounts of three major ginsenosides, Rg3, Rg5, and Rk1, in a higher concentration than TRG. One study (Kim et al., 2000) first reported this processed red ginseng, which is more potent in its ability to induce endothelium-dependent relaxation and free radical scavenging activity.

Compared with TRG, SG contains an abundance of ginsenosides Rg3, Rg5, and Rk1 (Kwon et al., 2001). In addition, new acetylated ginsenosides (Rs4, Rs5, Rs6, and Rs7) and dammarane glycosides (Rk1, Rk2, and Rk3) have been isolated from SG (Park et al., 2002a; Park et al., 2002b). These different types and amounts of ginsenosides endow SG with more potent pharmacological effects than TRG under certain pathological conditions. SG reportedly serves several functions, including free radical scavenging (Kang et al., 2006), peroxynitrite scavenging (Kang et al., 2009), antitumor-promoting (Song et al., 2012), antihyperglycemic (Jiao et al., 2014), and cytoprotection activities (Lee C. S. et al., 2012). Moreover, SG has memory-enhancing activities (Lee C. H. et al., 2013), and enhances cognitive function in patients with moderately severe Alzheimer’s disease (Heo et al., 2012).

2.3 BG

Due to repeated steaming and drying processes, BG is named according to its surface color change (Figure 1). The nine-time repeated steaming and drying is a typical method in TCM, mainly to rectify the properties and increase the components of Chinese medicine. Observations indicate that BG improves at temperatures of 95°C or higher, at least from the appearance (Oh et al., 2021). Multiple steaming and drying can enhance the antibacterial activity of BG (Lee H. W. et al., 2021). Compared with the TRG, the total saponins of BG are absorbed faster in the gut and exposed more widely (Yoo et al., 2021). In recent years, this method has been used to manufacture functional food.

After nine repeated steaming cycles, ginseng gradually becomes BG, with increased Rg3 content (Kim, 2015). Nineteen ginsenosides (Rg1, Re, Rf, Rb1, Rc, Rb2, Rd, F4, Rg6, Rk3, Rh4, 20(S)-, 20(R)-Rg3, 20(S)-, 20(R)-Rs3, Rk1, Rg5, Rs4, and Rs5) have been determined to be found in BG; among them, the ginsenosides Rg3, Rg5, and Rk1 are the main components (Sun et al., 2009). Research has also revealed that the total ginsenosides increase with number of steam cycles (Lee H. et al., 2012), and that the fructose in BG is 44 times that present in WG and 18.3 times that in RG (Zhu et al., 2019).

In animal and cell culture models, BG plays a role in the prevention and treatment of diseases such as cognitive impairment (Park et al., 2011), obesity (Park et al., 2019), fetal alcohol syndrome (Lee et al., 2009), and breast cancer (Kim and Kim, 2015). BG is a potential anti-aging supplementation, which can reduce the activation of p53-dependent p21 and p16 classical aging pathways in the liver, skeletal muscle, and white fat (Lee S. et al., 2022). BG can prevent liver injury by resisting oxidative stress, regulating lipid and glucose metabolism, and reducing inflammation and TLR4/NF-κB axis (Jiang et al., 2021; Wei et al., 2022). BG may maintain endothelial integrity by activating Akt, reducing vascular protein leakage, leukocyte infiltration, and proinflammatory factor release in alveolar lavage fluid, and thus plays a protective role against particle-induced lung injury and vascular hyperpermeability (Lee et al., 2019; Kim et al., 2022).

2.4 FRG

Fermenting food through edible microorganisms can produce other active small molecular compounds, which has aroused the interest of most food scientists and dietitians (Oh et al., 2015). Similarly, the fermentation applied in the red ginseng process, which is called FRG, produces a significant change in ginsenoside derivatives and possesses great pharmacological activity (Irfan et al., 2022), such as anti-inflammatory (Kim et al., 2018; Bae et al., 2021), antioxidant (Saba et al., 2018), anti-allergy (Kim et al., 2019), anti-diabetes (Jang et al., 2017), anti-anxiety (Han et al., 2020), and so on. There is another FRG, which involves ginseng steaming, extracting, fermenting, and freeze-drying. The pharmacodynamic function of ginseng after fermentation is improved, which may be related to the increase in ingestion rates and absorption levels of P. ginseng (Lee et al., 2015).

Bacteria and fungi, such as Lactobacillus, Bifidobacterium, Saccharomyces cerevisiae, and Red-Koji, mainly perform the fermentation. These microbes are essential in transforming ginseng components (Lee, et al., 2015; Choi et al., 2016). Moreover, mushroom mycelia can provide the microbial environment needed in ginseng fermentation (Bae et al., 2011). Interestingly, fermentation creates a new small molecule, Compound K (CK). CK is transformed from Rb1, Rb2, Rc, and Rd, the metabolite of FRG digested by intestinal microorganisms, and has considerable activity against diabetes, cancer, and immune stimulation (Jung et al., 2019; Kim et al., 2020). In addition, a large amount of Rg3 is fermented into Rh2 by lactic acid bacteria, which have obvious anti-tumor, anti-allergic, and anti-inflammatory activities (Bae et al., 2004; Trinh et al., 2007).

Purple-red ginseng is prepared by fresh ginseng and then through fermentation, steam ripening, and low-temperature ripening. This processing uses bilberry, sugar, and bulgaricus. The ginsenoside, Rg5, is the most abundant ingredient of this FRG. In addition, this FRG might be a beneficial therapeutic supplementary substitute for metabolic syndrome with the efficacy to improve insulin sensitivity and lower postprandial glucose levels (Kho et al., 2016; Lee S. J. et al., 2022).

2.5 PRG

After puffing, the texture of ginseng is loose and porous, and easy to dehydrate. The process goes through rapid heating at atmospheric pressure and immediate pressure reduction, and the product is what we often call PRG (Hoseney, 1986).

The red ginseng was puffed with rice to avoid a burning phenomenon at high temperatures (An et al., 2011). PRG shows higher extraction yield and crude saponin content than non-puffed ginseng (Kim et al., 2008). The primary ginsenosides (Rb1, Rb2, Rc, Rd, Re, and Rg1) are effectively converted into minor ginsenosides (F2, Rg3, Rk1, and Rg5) by puffing, which is similar to the effect of the steaming process on the content of transformed ginsenosides. However, the time required for the transformation of ginsenosides by steaming (4–36 days) is much longer than by puffing (less than 30 min) (Shin et al., 2019). During in vitro experiments, the ethanolic extract of PRG shows a more vital antioxidant capacity than TRG. In addition, more robust antioxidant properties are observed in bulk oil and oil-water emulsions (Lee et al., 2018).

3 Ginsenosides in red ginseng and transformation in processing

3.1 Ginsenosides in red ginseng

Ginsenosides are the main active components in red ginseng. So far, about 100 ginsenosides have been discovered in red ginseng (Table 1), and the number is still increasing. In red ginseng products, protopanaxadiol (PPD), protopanaxatriol (PPT), and oleanane (OLE) ginsenosides are the main classification of ginsenosides (Figure 2). Ginsenoside is a triterpenoid saponin with a dammarane skeleton. The ether bond combines carbon-3, carbon-6, or carbon-20 with glycosyl residue. Carbon-20 is a chiral carbon atom presenting the 20(S) and 20(R) epimers, such as ginsenoside Rg3, Rh2, Rs3, Rg2, and Rh1. Furthermore, dehydration in the carbon-20 can present positional isomers of the double bond at carbon-20 (21) or carbon-20 (22), while the double bond at carbon-20 (22) also presents cis-trans isomers, such as Rg9, F4, Rs6, Rh4, Rs4, Rg5, and Rh3.

TABLE 1
www.frontiersin.org

TABLE 1. Ginsenosides in red ginseng products.

FIGURE 2
www.frontiersin.org

FIGURE 2. Structure of protopanaxadiol (PPD), protopanaxatriol (PPT), and oleanolic acid (OA).

The structure of ginsenoside is directly related to anticancer activities. Interestingly, in red ginseng processing, the position of sugar chain hydrolysis (carbon-3 > carbon-6 > carbon-20) results in different degrees of anticancer activity (Kai et al., 2015). The 20(S)-ginsenosides have more substantial anticancer potential than their 20(R)-stereoisomers (Wang et al., 2007; Seoyoung et al., 2009). Ginsenosides with a double bond at C-20 (21) exhibit more effective anticancer activities than those at C-20 (22) (Kai, et al., 2015).

During red ginseng processing, the polar compounds transform into less polar compounds, which exhibit different pharmacological activity from other red ginseng products. TRG, SG, BG, FRG, and PRG have many ginsenosides in common, such as the major ginsenosides Rb1, Rb2, Rc, Rd, Re, and Rg1. In contrast, the minor ginsenosides are different in content and variety. So, red ginseng product pharmacological activity is significantly different from the diverse ingredients.

3.2 Transformation of ginsenosides

Ginsenosides can cause a structure change during red ginseng processing. Malonyl-ginsenoside is an initial form of ginseng before processing, after which deglycosylation, decarboxylation, and dehydration will occur. For example, Rg2, Rh1, and Rg3 are deglycosylated from Re, Rg1, and Rd; Rg3 and Rg2 are dehydrated to form Rg5 and Rg6; and malonyl-ginsenoside Rb2 and Rc are decarboxylated to create Rs1 and Rs2 (Chen et al., 2020). Previous research has indicated that ginsenoside Rb1 and Rb2 can transform into ginsenoside Rg3, and ginsenoside Re can transform into ginsenoside Rg2 (Lee et al., 2008; Kim Y. J. et al., 2014). The Maillard reaction is also the primary chemical reaction in red ginseng processing, whereby products are generated whenever reducing sugars are heated with amino acids, peptides, or proteins (Chen and Kitts, 2012). These products are a significant source of compounds that enhance antioxidant activity through heat treatment (Chen and Kitts, 2011). In the Maillard reaction, ginsenosides provide the reducing sugars and then transform into other ginsenosides (Yamabe et al., 2013).

Before being processed into red ginseng, malonyl-20(S)-ginsenoside is a natural compound of ginseng. The deglycosylate in carbon-20 easily occurs after processing; however, the location of the 20(S) and 20(R) epimers remains to be determined. The 20(R)-ginsenosides are present through the OH group selective attack after the glycosyl residue elimination at carbon-20 during red ginseng processing (Kang et al., 2007). For example, the ginsenoside Rg3, Rh2, Rs3, Rg2, Rh1, and Rf both have the 20(S) and 20(R) epimers.

TRG processing is mainly conducted by decarbonylation, decarboxylation, and the first deglycosylation in carbon-20 of the dammarane skeleton. SG, BG, and PRG promote these reactions. The second deglycosylation is mainly conducted in carbon-20 of the dammarane skeleton and dehydration in this position. However, the FRG processes some characteristic ginsenosides, such as ginsenoside CK, for microbial metabolism. At the same time, the amount of some minor ginsenosides, such as ginsenoside Rh2, Rg1, and Rh1, also increase with the fermentation process. The conversion pathway of oleanane ginsenosides is similar to dammarane ginsenosides. The demethylation, debutylization, and deglycosylation of ester bonds can easily occur at carbon-3 and carbon-28. The ether bond is stable at carbon-3 in the oleanane skeleton, which is also stable between sugars in this position (Figures 36).

FIGURE 3
www.frontiersin.org

FIGURE 3. Structure of protopanaxadiol ginsenosides. (ara(f):α-L-arabinofuranosyl; ara(p):α-L-arabinopyranosyl; rha:α-L-ahamnopyranosyl; glc:β-D-glucopyranosyl; xyl:β-D-xylopyranosyl; mal:malonyl; Ac:acetyl).

FIGURE 4
www.frontiersin.org

FIGURE 4. Structure of protopanaxatriol ginsenosides. (ara(f):α-L-arabinofuranosyl; rha:α-L-ahamnopyranosyl; glc:β-D-glucopyranosyl; xyl:β-D-xylopyranosyl; mal:malonyl; bu:butyl; Ac:acetyl).

FIGURE 5
www.frontiersin.org

FIGURE 5. Structure of oleanolic acid ginsenosides. (glcA: β-D-glucuronosyl; glc: β-D-glucopyranosyl; me: methyl; bu: butyl).

FIGURE 6
www.frontiersin.org

FIGURE 6. Transformation of ginsenosides in red ginseng products. (ara(f):α-L-arabinofuranosyl; ara(p):α-L-arabinopyranosyl; rha:α-L-ahamnopyranosyl; glc:β-D-glucopyranosyl; glcA: β-D-glucuronosyl; xyl:β-D-xylopyranosyl; mal:malonyl; bu: butyl. The red arrow represents biotransformation).

4 20(R)- and 20(S)-ginsenoside isomerism

Ginsenosides, active ingredients of P. ginseng, like 20(R)-ginsenoside and 20(S)-ginsenoside, exist as stereoisomers depending on the position of the hydroxyl group on carbon-20. A literature survey shows that ginsenoside Rg3 and Rh2 are stereospecific in the stimulation of the pharmacological activity. 20 (S)-ginsenoside is the dominant conformation relative to 20(R)-ginsenoside in terms of some activity experiments.

In an in vitro study, the stereochemistry of the hydroxyl group at C-20 may play an important role in preventing rotavirus infection, and anticancer and osteoclastogenesis inhibitory activity. Ginsenoside Rb2 and its hydrolytic product; 20(S)-ginsenoside Rg3, but not 20(R)-ginsenoside Rg3, prevent rotavirus infection (Yang et al., 2018). The regulation of DNA methylation may play an important role in the inhibitory effect of ginsenoside Rg3 on the growth of the HepG2 cell line. The inhibitory effect of 20(S)-ginsenoside Rg3 is stronger than that of 20(R)-ginsenoside Rg3 (Teng et al., 2017). 20(S)-Ginsenoside Rh2 potently protects HepG2 cells cytotoxicity treated with tert-butyl hydroperoxide, but 20(S)-ginsenoside Rg3 weakly protects it (Lee et al., 2005). The ginsenoside 20(R)-ginsenoside Rh2, but not ginsenoside 20(S)-ginsenoside Rh2, shows selective osteoclastogenesis inhibitory activity without any cytotoxicity on osteoclastogenesis using RAW264 cells (Liu et al., 2009).

During in vivo experiments, ginsenoside isomers showed significant differences in improving immunity, survival ability, and alleviating diabetes symptoms. 20(R)- ginsenoside Rg3 has a more potent adjuvant activity of the immune response than 20(S)-ginsenoside Rg3 with highly upregulated serum IFN-γ and IL-5 (Wei et al., 2012). The novel characteristics of 20(S)-ginsenoside Rg3 exhibited higher pharmacological effects in insulin secretion and AMPK activation than 20(R)-ginsenoside Rg3, suggesting that ginsenoside Rg3 epimers show differential activities; 20(S)- ginsenoside Rg3 may be a valuable candidate for an anti-diabetic agent (Park et al., 2008). 20(S)-ginsenoside Rg3 promotes angiogenesis by activating the AKT/ERK-eNOS signal pathway, and its activity is significantly more potent than that of 20(R)-ginsenoside Rg3 (Kwok et al., 2012). In addition, ginsenosides can also maintain body growth with a supplement of 20(S)-, but not 20(R)-ginsenoside Rg3 in a cholesterol-deprived medium (Lee et al., 2011).

The hydroxyl stereochemistry at C-20 has a great influence on the activity of ginsenosides. In the production process, the processing method of red ginseng should be changed according to the actual demand to achieve the maximum transformation of the target ginsenosides. For example, under optimum reaction conditions, the actual 20(R)-ginsenoside Rg3 converts PPD ginsenosides (Sun et al., 2013). As mentioned, ginsenoside Rg3, Rh2, Rs3, Rg2, Rh1, and Rf have the 20(S) and 20(R) epimers. There are few studies regarding the structure-activity relationship of other ginsenosides, which is still an urgent problem to be solved. In the future, these problems should be explored to determine the greater value of red ginseng.

5 Clinical trials of red ginseng

ClinicalTrials.gov is a database of privately and publicly funded clinical studies conducted worldwide. Through the ClinicalTrials.gov platform (https://clinicaltrials.gov/), a search of the term “red ginseng” found 78 research projects; while the exclusion of all other forms of ginseng other than red ginseng obtained 39 clinical studies (Figure 7); only six of these research projects were successfully concluded and the results were reported. These results suggest that KRG is efficacious as an adjuvant treatment for patients experiencing residual symptoms of major depression (Jeong et al., 2015), reducing proinflammatory cytokines and fatigue in overweight patients with non-alcoholic fatty liver disease (Hong et al., 2016), and protecting subjects from contracting acute respiratory illness (Lee S. A. et al., 2012). KRG may have beneficial effects for dry mouth in women, especially in those of menopausal age, but not in men (Park et al., 2010). There is no evidence that KRG has an effect on blood pressure, fasting blood glucose, or arterial stiffness in subjects with metabolic syndrome (Park et al., 2012). Based on mostly low certainty evidence, ginseng may only have trivial effects on erectile function or satisfaction with intercourse compared to a placebo when assessed using validated instruments (Lee Y. S. et al., 2021). South Korea and Canada lead the studies. Twenty-eight institutions from seven countries participated, including the Clinical Trial Center for Functional Foods Chonbuk National University Hospital, Clinical Nutrition and Risk Factor Modification Centre, and Clinical Trial Center for Functional Foods. Red ginseng is mainly used as a dietary supplement, and a small part is used in drug research. There are 31 research directions, primarily focusing on diabetes mellitus type 2, health, hypertension, and blood pressure, etc. Given this, clinical studies of red ginseng mainly focus on Korea and Canada and the use of dietary supplements in human clinical studies of type II diabetes, health, hypertension, and other neurological diseases.

FIGURE 7
www.frontiersin.org

FIGURE 7. Analysis of clinical trials of red ginseng.

6 Discussion

Red ginseng products derived from fresh ginseng include TRG, SG, BG, FRG, and PRG. Their chemical compositions are similar because all of them were derived from the same original plant, P. ginseng. The chemical composition of these products has been extensively reported. However, the research mainly focuses on TRG. The chemical composition differences between other red ginseng products remain undiscovered. Interestingly, the transformation law of ginsenosides in different red ginseng processing is similar. Some identical rare ginsenosides make other red ginseng products with similar pharmacological activities. For example, ginsenosides (Rg3, Rg5, and Rk1) of SG have been reported to improve cognitive function; ginsenosides (Rg3, Rg5, and Rk1) of BG are the same as SG, and many studies have been conducted on the prevention and treatment of obesity, breast cancer, cognitive impairment, and fetal a syndrome, etc. The composition of FRG by microorganisms is similar to that of other red ginseng products, which convert normal ginsenosides into minor ginsenosides and produce a unique component CK, which is mainly used in improving allergies, diabetes, anxiety, and other aspects, and may play an essential role in metabolic syndrome and other elements. PRG significantly reduces the conversion time of normal ginsenosides into minor ginsenosides. According to current in vitro studies, the anticancer effects of PRG are worthy of further studies.

Unfortunately, chemical constituents and efficacy studies on red ginseng products are still lacking, especially for PRG. The pharmacological activities focus on TRG and another new type of red ginseng. In some pharmacological activities, the new kind of red ginseng is more vital than TRG. There is little systematic research comparing the differences in pharmacological activities between the new types of red ginseng, and further studies should be conducted on the mechanism of pharmacodynamics in different red ginseng products applied appropriately in the clinic.

During red ginseng processing, the polar compounds are transformed into less polar compounds; demalonylation and decarboxylation easily occur, and the malonyl-ginsenosides decarboxylate into acetyl-ginsenosides; the Maillard reaction is also a reaction in red ginseng processing (Figure 1). The different process influences the content and variety of minor ginsenosides. Usually, the ginsenoside transformation of the new type of red ginseng is more vital than TRG. So, the amount of minor ginsenosides is minimal in TRG but abundant in the new type of red ginseng. With a further understanding of the structure and efficacy of ginsenosides, rare ginsenosides, such as Rg3 and Rh2, have been found to have significant pharmacological activities. The species and proportion of ginsenosides are different during red ginseng processing, which suggests that processing is crucial for the efficacy of red ginseng products. The processing of TCM expands the scope of the application thereof, which is the charm of TCM.

7 Conclusion

Modern technology applied to red ginseng processing provides further specifications. At the same time, modern technology can adapt to determine the development law of modern diseases. Red ginseng products are used in the treatment of AD, diabetes, and other conditions, and further systematic comparative studies of the chemical composition can promote full utilization thereof. The types and proportions of rare ginsenosides require further in-depth comparative analyses in pharmacological activity for better selection of drugs according to different clinical needs. Many researchers are focused on red ginseng processing, especially with regards the steam method and the detailed parameters of processing. In contrast, FRG has the advantage of microbial transformation in the ginsenosides to enhance the amount and variety of minor ginsenosides. To accurately control the processing conditions of different red ginseng products, we can maintain the transformation of ginsenosides and produce red ginseng products for specific diseases. This difference in ginsenosides results in red ginseng products being selectively applied in the clinic.

The processing of red ginseng can be developed in the direction of accurate processing and precise treatment. Moreover, ginsenoside transformation of red ginseng and the structure–functional relationship of ginsenoside derivatives can illustrate the medicinal compositions of this different pharmacological activity, which make the application of red ginseng practical and facilitate the development of new types of red ginseng in the future.

Author contributions

X-FX and X-RL developed a major research plan. JW, C-SL, and XW analyze data. S-QC, and H-XZ draw charts. X-WY write manuscripts. QL and H-MR helped collect data and references. X-XW and L-JX implemented corrections in the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This study was supported by the National Natural Science Foundation of China (No. 81973480), and the Beijing University of Chinese Medicine new teachers start the fund project (2022-JYB-XJSJJ-021).

Acknowledgments

The authors thank the Beijing University of Chinese Medicine for their assistance in conducting this study.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

TRG, Traditional red ginseng; SG, Sun ginseng; BG, Black ginseng; FRG, Fermented red ginseng; PRG, Puffed red ginseng; WG, White ginseng; P. ginseng A.C. Meyer, Panax ginseng; KRG, Korean red ginseng; TCM, Traditional Chinese medicine; CK, Compound K; PPD, Protopanaxadiol; PPT, Protopanaxatriol; OLE, Oleanane.

References

An, Y. E., Ahn, S. C., Yang, D. C., Park, S. J., Kim, B. Y., and Baik, M. Y. (2011). Chemical conversion of ginsenosides in puffed red ginseng. Lwt-Food Sci. Technol. 44 (2), 370–374. doi:10.1016/j.lwt.2010.09.013

CrossRef Full Text | Google Scholar

Bae, C. H., Kim, J., Nam, W., Kim, H., Kim, J., Nam, B., et al. (2021). Fermented red ginseng alleviates ovalbumin-induced inflammation in mice by suppressing interleukin-4 and immunoglobulin E expression. J. Med. Food 24 (6), 569–576. doi:10.1089/jmf.2020.4854

PubMed Abstract | CrossRef Full Text | Google Scholar

Bae, E. A., Han, M. J., Kim, E. J., and Kim, D. H. (2004). Transformation of ginseng saponins to ginsenoside Rh-2 by acids and human intestinal bacteria and biological activities of their transformants. Arch. Pharm. Res. 27(1), 61–67. doi:10.1007/Bf02980048

PubMed Abstract | CrossRef Full Text | Google Scholar

Bae, S. H., Lee, H. S., Kim, M. R., Kim, S. Y., Kim, J. M., and Suh, H. J. (2011). Changes of ginsenoside content by mushroom mycelial fermentation in red ginseng extract. J. Ginseng Res. 35 (2), 235–242. doi:10.5142/jgr.2011.35.2.235

PubMed Abstract | CrossRef Full Text | Google Scholar

Baek, N. I., Kim, D. S., Lee, Y. H., Park, J. D., Lee, C. B., and Kim, S. I. (1996). Ginsenoside Rh4, a genuine dammarane glycoside from Korean red ginseng. Planta Med. 62 (1), 86–87. doi:10.1055/s-2006-957816

PubMed Abstract | CrossRef Full Text | Google Scholar

Baek, N. I., Kim, J. M., Park, J. H., Ryu, J. H., Kim, D. S., Lee, Y. H., et al. (1997). Ginsenoside Rs(3), a genuine dammarane-glycoside from Korean red ginseng. Arch. Pharm. Res. 20 (3), 280–282. doi:10.1007/BF02976158

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, W., Balan, P., and Popovich, D. G. (2020). Changes of ginsenoside composition in the creation of black ginseng leaf. Molecules 25 (12), 2809. doi:10.3390/molecules25122809

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, X. M., and Kitts, D. D. (2011). Antioxidant and anti-inflammatory activities of Maillard reaction products isolated from sugar-amino acid model systems. J. Agric. Food Chem. 59 (20), 11294–11303. doi:10.1021/jf2031583

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, X. M., and Kitts, D. D. (2012). Characterization of antioxidant and anti-inflammatory activities of bioactive fractions recovered from a glucose-lysine Maillard reaction model system. Mol. Cell Biochem. 364 (1-2), 147–157. doi:10.1007/s11010-011-1213-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Cho, J. G., Lee, D. Y., Shrestha, S., Lee, S. K., Kang, H. M., Son, S. H., et al. (2013) Three new ginsenosides from the heat-processed roots of Panax ginseng Chem. Nat. Compd. 49 (5), 882–887. doi:10.1007/s10600-013-0769-8

CrossRef Full Text | Google Scholar

Choi, I. D., Ryu, J. H., Lee, D. E., Lee, M. H., Shim, J. J., Ahn, Y. T., et al. (2016) Enhanced absorption study of ginsenoside compound K (20-O-beta-(D-Glucopyranosyl)-20(S)-protopanaxadiol) after oral administration of fermented red ginseng extract (HYFRG) in healthy Korean volunteers and rats. Evid. Based Complement. Altern. Med. 2016, 3908142. doi:10.1155/2016/3908142

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, Y. J., Kang, L. J., and Lee, S. G. (2014). Stimulation of DDX3 expression by ginsenoside Rg3 through the Akt/p53 pathway activates the innate immune response via TBK1/IKKε/IRF3 signalling. Curr. Med. Chem. 21 (8), 1050–1060. doi:10.2174/09298673113206660306

PubMed Abstract | CrossRef Full Text | Google Scholar

Chung, I. M., Kim, Y. O., Ali, M., Kim, S. H., Park, I., Kim, E. H., et al. (2014). Triterpene glycosides from red ginseng marc and their anti-inflammatory activities. Bioorg Med. Chem. Lett. 24 (17), 4203–4208. doi:10.1016/j.bmcl.2014.07.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, S. K., Joo, M. K., Kim, J. K., Jeung, W., Kang, H., and Kim, D. H. (2020). Bifidobacteria-fermented red ginseng and its constituents ginsenoside Rd and protopanaxatriol alleviate anxiety/depression in mice by the amelioration of gut dysbiosis. Nutrients 12 (4), 901. doi:10.3390/nu12040901

PubMed Abstract | CrossRef Full Text | Google Scholar

Hasegawa, H. (2004). Proof of the mysterious efficacy of ginseng: Basic and clinical trials: Metabolic activation of ginsenoside: Deglycosylation by intestinal bacteria and esterification with fatty acid. J. Pharmacol. Sci. 95 (2), 153–157. doi:10.1254/jphs.fmj04001x4

PubMed Abstract | CrossRef Full Text | Google Scholar

Heo, J. H., Lee, S. T., Chu, K., Oh, M. J., Park, H. J., Shim, J. Y., et al. (2012). Heat-processed ginseng enhances the cognitive function in patients with moderately severe Alzheimer's disease. Nutr. Neurosci. 15 (6), 278–282. doi:10.1179/1476830512Y.0000000027

PubMed Abstract | CrossRef Full Text | Google Scholar

Hong, M., Lee, Y. H., Kim, S., Suk, K. T., Bang, C. S., Yoon, J. H., et al. (2016). Anti-inflammatory and antifatigue effect of Korean Red Ginseng in patients with nonalcoholic fatty liver disease. J. Ginseng Res. 40 (3), 203–210. doi:10.1016/j.jgr.2015.07.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoseney, R. C. (1986). Principles of cereal science and technology. A general reference cereal foods 51, 415.

Google Scholar

Irfan, M., Lee, Y. Y., Lee, K. J., Kim, S. D., and Rhee, M. H. (2022). Comparative antiplatelet and antithrombotic effects of red ginseng and fermented red ginseng extracts. J. Ginseng Res. 46 (3), 387–395. doi:10.1016/j.jgr.2021.05.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Jang, D. J., Lee, M. S., Shin, B. C., Lee, Y. C., and Ernst, E. (2008). Red ginseng for treating erectile dysfunction: A systematic review. Br. J. Clin. Pharmacol. 66 (4), 444–450. doi:10.1111/j.1365-2125.2008.03236.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Jang, S. H., Park, J., Kim, S. H., Choi, K. M., Ko, E. S., Cha, J. D., et al. (2017). Red ginseng powder fermented with probiotics exerts antidiabetic effects in the streptozotocin-induced mouse diabetes model. Pharm. Biol. 55 (1), 317–323. doi:10.1080/13880209.2016.1237978

PubMed Abstract | CrossRef Full Text | Google Scholar

Jeong, H. G., Ko, Y. H., Oh, S. Y., Han, C., Kim, T., and Joe, S. H. (2015). Effect of Korean Red Ginseng as an adjuvant treatment for women with residual symptoms of major depression. Asia Pac Psychiatry 7 (3), 330–336. doi:10.1111/appy.12169

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, G., Ramachandraiah, K., Murtaza, M. A., Wang, L., Li, S., and Ameer, K. (2021). Synergistic effects of black ginseng and aged garlic extracts for the amelioration of nonalcoholic fatty liver disease (NAFLD) in mice. Food Sci. Nutr. 9 (6), 3091–3099. doi:10.1002/fsn3.2267

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiao, L., Zhang, X., Wang, M., Li, B., Liu, Z., and Liu, S. (2014). Chemical and antihyperglycemic activity changes of ginseng pectin induced by heat processing. Carbohydr. Polym. 114, 567–573. doi:10.1016/j.carbpol.2014.08.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Jung, J., Jang, H. J., Eom, S. J., Choi, N. S., Lee, N. K., and Paik, H. D. (2019). Fermentation of red ginseng extract by the probiotic Lactobacillus plantarum KCCM 11613P: Ginsenoside conversion and antioxidant effects. J. Ginseng Res. 43 (1), 20–26. doi:10.1016/j.jgr.2017.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Kai, Q., Liu, Q., Wan, J. Y., Zhao, Y. J., Guo, R. Z., Alolga, R. N., et al. (2015). Rapid preparation of rare ginsenosides by acid transformation and their structure-activity relationships against cancer cells. Sci. Rep. 5, 8598. doi:10.1038/srep08598

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, K. S., Kim, H. Y., Pyo, J. S., and Yokozawa, T. (2006). Increase in the free radical scavenging activity of ginseng by heat-processing. Biol. Pharm. Bull. 29 (4), 750–754. doi:10.1248/bpb.29.750

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, K. S., Tanaka, T., Cho, E. J., and Yokozawa, T. (2009) Evaluation of the peroxynitrite scavenging activity of heat-processed ginseng J. Med. Food 12 (1), 124–130. doi:10.1089/jmf.2007.0646

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, K. S., Yamabe, N., Kim, H. Y., Okamoto, T., Sei, Y., and Yokozawa, T. (2007). Increase in the free radical scavenging activities of American ginseng by heat processing and its safety evaluation. J. Ethnopharmacol. 113 (2), 225–232. doi:10.1016/j.jep.2007.05.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Kasai, R., Besso, H., Tanaka, O., Saruwatari, Y., and Fuwa, T. (1983). Saponins of red ginseng. Chem. Pharm. Bull. (Tokyo) 31 (6), 2120–2125. doi:10.1248/cpb.31.2120

CrossRef Full Text | Google Scholar

Kennedy, D. O., and Scholey, A. B. (2003). Ginseng: Potential for the enhancement of cognitive performance and mood. Pharmacol. Biochem. Behav. 75 (3), 687–700. doi:10.1016/s0091-3057(03)00126-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Kho, M. C., Lee, Y. J., Park, J. H., Kim, H. Y., Yoon, J. J., Ahn, Y. M., et al. (2016). Fermented red ginseng potentiates improvement of metabolic dysfunction in metabolic syndrome rat models. Nutrients 8 (6), 369. doi:10.3390/nu8060369

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, A. J. (2015). Physiological activities of 9 cycle steaming and drying black ginseng using Makgeolli. Food Sci. Biotechnol. 24 (5), 1867–1873. doi:10.1007/s10068-015-0244-3

CrossRef Full Text | Google Scholar

Kim, D. S., Baek, N. I., Lee, Y. H., Park, J. D., and Kim, S. I. (1996). Preparation and structure determination of a new glycoside, (20E)-ginsenoside Rh3, and its isomer from dioltype ginseng saponins. Yakhak Hoeji 39, 86–93.

CrossRef Full Text | Google Scholar

Kim, H. I., Kim, J. K., Kim, J. Y., Han, M. J., and Kim, D. H. (2019). Fermented red ginseng and ginsenoside Rd alleviate ovalbumin-induced allergic rhinitis in mice by suppressing IgE, interleukin-4, and interleukin-5 expression. J. Ginseng Res. 43 (4), 635–644. doi:10.1016/j.jgr.2019.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, I. K., Lee, K. Y., Kang, J., Park, J. S., and Jeong, J. (2021). Immune-modulating effect of Korean red ginseng by balancing the ratio of peripheral T lymphocytes in bile duct or pancreatic cancer patients with adjuvant chemotherapy. Vivo 35 (3), 1895–1900. doi:10.21873/invivo.12454

CrossRef Full Text | Google Scholar

Kim, J. H., Ahn, S. C., Choi, S. W., Hur, N. Y., Kim, B. Y., and Baik, M. Y. (2008). Changes in effective components of ginseng by puffing. J. Korean Soc. Appl. Biol. Chem. 51 (3), 188–193.

CrossRef Full Text | Google Scholar

Kim, J. H., Han, I. H., Yamabe, N., Kim, Y. J., Lee, W., Eom, D. W., et al. (2014). Renoprotective effects of Maillard reaction products generated during heat treatment of ginsenoside Re with leucine. Food Chem. 143, 114–121. doi:10.1016/j.foodchem.2013.07.075

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J. K., Choi, M. S., Jeung, W., Ra, J., Yoo, H. H., and Kim, D. H. (2020). Effects of gut microbiota on the pharmacokinetics of protopanaxadiol ginsenosides Rd, Rg3, F2, and compound K in healthy volunteers treated orally with red ginseng. J. Ginseng Res. 44 (4), 611–618. doi:10.1016/j.jgr.2019.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J. K., Kim, J. Y., Jang, S. E., Choi, M. S., Jang, H. M., Yoo, H. H., et al. (2018). Fermented red ginseng alleviates cyclophosphamide-induced immunosuppression and 2,4,6-trinitrobenzenesulfonic acid-induced colitis in mice by regulating macrophage activation and T cell differentiation. Am. J. Chin. Med. 46 (8), 1879–1897. doi:10.1142/S0192415X18500945

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, M. O., Lee, J. W., Lee, J. K., Song, Y. N., Oh, E. S., Ro, H., et al. (2022). Black ginseng extract suppresses airway inflammation induced by cigarette smoke and lipopolysaccharides in vivo. Antioxidants (Basel) 11 (4), 679. doi:10.3390/antiox11040679

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, S. J., and Kim, A. K. (2015). Anti-breast cancer activity of fine black ginseng (Panax ginseng meyer) and ginsenoside Rg5. J. Ginseng Res. 39 (2), 125–134. doi:10.1016/j.jgr.2014.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, W. Y., Kim, J. M., Han, S. B., Lee, S. K., Kim, N. D., Park, M. K., et al. (2000). Steaming of ginseng at high temperature enhances biological activity. J. Nat. Prod. 63 (12), 1702–1704. doi:10.1021/np990152b

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y. J., Choi, W. I., Jeon, B. N., Choi, K. C., Kim, K., Kim, T. J., et al. (2014). Stereospecific effects of ginsenoside 20-Rg3 inhibits TGF-β1-induced epithelial-mesenchymal transition and suppresses lung cancer migration, invasion and anoikis resistance. Toxicology 322, 23–33. doi:10.1016/j.tox.2014.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Kitagawa, I., Yoshikawa, M., Yoshihara, M., Hayashi, T., and Taniyama, T. (1983). Chemical studies on crude drug precession. I. On the constituents of ginseng radix rubra (1). Yakugaku Zasshi-journal Pharm. Soc. Jpn. 103 (6), 612–622. doi:10.1248/yakushi1947.103.6_612

CrossRef Full Text | Google Scholar

Kwok, H. H., Guo, G. L., Lau, J. K., Cheng, Y. K., Wang, J. R., Jiang, Z. H., et al. (2012). Stereoisomers ginsenosides-20(S)-Rg3 and -20(R)-Rg3 differentially induce angiogenesis through peroxisome proliferator-activated receptor-gamma. Biochem. Pharmacol. 83 (7), 893–902. doi:10.1016/j.bcp.2011.12.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Kwon, S. W., Han, S. B., Park, I. H., Kim, J. M., Park, M. K., and Park, J. H. (2001). Liquid chromatographic determination of less polar ginsenosides in processed ginseng. J. Chromatogr. A 921 (2), 335–339. doi:10.1016/s0021-9673(01)00869-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, B., Sur, B., Lee, H., and Oh, S. (2020). Korean Red Ginseng prevents posttraumatic stress disorder-triggered depression-like behaviors in rats via activation of the serotonergic system. J. Ginseng Res. 44 (4), 644–654. doi:10.1016/j.jgr.2019.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, C. H., Kim, J. M., Kim, D. H., Park, S. J., Liu, X., Cai, M., et al. (2013). Effects of Sun ginseng on memory enhancement and hippocampal neurogenesis. Phytother. Res. 27 (9), 1293–1299. doi:10.1002/ptr.4873

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, C. S., Lee, J. H., Oh, M., Choi, K. M., Jeong, M. R., Park, J. D., et al. (2012). Preventive effect of Korean red ginseng for acute respiratory illness: A randomized and double-blind clinical trial. J. Korean Med. Sci. 27 (12), 1472–1478. doi:10.3346/jkms.2012.27.12.1472

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H., Lee, J. Y., Song, K. C., Kim, J., Park, J. H., Chun, K. H., et al. (2012). Protective effect of processed Panax ginseng, sun ginseng on UVB-irradiated human skin keratinocyte and human dermal fibroblast. J. Ginseng Res. 36 (1), 68–77. doi:10.5142/jgr.2012.36.1.68

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. U., Bae, E. A., Han, M. J., and Kim, D. H. (2005). Hepatoprotective effect of 20(S)-ginsenosides Rg3 and its metabolite 20(S)-ginsenoside Rh2 on tert-butyl hydroperoxide-induced liver injury. Biol. Pharm. Bull. 28 (10), 1992–1994. doi:10.1248/bpb.28.1992

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. W., Lee, M. S., Kim, T. H., Alraek, T., Zaslawski, C., Kim, J. W., et al. (2021). Ginseng for erectile dysfunction. Cochrane Database Syst. Rev. 4 (4), CD012654. doi:10.1002/14651858.CD012654.pub2

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, J. H., Ahn, J. Y., Shin, T. J., Choi, S. H., Lee, B. H., Hwang, S. H., et al. (2011). Effects of minor ginsenosides, ginsenoside metabolites, and ginsenoside epimers on the growth of Caenorhabditis elegans. J. Ginseng Res. 35 (3), 375–383. doi:10.5142/jgr.2011.35.3.375

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S., Jung, S., You, H., Lee, Y., Park, Y., Lee, H., et al. (2022). Effect of fermented red ginseng concentrate intake on stool characteristic, biochemical parameters, and gut microbiota in elderly Korean women. Nutrients 14 (9), 1693. doi:10.3390/nu14091693

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. A., Jo, H. K., Im, B. O., Kim, S., Whang, W. K., and Ko, S. K. (2012). Changes in the contents of prosapogenin in the red ginseng (Panax ginseng) depending on steaming batches. J. Ginseng Res. 36 (1), 102–106. doi:10.5142/jgr.2012.36.1.102

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. H., Oh, M., Park, J., Jang, S. Y., Cheong, S. H., Lee, H., et al. (2015). Effects of µ-opioid receptor gene polymorphism on postoperative nausea and vomiting in patients undergoing general anesthesia with remifentanil: Double blinded randomized trial. Food Sci. Biotechnol. 24 (2), 651–657. doi:10.3346/jkms.2015.30.5.651

CrossRef Full Text | Google Scholar

Lee, S. J., Lee, D. Y., O'Connell, J. F., Egan, J. M., and Kim, Y. (2022). Black ginseng ameliorates cellular senescence via p53-p21/p16 pathway in aged mice. Biol. (Basel) 11 (8), 1108. doi:10.3390/biology11081108

CrossRef Full Text | Google Scholar

Lee, S. J., Oh, S., Kim, M. J., Sim, G. S., Moon, T. W., and Lee, J. (2018). Oxidative stability of extracts from red ginseng and puffed red ginseng in bulk oil or oil-in-water emulsion matrix. J. Ginseng Res. 42 (3), 320–326. doi:10.1016/j.jgr.2017.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. M., Kim, S. C., Oh, J., Kim, J. H., Na, M., Seong, S. C., et al. (2013a). 20(R)-Ginsenoside Rf: A new ginsenoside from red ginseng extract. Phytochem. Lett. 6 (4), 620–628. doi:10.1007/s00167-012-1998-2

CrossRef Full Text | Google Scholar

Lee, S. M., Seo, H. K., Oh, J., and Na, M. (2013b). Updating chemical profiling of red ginseng via the elucidation of two geometric isomers of ginsenosides Rg9 and Rg10. Food Chem. 141 (4), 3920–3924. doi:10.1016/j.foodchem.2013.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. R., Kim, M. R., Yon, J. M., Baek, I. J., Park, C. G., Lee, B. J., et al. (2009). Black ginseng inhibits ethanol-induced teratogenesis in cultured mouse embryos through its effects on antioxidant activity. Toxicol Vitro 23 (1), 47–52. doi:10.1016/j.tiv.2008.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, W., Ku, S. K., Kim, J. E., Cho, S. H., Song, G. Y., and Bae, J. S. (2019). Inhibitory effects of black ginseng on particulate matter-induced pulmonary injury. Am. J. Chin. Med. 47 (6), 1237–1251. doi:10.1142/S0192415X19500630

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, Y. J., Kim, H. Y., Kang, K. S., Lee, J. G., Yokozawa, T., and Park, J. H. (2008). The chemical and hydroxyl radical scavenging activity changes of ginsenoside-Rb1 by heat processing. Bioorg Med. Chem. Lett. 18 (16), 4515–4520. doi:10.1016/j.bmcl.2008.07.056

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, Y. S., Kim, K. W., Yoon, D., Kim, G. S., Kwon, D. Y., Kang, O. H., et al. (2021). Comparison of antivirulence activities of black ginseng against methicillin-resistant Staphylococcus aureus according to the number of repeated steaming and drying cycles. Antibiot. (Basel) 10 (6), 617. doi:10.3390/antibiotics10060617

CrossRef Full Text | Google Scholar

Li, B. H., Zhao, J. O., Wang, C. Z., Searle, J., He, T. C., Yuan, C. S., et al. (2011). Ginsenoside Rh2 induces apoptosis and paraptosis-like cell death in colorectal cancer cells through activation of p53. Cancer Lett. 301 (2), 185–192. doi:10.1016/j.canlet.2010.11.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Zhong, W., Wang, W., Hu, S., Yuan, J., Zhang, B., et al. (2014). Ginsenoside metabolite compound K promotes recovery of dextran sulfate sodium-induced colitis and inhibits inflammatory responses by suppressing NF-κB activation. PLoS One 9 (2), e87810. doi:10.1371/journal.pone.0087810

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, H., Lu, X., Hu, Y., and Fan, X. (2020). Chemical constituents of Panax ginseng and Panax notoginseng explain why they differ in therapeutic efficacy. Pharmacol. Res. 161, 105263. doi:10.1016/j.phrs.2020.105263

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Shiono, J., Shimizu, K., Yu, H., Zhang, C., Jin, F., et al. (2009). 20(R)-ginsenoside Rh2, not 20(S), is a selective osteoclastgenesis inhibitor without any cytotoxicity. Bioorg Med. Chem. Lett. 19 (12), 3320–3323. doi:10.1016/j.bmcl.2009.04.054

PubMed Abstract | CrossRef Full Text | Google Scholar

Min, J. H., Cho, H. J., and Yi, Y. S. (2022). A novel mechanism of Korean Red Ginseng-mediated anti-inflammatory action via targeting caspase-11 non-canonical inflammasome in macrophages. J. Ginseng Res. 46 (5), 675–682. doi:10.1016/j.jgr.2021.12.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Oh, H. B., Lee, J. W., Lee, D. E., Na, S. C., Jeong, D. E., Hwang, D. I., et al. (2021). Characteristics of black ginseng (Panax ginseng C.A. Mayer) production using ginseng stored at low temperature after harvest. Metabolites 11 (2), 98. doi:10.3390/metabo11020098

PubMed Abstract | CrossRef Full Text | Google Scholar

Oh, J., Jeon, S. B., Lee, Y., Lee, H., Kim, J., Kwon, B. R., et al. (2015). Fermented red ginseng extract inhibits cancer cell proliferation and viability. J. Med. Food 18 (4), 421–428. doi:10.1089/jmf.2014.3248

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, B. J., Lee, Y. J., Lee, H. R., Jung, D. H., Na, H. Y., Kim, H. B., et al. (2012). Effects of Korean red ginseng on cardiovascular risks in subjects with metabolic syndrome: A double-blind randomized controlled study. Korean J. Fam. Med. 33 (4), 190–196. doi:10.4082/kjfm.2012.33.4.190

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, H. J., Shim, H. S., Kim, K. S., and Shim, I. (2011). The protective effect of black ginseng against transient focal ischemia-induced neuronal damage in rats. Korean J. Physiol. Pharmacol. 15 (6), 333–338. doi:10.4196/kjpp.2011.15.6.333

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, I. H., Han, S. B., Kim, J. M., Piao, L., Kwon, S. W., Kim, N. Y., et al. (2002a). Four new acetylated ginsenosides from processed ginseng (sun ginseng). Arch. Pharm. Res. 25 (6), 837–841. doi:10.1007/BF02977001

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, I. H., Kim, N. Y., Han, S. B., Kim, J. M., Kwon, S. W., Kim, H. J., et al. (2002b). Three new dammarane glycosides from heat processed ginseng. Arch. Pharm. Res. 25 (4), 428–432. doi:10.1007/BF02976595

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, J. D., Lee, Y. H., and Kim, S. I. (1998). Ginsenoside Rf2, a new dammarane glycoside from Korean red ginseng (Panax ginseng). Arch. Pharm. Res. 21 (5), 615–617. doi:10.1007/BF02975384

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, J. W., Lee, B. J., Bu, Y., Yeo, I., Kim, J., and Ryu, B. H. (2010). Effects of Korean red ginseng on dry mouth: A randomized, double-blind, placebo-controlled trial. J. Ginseng Res. 34 (3), 183–191. doi:10.5142/jgr.2010.34.3.183

CrossRef Full Text | Google Scholar

Park, M. W., Ha, J., and Chung, S. H. (2008). 20(S)-ginsenoside Rg3 enhances glucose-stimulated insulin secretion and activates AMPK. Biol. Pharm. Bull. 31 (4), 748–751. doi:10.1248/bpb.31.748

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, S. J., Park, M., Sharma, A., Kim, K., and Lee, H. J. (2019). Black ginseng and ginsenoside Rb1 promote browning by inducing UCP1 expression in 3T3-L1 and primary white adipocytes. Nutrients 11 (11), 2747. doi:10.3390/nu11112747

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, X., Hao, M., Zhao, Y., Cai, Y., Chen, X., Chen, H., et al. (2021). Red ginseng has stronger anti-aging effects compared to ginseng possibly due to its regulation of oxidative stress and the gut microbiota. Phytomedicine 93, 153772. doi:10.1016/j.phymed.2021.153772

PubMed Abstract | CrossRef Full Text | Google Scholar

Ryu, J. H., Park, J. H., Kim, T. H., Dong, H. S., Kim, J. M., and Park, J. H. (1996). A genuine dammarane glycoside, (20E)-ginsenoside F 4 from Korean red ginseng. Arch. Pharm. Res. 19 (4), 335–336. doi:10.1007/bf02976251

CrossRef Full Text | Google Scholar

Saba, E., Lee, Y. Y., Kim, M., Kim, S. H., Hong, S. B., and Rhee, M. H. (2018). A comparative study on immune-stimulatory and antioxidant activities of various types of ginseng extracts in murine and rodent models. J. Ginseng Res. 42 (4), 577–584. doi:10.1016/j.jgr.2018.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Seoyoung, L., Geuntae, K., Sihun, R., Song, J. S., Hiejoon, K., Hong, S. S., et al. (2009). Proteomic analysis of the anti-cancer effect of 20S-ginsenoside Rg3 in human colon cancer cell lines. Biosci. Biotech. BIOCH 73 (4), 811–816. doi:10.1271/bbb.80637

CrossRef Full Text | Google Scholar

Shin, J. H., Park, Y. J., Kim, W., Kim, D. O., Kim, B. Y., Lee, H., et al. (2019). Change of ginsenoside profiles in processed ginseng by drying, steaming, and puffing. J. Microbiol. Biotechnol. 29 (2), 222–229. doi:10.4014/jmb.1809.09056

PubMed Abstract | CrossRef Full Text | Google Scholar

Shin, S. J., Park, Y. H., Jeon, S. G., Kim, S., Nam, Y., Oh, S. M., et al. (2020). Red ginseng inhibits tau aggregation and promotes tau dissociation in vitro. Oxid. Med. Cell Longev. 2020, 7829842. doi:10.1155/2020/7829842

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, K. C., Chang, T. S., Lee, H., Kim, J., Park, J. H., and Hwang, G. S. (2012). Processed Panax ginseng, sun ginseng increases type I collagen by regulating MMP-1 and TIMP-1 expression in human dermal fibroblasts. J. Ginseng Res. 36 (1), 61–67. doi:10.5142/jgr.2012.36.1.61

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, B. S., Gu, L. J., Fang, Z. M., Wang, C. Y., Wang, Z., Lee, M. R., et al. (2009). Simultaneous quantification of 19 ginsenosides in black ginseng developed from Panax ginseng by HPLC-ELSD. J. Pharm. Biomed. Anal. 50 (1), 15–22. doi:10.1016/j.jpba.2009.03.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, C., Gao, W., Zhao, B., and Cheng, L. (2013). Optimization of the selective preparation of 20(R)-ginsenoside Rg3 catalyzed by d, l-tartaric acid using response surface methodology. Fitoterapia 84, 213–221. doi:10.1016/j.fitote.2012.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Teng, S., Wang, Y., Li, P., Liu, J., Wei, A., Wang, H., et al. (2017). Effects of R type and S type ginsenoside Rg3 on DNA methylation in human hepatocarcinoma cells. Mol. Med. Rep. 15 (4), 2029–2038. doi:10.3892/mmr.2017.6255

PubMed Abstract | CrossRef Full Text | Google Scholar

Trinh, H. T., Han, S. J., Kim, S. W., Lee, Y. C., and Kim, D. H. (2007). Bifidus fermentation increases hypolipidemic and hypoglycemic effects of red ginseng. J. Microbiol. Biotechnol. 17 (7), 1127–1133.

PubMed Abstract | Google Scholar

Wang, W., Zhao, Y., Rayburn, E. R., Hill, D. L., Wang, H., and Zhang, R. (2007). In vitro anti-cancer activity and structure-activity relationships of natural products isolated from fruits of Panax ginseng. Cancer Chemother. Pharmacol. 59 (5), 589–601. doi:10.1007/s00280-006-0300-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, W., Liu, L., Liu, X., Tao, Y., Gong, J., Wang, Y., et al. (2022). Black ginseng protects against Western diet-induced nonalcoholic steatohepatitis by modulating the TLR4/NF-κB signaling pathway in mice. J. Food Biochem. 46 (12), e14432. doi:10.1111/jfbc.14432

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, X., Chen, J., Su, F., Su, X., Hu, T., and Hu, S. (2012). Stereospecificity of ginsenoside Rg3 in promotion of the immune response to ovalbumin in mice. Int. Immunol. 24 (7), 465–471. doi:10.1093/intimm/dxs043

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, Y., Zhao, W., Zhang, Q., Zhao, Y., and Zhang, Y. (2011). Purification and characterization of a novel and unique ginsenoside Rg1-hydrolyzing β-d-Glucosidase from Penicillium sclerotiorum. Acta biochimica biophysica Sinica 43 (3), 226–231. doi:10.1093/abbs/gmr001

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, Y. Y., Luo, D., Cheng, Y. J., Ma, J. F., Wang, Y. M., Liang, Q. l., et al. (2012). Steaming-induced chemical transformations and holistic quality assessment of red ginseng derived from Panax ginseng by means of HPLC-ESI-MS/MS(n)-based multicomponent quantification fingerprint. J. Agric. Food Chem. 60 (33), 8213–8224. doi:10.1021/jf301116x

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, Y., Halima, M., Che, X., Zhang, Y., Schaaf, M. J. M., Li, M., et al. (2022). Steamed Panax notoginseng and its saponins inhibit the migration and induce the apoptosis of neutrophils in a zebrafish tail-fin amputation model. Front. Pharmacol. 13, 946900. doi:10.3389/fphar.2022.946900

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, F. L., Zhang, Q. Y., Jiang, L., Peng, G. M., Yao, R., Li, B. T., et al. (2016). Study on chemical constituents of radix ginseng destillata alcohol extract by UHPLC-Q-TOF/MS. Traditional Chin. Drug Res. Clin. Pharmacol. 26 (4), 529–534.

Google Scholar

Xu, X. F., Gao, Y., Xu, S. Y., Liu, H., Xue, X., Zhang, Y., et al. (2018). Remarkable impact of steam temperature on ginsenosides transformation from fresh ginseng to red ginseng. J. Ginseng Res. 42 (3), 277–287. doi:10.1016/j.jgr.2017.02.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, X. F., Nie, L. X., Pan, L. L., Hao, B., Yuan, S. X., Lin, R. C., et al. (2014). Quantitative analysis of Panax ginseng by FT-NIR spectroscopy. J. Anal. Methods Chem. 2014 (8), 741571. doi:10.1155/2014/741571

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamabe, N., Kim, Y. J., Lee, S., Cho, E. J., Park, S. H., Ham, J., et al. (2013). Increase in antioxidant and anticancer effects of ginsenoside Re-lysine mixture by Maillard reaction. Food Chem. 138 (2-3), 876–883. doi:10.1016/j.foodchem.2012.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, H., Oh, K. H., Kim, H. J., Cho, Y. H., and Yoo, Y. C. (2018). Ginsenoside-Rb2 and 20(S)-Ginsenoside-Rg3 from Korean red ginseng prevent rotavirus infection in newborn mice. J. Microbiol. Biotechnol. 28 (3), 391–396. doi:10.4014/jmb.1801.01006

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoo, S., Park, B. I., Kim, D. H., Lee, S., Lee, S. H., Shim, W. S., et al. (2021). Ginsenoside absorption rate and extent enhancement of black ginseng (CJ EnerG) over red ginseng in healthy adults. Pharmaceutics 13 (4), 487. doi:10.3390/pharmaceutics13040487

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H. M., Li, S. L., Zhang, H., Wang, Y., Zhao, Z. L., Chen, S. L., et al. (2012). Holistic quality evaluation of commercial white and red ginseng using a UPLC-QTOF-MS/MS-based metabolomics approach. J. Pharm. Biomed. Anal. 62, 258–273. doi:10.1016/j.jpba.2012.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Z., Zhang, Y., Gao, M., Cui, X., Yang, Y., van Duijn, B., et al. (2019). Steamed Panax notoginseng attenuates anemia in mice with blood deficiency syndrome via regulating hematopoietic factors and JAK-STAT pathway. Front. Pharmacol. 10, 1578. doi:10.3389/fphar.2019.01578

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, Q., Li, Z., Liu, J., Han, L., Zhang, N., Zhang, H., et al. (2016). Two new dammarane-type triterpene sapogenins from Chinese red ginseng. Nat. Prod. Res. 30 (1), 95–99. doi:10.1080/14786419.2015.1038538

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q. L., Xu, W., and Yang, X. W. (2016). Chemical constituents of Chinese red ginseng. Zhongguo Zhong Yao Za Zhi 41 (2), 233–249. doi:10.4268/cjcmm20160214

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q. L., and Yang, X. W. (2015). Four new ginsenosides from red ginseng with inhibitory activity on melanogenesis in melanoma cells. Bioorg Med. Chem. Lett. 25 (16), 3112–3116. doi:10.1016/j.bmcl.2015.06.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, L., Luan, X., Dou, D., and Huang, L. (2019). Comparative analysis of ginsenosides and oligosaccharides in white ginseng (WG), red ginseng (RG) and black ginseng (BG). J. Chromatogr. Sci. 57 (5), 403–410. doi:10.1093/chromsci/bmz004

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: red ginseng products, ginsenosides, transformation rule, herbal medicine, pharmacological activities

Citation: Ye X-W, Li C-S, Zhang H-X, Li Q, Cheng S-Q, Wen J, Wang X, Ren H-M, Xia L-J, Wang X-X, Xu X-F and Li X-R (2023) Saponins of ginseng products: a review of their transformation in processing. Front. Pharmacol. 14:1177819. doi: 10.3389/fphar.2023.1177819

Received: 02 March 2023; Accepted: 17 April 2023;
Published: 28 April 2023.

Edited by:

Yin Xiong, Kunming University of Science and Technology, China

Reviewed by:

Shengyuan Xiao, Jilin Agricultural University, China
Kunming Qin, Jiangsu Ocean Universiity, China
Yulin Dai, Changchun University of Chinese Medicine, China
Deqiang Dou, Liaoning University of Traditional Chinese Medicine, China

Copyright © 2023 Ye, Li, Zhang, Li, Cheng, Wen, Wang, Ren, Xia, Wang, Xu and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xin-Fang Xu, xuxinfang007@163.com; Xiang-Ri Li, lixiangri@sina.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.