Next Article in Journal / Special Issue
A Review of Seven Applications of Neutrosophic Logic: In Cultural Psychology, Economics Theorizing, Conflict Resolution, Philosophy of Science, etc.
Previous Article in Journal
Concept, Possibilities and Pilot-Testing of a New Smartphone Application for the Social and Life Sciences to Study Human Behavior Including Validation Data from Personality Psychology
Previous Article in Special Issue
The 2016 US Presidential Elections: What Went Wrong in Pre-Election Polls? Demographics Help to Explain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Red Ginseng on Neural Injuries with Reference to the Molecular Mechanisms

Department of Functional Histology, Ehime University Graduate School of Medicine, Ehime 791-0295, Japan
*
Author to whom correspondence should be addressed.
J 2019, 2(2), 116-127; https://doi.org/10.3390/j2020009
Submission received: 13 March 2019 / Revised: 24 March 2019 / Accepted: 26 March 2019 / Published: 28 March 2019
(This article belongs to the Special Issue Feature Papers for J-Multidisciplinary Scientific Journal)

Abstract

:
Red ginseng, as an effective herbal medicine, has been traditionally and empirically used for the treatment of neuronal diseases. Many studies suggest that red ginseng and its ingredients protect the brain and spinal cord from neural injuries such as ischemia, trauma, and neurodegeneration. This review focuses on the molecular mechanisms underlying the neuroprotective effects of red ginseng and its ingredients. Ginsenoside Rb1 and other ginsenosides are regarded as the active ingredients of red ginseng; the anti-apoptotic, anti-inflammatory, and anti-oxidative actions of ginsenosides, together with a series of bioactive molecules relevant to the above actions, appear to account for the neuroprotective effects in vivo and/or in vitro. Moreover, in this review, the possibility is raised that more effective or stable neuroprotective derivatives based on the chemical structures of ginsenosides could be developed. Although further studies, including clinical trials, are necessary to confirm the pharmacological properties of red ginseng and its ingredients, red ginseng and its ingredients could be promising candidate drugs for the treatment of neural injuries.

1. Introduction

As a variety of traditional medicine, ginseng root (Panax ginseng CA Meyer) has been utilized for centuries and is one of the most trusted herbs in Asia. There are two types of ginseng products: red ginseng (RG), which is steamed and dried ginseng root as described in the Japanese Pharmacopoeia, and white ginseng (WG), which is dried but not steamed. Following vapor steaming, RG can not only be preserved for a long time but also endowed with new ingredients not present in fresh ginseng. Many studies have demonstrated that RG and its ingredients show broad pharmacological effects in human and animal models. These effects include anti-aging, anti-oxidant, anti-inflammation, anti-diabetes, anti-depression, among others [1,2,3].
Injury to the nervous tissue is commonly caused by ischemia, trauma, and neuro-degeneration [4]. Historically, RG was thought to be beneficial to the nervous system, and modern researchers have shown increasing evidence for the favorable effects of RG and its ingredients on the nervous system under pathological conditions [5]. This review aims to summarize the effects of RG and its ingredients on neural injuries and to discuss the possible molecule mechanisms underlying the neuroprotective effects of RG, thereby raising new possibilities for the treatment of neural injuries.

2. The Main Active Ingredients of Red Ginseng (RG)

The main active components of ginseng are ginsenosides, a special group of triterpenoid saponins [6]; there are more than 100 kinds of ginsenosides which have been isolated from the panax species. Protopanaxadiol (PPD) and protopanaxatriol (PPT) are the main structures of ginsenosides (Figure 1) [7]. Some studies have characterized the chemical compositions of RG and WG, indicating the differences between RG and WG at the molecular level [8,9,10]. Ginsenosides in RG are known to increase in content and new ginsenosides can be generated within RG with steaming (Table 1) [10,11]. We investigated an RG extract with high-performance liquid chromatography (HPLC) to ascertain the presence of the main ginsenosides as the active molecules in RG (Figure 2) [12].
Additional components, such as flavonoids, polysaccharides, volatile oils, and gintonin, as well as nonsaponin compounds, are also found in RG [13]. Although reports have suggested pharmacological properties of these compounds, more evidence is needed to confirm these effects.

3. Effects of RG on Brain Ischemia

When an artery leading to the brain is occluded, insufficient blood supply causes cerebral hypoxia and subsequent damage to nerve cells in situ. This pathological condition is known as brain ischemia. The injured area with irreversible damage is termed the ischemic core, and the surrounding tissue is called the ischemic penumbra. Because of the collateral circulation, the penumbra is salvageable during a short period after ischemia and can become a target for the treatment of acute ischemic stroke [14]. Effective treatment for ischemic stroke should include resuming the blood supply as soon as possible and keeping the penumbra viable until and after the blood supply is regained. Intravenous administration of recombinant tissue plasminogen activator(r-tPA) is the only treatment for acute ischemic stroke that has been approved by the U.S. FDA [15]. Numerous studies have been conducted to search for effective neuroprotective compound(s) or drug(s) in the last 30 years. RG was reported to exhibit neuroprotective effect by Wen et al. [16]. They showed that RG powder prevent learning disabilities and neuronal loss in gerbils with 5-minute forebrain ischemia. In this animal model, both orally administered RG powder and intraperitoneally injected crude ginseng saponin significantly protected hippocample CA1 neurons against delayed neuronal death. The neuroprotective molecule in RG powder was identified as ginsenoside Rb1(gRb1) on the basis of in vitro and in vivo studies [16,17]. Zhang et al. proved the neuroprotective action of gRb1 using spontaneous hypotensive rats with permanent occlusion of a unilateral middle cerebral artery distal to the striate branches [18]. In this study, gRb1 was administered directly into the left lateral ventricle with an osmotic minipump to avoid the obstacle of the blood-brain barrier; as a result, gRb1 significantly reduced the infarct area within the temporoparietal cortex. Another active molecule in RG, ginsenoside Rh2, was reported to have a protective effect on ischemia-reperfusion cerebral injury in rats [19]. Ginsenoside Rh2 is a breakdown product of gRb1, and is increased in content when RG is fermented [20]. Intravenous infused gRb1 was reported to decrease infarct volume in the same ischemia model as used in the study of Zhang et al. [18], as well as ameliorated the space navigation ability of the rats with cerebral infarction [21]. This study demonstrated the anti-apoptotic effect of gRb1, together with a molecular mechanism involving Bcl-xL and its promotor STAT5. The Bcl-xL protein is a member of the Bcl-2 family and has anti-apoptotic activity by suppressing the release of cytochrome c [22]. Upregulation of Bcl-xL expression in neurons around ischemic penumbra has been reported [23], and overexpression of Bcl-xL is known to diminish the infarct volume in mice with permanent occlusion of the unilateral middle cerebral artery (MCAO) [24]. These studies proved that Bcl-xL is involved in the apoptotic processes caused by ischemic stroke, and that upregulation of Bcl-xL expression could prevent the apoptotic processes, thus rescuing a population of neurons from ischemic damage. A stable chemical derivative of gRb1, dihydroginsenoside Rb1 (dgRb1), was also reported to alleviate ischemic brain damage through upregulation of Bcl-xL and VEGF when infused intravenously [25]. The effective dose of dgRb1 was ten times lower than that of gRb1, possibly because dgRb1 is chemically more stable than gRb1 and thus less easily bio-degradable (Figure 3). Another Bcl-2 family member, Bcl-2, was also reported to be upregulated by gRb1 and ginsenoside Rh2 in ischemic animal models [19,26,27]. Cells initiating apoptosis show an increase in cytosolic cytochrome c, which converts pro-capase-3 into active caspase-3 to induce apoptosis. The Bcl-2 family could prevent this upregulation of cytochrome c, thereby interrupting apoptosis [28]. Several studies have reported that cytochrome c and caspase-3 may be inhibited by gRb1 administration in vivo and in vitro [29,30]. In summary, RG and its active components prevent neuronal apoptosis through the interruption of the caspase cascade (Figure 4). Furthermore, gRb1 appears to upregulate the expression of certain neurotrophic factors, such as BDNF and GDNF, during transient MCAO in rats [27,30]. These neurotrophic factors are necessary for the growth, maintenance, and survival of cells in the brain, and thus they are likely to protect neurons from ischemic stroke.
In addition to its anti-apoptotic effect, RG and its ingredients have been reported to inhibit inflammatory responses after brain ischemia. Ischemic insult to the brain immediately induces the response of microglia and the secretion of pro-inflammatory cytokines, which makes the ischemic lesion larger as time progresses [31]. Intranasally applied gRb1 reduced infarct volume and alleviated the neurological severity score in an MCAO rat model by suppressing the activation of microglia in the penumbra; it also reduced the expression of pro-inflammatory cytokines, such as TNF-α and IL-6 [32]. Nuclear factor-κB (NF-κB) is a protein complex which plays a key role in immune responses [33]. In focal cerebral ischemia, NF-κB appears to be activated to promote cell death [34]. Administration of gRb1 has been shown to inhibit the activation of NF-κB, to suppress its binding to DNA and to decrease the TNF-α and IL-6 levels, thereby preventing neuron death [32].
A goal of treatment for brain ischemia is the restoration of the cerebral blood supply, which is called reperfusion. Reperfusion after ischemia can bring oxygen and glucose to the brain; however, rapid reperfusion could induce unexpected injuries to the brain. One mechanism underlying ischemia/reperfusion injuries is oxidative stress [35]. An excess of reactive oxygen species (ROS) such as peroxides and free radicals could directly damage the DNA, RNA, protein, and lipids in neurons, inducing neural dysfunction. The nuclear factor erythroid 2-related factor 2 (Nrf2) is a key molecule in the control of cellular anti-oxidative responses [36]. When activated by ROS, Nrf2 can translocate to the nucleus and bind to the antioxidant response element (ARE) to regulate gene expressions involved in anti-oxidation. Pre-activation of Nrf2 has been shown to reduce the liver damage caused by ischemia/reperfusion in mice [37]. Several studies have shown that gRb1 can protect neurons through activation of the Nrf2/ARE pathway in human SK-N-SH dopaminergic cells, SH-SY5Y cells, and neural progenitor cells [38,39,40]. Shi et al. indicated that gRb1 protected the brain from neural injury through Nrf2 activation, and that the knockdown of Nrf2 counteracted the neuroprotective effect of gRb1 [41]. Liu et al. indicated that gRb1 protected the spinal cord from oxidative stress by activating the Nrf2/ARE signal pathway [42]. Taken together, the neuroprotection achieved by RG and its active ingredients, especially gRb1, is attributed to their anti-apoptotic, anti-inflammatory, and anti-oxidative actions.

4. Effects of RG on Neurotrauma

Neurotrauma, including spinal cord injury (SCI) and traumatic brain injury (TBI), is a serious issue that adversely impacts public health around the world. Traumatic injury to the brain or spinal cord causes primary damage to the neurons and axons in situ, and the downstream consequences are known as secondary damage due to ensuing ischemia, inflammatory responses, and excessive release of excitatory neurotransmitters [43,44]. GRb1 and dihydroginsenoside Rb1(dgRb1), as a chemical derivative of gRb1, have been reported to ameliorate compressive spinal cord injury through the upregulation of Bcl-xL and VEGF expression [25,45]. VEGF promotes nerve repair after ischemic stroke or traumatic injury. There are presumably two mechanisms underlying neuroprotection by VEGF: direct neurotrophic action and the angiogenesis-facilitating effect which results in an increase in blood flow within the injured area [46]. Oral administration of an RG extract has also been reported to promote neurorestoration after SCI in rats [12]. The RG extract was shown to prevent neuron loss and to facilitate restoration of white matter, promoting recovery from motor and behavioral abnormalities. Moreover, the RG extract reduced the expression of TNFα and IL-1β and decreased the aggregation of Iba-1 positive microglia in the injured spinal cord [12]. Similar studies have reported that RG extract improved functional recovery and neural damage after SCI by inhibiting inflammatory responses [47,48]. In addition, Wang et al. reported that gRb1 treatment decreased neuron loss and promoted functional recovery through inhibition of autophagy following spinal cord injury both in vivo and in vitro [49].
In addition to the neuroprotective effects of RG and its ingredients on experimental animals with SCI, natural products have been shown to exhibit similar favorable effects in the case of TBI. Treatment with ginseng total saponins decreased contusion damage in the cerebrum and improved neurological deficits after experimental TBI [50]. Xia et al. reported that ginseng total saponins attenuated secondary damage after TBI by alleviating oxidative stress in the brain and by downregulating the expression of IL-6 and IL-1β [51]. Another study demonstrated that ginseng ameliorated cognitive deficits after TBI through inhibition of microglia-dependent neuro-inflammation [52]. These studies suggest that RG and its ingredients protect neurons from traumatic damage through suppression of neuroinflammation and neuronal apoptosis.

5. Effect of RG on Neurodegeneration

Neurodegeneration is characterized by the progressive loss of neural functions and neurons; it is responsible for motor disorders such as rigidity, ataxias, and/or cognitive impairment. Neurodegenerative diseases, including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS), are not completely curable as neurons in the affected lesions fall into apoptosis in a time-dependent manner, regardless of medical treatment. Numerous efforts have been made to find effective therapies, although the etiology of each neurodegenerative disease has not yet been clearly determined. Several studies have shown that RG and its ingredients ameliorate cognitive disorders in AD animal models [53,54,55]. Aβ and phosphorylated-tau content were also decreased in the brain through use of RG or its ingredients in animal models of AD [55,56]. There are reports that AD patients treated with RG showed significant improvement in cognitive function and an increase in the relative ratio of alpha waves when examined with quantitative electroencephalogram (EEG) [57,58]. Because these clinical studies only included a small number of patients without placebo controls, well-designed and more extensive studies are needed to ascertain the favorable effects of RG on patients of AD [59]. Several RG extracts were shown to have beneficial effects on animal models of PD [60,61] and HD [62]. The putative active molecules of RG available for the treatment of neurodegenerative diseases were identified as the ginsenosides Rb1 and Rg1 [63,64,65]. In addition to their anti-apoptotic and anti-inflammatory actions, RG and its ingredients were proven to reduce Aβ production in an AD model [53] and to inhibit α-synuclein aggregation in a PD model [66]. Further studies are needed to identify the molecular mechanisms underlying the effects of RG and its ingredients on neurodegenerative diseases.

6. Conclusion and Perspectives of RG Research

The studies reviewed above suggest that RG and its ingredients could protect neurons from a variety of injuries, such as ischemia, trauma, and degeneration. Numerous studies have indicated that ginsenoside Rb1 and other ginsenosides are the active molecules responsible for the neuroprotective effects of RG in neural injuries (Table 2). Further studies, including clinical trials, are necessary to confirm the pharmacological properties of RG and its ingredients. Additionally, this review has raised the possibility that more effective or stable neuroprotective derivatives based on the chemical structures of ginsenosides could be developed. In support of this speculation, dihydroginsenoside Rb1 is endowed with a more stable chemical structure compared with gRb1 and was shown to be as effective as gRb1 in terms of neuroprotection at a dose lower than those typically used for gRb1. Overall, RG and its ingredients could be promising candidate drugs for the treatment of neural injuries.

Author Contributions

Conceptualization and writing (original draft preparation), P.Z.; Writing (review and editing), M.S.

Funding

This work was funded by JSPS KAKENHI, Grant Numbers: JP16K11407 and JP18K08920.

Acknowledgments

The authors appreciate Japan Red ginseng Research Association for providing valuable information of ginseng. The secretarial assistance of Kaori Hiraoka is acknowledged.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

RGred ginseng
WGwhite ginseng
PPDprotopanaxadiol
PPTprotopanaxatriol
HPLChigh performance liquid chromatography
r-tPArecombinant tissue plasminogen activator
gRb1ginsenoside Rb1
STAT5signal transducer and activator of transcription 5
MCAOmiddle cerebral artery occlusion
dgRb1dihydroginsenoside Rb1
VEGFvascular endothelial growth factor
BDNFbrain-derived neurotrophic factor
GDNFglial cell line-derived neurotrophic factor
NF-κBnuclear factor-κB
ROSreactive oxygen species
Nrf2nuclear factor erythroid 2-related factor
AREantioxidant response element
SCIspinal cord injury
TBItraumatic brain injury
PDParkinson’s disease
ADAlzheimer’s disease
HDHuntington’s disease
ALSamyotrophic lateral sclerosis
EEGelectroencephalogram

References

  1. Cheng, Y.; Shen, L.H.; Zhang, J.T. Anti-amnestic and anti-aging effects of ginsenoside Rg1 and Rb1 and its mechanism of action. Acta Pharmacol. Sin. 2005, 26, 143–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kim, D.Y.; Yang, W.M. Panax ginseng ameliorates airway inflammation in an ovalbumin-sensitized mouse allergic asthma model. J. Ethnopharmacol. 2011, 136, 230–235. [Google Scholar] [CrossRef]
  3. Liu, T.-P.; Liu, I.-M.; Cheng, J.-T. Improvement of Insulin Resistance by Panax Ginseng in Fructose-rich Chow-fed Rats. Horm Metab Res. 2005, 37, 146–151. [Google Scholar] [CrossRef]
  4. Purves, D.; Augustine, G.J.; Fitzpatrick, D.; Katz, L.C.; LaMantia, A.-S.; McNamara, J.O.; Williams, S.M. Neuroscience, 2nd ed.; Sinauer Associates, Inc.: Sunderland, MA, USA, 2001. [Google Scholar]
  5. Radad, K.; Moldzio, R.; Rausch, W.-D. Ginsenosides and Their CNS Targets. CNS Neurosci. Ther. 2011, 17, 761–768. [Google Scholar] [CrossRef]
  6. Christensen, L.P. Chapter 1 Ginsenosides: Chemistry, Biosynthesis, Analysis, and Potential Health Effects. In Advances in Food and Nutrition Research; Academic Press: Cambridge, MA, USA, 2008; Volume 55, pp. 1–99. [Google Scholar]
  7. Baek, S.-H.; Bae, O.-N.; Park, J.H. Recent Methodology in Ginseng Analysis. J. Ginseng Res. 2012, 36, 119–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Lim, C.-Y.; Moon, J.-M.; Kim, B.-Y.; Lim, S.-H.; Lee, G.-S.; Yu, H.-S.; Cho, S.-I. Comparative study of Korean White Ginseng and Korean Red Ginseng on efficacies of OVA-induced asthma model in mice. J. Ginseng Res. 2015, 39, 38–45. [Google Scholar] [CrossRef] [PubMed]
  9. So, S.-H.; Lee, J.W.; Kim, Y.-S.; Hyun, S.H.; Han, C.-K. Red ginseng monograph. J. Ginseng Res. 2018, 42, 549–561. [Google Scholar] [CrossRef] [PubMed]
  10. Ki-Yeul, N. The Comparative Understanding between Red Ginseng and White Ginsengs, Processed Ginsengs (Panax ginseng C. A. Meyer). J. Ginseng Res. 2005, 29, 1–18. [Google Scholar]
  11. Lee, S.M.; Bae, B.-S.; Park, H.-W.; Ahn, N.-G.; Cho, B.-G.; Cho, Y.-L.; Kwak, Y.-S. Characterization of Korean Red Ginseng (Panax ginseng Meyer): History, preparation method, and chemical composition. J. Ginseng Res. 2015, 39, 384–391. [Google Scholar] [CrossRef] [Green Version]
  12. Zhu, P.; Samukawa, K.; Fujita, H.; Kato, H.; Sakanaka, M. Oral Administration of Red Ginseng Extract Promotes Neurorestoration after Compressive Spinal Cord Injury in Rats. J. Evid.-Based Complementary Altern. Med. 2017, 2017, 1265464. [Google Scholar] [CrossRef]
  13. Jia, L.; Zhao, Y. Current Evaluation of the Millennium Phytomedicine-Ginseng (I): Etymology, Pharmacognosy, Phytochemistry, Market and Regulations. Curr. Med. Chem. 2009, 16, 2475–2484. [Google Scholar] [CrossRef]
  14. Astrup, J.; Siesjö, B.K.; Symon, L. Thresholds in cerebral ischemia–the ischemic penumbra. Stroke 1981, 12, 723–725. [Google Scholar] [CrossRef] [PubMed]
  15. Jahan, R.; Vinuela, F. Treatment of acute ischemic stroke: Intravenous and endovascular therapies. Expert Rev. Cardiovasc. Ther. 2009, 7, 375–387. [Google Scholar] [CrossRef]
  16. Wen, T.C.; Yoshimura, H.; Matsuda, S.; Lim, J.-H.; Sakanaka, M. Ginseng root prevents learning disability and neuronal loss in gerbils with 5-minute forebrain ischemia. Acta Neuropathol. 1995, 91, 15–22. [Google Scholar] [CrossRef]
  17. Lim, J.-H.; Wen, T.-C.; Matsuda, S.; Tanaka, J.; Maeda, N.; Peng, H.; Aburaya, J.; Ishihara, K.; Sakanaka, M. Protection of ischemic hippocampal neurons by ginsenoside Rb1, a main ingredient of ginseng root. Neurosci. Res. 1997, 28, 191–200. [Google Scholar] [CrossRef]
  18. Zhang, B.; Matsuda, S.; Tanaka, J.; Tateishi, N.; Maeda, N.; Wen, T.-C.; Peng, H.; Sakanaka, M. Ginsenoside Rb1 prevents image navigation disability, cortical infarction, and thalamic degeneration in rats with focal cerebral ischemia. J. Stroke Cerebrovasc. Dis. 1998, 7, 1–9. [Google Scholar] [CrossRef]
  19. Bae, E.-A.; Hyun, Y.-J.; Choo, M.-K.; Oh, J.K.; Ryu, J.H.; Kim, D.-H. Protective effect of fermented Red ginseng on a transient focal ischemic rats. Arch. Pharm Res. 2004, 27, 1136–1140. [Google Scholar] [CrossRef] [PubMed]
  20. Popovich, D.G.; Kitts, D.D. Generation of ginsenosides Rg3 and Rh2 from North American ginseng. Phytochemistry 2004, 65, 337–344. [Google Scholar] [CrossRef] [PubMed]
  21. Zhang, B.; Hata, R.; Zhu, P.; Sato, K.; Wen, T.-C.; Yang, L.; Fujita, H.; Mitsuda, N.; Tanaka, J.; Samukawa, K.; et al. Prevention of Ischemic Neuronal Death by Intravenous Infusion of a Ginseng Saponin, Ginsenoside Rb1, That Upregulates Bcl-xL Expression. J. Cereb. Blood Flow Metab. 2006, 26, 708–721. [Google Scholar] [CrossRef] [PubMed]
  22. Chao, D.T.; Korsmeyer, S.J. BCL-2 FAMILY: Regulators of Cell Death. Annu. Rev. Immunol. 1998, 16, 395–419. [Google Scholar] [CrossRef]
  23. Isenmann, S.; Stoll, G.; Schroeter, M.; Krajewski, S.; Reed, J.C.; Bähr, M. Differential Regulation of Bax, Bcl-2, and Bcl-X Proteins in Focal Cortical Ischemia in the Rat. Brain Pathol. 1998, 8, 49–62. [Google Scholar] [CrossRef] [PubMed]
  24. Wiessner, C.; Allegrini, P.R.; Rupalla, K.; Sauer, D.; Oltersdorf, T.; McGregor, A.L.; Bischoff, S.; Böttiger, B.W.; van der Putten, H. Neuron-specific transgene expression of Bcl-XL but not Bcl-2 genes reduced lesion size after permanent middle cerebral artery occlusion in mice. Neurosci. Lett. 1999, 268, 119–122. [Google Scholar] [CrossRef]
  25. Sakanaka, M.; Zhu, P.; Zhang, B.; Wen, T.-C.; Cao, F.; Ma, Y.-J.; Samukawa, K.; Mitsuda, N.; Tanaka, J.; Kuramoto, M.; et al. Intravenous Infusion of Dihydroginsenoside Rb1 Prevents Compressive Spinal Cord Injury and Ischemic Brain Damage through Upregulation of VEGF and Bcl-xL. J. Neurotrauma 2007, 24, 1037–1054. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, G.; Liu, A.; Zhou, Y.; San, X.; Jin, T.; Jin, Y. Panax ginseng ginsenoside-Rg2 protects memory impairment via anti-apoptosis in a rat model with vascular dementia. J. Ethnopharmacol. 2008, 115, 441–448. [Google Scholar] [CrossRef] [PubMed]
  27. Yuan, Q.-L.; Yang, C.-X.; Xu, P.; Gao, X.-Q.; Deng, L.; Chen, P.; Sun, Z.-L.; Chen, Q.-Y. Neuroprotective effects of ginsenoside Rb1 on transient cerebral ischemia in rats. Brain Res. 2007, 1167, 1–12. [Google Scholar] [CrossRef]
  28. Yang, J.; Liu, X.; Bhalla, K.; Kim, C.N.; Ibrado, A.M.; Cai, J.; Peng, T.-I.; Jones, D.P.; Wang, X. Prevention of Apoptosis by Bcl-2: Release of Cytochrome c from Mitochondria Blocked. Science 1997, 275, 1129. [Google Scholar] [CrossRef] [PubMed]
  29. Liang, J.; Yu, Y.; Wang, B.; Lu, B.; Zhang, J.; Zhang, H.; Ge, P. Ginsenoside Rb1 Attenuates Oxygen-Glucose Deprivation-Induced Apoptosis in SH-SY5Y Cells via Protection of Mitochondria and Inhibition of AIF and Cytochrome c Release. Molecules 2013, 18, 12777–12792. [Google Scholar] [CrossRef] [Green Version]
  30. Gao, X.-Q.; Yang, C.-X.; Chen, G.-J.; Wang, G.-Y.; Chen, B.; Tan, S.-K.; Liu, J.; Yuan, Q.-L. Ginsenoside Rb1 regulates the expressions of brain-derived neurotrophic factor and caspase-3 and induces neurogenesis in rats with experimental cerebral ischemia. J. Ethnopharmacol. 2010, 132, 393–399. [Google Scholar] [CrossRef]
  31. Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J.V.; Zuo, Y.; Jung, S.; Littman, D.R.; Dustin, M.L.; Gan, W.-B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005, 8, 752. [Google Scholar] [CrossRef]
  32. Zhu, J.; Jiang, Y.; Wu, L.; Lu, T.; Xu, G.; Liu, X. Suppression of local inflammation contributes to the neuroprotective effect of ginsenoside Rb1 in rats with cerebral ischemia. Neuroscience 2012, 202, 342–351. [Google Scholar] [CrossRef]
  33. Vallabhapurapu, S.; Karin, M. Regulation and Function of NF-κB Transcription Factors in the Immune System. Annu. Rev. Immunol. 2009, 27, 693–733. [Google Scholar] [CrossRef] [PubMed]
  34. Schneider, A.; Martin-Villalba, A.; Weih, F.; Vogel, J.; Wirth, T.; Schwaninger, M. NF-κB is activated and promotes cell death in focal cerebral ischemia. Nature Medicine 1999, 5, 554–559. [Google Scholar] [CrossRef]
  35. Lin, L.; Wang, X.; Yu, Z. Ischemia-reperfusion Injury in the Brain: Mechanisms and Potential Therapeutic Strategies. Biochem. Pharmacol. Open Access 2016, 5, 1–16. [Google Scholar]
  36. Ma, Q. Role of Nrf2 in Oxidative Stress and Toxicity. Annu. Rev. Pharmacol Toxicol 2013, 53, 401–426. [Google Scholar] [CrossRef] [Green Version]
  37. Xu, D.; Xu, M.; Jeong, S.; Qian, Y.; Wu, H.; Xia, Q.; Kong, X. The Role of Nrf2 in Liver Disease: Novel Molecular Mechanisms and Therapeutic Approaches. Front. Pharmacol. 2019, 9, 1428. [Google Scholar] [CrossRef] [PubMed]
  38. Hwang, Y.P.; Jeong, H.G. Ginsenoside Rb1 protects against 6-hydroxydopamine-induced oxidative stress by increasing heme oxygenase-1 expression through an estrogen receptor-related PI3K/Akt/Nrf2-dependent pathway in human dopaminergic cells. Toxicol. Appl. Pharmacol. 2010, 242, 18–28. [Google Scholar] [CrossRef]
  39. Fernández-Moriano, C.; González-Burgos, E.; Iglesias, I.; Lozano, R.; Gómez-Serranillos, M.P. Evaluation of the adaptogenic potential exerted by ginsenosides Rb1 and Rg1 against oxidative stress-mediated neurotoxicity in an in vitro neuronal model. PLoS ONE 2017, 12, e0182933. [Google Scholar] [CrossRef]
  40. Ni, N.; Liu, Q.; Ren, H.; Wu, D.; Luo, C.; Li, P.; Wan, J.-B.; Su, H. Ginsenoside Rb1 Protects Rat Neural Progenitor Cells against Oxidative Injury. Molecules 2014, 19, 3012–3024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Shi, Y.; Miao, W.; Teng, J.; Zhang, L. Ginsenoside Rb1 Protects the Brain from Damage Induced by Epileptic Seizure via Nrf2/ARE Signaling. CPB 2018, 45, 212–225. [Google Scholar] [CrossRef] [Green Version]
  42. Liu, X.; Gu, X.; Yu, M.; Zi, Y.; Yu, H.; Wang, Y.; Xie, Y.; Xiang, L. Effects of ginsenoside Rb1 on oxidative stress injury in rat spinal cords by regulating the eNOS/Nrf2/HO-1 signaling pathway. Exp. Ther. Med. 2018, 16, 1079–1086. [Google Scholar] [CrossRef]
  43. Dusart, I.; Schwab, M.E. Secondary Cell Death and the Inflammatory Reaction After Dorsal Hemisection of the Rat Spinal Cord. Eur. J. Neurosci. 1994, 6, 712–724. [Google Scholar] [CrossRef]
  44. Oyinbo, C.A. Secondary injury mechanisms in traumatic spinal cord injury: A nugget of this multiply cascade. Acta Neurobiol. Exp. 2011, 71, 281–299. [Google Scholar]
  45. Zhu, P. Intravenous Infusion of Ginsenoside Rb1 Ameliorates Compressive Spinal Cord Injury through Upregulation of Bcl-xL and VEGF. Int. J. Neurol. Neurother. 2015, 2, 1. [Google Scholar] [CrossRef]
  46. Ruiz de Almodovar, C.; Lambrechts, D.; Mazzone, M.; Carmeliet, P. Role and Therapeutic Potential of VEGF in the Nervous System. Phys. Rev. 2009, 89, 607–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Kim, Y.O.; Kim, Y.; Lee, K.; Na, S.W.; Hong, S.P.; Valan Arasu, M.; Yoon, Y.W.; Kim, J. Panax ginseng Improves Functional Recovery after Contusive Spinal Cord Injury by Regulating the Inflammatory Response in Rats: An In Vivo Study. Available online: https://www.hindawi.com/journals/ecam/2015/817096/ (accessed on 26 February 2019).
  48. Wang, W.; Shen, H.; Xie, J.-J.; Ling, J.; Lu, H. Neuroprotective effect of ginseng against spinal cord injury induced oxidative stress and inflammatory responses. Int. J. Clin. Exp. Med. 2015, 8, 3514–3521. [Google Scholar]
  49. Wang, P.; Lin, C.; Wu, S.; Huang, K.; Wang, Y.; Bao, X.; Zhang, F.; Huang, Z.; Teng, H. Inhibition of Autophagy is Involved in the Protective Effects of Ginsenoside Rb1 on Spinal Cord Injury. Cell Mol. Neurobiol. 2018, 38, 679–690. [Google Scholar] [CrossRef] [PubMed]
  50. Ji, Y.C.; Kim, Y.B.; Park, S.W.; Hwang, S.N.; Min, B.K.; Hong, H.J.; Kwon, J.T.; Suk, J.S. Neuroprotective Effect of Ginseng Total Saponins in Experimental Traumatic Brain Injury. J. Korean Med. Sci. 2005, 20, 291–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Xia, L.; Jiang, Z.-L.; Wang, G.-H.; Hu, B.-Y.; Ke, K.-F. Treatment with ginseng total saponins reduces the secondary brain injury in rat after cortical impact. J. Neurosci. Res. 2012, 90, 1424–1436. [Google Scholar] [CrossRef]
  52. Kumar, A.; Rinwa, P.; Dhar, H. Microglial inhibitory effect of ginseng ameliorates cognitive deficits and neuroinflammation following traumatic head injury in rats. Inflammopharmacol 2014, 22, 155–167. [Google Scholar] [CrossRef] [PubMed]
  53. Fang, F.; Chen, X.; Huang, T.; Lue, L.-F.; Luddy, J.S.; Yan, S.S. Multi-faced neuroprotective effects of Ginsenoside Rg1 in an Alzheimer mouse model. Biochim. Biophys. Acta Mol. Basis Dis. 2012, 1822, 286–292. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, Y.; Kan, H.; Yin, Y.; Wu, W.; Hu, W.; Li, W.; Li, W. Protective effects of ginsenoside Rg1 on chronic restraint stress induced learning and memory impairments in male mice. Pharmacol. Biochem. Behav. 2014, 120, 73–81. [Google Scholar] [CrossRef] [PubMed]
  55. Li, H.; Kang, T.; Qi, B.; Kong, L.; Jiao, Y.; Cao, Y.; Zhang, J.; Yang, J. Neuroprotective effects of ginseng protein on PI3K/Akt signaling pathway in the hippocampus of D-galactose/AlCl3 inducing rats model of Alzheimer’s disease. J. Ethnopharmacol. 2016, 179, 162–169. [Google Scholar] [CrossRef]
  56. Li, H.; Song, J.; Zhang, J.; Wang, T.; Yan, Y.; Tao, Z.; Li, S.; Zhang, H.; Kang, T.; Yang, J. Ginseng Protein Reverses Amyloid Beta Peptide and H2O2 Cytotoxicity in Neurons, and Ameliorates Cognitive Impairment in AD Rats Induced by a Combination of D-Galactose and AlCl3. Phytother. Res. 2017, 31, 284–295. [Google Scholar] [CrossRef]
  57. Heo, J.-H.; Lee, S.-T.; Chu, K.; Oh, M.J.; Park, H.-J.; Shim, J.-Y.; Kim, M. An open-label trial of Korean red ginseng as an adjuvant treatment for cognitive impairment in patients with Alzheimer’s disease. Eur. J. Neurol. 2008, 15, 865–868. [Google Scholar] [CrossRef] [PubMed]
  58. Heo, J.-H.; Park, M.-H.; Lee, J.-H. Effect of Korean Red Ginseng on Cognitive Function and Quantitative EEG in Patients with Alzheimer’s Disease: A Preliminary Study. J. Altern. Complement. Med. 2016, 22, 280–285. [Google Scholar] [CrossRef]
  59. Lee, M.S.; Yang, E.J.; Kim, J.I.; Ernst, E. Ginseng for Cognitive Function in Alzheimer’s Disease: A Systematic Review; Centre for Reviews and Dissemination: Heslington, York, UK, 2009. [Google Scholar]
  60. Van Kampen, J.; Robertson, H.; Hagg, T.; Drobitch, R. Neuroprotective actions of the ginseng extract G115 in two rodent models of Parkinson’s disease. Exp. Neurol. 2003, 184, 521–529. [Google Scholar] [CrossRef] [PubMed]
  61. Van Kampen, J.M.; Baranowski, D.B.; Shaw, C.A.; Kay, D.G. Panax ginseng is neuroprotective in a novel progressive model of Parkinson’s disease. Exp. Gerontol. 2014, 50, 95–105. [Google Scholar] [CrossRef]
  62. Kim, J.-H.; Kim, S.; Yoon, I.-S.; Lee, J.-H.; Jang, B.-J.; Jeong, S.M.; Lee, J.-H.; Lee, B.-H.; Han, J.-S.; Oh, S.; et al. Protective effects of ginseng saponins on 3-nitropropionic acid-induced striatal degeneration in rats. Neuropharmacology 2005, 48, 743–756. [Google Scholar] [CrossRef]
  63. Liu, L.; Hoang-Gia, T.; Wu, H.; Lee, M.-R.; Gu, L.; Wang, C.; Yun, B.-S.; Wang, Q.; Ye, S.; Sung, C.-K. Ginsenoside Rb1 improves spatial learning and memory by regulation of cell genesis in the hippocampal subregions of rats. Brain Res. 2011, 1382, 147–154. [Google Scholar] [CrossRef]
  64. Yang, L.; Zhang, J.; Zheng, K.; Shen, H.; Chen, X. Long-term Ginsenoside Rg1 Supplementation Improves Age-Related Cognitive Decline by Promoting Synaptic Plasticity Associated Protein Expression in C57BL/6J Mice. J. Gerontol. Ser. A 2013, 69A, 282–294. [Google Scholar] [CrossRef]
  65. Rokot, N.T.; Kairupan, T.S.; Cheng, K.-C.; Runtuwene, J.; Kapantow, N.H.; Amitani, M.; Morinaga, A.; Amitani, H.; Asakawa, A.; Inui, A. A Role of Ginseng and Its Constituents in the Treatment of Central Nervous System Disorders. Available online: https://www.hindawi.com/journals/ecam/2016/2614742/ (accessed on 25 February 2019).
  66. Radad, K.; Gille, G.; Moldzio, R.; Saito, H.; Ishige, K.; Rausch, W.-D. Ginsenosides Rb1 and Rg1 effects on survival and neurite growth of MPP+-affected mesencephalic dopaminergic cells. J. Neural. Transm. 2004, 111, 37–45. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of ginsenosides in Panax ginseng, reproduced with permission from Baek, S.-H, et al. [7]. PPD: protopanaxadiol; PPT: protopanaxatriol; DH: dehydro; Glc: glucose; Ara: abrabinose; Xyl: xylose; Rha: rhamnose; Ac: acetyl; pyr: pyranosyl; fur: furanosyl.
Figure 1. Chemical structures of ginsenosides in Panax ginseng, reproduced with permission from Baek, S.-H, et al. [7]. PPD: protopanaxadiol; PPT: protopanaxatriol; DH: dehydro; Glc: glucose; Ara: abrabinose; Xyl: xylose; Rha: rhamnose; Ac: acetyl; pyr: pyranosyl; fur: furanosyl.
J 02 00009 g001
Figure 2. HPLC separation of ginsenosides in a red ginseng extract, reproduced with permission from Zhu PX, et al. [12]. Column: Superspher RP-18(e) (4.0 mm i.d. × 250 mm, Merck); mobile phase: (A) CH3CN-H2O-0.1% H3PO4 (21:72:8, v/v), (B) CH3CN; flow rate: total flow 0.8 mL/min (flow program: (A) 0 →19 min: 100%; 19 → 20 min: 100 → 90%; 20 → 73 min: 90%; 73 →103 min: 90→70%; 103→120 min: 70%); Temp.: Temp. program (0→30 min: 35 °C; 30→60 min: 55 °C; 60→120 min: 35 °C); detection: UV 202 nm.
Figure 2. HPLC separation of ginsenosides in a red ginseng extract, reproduced with permission from Zhu PX, et al. [12]. Column: Superspher RP-18(e) (4.0 mm i.d. × 250 mm, Merck); mobile phase: (A) CH3CN-H2O-0.1% H3PO4 (21:72:8, v/v), (B) CH3CN; flow rate: total flow 0.8 mL/min (flow program: (A) 0 →19 min: 100%; 19 → 20 min: 100 → 90%; 20 → 73 min: 90%; 73 →103 min: 90→70%; 103→120 min: 70%); Temp.: Temp. program (0→30 min: 35 °C; 30→60 min: 55 °C; 60→120 min: 35 °C); detection: UV 202 nm.
J 02 00009 g002
Figure 3. Molecular structures of gRb1 and dgRb1. The arrow indicates the difference between gRb1 and dgRb1, reproduced with permission from Sakanaka, M. et al. [25].
Figure 3. Molecular structures of gRb1 and dgRb1. The arrow indicates the difference between gRb1 and dgRb1, reproduced with permission from Sakanaka, M. et al. [25].
J 02 00009 g003
Figure 4. Diagram of the effect of RG and its ingredients on the caspase cascade.
Figure 4. Diagram of the effect of RG and its ingredients on the caspase cascade.
J 02 00009 g004
Table 1. Variation of ginsenoside content (mg/g) in ginseng root during the steaming process, reproduced with permission from Ki-Yeul, N. [10]. ↑: increased. *: Newly produced.
Table 1. Variation of ginsenoside content (mg/g) in ginseng root during the steaming process, reproduced with permission from Ki-Yeul, N. [10]. ↑: increased. *: Newly produced.
Steaming Time (Hours)
0123
Total35.43942.113↑57.974↑50.332↑
Diol21.76828.488↑43.063↑36.974↑
Triol11.56911.50512.533↑11.237
Diol/Triol1.8672.476↑3.436↑3.29↑
Ginsenoside-Ro2.0122.12↑2.378↑2.121↑
Rb18.09710.88↑16.016↑14.316↑
Rb25.5317.23↑11.352↑9.282↑
Rc4.9896.495↑9.943↑8.119↑
Rd1.2511.629↑2.31↑1.864↑
Re4.7794.1055.223↑3.72
Rf2.542.5332.669↑2.413
Rg13.8594.335↑3.7233.971↑
20(S)-Ginsenoside Rg10.4810.4680.7↑0.589↑
20(R)-Ginsenoside Rg2--0.042 *0.125 *
20(S)-Ginsenoside Rg3-0.093 *0.221 *0.408 *
20(R)-Ginsenoside Rg3-0.054 *0.14 *0.262 *
20(S)-Ginsenoside Rh1-0.064 *0.134 *0.277 *
20(R)-Ginsenoside Rh1--0.042 *0.142 *
Ginsenoside-Rs11.1431.1051.577↑1.233↑
quinquenoside-R10.7571.002↑1.504↑1.49↑
Table 2. Neuroprotective effects of RG and its ingredients in neural injury and the molecular mechanisms.
Table 2. Neuroprotective effects of RG and its ingredients in neural injury and the molecular mechanisms.
Types of Neural InjuryIschemiaTraumaNeurodegeneration
Types of studyIn vivo and in vitro studies. In vivo and in vitro studies.In vivo and in vitro studies, clinical studies.
Active componentsRG powder, RG extract, gRb1, gRh2, dgRb1RG extract, ginseng total saponin, gRb1, dgRb1RG powder, gRb1, gRg1
EffectPrevention of learning disability and neuron loss; reduction of infarct volume. Prevention of neuron loss and promotion of restoration of white matter after SCI;
amelioration of cognitive deficits; decrement of contusion damage after TBI.
Amelioration of cognitive disorders; improvement of cognitive function; increment of relative ratio of alpha waves;
reduction of Aβ and phosphorylated-tau;
inhibition of α-synuclein aggregation.
Proposed mechanismsAnti-apoptosis: up-regulation of Bcl-xL and Bcl-2; inhibition of cytochrome-c and caspase-3.
Neurotrophic action: up-regulation of BDNF and GDNF.
Anti-inflammation: inhibition of pro-inflammatory cytokines; inhibition of microglia activation; inhibition of NF-κB activation.
Anti-oxidation: activation of Nrf2/ARE pathway.
Anti-apoptosis: up-regulation of Bcl-xL
Neurotrophic action: up-regulation of VEGF.
Anti-inflammation: inhibition of pro-inflammatory cytokines; inhibition of microglia activation;
inhibition of autophagy.
Anti-oxidation.
Anti-apoptosis: upregulation of Bcl-xL.
Anti-inflammation: inhibition of pro-inflammatory cytokines; inhibition of microglia activation.
Reference[16,17,18,19,20,21,25,26,27,29,30,32,38,39,40,41,42] [12,25,45,46,47,48,49,50,51,52] [53,54,55,56,57,58,60,61,62,63,64,65,66]

Share and Cite

MDPI and ACS Style

Zhu, P.; Sakanaka, M. Effects of Red Ginseng on Neural Injuries with Reference to the Molecular Mechanisms. J 2019, 2, 116-127. https://doi.org/10.3390/j2020009

AMA Style

Zhu P, Sakanaka M. Effects of Red Ginseng on Neural Injuries with Reference to the Molecular Mechanisms. J. 2019; 2(2):116-127. https://doi.org/10.3390/j2020009

Chicago/Turabian Style

Zhu, Pengxiang, and Masahiro Sakanaka. 2019. "Effects of Red Ginseng on Neural Injuries with Reference to the Molecular Mechanisms" J 2, no. 2: 116-127. https://doi.org/10.3390/j2020009

Article Metrics

Back to TopTop