Skip to main content

Panax japonicus C.A. Meyer: a comprehensive review on botany, phytochemistry, pharmacology, pharmacokinetics and authentication

Abstract

Background

Panax japonicus C.A. Meyer (Zhujieshen) is widely used in traditional medicine as a tonic hemostatic and anti-inflammatory agent in China, Japan, and Korea. Furthermore, it is used as an important substitute for ginseng roots by minority ethnic groups in China. The purpose of this review is to summarize the latest research on Zhujieshen in recent years, aiming at providing a systematic overview of the current knowledge, and perspectives for future research and exploitation.

Main body

This review examines the research advances in botanical profile, phytochemicals, pharmacology, pharmacokinetics, and authentication of Zhujieshen. Various compounds have been reported as active components, mainly including saponins, volatile oils, and polysaccharides. Pharmacological investigations have demonstrated that Zhujieshen is an important herb with significant bioactivities, such as anti-inflammatory, hepato-protective, cardio-protective, neuro-protective, anti-tumor, anti-oxidant, anti-thrombotic and immunomodulatory activities.

Conclusion

Currently, research on Zhujieshen is in the preliminary stages, and further research is required to understand the active compounds present and mechanisms of action. We hope that this comprehensive review of Zhujieshen will serve as a background for future research and exploitation.

Introduction

Panax japonicus C.A. Meyer is a perennial herb belonging to the genus Panax in the Araliaceae family that mainly grows wild in China, Japan, and Korea [1,2,3]. P. japonicus is also known as “Zhujieshen” (竹节参 in Chinese) due to its bamboo-like long horizontally creeping rhizome. The rhizome of Zhujieshen used as a traditional Chinese medicine for a thousand years, is the king of herbs in traditional Tujia and Hmong medicine. It has been used as an important substitute for ginseng roots by minority ethnic groups [1, 4, 5]. Its pharmacological effects include promotion of blood flow, similarly to P. notoginseng, and as a strengthening tonic, similarly to P. ginseng. Zhujieshen has mainly been used as a tonic and hemostatic and anti-inflammatory agent in China for treatment of fracture, hematemesis, cough, bleeding wounds, arthralgia, and weakness after illness [3, 6]. The earliest history of Zhujieshen was recorded in A Supplement to the Compendium of Materia Medica (Qing Dynasty, Ben Cao Gang Mu Shi Yi, 本草纲目拾遗) in the Qing Dynasty. It has been recorded in the Pharmacopoeia of the People's Republic of China, which attributes various pharmacological activities to it.

Many recent scientific studies have reported on Zhujieshen, suggesting that it would be timely to conduct a systematic overview of the current knowledge. This review is intended to address research advances in botanical profile, phytochemicals, pharmacology, and pharmacokinetics of the extracts and isolated compounds from Zhujieshen. Moreover, this review aims to provide novel perspectives to further expand the therapeutic applications and exploitation of Zhujieshen for the treatment of various human diseases.

Botany

The bamboo-root-like rhizome of Zhujieshen (Fig. 1) is horizontal, plump, and white with short internodes, each node having a deep concave stem mark (concave eye). The stem is erect, 30–60 cm high, and cylindrical with longitudinal stripes. Three to five palmately compound leaves form one whorl at the top of the stem. The petiole is delicate and leaflets (usually 3 ~ 5) are obovate to obovate-elliptic, 8–12 cm long, and 3–5 cm wide. The apex of the leaflet is long and tapering. The base is wedge-shaped, extends downward and the edges are serrated. A few bristles are scattered on the upper vein, while the lower side is glabrous. The outermost pair of lateral leaflets is smaller. The microparticle is up to 2.5 cm long. An umbel is formed at the center of the top of the stem, about 15 cm in length, composed of many small flowers. The flowers are yellow-green, the calyx margin has five teeth, there are five petals, five stamens, and perigyny, and the ovary has two compartments. The fruits are kidney-shaped and bright red, each containing two seeds.

Fig.1
figure 1

The medicinal plant Zhujieshen. a The whole plants, b dry bamboo-root-like rhizome

Phytochemistry

Zhujieshen is a rich source of many bioactive phytochemicals that have been isolated and identified from rhizomes, taproots, lateral roots, leaves, and fruits [7,8,9,10]. Triterpenoid saponins are the main bioactive components of Zhujieshen, including oleanolane type (A) and dammarane-type saponins. The dammarane type saponins can be divided into 20(S)-protopanaxadiol type (B) and 20(S)-protopanaxatriol type (C) triterpenoid saponins according to whether the C6 position of the dammarane nucleus contains a hydroxyl group or an O-glycoside substituent [11]. Additionally, there is ocotillol type (D) saponins in Zhujieshen, in which a furan ring is introduced by an oxygen atom connection between C20 and C24 of the dammarane skeleton. These are also known as 20(S), 24(S)-epoxydammarane-3β,6α,12β,25-tetraol type tetracyclic triterpenoid saponins [11]. The parent nuclei of different saponins are shown in Fig. 2. The main saponins in Zhujieshen are shown in Table 1 and Fig. 3. Moreover, other interesting compounds, such as volatile oils and polysaccharides have been reported to contribute to the biological activities of Zhujieshen [1, 7, 25,26,27]. Analysis of plant volatiles is a significant and growing area of research, in which gas chromatography combined with mass spectrometry has been used to analyze the volatile oils. Table 2 shows the constituents of volatile oils from Zhujieshen [7, 28, 29]. Carbohydrates are important chemical components isolated from Zhujieshen, which include polysaccharides, oligosaccharides, and reducing sugars. Two polysaccharides that activate the reticuloendothelial system, Tachibana-A and B, were isolated from Zhujieshen by Ohtani et al. in 1989 [76]. Huang et al. isolated five water-soluble polysaccharides (RPS1–RPS5) from Zhujieshen. They also determined their average molecular masses and characterized their morphologies. In addition, it is rich in amino acids, and contains a small quantity of inorganic elements and other ingredients [26].

Fig.2
figure 2

The parent nuclei of different saponins in Zhujieshen. a oleanolane type; b 20(S)-protopanaxadiol type; c 20(S)-protopanaxatriol type; d ocotillol type; e other promiscuous subtype saponins core; f other promiscuous subtype saponins core; g 24-hydroxy-20(S)protopanaxadiol saponins core; h other promiscuous subtype saponins core

Table 1 The saponins isolated from Zhujieshen
Fig. 3
figure 3

The chemical structures of compounds 101–106 from Zhujieshen

Table 2 Constituents of the volatile oils from Zhujieshen

Pharmacological activities

Anti-inflammatory activity

Increasing lines of evidence indicate the potential of Zhujieshen for effective treatment of inflammatory diseases [30,31,32,33]. Saponins from Zhujieshen extract have been found to regulate MAPK and NF-κB signaling pathways to attenuate age-related neuroinflammation [34]. In another study, saponins isolated from Zhujieshen were shown to inhibit inflammation in the colon of natural aging rats, possibly by modulation of the neuraminidase 3/intestinal alkaline phosphatase signaling pathway [35]. Chikusetsusaponin V exhibited anti-inflammatory activity in LPS-stimulated RAW 264.7 macrophages via inhibition of NF-κB and MAPK signaling pathways [36]. Chikusetsusaponin IVa effectively inhibited high-fat diet-induced inflammation in mouse adipose tissue by inhibiting NLRP3 inflammasome activation and NF-κB signaling [37]. Additionally, polysaccharides from Zhujieshen reduced LPS-induced microglia inflammation, possibly by inhibition of NF-κB signaling [38].

Hepato-protective activity

With the deeper studies of Zhujieshen, it is gradually accepted that it also has hepato-protective activity [39,40,41,42]. The hepato-protective activity of total saponins from Zhujieshen was investigated in a mouse model of fatty liver fibrosis, in which the mice were fed a high-fat diet combined with an intraperitoneal injection of porcine serum [43]. The low-dose (100 mg/kg) and high-dose (300 mg/kg) groups of mice on a high-fat diet were treated by oral gavage once every 2 days for 11 weeks. The mice were given porcine serum by intraperitoneal injection from the fifth week. Compared with animals in the model group, the final body weight, liver weight, and liver index were markedly decreased by treatment with total saponins, especially in the high-dose group (P < 0.05). Liver steatosis, inflammatory cell infiltration, and collagen fiber generation were significantly improved, which might be due to inhibition of CHOP, endoplasmic reticulum stress response, and JNK-mediated apoptosis and inflammation. Another study found that saponin from Zhujieshen, chikusetsusaponin V, attenuated LPS-induced acute liver injury in mice as a consequence of its potent anti-inflammatory activity [44]. The polysaccharides isolated from Zhujieshen had a protective effect on liver injury induced by acetaminophen and relieved acute liver injury induced by LPS and D-galactosamine. The mechanism for this protective effect may be related to its anti-inflammatory and antioxidant activities [45, 46]. Additionally, studies have indicated that extracts of Zhujieshen could improve non-alcoholic fatty liver induced in mice by high-fat diet [47] and have a preventive effect on alcoholic liver injury [48]. These studies demonstrate that Zhujieshen has potential in the prevention and treatment of liver diseases.

Cardio-protective effects

Different fractions extracted from Zhujieshen have exhibited highly effective cardiac protection [49]. The cardioprotective effects of saponins from Zhujieshen were evaluated in a rat model of acute myocardial ischemia injury via ligation of the left anterior descending branch [50]. Multiple indicators were utilized to evaluate cardiac protection, including infarct size, biochemical indicators, hemodynamics, influences on myocardial pathology, and mRNA expression of superoxide dismutase, caspase-3, and Bcl-2 families. The results demonstrated that saponins from Zhujieshen exerted beneficial cardiac protection by scavenging oxidative stress-triggered accumulation and overproduction of reactive oxygen species, alleviating myocardial ischemia injury and decreasing cardiac cell apoptosis. The mechanism involved the inhibition of NF-kB, ERK1/2, and p38 MAPK activation, increasing the expression of sirtuin1 to alleviate myocardial infarction injury and cardiac cell apoptosis [51]. Furthermore, chikusetsusaponin IVa, one of the main saponins from Zhujieshen, attenuated myocardial fibrosis induced by isoprenaline, mainly by activating autophagy through the AMPK/mTOR/ULK1 pathway [52]. These results suggest that Zhujieshen could be considered a potential candidate for the treatment of cardiac diseases.

Neuro-protective effects

The saponins from Zhujieshen conferred neuro-protection in natural aging rats and Alzheimer’s disease rats [53,54,55]. The mechanism may be partly through the regulation of oxidative stress and mitochondria-related pathways [56]. Saponins, the major components of Zhujieshen, have been reported to exhibit neuroprotective effects against D-galactose-induced neuronal injury by decreasing apoptosis and oxidative stress, ultimately improving cognitive performance [57]. The research revealed that this neuroprotective activity is closely correlated with Nrf2 and SIRT1-mediated anti-oxidant signaling pathways. Chikusetsusaponin V, the most abundant saponin from Zhujieshen, exhibited neuroprotective effects, possibly by modulation of SIRT1/PGC-1α/Mn-SOD signaling pathways [58]. In another study, chikusetsusaponin IVa attenuated isoflurane-induced neurotoxicity and cognitive deficits through SIRT1/ERK1/2 in developmental rats [59]. These findings suggested that Zhujieshen may have great promise for the treatment of neurodegenerative diseases.

Anti-tumor activity

Cancer is one of the biggest causes of morbidity and mortality in the world, making it an important target for natural medicines with less toxicity [60]. Zhujieshen is well-known for its potential anti-cancer effects. Studies have shown that chikusetsusaponins IVa can also inhibit prostate cancer cell proliferation and induce cell death without cytotoxicity in prostate normal cells. Its mechanism may be through the promotion of intracellular reactive oxygen species (ROS) production, thereby inducing mitochondrial-regulated apoptosis [61]. Furthermore, another study has indicated chikusetsusaponins IV and V exhibit anti-hepatoma effects by influencing apoptosis-related proteins, intracellular calcium levels, and cell proliferation analyzed through CCK-8 [62]. Moreover, Yuan D et al. further verified the inhibitory effect of total saponins of Panax japonicus on the growth of HL-60 cells in vitro by culturing HL-60 cells in vitro and detecting the viability and number of cancer cells [63]. In addition to that, the human gastric cancer cell line SGC-7901 [64], murine colon adenocarcinoma CT26 cells [65], mouse H22 hepatoma cells [66, 67], human ovarian cancer A2780 cells [68], human A549 lung cancer cells [69, 70], and human cervical cancer HeLa cells [71, 72] have been used to investigate the anti-tumor activity of Zhujieshen. All of these studies gave positive results that could be attributed to chikusetsusaponins IV, IVa, and V, total saponins, polysaccharides, and their derivatives. The most likely mechanisms include triggering of apoptosis, suppression of migration and invasion of cancer cells, and regulation of oncogene expression [68]. Therefore, Zhujieshen is proposed as a potential adjuvant therapy for protecting against human tumors in the future.

Antioxidant activity

The extracts obtained from the root of Zhujieshen have exhibited very promising antioxidant activities. The total extracts could eliminate excessive free radicals produced by the body to improve antioxidant capacity and exert anti-apoptotic effects through the Bcl-2 family protein, thereby delaying aging [73]. Similarly, polysaccharides from Zhujieshen showed good potential antioxidant activities by analysis of scavenging capacity for DPPH, hydrogen peroxide, and free radicals of superoxide anion in vitro [74]. Additionally, the essential oil extracted from Zhujieshen has also demonstrated antioxidant activity [75]. These findings suggest that Zhujieshen could be used as a potential treatment for oxidative stress-dependent disorders such as Alzheimer's disease, diabetes mellitus, and arteriosclerosis. However, further research should be conducted in vivo to fully understand its potential and the underlying mechanisms.

Effect on the immune system

In 1989, it was reported that two polysaccharides from Zhujieshen (Tachibana-A and Tachibana-B) exhibited reticuloendothelial-potentiating activity to improve the activity of macrophages in the reticuloendothelial system [76]. Huang et al. found that novel water-soluble highly branched heteropolysaccharides isolated from Zhujieshen were potential immunopotentiators, inhibited the proliferation of S-180 tumor cells, and protected essential organs in BALB/c mice [27]. Moreover, polysaccharides from Zhujieshen significantly improved immune function in Kunming mice with low immunity induced by cyclophosphamide [77,78,79]. Other immunopharmacological studies revealed that saponins from Zhujieshen exhibited potent stimulating effects in immunosuppressed mice via specific and nonspecific immunity [77,78,79,80]. Additionally, a composition of saponins and polysaccharides showed higher immunomodulatory effects than those of saponins or polysaccharides alone [81].

Effect on the hematological system

In ancient times, Panax japonicus had a good effect on diseases caused by “qi” stagnation and blood stasis. Modern studies have shown that Zhujieshen bipinnatifidus could be considered a potential substitute for P. notoginseng as a hemostatic herbs. It is mainly used for the treatment of trauma and ischemic cardiovascular diseases [82]. Matsuda et al. reported that chikusetsusaponin III, IV, and V showed the promotional effect of the fibrinolytic system [83]. Chikusetsusaponin IVa exerts antithrombotic effects, including minor hemorrhagic events. Research showed that chkusetsusaponin IVa could prolong the recalcification time, prothrombin time, activated partial thromboplastin time, and thrombin time of normal human plasma in a dose-dependent manner, thereby exerting a certain anti-thrombotic effect, including mild bleeding events [84]. Zhang and his team further extract and separate the crude extract of zhujieshen into polysaccharides and small molecular compounds. The research found that both extracted substances could accelerate the recovery of the white blood cell, red blood cell, and hemoglobin levels in the blood deficiency model mice. Hematopoietic activity may result from stimulating the secretion of interleukin-3, interleukin-6, erythropoietin, GM colony-stimulating factor (CSF), and M-CSF and by the resistance of spleen cells to apoptosis [85].

Other pharmacological effects

The saponins and polysaccharides from Zhujieshen exerted anti-hyperlipidemic potential in a mouse model of hyperlipidemia induced by celiac injection of 75% egg-yolk emulsion [86, 87]. Han et al. reported that chikusetsusaponins isolated from Zhujieshen had significant anti-obesity activity in mice fed a high-fat diet, which was partly related to delayed intestinal absorption of fat through inhibition of pancreatic lipase activity [88]. It was found that saponins from Zhujieshen significantly improved reproductive dysfunction in male mice fed a high-fat diet, possibly due to the reduction of macrophage infiltration and inhibition of testicular inflammation mediated by the NF-κB pathway [89]. Some pharmacological studies have found that extracts of Zhujieshen exhibited protective effects on gastric and intestinal mucous membranes [90,91,92]. It could also alleviate the renal injury of diabetic mice by reducing blood lipids, and regulating the biosynthesis of unsaturated fatty acids and purine metabolism [93]. Furthermore, it has been reported that saponins from Zhujieshen protect against cerebral ischemia injury [94, 95].

Pharmacokinetics

Studying the pharmacokinetic characteristics of Zhujieshen helps to understand its in vivo behavior and mechanism of action. The pharmacokinetic behaviors of six bioactive saponins (ginsenosides Rb1, Rg1, and Re, chikusetsusaponins V and IV, and hemsgiganoside B) from Zhujieshen were investigated by UHPLC-MS/MS [96]. After oral administration of total saponins at a dose of 500 mg/kg, the main non-compartmental parameters of the six analytes were calculated using DAS 3.2.8 software, including T1/2, Cmax, Tmax, AUC0−t, CLz/F, and apparent volume of distribution. The six analytes were quickly absorbed into the blood, and Cmax was reached within 0.73 h, except for ginsenoside Rb1 with a Cmax of 7.60 h. The values of Cmax, T1/2, and AUC0−t of ginsenoside Rb1 were significantly greater than those of the others, mainly due to a considerably lower value of CLz/F. When saponins extracted from Zhujieshen were orally administered at a dose of 173 mg/kg (equivalent to chikusetsusaponin V 100.3 mg/kg and chikusetsusaponinIV 36.7 mg/kg), similar values of T1/2 and Tmax were obtained for chikusetsusaponin V and chikusetsusaponin IV [97].

Authentication

Zhujieshen, one of the Panax species, is an expensive herbal medicine that is used in China, Korea, Japan, and Vietnam. As a consequence of its high pharmacological and economic value, many plants, including Phedimus aizoon (L.)’t Hart (景天三七), Panax stipuleanatus H. T. Tsai & K. M. Feng (屏边三七), Panax japonicus C. A. Mey. Var. angustifolius (Burk) Cheng et Chu (狭叶竹节参) and Panax japonicus C. A. Mey. Var. bipinnatifada (Seem.) C. Y. Wu et K. M. Feng (羽叶三七), with similar root morphology, have been used to adulterate Zhujieshen [98, 99]. Morphological and histological characteristics are the traditional methods for authentication of herbal medicines, which could be used to authenticate Zhujieshen and its common adulterant (Phedimus aizoon (L.)’t Hart.) [100]. However, these methods may not always be precise enough, especially when dealing with closely related species with similar appearances or intra-species morphological variations. Additionally, most commercial ginseng products are solid forms, including powder, granule, pill, capsule, or other processed products, which makes identification difficult. Following advances in the understanding of gene function, DNA barcoding, a biological technique, has been widely used in species identification, breeding, and evolutionary studies [101, 102]. The identification of species-specific DNA markers has played an important role in the authentication of Zhujieshen and related species or adulterants [99, 103, 104]. ITS2 has been successfully established to differentiate Zhujieshen and its non-identical adulterants with a high-rate identification [99]. Choi et al. determined a species-specific amplified fragment length polymorphism-derived sequence for rapid authentication of Zhujieshen among other related Panax species [105]. Nguyen et al. developed 18 coding DNA sequence-derived, species-specific, single nucleotide polymorphism markers from chloroplast genomes to authenticate seven Panax species (Panax japonicus, Panax ginseng, Panax notoginseng, Panax stipuleanatus, Panax vietnamensis, Panax quinquefolius, and Panax trifolius), enabling differentiation of these seven species from the others [106].

Predictive analysis on quality marker

The combination of microscopic and physicochemical identification is an important tool for identification when Zhujieshen is processed into different products in the Chinese Pharmacopoeia Commission (2020). Given that ginsenoside Ro and chikusetsusaponin IVa are the characteristic ingredients of Zhujieshen, many investigations have used them as biomarkers to determine its quality. According to the reports, triterpenoid saponins compounds including chikusetsusaponin V, chikusetsusaponin IVa, and chikusetsusaponin IV, have multiple activities associated with the efficacy of Zhujieshen, including anti-tumor, anti-inflammatory, and anti-myocardial ischemia effects. Moreover, these compounds have stable activity, can be quantitatively measured, and possess traceability properties [107, 108]. According to the concept of quality markers (Quality markers) proposed by Academician Liu Changxiao, triterpenoids can be the Q-marker of Zhujieshen [109].

Conclusion and future prospects

It is reported that Zhujieshen possesses both the conserving vitality activities of Panax ginseng C. A. Mey and the replenishing blood activities of Panax notoginseng (Burkill) F. H. Chen ex C. H. simultaneously [110]. It was traditionally used as Panax ginseng to enhance immunity. Additionally, it can be used in the treatment of rheumatic arthritis as Panax notoginseng. This is why Zhujieshen was also referred to as the "king of herbs" in traditional Tujia and Hmong medicines. Also, it has been successfully used in clinical practice for centuries to treat fractures, hematemesis, cough, bleeding wounds, arthralgia, and weakness after illness. Phytochemical research has indicated that saponins and polysaccharides are the major active constituents. The total saponin content in the roots of Zhujieshen can reach 15%, which is 2 to sevenfold higher than that of P. ginseng. Various types of saponins have been isolated and a range of pharmacological studies have focused on these components. The crude extracts and pure compounds from Zhujieshen have been shown to have diverse pharmacological activities including anti-inflammatory, hepato-protective, cardio-protective, neuro-protective, anti-tumor, anti-oxidant, anti-thrombotic and immunomodulatory activities (Fig. 4). Recent studies have also focused on the potential of Zhujieshen in oxidative stress-dependent disorders such as Alzheimer’s disease, diabetes mellitus, and arteriosclerosis. Although there are many promising results, most studies have only been conducted in cell lines in vitro or animal models. Mechanism of action studies are very limited and no clinical trials of Zhujieshen extracts have been reported yet. Further research on components and their mechanisms of action is needed to advance the therapeutic role of Zhujieshen. Additionally, pharmacological investigation of new saponins and volatile oils, and the exact structural characteristics of polysaccharides and trace compounds isolated from Zhujieshen have mostly been ignored, limiting the diversity of research and application of Zhujieshen. Furthermore, few modern studies have been conducted to confirm any side effects or toxicity. Finally, future research work needs to be focused on the identification of components, further pharmacological studies, and therapeutic mechanisms, together with links between traditional uses, active compounds, and reported pharmacological activities. Such studies would help confirm clinical efficacy and enable improved quality control based on the active compounds. This review might be helpful to summarize the current status of research on Zhujieshen and point out directions for future research.

Fig. 4
figure 4

Pharmacological properties of Zhujieshen

Availability of data and materials

Not applicable.

Abbreviations

AMPK:

Adenosine monophosphate-activated protein kinase

AUC0−t :

Mean residence time, area under the concentration–time curve

CHOP:

C/EBP homologous protein

CLz/F :

Apparent clearance

Cmax :

Maximum plasma concentration

CSF:

Colony-stimulating factor

DPPH:

2,2-Diphenyl-1-picryl-hydrazyl

ERK1/2:

Extracellular regulated protein kinases 1/2

JNK:

P-Jun N-terminal kinase

LPS:

Lipopolysaccharide

MAPK:

Mitogen-activated protein kinase

Mn-SOD:

Manganese superoxide dismutase

mTOR:

Mammalian target of rapamycin

NF-κB:

Nuclear factor-kappa B

NLRP3:

Nod-like receptor protein 3

Nrf2:

Nuclear factor erythroid 2-related factor 2

PGC-1α:

Peroxisome proliferator-activated receptor gamma-coactivator-1α

Q-marker:

Quality marker

ROS:

Reactive oxygen species

SIRT1:

Silent mating type information regulation 2 homologue

T1/2 :

Elimination half-life

Tmax :

Time to reach Cmax

ULK1:

Uncoordinated-51-like kinase1

References

  1. Yang X, Wang R, Zhang S, et al. Polysaccharides from Panax japonicus C.A. Meyer and their antioxidant activities. Carbohydr Polym. 2014;101:386–91.

    Article  CAS  PubMed  Google Scholar 

  2. Morita T, Tanaka O, Kohda H. Saponin composition of rhizomes of Panax japonicus collected in South Kyushu, Japan, and its significance in oriental traditional medicine. Chem pharm bull. 1985;33:3852–8.

    Article  CAS  Google Scholar 

  3. Zhang S, Wu Y, Jin J, et al. De novo characterization of Panax japonicus C. A. Mey transcriptome and genes related to triterpenoid saponin biosynthesis. Biochem Biophys Res Co. 2015;466(3):450–5.

    Article  CAS  Google Scholar 

  4. Wang R, Chen P, Jia F, et al. Characterization and antioxidant activities of polysaccharides from Panax japonicus C.A. Meyer. Carbohydr Polym. 2012;88(4):1402–6.

    Article  CAS  Google Scholar 

  5. You XL, Han JY, Choi YE. Plant regeneration via direct somatic embryogenesis in Panax japonicus. Plant Biotechnol Rep. 2007;1(1):5–9.

    Article  Google Scholar 

  6. Ngan F, Shaw P, But P, et al. Molecular authentication of Panax species. Phytochemistry. 1999;50(5):787–91.

    Article  CAS  PubMed  Google Scholar 

  7. Xia P, Li J, Wang R, et al. Comparative study on volatile oils of four Panax genus species in Southeast Asia by gas chromatography–mass spectrometry. Ind Crops Prod. 2015;74:478–84.

    Article  CAS  Google Scholar 

  8. Yoshizaki K, Devkota HP, Yahara S. Four new triterpenoid saponins from the leaves of Panax japonicus grown in southern Miyazaki Prefecture (4). Chem Pharm Bull. 2013;61(3):273–8.

    Article  CAS  Google Scholar 

  9. Yoshizaki K, Devkota HP, Fujino H, et al. Saponins composition of rhizomes, taproots, and lateral roots of Satsuma-ninjin (Panax japonicus). Chem Pharm Bull. 2013;61(3):344–50.

    Article  CAS  Google Scholar 

  10. Yoshizaki K, Murakami M, Fujino H, et al. New triterpenoid saponins from fruit specimens of Panax japonicus collected in Toyama Prefecture and Hokkaido (2). Chem Pharm Bull. 2012;60(6):728–35.

    Article  CAS  Google Scholar 

  11. Ouyang LN, Xiang DW, Wu X, et al. Research progress on chemical constituents and pharmacological activities of Panax japonicus. Chin Tradit Herbal Drugs. 2010;41(6):1023–7.

    CAS  Google Scholar 

  12. Zhou M, Xu M, Zhu HT, et al. New dammarane-type saponins from the rhizomes of Panax japonicus. Helv Chim Acta. 2011;94(11):2010–9.

    Article  CAS  Google Scholar 

  13. Tanaka K, Kubota M, Zhu S, et al. Analysis of ginsenosides in Ginseng drugs using liquid chromatography Fourier transform ion cyclotron resonance mass spectrometry. Nat Prod Commun. 2007;2(6):625–32.

    CAS  Google Scholar 

  14. Chen J, Tan M, Zou L, et al. Qualitative and quantitative analysis of the saponins in Panacis Japonici Rhizoma using ultra-fast liquid chromatography coupled with triple quadrupole-time of flight tandem mass spectrometry and ultra-fast liquid chromatography coupled with triple quadrupole-linear ion trap tandem mass spectrometry. Chem Pharm Bull. 2019;67(8):839–48.

    Article  CAS  Google Scholar 

  15. Wu QS, Chen P, Zhang QW, et al. Advances in research of chemical constituents, pharmacological activities and analytical methods of Panax japonicus. Asia Pac Tradit Med. 2016;12(06):46–54.

    Google Scholar 

  16. Du Z, Li J, Zhang X, et al. An integrated LC-MS-based strategy for the quality assessment and discrimination of three Panax Species. Molecules. 2018;23(11):2988.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Chen JL, Tan MX, Zou LS, et al. Stimultaneous determination of multiple bioactive constituents in Panacis Japonici Rhizoma processed by different methods and grey relational analysis. Chin J of Chin Mater Med. 2018;43(21):4274–82.

    Google Scholar 

  18. Tanaka O, Morita T, Kasai R, et al. Study on saponins of rhizomes of Panax pseudo-ginseng subsp. himalaicus collected at Tzatogang and Pari-la, Bhutan-Himalaya. Chem Pharm Bull. 1985;33(6):2323–30.

    Article  CAS  Google Scholar 

  19. Cai P, Xiao ZY, Wei JX. Chemical constituents of Panax japonicus (I). Chin Tradit Herbal Drugs. 1982;13(3):1–2.

    CAS  Google Scholar 

  20. Cai P, Xiao ZY. Chemical constituents of Panax japonicus (II). Chin Tradit Herbal Drugs. 1984;15(6):1–6.

    Google Scholar 

  21. Atopkina LN, Denisenko VA. Synthesis of 20S-protopanaxatriol β-d-glucopyranosides. Chem Nat Compd. 2019;55(1):82–7.

    Article  CAS  Google Scholar 

  22. Jia L, Zhao Y. Current evaluation of the millennium phytomedicine–ginseng (I): etymology, pharmacognosy, phytochemistry, market and regulations. Curr Med Chem. 2009;16(19):2475.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zou K, Zhu S, Meselhy MR, et al. Dammarane-type saponins from Panax japonicus and their neurite outgrowth activity in SK-N-SH cells. J Nat Prod. 2002;65(9):1288–92.

    Article  CAS  PubMed  Google Scholar 

  24. Zou K, Zhu S, Tohda C, et al. Dammarane-type triterpene saponins from Panax japonicus. J Nat Prod. 2002;65(3):346–51.

    Article  CAS  PubMed  Google Scholar 

  25. Huang Z, Huang Y, Li X, et al. Molecular mass and chain conformations of Rhizoma Panacis Japonici polysaccharides. Carbohydr Polym. 2009;78(3):596–601.

    Article  CAS  Google Scholar 

  26. Huang Z, Zhang L. Chemical structures of water-soluble polysaccharides from Rhizoma Panacis Japonici. Carbohydr Res. 2009;344(9):1136–40.

    Article  CAS  PubMed  Google Scholar 

  27. Huang Z, Zhang L, Duan X, et al. Novel highly branched water-soluble heteropolysaccharides as immunopotentiators to inhibit S-180 tumor cell growth in BALB/c mice. Carbohyd Polym. 2012;87(1):427–34.

    Article  CAS  Google Scholar 

  28. Meyer C, Zhang L, Zhang X, et al. Comparative analysis of the essential oils from normal and hairy roots of Panax japonicas C.A. Meyer. Afr J Biotechnol. 2011;10:2440–5.

    Google Scholar 

  29. Yang LB, Liu SJ, Da LL, et al. Research of fat-soluble components of Panax japonicus C. A. Mey. J Anhui Agric Sci. 2011;39(20):12145–6.

    Google Scholar 

  30. Chen L, Ren H, Xu R, et al. The effect of Fufang Zhujieshen tablets on inflammatory factors in osteoarthritis. Chin Hosp Pharm J. 2019;39(06):580–5.

    Google Scholar 

  31. Tan QL. The effects of anti -inflammatory of compound Panax japonicus Tablet in rheumatoid arthritis mice. Chin Med Herald. 2011;8(28):27–8.

    Article  Google Scholar 

  32. Wang ZF, Tan QL, Zhang H, et al. Experimental studies on the mechanism of compound Japanese Ginseng pill in treatment of Rheumatoid arthritis. Lishizhen Med Mater Med Res. 2009;20(7):1611–3.

    Google Scholar 

  33. Wen DJ, Chen GD, Zhang CL, et al. Study on the anti-inflammatory effects of total Panax japonicus saponins. Lishizhen Med Mater Med Res. 2008;19(5):1155–6.

    CAS  Google Scholar 

  34. Deng L, Yuan D, Zhou Z, et al. Saponins from Panax japonicus attenuate age-related neuroinflammation via regulation of the mitogen-activated protein kinase and nuclear factor kappa B signaling pathways. Neural Regen Res. 2017;12(11):1877–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cheng ZH, Dun YY, Liu J, et al. Effects of saponins from Panax japonicus on colonic inflammation through Neu3/IAP signaling pathway in aging rats. Lishizhen Med Mater Med Res. 2019;30(7):1597–601.

    Google Scholar 

  36. Wang T, Dai Y, Dun Y, et al. Chikusetsusaponin V inhibits inflammatory responses via NF-κB and MAPK signaling pathways in LPS-induced RAW 264.7 macrophages. Immunopharm Immunot. 2014;36(6):404–11.

    Article  CAS  Google Scholar 

  37. Yuan C, Liu C, Wang T, et al. Chikusetsu saponinIVa ameliorates high fat diet-induced inflammation in adipose tissue of mice through inhibition of NLRP3 inflammasome activation and NF-κB signaling. Oncotarget. 2017;8(19):31023.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Zhao QQ, Wang T, Yuang D, et al. Effect of Panax japonicus polysaccharide on LPS induced microglial inflammatory response. J Chin Med Mater. 2019;42(6):1409–12.

    Google Scholar 

  39. Duan L, Liu CQ, Wu LC, et al. Effects of Panax japonicus hypolipidemic compound on non-alcoholic fatty liver disease in mice and its mechanism. Med J Chin PLA. 2017;42(9):764–8.

    Google Scholar 

  40. Qin YE, Cui QQ, Zhang CC, et al. Effects of total saponins from Panax japonicus on acute hepatic injury induced by carbon tetrachloride. Chin J Inf Tradit Chin Med. 2014;21(10):47–9.

    Google Scholar 

  41. Qin YE, Zhang CC, Wang T, et al. Effect of polysaccharide from Panax japonicus on hepatic cell injury. Chin J Inf Tradit Chin Med. 2014;21(11):59–62.

    CAS  Google Scholar 

  42. Yang XL, Chen P. Protective Effects of polysaccharide and total Saponins from Panax japonicus on acute hepatic injury. Chin J Inf Tradit Chin Med. 2011;17(1):65–6.

    Google Scholar 

  43. Yuan D, Xiang T, Huo Y, et al. Preventive effects of total saponins of Panax japonicus on fatty liver fibrosis in mice. Arch Med Sci. 2018;14(2):396–406.

    Article  CAS  PubMed  Google Scholar 

  44. Dai YW, Zhang CC, Zhao HX, et al. Chikusetsusaponin V attenuates lipopolysaccharide-induced liver injury in mice. Immunopharm Immunot. 2016;38(3):167–74.

    Article  CAS  Google Scholar 

  45. Xu R, Liu Z, Fu Q, et al. Protective effects of polysaccharides from Panax japonicus on mice with liver injury induced by acetaminophen. J South-Central Univ Nat (Nat Sci Ed). 2020;39(1):51–5.

    Google Scholar 

  46. Jiang SQ, Duan H, Shu GW, et al. Protective effects of polysaccharides from Panax japonicus on mice with acute liver injury induced by LPS/D-GalN. Chin Med Mat. 2017;40(5):1170–3.

    Google Scholar 

  47. Zhou Q, Duan L, Wu LC, et al. Experimental study of the protective effects of extracts of Panax japonica rhizoma, Salviae Miltiorrhiz radix Et Rhizoma and Crataegi Fructus compound on the hypolipidaemic in nonalcoholic fatty liver of mice. Chin J Clin Pharmacol. 2018;34(13):1532–5.

    Google Scholar 

  48. He ZG, Wang YP, Liu L, et al. Effects of Panax japonicus extractions on serum biochemical indices and inflammatory factors in mice with alcoholic liver injury. Zhejiang J Integr Tradit Chin West Med. 2018;28(1):21–4.

    Google Scholar 

  49. He HB, Xu J, Wang HW, et al. Effects of preconditioning with different fractions extracted from Panax japonicus C. A. Mey on acute myocardial ischemia in rats induced by ligation of left anterior descending branch. J Third Mil Med Univ. 2011;33(14):1462–6.

    Google Scholar 

  50. He H, Xu J, Xu Y, et al. Cardioprotective effects of saponins from Panax japonicus on acute myocardial ischemia against oxidative stress-triggered damage and cardiac cell death in rats. J Ethnopharmacol. 2012;140(1):73–82.

    Article  CAS  PubMed  Google Scholar 

  51. Wei N, Zhang C, He H, et al. Protective effect of saponins extract from Panax japonicus on myocardial infarction: involvement of NF-κB, Sirt1 and mitogen-activated protein kinase signalling pathways and inhibition of inflammation. J Pharm Pharmacol. 2014;66(11):1641–51.

    Article  CAS  PubMed  Google Scholar 

  52. Wang L, Yuan D, Zheng J, et al. Chikusetsu saponin IVa attenuates isoprenaline-induced myocardial fibrosis in mice through activation autophagy mediated by AMPK/mTOR/ULK1 signaling. Phytomedicine. 2019;58:152764.

    Article  CAS  PubMed  Google Scholar 

  53. Zeng CH, Wang WS, Lu WJ, et al. Effects of Panax japonicus on the expression of Drd-2 and GFAP, TNF-α in hippocampus of AD Rats. Genomics Appl Biol. 2019;38(4):1560–5.

    Google Scholar 

  54. Zeng CH, Min Y, Lu WJ, et al. Effects of Panax japonicus on the expression of Inos, Arg-1, Aβ1-42, TNF-α in hippocampal microglia of AD Rats. Lishizhen Med Mater Med Res. 2018;29(9):2097–100.

    Google Scholar 

  55. Zhao H, Wang L, Zhang QX, et al. Effects of Panax japonicus on transmitter amino acid and free radical metabolism in vascular dementia rat. Chin J Gerontol. 2010;30(21):3096–8.

    Google Scholar 

  56. Wan JZ, Wang R, Zhou ZY, et al. Saponins of Panax japonicus confer neuroprotection against brain aging through mitochondrial related oxidative stress and autophagy in rats. Curr Pharm Biotechnol. 2020;21(8):667–80.

    Article  CAS  PubMed  Google Scholar 

  57. Wang T, Di G, Yang L, et al. Saponins from Panax japonicus attenuate D-galactose-induced cognitive impairment through its anti-oxidative and anti-apoptotic effects in rats. J Pharm Pharmacol. 2015;67(9):1284–96.

    Article  CAS  PubMed  Google Scholar 

  58. Wan J, Deng L, Zhang C, et al. Chikusetsu saponin V attenuates H2O2- induced oxidative stress in human neuroblastoma SH-SY5Y cells through Sirt1/PGC-1α/Mn-SOD signaling pathways. Can J Physiol Pharm. 2016;94(9):919–28.

    Article  Google Scholar 

  59. Fang X, Han Q, Li S, et al. Chikusetsu saponin IVa attenuates isoflurane-induced neurotoxicity and cognitive deficits via SIRT1/ERK1/2 in developmental rats. Am J Transl Res. 2017;9(9):4288–99.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Kim H, Lee H, Kim DJ, et al. Panax ginseng exerts antiproliferative effects on rat hepatocarcinogenesis. Nutr Res. 2013;33(9):753–60.

    Article  CAS  PubMed  Google Scholar 

  61. Zhu WB, Tian FJ, Liu LQ. Chikusetsu (CHI) triggers mitochondria-regulated apoptosis in human prostate cancer via reactive oxygen species (ROS) production. Biomed Pharmacother. 2017;90:446–54.

    Article  CAS  PubMed  Google Scholar 

  62. Zuo T, Zhang Z, Jiang P, Zhang R, et al. Characterization of chikusetsusaponinIV and V induced apoptosis in HepG2 cancer cells. Mol Biol Rep. 2022;49(6):4247–55.

    Article  CAS  PubMed  Google Scholar 

  63. Yuan D, Zuo R, Zhang CC. Effects of total saponins of Panax japonicus on human leukemic HL-60 cells. J Integr Med. 2007;5(5):570–2.

    CAS  Google Scholar 

  64. Zhang Y, Wang G, Zuo T, et al. Effects of Chikusetsusaponin IV, IVa and V on the proliferation, migration, invasion and apoptosis of human gastric cancer SGC-7901 cells. Tradit Chin Drug Res Clin Pharmacol. 2019;30(7):796–801.

    Google Scholar 

  65. Zhou ZY, Chen YX, Li DH, et al. Total saponins of Panax japonicus improve cancer cachexia in mice through inhibiting inflammatory response mediated by NF- κB. Chin Pharmacol Bull. 2018;34(4):532–7.

    Google Scholar 

  66. Shu G, Jiang S, Mu J, et al. Antitumor immunostimulatory activity of polysaccharides from Panax japonicus C. A. Mey: Roles of their effects on CD4+ T cells and tumor associated macrophages. Int J Biol Macromol. 2018;111:430–9.

    Article  CAS  PubMed  Google Scholar 

  67. Chen C, Wu W, Xu X, et al. Chain conformation and anti-tumor activity of derivatives of polysaccharide from Rhizoma Panacis Japonici. Carbohydr Polym. 2014;105:308–16.

    Article  CAS  PubMed  Google Scholar 

  68. Chen X, Wu Q, Meng F, et al. ChikusetsusaponinIVa methyl ester induces G1 cell cycle arrest, triggers apoptosis and inhibits migration and invasion in ovarian cancer cells. Phytomedicine. 2016;23(13):1555–65.

    Article  CAS  PubMed  Google Scholar 

  69. Jiang HY, Deng QH, Zheng XH, et al. Effects of total saponins of Panax japonicus on apoptosis and activity of caspase-3 on A549 lung cancer cell. Sci Technol Eng. 2015;15(25):100–3.

    Google Scholar 

  70. Gao GZ, Zhang HR, Zhang T, et al. Study on the mechanism of saponins from Panax japonicus on inhition of A549 cell proliferation and migration through regulating PTEN/PI3K/Akt Pathway. Prog Mod Biomed. 2020;20(2):242–7.

    Google Scholar 

  71. Ni GF. Experimental study on antitumor activity of different extracts from Panax japonicus in vitro. Zhejiang J Tradit Chin Med. 2007;42(4):230–1.

    Google Scholar 

  72. Liu Y, Huang Z. Effect of water extract of Panax japonicus on apoptosis of cervical cancer Hela cells. Chin Tradit Pat Med. 2021;43(01):224–7.

    Google Scholar 

  73. Xi M, Hai C, Tang H, et al. Antioxidant and antiglycation properties of total saponins extracted from traditional Chinese medicine used to treat diabetes mellitus. Phytother Res. 2008;22(2):228–37.

    Article  CAS  PubMed  Google Scholar 

  74. Wang R, Chen P, Jia F, et al. Optimization of polysaccharides from Panax japonicus C.A. Meyer by RSM and its anti-oxidant activity. Int J Biol Macromol. 2012;50(2):331–6.

    Article  CAS  PubMed  Google Scholar 

  75. Zhang L, Zhao RF. Essential oil and antioxidant activity of tuber from Panax japonicus in Guizhou. Guizhou Agri Sci. 2010;38(6):44–6.

    Google Scholar 

  76. Ohtani K, Hatono S, Mizutani K, et al. Reticuloendothelial system-activating polysaccharides from rhizomes of Panax japonicas. I. Tochibanan-A and -B. Chem Pharm Bull. 1989;37(10):2587.

    Article  CAS  Google Scholar 

  77. Zhang J, Li CY, Li JP, et al. Immunoregulation on mice of low immunity and effects on five kinds of human cancer cells of Panax japonicus polysaccharide. Evid-Based Compl Alt. 2015;2015:839697.

    Google Scholar 

  78. Zhang J, Li CY, Li JP, et al. Immunoregulative effects of Panax japonicus polysaccharide on mice of low immunity. In: 2014 China Pharmaceutical Conference and the 14th China Pharmacist Week Proceedings. 2014; 1–6.

  79. Zhang CC, Zhao HX, Jiang MJ, et al. Effects of polysaccharides isolated from Panax japonicus on immunosuppression mice. J Chin Med Mater. 2011;34(1):91–4.

    Google Scholar 

  80. Zhang CC, Jiang MJ, Zhao HX, et al. Effects of total saponins of Panax japonicus Rhizoma on cyclophosphamide-in-duced immunosuppressed mice. Chin Tradit Pat Med. 2011;33(7):1134–8.

    Google Scholar 

  81. Wang HW, Jiang MJ, Zhao HX, et al. Immunomodulatory effects of saponin-polysaccharide and Panax japonicus composition on cyclophosphamide-induced immunosuppressed mice. Guangdong Med J. 2010;31(20):2620–2.

    CAS  Google Scholar 

  82. Yang BR, Yuen SC, Fan GY, et al. Identification of certain Panax species to be potential substitutes for Panax notoginseng in hemostatic treatments. Pharmacol Res. 2018;134:1–15.

    Article  CAS  PubMed  Google Scholar 

  83. Matsuda H, Samukawa KL, Fukuda S, et al. Studies of Panax japonicus fibrinolysis. Planta Med. 1989;55(1):18–21.

    Article  CAS  PubMed  Google Scholar 

  84. Dahmer T, Berger M, Barlette AG, et al. Antithrombotic effect of Chikusetsusaponin IVa isolated from Ilex paraguariensis (Mate). J Med Food. 2012;15(12):1073–80.

    Article  CAS  PubMed  Google Scholar 

  85. Zhang H, Wang HF, Liu Y, et al. The haematopoietic effect of Panax japonicus on blood deficiency model mice. J Ethnopharmacol. 2014;154(3):818–24.

    Article  PubMed  Google Scholar 

  86. Yang XL, Chen P, Wang RF, et al. Experimental study on the antihyperlipidemia effect of the polysaccharides of Panax japonicus in mice. Chin Hosp Pharm J. 2011;31(6):433–5.

    CAS  Google Scholar 

  87. Yang XL, Chen P. Experimental study on the anti hyperlipidemia effect of the total rhizoma Panacis japonica saponins. Acta Chin Med Pharmacol. 2010;38(6):22–4.

    Google Scholar 

  88. Han L, Zheng Y, Yoshikawa M, et al. Anti-obesity effects of chikusetsusaponins isolated from Panax japonicus rhizomes. BMC Complem Altern M. 2005;5(1):9.

    Article  Google Scholar 

  89. Ma QY, Zhang CC, Yang SQ, et al. Protective mechanism of saponins of Panax japonicus on high-fat diet induced reproductive dysfunction in mice. Chin Pharmacol Bull. 2019;35(10):1375–80.

    Google Scholar 

  90. Yamahara J, Kubomura Y, Miki K, et al. Anti-ulcer action of Panax japonicus rhizome. J Ethnopharmacol. 1987;19(1):95–101.

    Article  CAS  PubMed  Google Scholar 

  91. Mei Y, Xiao CC, Feng JW, et al. The preventive effect of total saponins of Panax japonicus on nonsteroidal anti-inflammatory drug-induced enteropathy. Chin J Cell Mol Immunol. 2016;32(6):734–8.

    Google Scholar 

  92. Borrelli F, Izzo AA. The plant kingdom as a source of anti-ulcer remedies. Phytother Res. 2000;14(8):581–91.

    Article  CAS  PubMed  Google Scholar 

  93. Wang T, Huang X, Zhai K, et al. Integrating metabolomics and network pharmacology to investigate Panax japonicus prevents kidney injury in HFD/STZ-induced diabetic mice. J Ethnopharmacol. 2023;303:115893.

    Article  CAS  PubMed  Google Scholar 

  94. Zhao H, Zhang QX, Mu Y. Effects of total rhizoma Panacis japonjica saponins (tRPJS) on nitric oxide synthase in hippocampus region following rat ischemic cerebral injury. Chin J Chin Mater Med. 2008;33(5):557–9.

    Google Scholar 

  95. Jia ZH, Zhao H. Effects of TSPJ on neuronal apoptosis and related gene expression in rat cerebral ischemia-reperfusion injury. Chin J Exp Tradit Med Form. 2011;17(21):168–72.

    CAS  Google Scholar 

  96. Zheng H, Qiu F, Zhao H, et al. Simultaneous determination of six bioactive saponins from Rhizoma Panacis Japonici in rat plasma by UHPLC-MS/MS: application to a pharmacokinetic study. J Chromatogr B. 2018;1092:199–206.

    Article  CAS  Google Scholar 

  97. Qi D, Yang X, Chen J, et al. Determination of chikusetsusaponin V and chikusetsusaponinIV in rat plasma by liquid chromatography-mass spectrometry and its application to a preliminary pharmacokinetic study. Biomed Chromatogr. 2013;27(11):1568–73.

    Article  CAS  PubMed  Google Scholar 

  98. Xu C, Zhang CC, Li J, et al. Identification of Panax japonicus and its related plant species based on RAPD markers. Lishizhen Med Mate Med Res. 2016;27(1):101–4.

    CAS  Google Scholar 

  99. Chen JA, Yang L, Li RZ, et al. Identification of Panax japonicus and its related species or adulterants using ITS2 sequence. China Tradit Herb Drugs. 2018;49(15):3672–80.

    Google Scholar 

  100. Xia L, Liang YS, Li SL, et al. Comparative identification of Panax japonicus and its adulterant Sedum notoginseng. J Chin Med Mater. 2014;37(5):797–800.

    Google Scholar 

  101. Duan YM, Duan CL. Application of DNA molecular markers in the research of Panax Ginseng. Res Pract Chin Med. 2009;23(6):74–6.

    CAS  Google Scholar 

  102. Zhu S, Fushimi H, Komatsu K. Development of a DNA microarray for authentication of Ginseng drugs based on 18S rRNA gene sequence. J Agr Food Chem. 2008;56(11):3953–9.

    Article  CAS  Google Scholar 

  103. Zhou M, Gong X, Pan Y. Panax species identification with the assistance of DNA data. Genet Resour Crop Ev. 2018;65(7):1839–56.

    Article  CAS  Google Scholar 

  104. Cheng Y, Zou P, Zhang CF, et al. Development and validation of SSR markers based on transcriptome of Panax japonicus. J Chin Med Mater. 2017;40(12):2805–9.

    Google Scholar 

  105. Choi Y, Ahn CH, Kim B, et al. Development of species specific AFLP-derived SCAR marker for authentication of Panax japonicus C. A. MEYER. Biol Pharm Bull. 2008;31(1):135–8.

    Article  PubMed  Google Scholar 

  106. Nguyen VB, Linh Giang VN, Waminal NE, et al. Comprehensive comparative analysis of chloroplast genomes from seven Panax species and development of an authentication system based on species-unique single nucleotide polymorphism markers. J Ginseng Res. 2020;44(1):135–44.

    Article  PubMed  Google Scholar 

  107. Wu Q, Wang C, Lu J, et al. Simultaneous determination of six saponins in Panacis japonici rhizoma using quantitative analysis of multi-components with single-marker method. Curr Pharm Anal. 2017;13:289–95.

    Article  CAS  Google Scholar 

  108. Meng F, Wu Q, Wang R, et al. A novel strategy for quantitative analysis of major ginsenosides in Panacis japonici rhizoma with a standardized reference fraction. Molecules. 2017;22(12):2067.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Wang XJ, Xie Q, Liu Y, et al. Panax japonicus and chikusetsusaponins: a review of diverse biological activities and pharmacology mechanism. Chin Herb Med. 2020;13(1):64–77.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Zhang SP, Wang RF, Zeng WY, et al. Resource investigation of traditional medicinal plant Panax japonicus (T. Nees) C. A. Mey and its varieties in China. J Ethnopharmacol. 2015;166:79–85.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Thanks Chao Jiang (National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences) for providing herbal pictures.

Funding

This work has not received any financial support.

Author information

Authors and Affiliations

Authors

Contributions

YC and ML wrote the manuscript. ML systemically revised the manuscript for important content. JW, ZY, and GL completed the Figures and Tables. XR and YC collected literature and checked data. LS proposed the conception and designed the structure of the manuscript.

Corresponding author

Correspondence to Lili Sun.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The manuscript is approved by all authors for publication.

Competing interests

The authors declare no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Liu, M., Wen, J. et al. Panax japonicus C.A. Meyer: a comprehensive review on botany, phytochemistry, pharmacology, pharmacokinetics and authentication. Chin Med 18, 148 (2023). https://doi.org/10.1186/s13020-023-00857-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13020-023-00857-y

Keywords