Skip to main content
Heliyon logoLink to Heliyon
. 2024 Mar 26;10(7):e28581. doi: 10.1016/j.heliyon.2024.e28581

Advancements in research on the effects of panax notoginseng saponin constituents in ameliorating learning and memory disorders

Shusen Liu a,b,c, Min Wang a,b, Haiyan Xiao a,b, Jingxue Ye a,b, Li Cao a,b, Wenlan Li c,, Guibo Sun a,b,⁎⁎
PMCID: PMC10998096  PMID: 38586351

Abstract

Learning and memory disorder is a cluster of symptoms caused by neuronal aging and other diseases of the central nervous system (CNS). Panax notoginseng saponins (PNS) are a series of saponins derived from the natural active ingredients of traditional Chinese medicine (TCM) that have neuroprotective effects on the central nervous system. In this paper, we review the ameliorative effects and mechanisms of Panax notoginseng saponin-like components on learning and memory disorders to provide valuable references and insights for the development of new drugs for the treatment of learning and memory disorders. Our summary results suggest that Panax ginseng saponins have significant effects on improving learning and memory disorders, and these effects and potential mechanisms are mediated by their anti-inflammatory, anti-apoptotic, antioxidant, β-amyloid lowering, mitochondrial homeostasis in vivo, neuronal structure and function improving, neurogenesis promoting, neurotransmitter release regulating, and probiotic homeostasis in vivo activities. These findings suggest the potential of Panax notoginseng saponin-like constituents as drug candidates for improving learning and memory disorders.

Keywords: Panax notoginseng saponin, Learning and memory disorders, Alzheimer's disease, Vascular cognitive impairment and dementia

1. Introduction

Learning and memory, as advanced cognitive functions of the brain, play essential roles in human survival, development, and cognition. Neuroscience has primarily focused on researching the mechanisms involved in the formation and degeneration of learning and memory. This line of inquiry is of paramount importance in the treatment of neurodegenerative diseases and enhancing the quality of human life. Specifically, learning refers to the cognitive process of adapting to the external environment, while memory encompasses neural activity that preserves and reproduces learned behaviors through interactions among neurons and synapses [1]. Current research has indicated that neuroinflammation [2], neuronal apoptosis [3], and altered synaptic plasticity [4] have significant impacts on learning and memory capacity. Moreover, neuronal injury resulting from normal aging or various diseases associated with the central nervous system (CNS) - including Alzheimer's disease (AD) [5], vascular cognitive impairment and dementia (VCID) [6], malnutrition dementia [7], depression [8], traumatic brain injury [8], post-traumatic stress disorder (PTSD) [9], and Kosakov syndrome [10], can lead to cognitive impairment with a decline in learning and memory ability.

Recently, medicinal plants have gained much attention, with Panax notoginseng, a well-known and popular Chinese medicine, being found to have potential as a neuroprotective agent [11]. Notoginseng Radix et Rhizoma, commonly referred to as Tianqi or Sanqi in East Asian nations, denotes the dried root and rhizome of Panax notoginseng (Burk.) F. H. Chen, a member of the Panax species within the Araliaceae family Panax notoginseng boasts a long-standing medicinal legacy that spans over four centuries. Saponins, as a widely present compound in medicinal plants, have pharmacological activities such as anti-inflammatory, antioxidant, and the inhibition of abnormal autophagy and apoptosis in hippocampal neurons [[12], [13], [14]]. These properties make them have a significant pharmacological effect in improving learning and memory. The main components isolated from Notoginseng Radix et Rhizoma are PNS, which have antioxidant, anti-apoptotic, and anti-endoplasmic reticulum stress effects [[15], [16], [17]]. Different anatomical parts (root, stem, leaf, bud, and seed) of Notoginseng Radix et Rhizoma contain various saponins, including ginsenoside (Rb1, Re, Rg1, Rg2, Rh1), notoginsenoside (R1-R6), and aescin (VII) [18]. The five saponins with the highest content in Notoginseng – notoginsenoside R1 (7%–10%), ginsenoside Rb1 (30%–36%), Rg1 (20%–40%), Rd (5%–8.4%), and Re (3.9%–6%) constitute up to 90% of the total PNS and are the most studied components in pharmaceutical experiments by far [19](Fig. 1).

Fig. 1.

Fig. 1

The chemical structures of the five saponins with the highest content in Panax notoginseng.

Notoginseng and Ginseng both belong to the Araliaceae Juss. family. They contain chemical components with a high degree of similarity [20]. In terms of pharmacological effects, Ginseng has a relatively good effect on improving learning and memory, and at the same time, there are also many studies proving the potential of Notoginseng and its saponins in improving learning and memory disorders [21]. Although some articles summarize the effects of Ginseng and ginsenoside on cognition [22] and the central nervous system [23], no review comprehensively summarizes the effects of Notoginseng and its saponins on improving learning and memory disorders. We searched the PubMed and China National Knowledge Infrastructure databases via using “Panax notoginseng saponins”, “Notoginseng saponins","Ginsenoside","Notoginsenoside” and “Learning and memory disorders” as search terms. The PubMed database was comprehensively searched up to August 2022, and it showed 85 pieces of literature; we excluded some irrelevant ones and searched for other potential and relevant references. and there were no limitations in the language of all publications. Therefore, this article provides a comprehensive review of the improvement effects and mechanisms of PNS in learning and memory disorders, aiming to provide a beneficial reference for future research.

2. Research on the etiology of learning and memory disorders

The ability to learn and form memories is crucial for organisms to adapt strategically to changing environmental demands [24]. Whether it depends on the learning and memory of the hippocampus, non-hippocampal memory, or non-neuronal immune memory, it requires a complete neuronal structure and a healthy brain physiological environment [[25], [26], [27]]. Learning and memory disorders are symptom clusters that can manifest in many diseases, such as Alzheimer's disease (AD) [5], vascular cognitive impairment and dementia (VCID) [6], malnutrition dementia [7], depression [8], traumatic brain injury (TBI) [28], post-traumatic stress disorder (PTSD) [9], and Korsakov syndrome (KS) [10] and so on.

There are direct or indirect relationships between these aforementioned disorders and learning and memory disorders. The main reason for the learning and memory impairments in AD patients is due to the accumulation of extracellular β-amyloid (Aβ), which affects neuronal functions [[29], [30], [31], [32]]. Meanwhile, Aβ promotes the abnormal phosphorylation of tau protein, leading to its aggregation into neurofibrillary tangles and generates toxic effects on neurons [[33], [34], [35]]. VCID is an age-related mental disability caused by a range of cerebrovascular disorders involving neurovascular dysfunction, blood-brain barrier (BBB) disruption, white matter damage, microRNA, oxidative stress, neuroinflammation, and gut microbiota alterations [6,36,37]. Malnutrition can lead to learning and memory disorders, and elderly people with dementia are at an increased risk of malnutrition due to various nutritional problems [[38], [39], [40], [41]]. While the mechanism of how depression leads to learning and memory impairments remains unclear [42,43], several studies have demonstrated that treating and repairing neurons in the hippocampus can improve cognitive disorders caused by depression [8,44,45]. Traumatic brain injuries can induce not only acute learning and memory disorders but also a number of neurodegenerative diseases [[46], [47], [48], [49], [50]]. Post-traumatic stress disorder (PTSD) can lead to dysregulation in multiple biological systems. These dysregulations include immune system dysregulation, heightened inflammation, oxidative stress, mitochondrial dysfunction, renin-angiotensin system dysregulation, and accelerated biological aging. These various dysregulations collectively contribute to an elevated risk of subsequent learning and memory disorders [9,[51], [52], [53]]. Korsakov syndrome (KS) is a severe neurological disorder characterized by anterograde amnesia [10] that results from alcoholism [53,54].

In current clinical trials (Phase III and Phase IV), drugs targeting learning and memory disorders include agonists or inhibitors of central nervous system receptors, including N-Methylaspartate and Memantine; opioids like Meperidine and Morphine; sleep aids and tranquilizers such as Suvorexant and Melatonin; Calcium channel blockers like Nilvadipine and Amlodipine; Hormones, for instance, Estradiol; Non-steroidal anti-inflammatory drugs (NSAIDs), such as Indomethacin; Anesthetics including Benzocaine and Cadexomer iodine; Anti-anxiety drugs and antidepressants including Opipramol and Buspirone; anticonvulsant medications like Topiramate and Lamotrigine; neuroprotectants such as 3-N-butylphthalide and Cerebrolysin; along with vitamins like Nicotinamide and B group vitamins. In addition, certain herbal-related drugs such as Black cohosh, Valerian, Piper methysticum, Chamomile, Passionflower, Erythrina mulungu, and Yohimbe have either entered or completed Phase III and Phase IV clinical trials alongside others [55,56]. (Fig. 2 for associated disorders as well as the main drug types).

Fig. 2.

Fig. 2

Associated disorders causing learning and memory disorders and the main types of drugs entering phase III and IV clinics.

3. The improvement of learning-memory disorder by notoginseng saponins

3.1. Notoginsenoside R1

Notoginsenoside R1 (R1) is a distinctive saponin found in the root of Panax notoginseng, comprising approximately 7–10% of its saponins [57,58]. Studies have shown that R1 can increase the membrane excitability of CA1 pyramidal neurons in hippocampal slices by lowering the spike threshold, possibly through a mechanism involving the inhibition of voltage-gated K (+) currents, thereby preventing Aβ-induced synaptic dysfunction and improving hippocampal-based memory performance in an AD mouse model [59]. Furthermore, voltage-gated sodium channels (Nav) play a crucial role in regulating cell excitability and the initiation and transmission of action potentials (APs) [60]. R1 modulates the abundance and/or spatial arrangement of voltage-gated sodium channels (Nav). Furthermore, R1 enhanced the viability of neurons damaged by Aβ1-42. The reduction of neuronal hyperexcitability caused by R1 could be associated with the inhibition of Navβ2 cleavage, resulting in the partial restoration of the aberrant localization of Nav 1.1α [61]. In addition, it has been demonstrated that R1 can alleviate impaired learning and memory in SD mice by regulating the Melatonin receptor type 1A (MTNR1A)-mediated phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway, thereby reducing excessive autophagy and apoptosis of hippocampal neurons [62]. Moreover, it protects against isoflurane-induced learning and cognitive dysfunction by promoting the expression of miR-29a and preventing inflammatory responses [63]. R1 promotes post-stroke angiogenesis by activating the Nicotinamide phosphoribosyltransferase- Nicotinamide adenine dinucleotide- Sirtuin 1 (NAMPT-NAD-SIRT1) cascade, which helps to improve vascular cognitive impairment and dementia [57].

3.2. Ginsenoside Rg1

Ginsenoside Rg1 (Rg1) has the highest content among PNS, accounting for approximately 20%–40% of PNS [58]. Studies targeting Rg1 to improve learning and memory disorder have also been conducted at a relatively early stage; an earlier study found that Rg1 can improve performance in a passive avoidance learning paradigm and enhance cholinergic metabolism [64].

Several studies have illustrated the efficacy of Rg1 in alleviating learning and memory deficits associated with Alzheimer's disease. This is attributed to its capacity to reduce amyloid-beta levels [65,66], modulate the activity of protein kinase A (PKA) and cAMP response element-binding protein (CREB) [67], as well as promote neuroprotection and memory enhancement through various pathways [68,69]. Moreover, Rg1 has demonstrated the ability to enhance cognitive abilities in aged mice, hinder Aβ formation, suppress inflammatory responses, regulate neurotransmitters, and boost neural stem cell proliferation, significantly improving stress-induced learning and memory impairments [[70], [71], [72], [73], [74]]. Furthermore, Rg1 exhibits potential therapeutic applications in treating learning and memory impairments in perimenopausal women [75,76], vascular cognitive impairment [77], sepsis-associated encephalopathy [78], and post-traumatic stress disorder [79,80]. For details, refer to Table 1.

Table 1.

Effects and mechanisms of ginsenoside Rg1 in ameliorating learning and memory impairment as reported and evidenced.

Effect Mechanism and Descriptions Cited references
Improving learning and memory disorder; improving performance in a passive avoidance learning paradigm Enhancing cholinergic metabolism [64]
Effective in improving learning and memory disorders caused by Alzheimer's disease Reducing the amount of Abeta detected in the brains of mice; reducing the level of amyloid beta; regulating the activity of PKA/CREB; improving cognitive performance in SAMP8 mice [65,66]
Inducing neuroprotection and ameliorating learning and memory disorders Ameliorating amyloid pathology; modulating the amyloid precursor protein process; improving cognition; activating PKA/CREB signaling [67]
Attenuating the generation of Aβ; improving learning and memory Enhancing the binding of PPARγ to the BACE1 promoter; suppressing the activity of BACE1; increasing the expression of IDE [81,82]
Reducing okadaic acid-induced spatial memory impairment; preventing Aβ formation Through the GSK3β/tau signaling pathway [68]
Restoring hippocampal long-term potentiation and memory Promoting the clearance of AD-related proteins; activating the BDNF-TrkB pathway [69]
Improving behavioral deficits in AD mice By regulating the expression of CPLX2, SYN2, and SNP25 proteins [83]
In response to the senescence of neuronal cells; ameliorating learning disabilities in aged rats Reversing tert-butyl hydroperoxide-induced morphological changes; promoting expression of synaptic plasticity-related proteins; regulating the PI3K/AKT pathway [[70], [71], [72]]
Protecting against neural stem cell senescence Ameliorating D-galactose-induced cognitive impairment; reducing oxidative stress; downregulating the Akt/mTOR signaling pathway [73]
Ameliorating cognitive deficits in aging mice induced by D-galactose and AlCl3 Restoring FGF2-Akt and BDNF-TrkB signaling axis; inhibiting apoptosis [74]
Alleviating learning and memory impairments induced by painkillers and other chemicals Improving spatial learning capacity impaired by morphine; restoring morphine-inhibited long-term potentiation [84]
Effectively improving memory impairment induced by scopolamine [85]
Inhibiting mitochondrial dysfunction; exerting antioxidant, anti-inflammatory, and anti-apoptotic effects Ameliorating isoflurane-induced caspase-3 activation; attenuating isoflurane/surgery induced neurocognitive impairment and Sirt3 dysfunction [[86], [87], [88]]
Ameliorating lipopolysaccharide-induced cognitive impairment Through regulation of the cholinergic system [89]
Protecting against neuronal degeneration induced by chronic dexamethasone By inhibiting mouse NLRP1 inflammatory cytokines [90]
Ameliorating cognitive deficits induced by repeated alcohol intoxication Modulating NR2B-containing NMDARs and excitotoxic signaling [91]
Rg1 showed good effects against neurological functions, neural structures, and neurophysiological aspects affecting learning and memory Ameliorated chronic restraint stress-induced learning and memory deficits by reducing reactive oxygen species (ROS) production, reducing neuronal oxidative damage in mouse frontal cortex and hippocampal cornu ammonis 1 (CA1), inhibiting the expression of NADPH oxidase 2 (NOX2), neutrophil cytosol factor 1 (p47phox), and ras-related c3 botulinum toxin substrate 1 (RAC1). [92]
Modulated firing in the medial prefrontal cortex of rats and inhibited hippocampo-medial prefrontal cortical long-term potentiation. Rg1 ameliorated the learning and memory deficits induced by chronic restraint stress in rats by mediating the BDNF/TrkB/extracellular signal-regulated kinase (Erk) pathway in the prefrontal cortex. [93,94]
Rg1 acts similarly to growth factors in promoting the proliferation and differentiation of neural stem cells. May reduce cognitive impairment induced by cardiac arrest by regulating neuroinflammation and hippocampal plasticity. [95,96]
Promotes remyelination and functional recovery in demyelinating diseases by enhancing oligodendrocyte progenitor cell-mediated remyelination, improving spatial memory, motor function, and anxiety-like behavior in mice. Promotes glutamate release, possibly through a Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent pathway, to regulate central nervous system neurotransmitters to enhance learning and memory. [97,98]
Reduces PTSD-like behaviors in mice by reducing corticosterone and corticotrophin-releasing hormone levels. Has a protective effect on PTSD-like behavior in mice by promoting synaptic proteins and reducing inward rectifying potassium channel 4.1 (Kir4.1) and tumor necrosis factor alpha (TNF-α) in the hippocampus. [79,80,99]
Potential application in the treatment of learning and memory disorders in postmenopausal women. Can prevent cognitive impairment and hippocampal neuronal apoptosis in vascular dementia mice, probably by promoting g protein-coupled receptor 30 (GPR30) expression. [[75], [76], [77]]

3.3. Ginsenoside Rb1

Ginsenoside Rb1(Rb1) accounts for 30–36% of the total saponins found in Notoginseng Radix et Rhizoma and has been studied extensively [58]. In vitro studies have demonstrated that Rb1 enhances choline uptake into nerve endings and promotes the release of acetylcholine (ACh) from hippocampal slices. This indicates that the potential of Rb1 to mitigate memory deficits may be linked to its facilitation of ACh metabolism in the central nervous system [78]. Further research reveals that Rb1 can increase the expression of phosphorylated Akt (P-Akt) and phosphorylated mTOR (P-mTOR), reduce the effect of phosphorylated phosphatase and tensin homolog (P-PTEN), and alleviate memory impairment, pyramidal cell necrosis, and apoptosis in the hippocampal CA1 region of rats after L-glutamate and Ca2+ microperfusion, in a dose-dependent manner [100]. In addition, intravenous administration of Rb1 can up-regulate the expression of B-cell lymphoma-extra large (Bcl-x(L)) and prevent the death of ischemic neurons. Intravenous administration of gRb1 exhibits a greater range of effective doses than intracerebroventricular injection, possibly because the blood-brain barrier regulates the central adsorption of Rb1 from the bloodstream, allowing for an optimal amount of Rb1 to be transported into the ischemic brain [101]. Furthermore, metabolite M1 of Rb1 has been shown to improve Aβ(25–35)-induced memory impairment, axonal atrophy, and synaptic loss [102]. The metabolite compound K of Rb1 may also improve learning and memory disorders by resisting inflammation and oxidation, promoting neurotransmitter release, and reducing Aβ deposition through various mechanisms [[103], [104], [105], [106], [107], [108]].

3.4. Ginsenoside Rd

Ginsenoside Rd (Rd) accounts for 5%–8.4% of the total saponins present in Notoginseng [58]. In vitro and in vivo studies suggest that Rd promotes the proliferation of neural stem cells (NSCs) without affecting their differentiation, which could explain how ginseng enhances intelligence [109].

Inflammation and apoptosis severity are highly correlated with cognitive decline. Rd has been shown to alleviate inflammation induced by β-amyloid peptide 1–40 and attenuate cognitive dysfunction in a rat model of Alzheimer's disease [110]. Moreover, subsequent studies confirmed that Rd can reduce tau phosphorylation and sequential cognition impairment after ischemic stroke [111]. Furthermore, another study found that Rd increases the expression of α-secretase and soluble amyloid precursor protein α while decreasing the expression of β-secretase and amyloid β by activating estrogen-like activity, thereby enhancing the learning and memory function of ovariectomized rats [112]. Another study revealed that Rd improves learning and memory ability in amyloid β-protein precursor transgenic mice, possibly through inhibiting the transcription activity of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) [113].

Administration of Rd has also been shown to ameliorate cognitive dysfunction induced by chronic cerebral hypoperfusion in mice, with induction of BDNF responsible for Rd-mediated neuroprotection both in vivo during chronic cerebral hypoperfusion and in vitro during oxygen-glucose deprivation/reperfusion [114]. Additionally, treatment with Rd reversed the decrease in phosphorylated PI3K, CREB, BDNF, and TrkB expression in the hippocampus caused by chronic restraint stress. The findings suggest that Rd can improve cognitive impairment in chronically stressed mice by mitigating oxidative stress and inflammation while upregulating the hippocampal BDNF-mediated CREB signaling pathway [115].

Finally, a study found that Rd can reduce anxiety and depression in mice by improving gut dysbiosis caused by alterations in intestinal microorganisms [116].

3.5. Ginsenoside Re

Ginsenoside Re(Re) is a minor component of the main saponins found in Panax notoginseng, accounting for 3.9%–6% of their total content [58]. Previous studies have shown that administration of Re before brake stress significantly improves helpless behavior and cognitive impairment by modulating the central noradrenergic system in rats [117]. Moreover, Re has been demonstrated to protect against chronic restraint stress-induced cognitive deficits by regulating NACHT, LRR and PYD domains-containing protein 3(NLRP3) and nuclear factor-erythroid 2-related factor 2 (Nrf2) pathways in mice [118]. Chronic treatment with Re can also reduce the levels of TNF-α and malondialdehyde (MDA) in both cerebral regions of diabetic rats and significantly improve their cognitive function [119].

A combined pharmacokinetic and pharmacodynamic study of Re suggested that increased levels of dopamine and acetylcholine outside the cells, particularly in the hippocampus, may contribute to its anti-dementia activity [120]. Additionally, Re can protect against phencyclidine-induced behavioral changes and mitochondrial dysfunction in the dorsolateral cortex of mice. This protective effect is achieved through the interactive modulation of glutathione peroxidase-1 and NADPH oxidase [121].

Furthermore, other studies have confirmed that Re can inhibit the interaction between platelet-activating factor receptor (PAFR), NF-κB, and microglia proliferation in the hippocampus and reduce memory impairment in aged Klotho-deficient mice through the mutual regulation of angiotensin II ⅱAT1 receptor, Nrf2, and glutathione peroxidase-1 (GPx-1) genes [122,123].

3.6. Other panax notoginseng saponins

In addition to the five main components of PNS mentioned above, other saponins comprise approximately 9.8% of PNS [58]. Although their content is relatively low, they exhibit significant pharmacological effects on learning and memory disorders that cannot be ignored, and their actions and mechanisms are shown in Table 2.

Table 2.

Effects and Mechanisms of other Panax notoginseng saponins in Ameliorating Learning and Memory Impairment as Reported and Evidenced.

Chemical Component Effects Mechanism and Descriptions Cited references
Ginsenoside Rb3 Antidepressant-like effects, modulates neurotransmitters [124]
Ginsenoside Rf Improves spatial learning and memory in AD mice [125]
Ginsenoside Rg2 Enhances cognitive behavior; protects against memory impairment; anti-apoptotic effects Autophagy induction, protein aggregate clearance, protective effects [[126], [127], [128], [129], [130]]
Ginsenoside Rg3 Attenuates learning and memory disorders; controls fear memory regression Anti-inflammatory activity, Aβ42 uptake and degradation, HPA axis, BDNF-TrkB pathway [[131], [132], [133], [134]]
Ginsenoside Rg5 Ameliorates cognitive dysfunction, prevents apoptosis, impacts gene expression related to cognitive impairment Neuroinflammatory attenuation, decreases β-amyloid accumulation, HO-1/Nrf2 signaling pathway, gene expression modulation [135,136]
Ginsenoside Rh1 Improves memory in mouse models, increases cell survival Cell survival, upregulates BDNF expression [137,138]
Ginsenoside Rh2 Reverses cognitive impairment from sleep deprivation, promotes spatial learning, protects against memory impairment, exhibits antidepressant effects Cholinergic transmission regulation, reduction of oxidative stress, ERK-CREB-BDNF signaling pathway modulation [[139], [140], [141], [142]]
Ginsenoside Rk3 Cognitive improvement Improves spatial learning and memory deficits in double transgenic mouse models of APP/PS1 [143]

4. Conclusion and remarks

PNS are a series of natural compounds that improve learning-memory disorders and cognition through their effects on the central nervous system. In this review, we summarize the therapeutic effects and molecular mechanisms by which various Panax ginseng saponin-like constituents improve learning-memory disorders (Fig. 3). The available data and our summarized results suggest that Panax ginseng saponins may improve learning-memory impairment through various pharmacological activities, such as their anti-inflammatory, anti-apoptotic, and antioxidant properties, reduction of amyloid beta levels, maintenance of mitochondrial homeostasis, improvement of neuronal structure and function, promotion of neurogenesis, modulation of neurotransmitter release, and maintenance of probiotic homeostasis. Based on these findings, Panax notoginseng saponin-like components, as a series of specific natural compounds, hold promising potential for improving learning and memory disorders.

Fig. 3.

Fig. 3

Mechanism of improving learning and memory impairments by PNS.

In ameliorating the learning-memory impairment associated with AD, PNS-like components play a role in improving learning-memory impairment by modulating the PKA/CREB pathway [65,66], GSK3β/tau pathway [67], regulating PPARγ [82], NFκB [123], and reducing amyloid β levels.

In enhancing the hippocampus, neurons, and their structure and function, PNS-like components have been shown to promote the value-added of neural stem cells [94,109], regulate the PI3K/Akt/mTOR pathway, reduce excessive autophagy and apoptosis of hippocampal neurons [62,100], regulate neuronal structure and function [61,97,140], and modulate neurotransmitters in the central nervous system [78,98,111]to improve learning and memory disorders.

In response to learning and memory disorders caused by chemicals such as anesthetics, painkillers, and alcohol, PNS-like components improve mitochondrial function [86,129] through anti-inflammation [63,84,90,131,144], reduce Sirt3 dysfunction 75, modulate the PI3K/AKT/GSK-3β signaling pathway [88], modulate the NF-κB signaling pathway [133], and modulate the ERK/CREB/BDNF signaling pathway [141].

In stroke and vascular dementia-induced learning and memory disorder, PNS-like components play ameliorative roles by promoting angiogenesis [57], reducing hippocampal neuronal apoptosis [77,101,130], and providing neuroprotection [114].

In neuronal cell aging, PNS-like components have been effective in ameliorating learning and memory disorder by improving morphological changes in cell aging [70], regulating the PI3K/AKT/mTOR pathway [[71], [72], [73]], and regulating the FGF2/Akt and BDNF/TrkB signaling pathways [73].

Due to post-traumatic stress disorder, depression, and external physical stimuli that lead to learning and memory disorder, PNS-like components may play a role in the amelioration of learning and memory disorder by reducing neuronal inflammation and oxidative damage [80,92,115,136], modulating the BDNF/TrkB/Erk signaling pathway [93,115,134,142], regulating the NLRP3/Nrf2 signaling pathway [118], regulating neurotransmitter and hormone levels [79,117,124,128,136], and regulating intestinal flora dysbiosis115.

In addition to the above, PNS also plays a role in the treatment of learning and memory disorder in postmenopausal women [75,76], ameliorating cognitive dysfunction in mice with sepsis-related encephalopathy [145], and improving cognitive function in diabetic rats [119].

PNS have been extensively studied for the treatment of cardiovascular and cerebrovascular diseases, yielding abundant results. As research in human brain science, behavior, and brain-related diseases, such as Alzheimer's disease, has progressed, there has been a gradual increase in studies exploring the potential of PNS in treating learning and memory disorders. In comparison to ongoing or completed clinical trials examining pharmacological interventions for learning and memory disorders, PNS have exhibited broader and more effective therapeutic effects. However, due to factors such as the cost and availability of PNS, there is a lack of comprehensive clinical research examining their efficacy in improving learning and memory disorders. With the advancement of professional expertise and technical capabilities, it is anticipated that future studies will contribute to a more thorough understanding of the role of PNS in treating and improving learning and memory disorders.

In conclusion, this review was conducted by discussing the ameliorative effects of PNS-like constituents on learning and memory disorders due to various causes. The summarized results and analyses suggest that Panax notoginseng saponin-like constituents hold promise as a series of natural active ingredients for improving learning and memory disorders.

Data availability statement

Data availability is not applicable to this article as no new data were created or analyzed in this study.

Funding Statement

The study received financial support from the CAMS Innovation Fund for Medical Sciences (CIFMS), 2021-I2M-1-031, National Key Research and Development Program of China, (No.2023YFD2201802), and the Key Research and Development Program Project of Heilongjiang Province (2022ZX02C08)

CRediT authorship contribution statement

Shusen Liu: Writing – original draft, Data curation. Min Wang: Writing – review & editing. Haiyan Xiao: Writing – review & editing, Data curation. Jingxue Ye: Writing – review & editing, Resources. L.I. Cao: Writing – review & editing, Project administration. Wenlan Li: Writing – review & editing, Visualization, Funding acquisition, Conceptualization. Guibo Sun: Writing – review & editing, Visualization, Project administration, Funding acquisition, Conceptualization.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Contributor Information

Wenlan Li, Email: lwldzd@163.com.

Guibo Sun, Email: sunguibo@126.com.

References

  • 1.Kim S.J., Linden D.J. Ubiquitous plasticity and memory storage. Neuron. 2007;56:582–592. doi: 10.1016/j.neuron.2007.10.030. [DOI] [PubMed] [Google Scholar]
  • 2.Zhang Y., Chu J.-M.-T., Wong G.-T.-C. Cerebral glutamate regulation and receptor changes in perioperative neuroinflammation and cognitive dysfunction. Biomolecules. 2022;12:597. doi: 10.3390/biom12040597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cao Y., Yang Y., Wu H., Lu Y., Wu S., Liu L., Wang C., Huang F., Shi H., Zhang B., Wu X., Wang Z. Stem-leaf saponins from Panax notoginseng counteract aberrant autophagy and apoptosis in hippocampal neurons of mice with cognitive impairment induced by sleep deprivation. J Ginseng Res. 2020;44:442–452. doi: 10.1016/j.jgr.2019.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ben Zablah Y., Zhang H., Gugustea R., Jia Z. LIM-kinases in synaptic plasticity, memory, and brain diseases. Cells. 2021;10:2079. doi: 10.3390/cells10082079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.McKhann G.M., Knopman D.S., Chertkow H., Hyman B.T., Jack C.R., Kawas C.H., Klunk W.E., Koroshetz W.J., Manly J.J., Mayeux R., Mohs R.C., Morris J.C., Rossor M.N., Scheltens P., Carrillo M.C., Thies B., Weintraub S., Phelps C.H. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7:263–269. doi: 10.1016/j.jalz.2011.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gorelick P.B., Scuteri A., Black S.E., DeCarli C., Greenberg S.M., Iadecola C., Launer L.J., Laurent S., Lopez O.L., Nyenhuis D., Petersen R.C., Schneider J.A., Tzourio C., Arnett D.K., Bennett D.A., Chui H.C., Higashida R.T., Lindquist R., Nilsson P.M., Roman G.C., Sellke F.W., Seshadri S. Vascular contributions to cognitive impairment and dementia. Stroke. 2011;42:2672–2713. doi: 10.1161/STR.0b013e3182299496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cai Shi D., Long C., Vardeman E., Kennelly E.J., Lawton M.A., Di R. Potential anti-alzheimer properties of mogrosides in vitamin B12-deficient Caenorhabditis elegans. Molecules. 2023;28:1826. doi: 10.3390/molecules28041826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Manji H.K., Quiroz J.A., Sporn J., Payne J.L., Denicoff K., A Gray N., Zarate C.A., Charney D.S. Enhancing neuronal plasticity and cellular resilience to develop novel, improved therapeutics for difficult-to-treat depression. Biol. Psychiatr. 2003;53:707–742. doi: 10.1016/s0006-3223(03)00117-3. [DOI] [PubMed] [Google Scholar]
  • 9.Miller M.W., Sadeh N. Traumatic stress, oxidative stress and post-traumatic stress disorder: neurodegeneration and the accelerated-aging hypothesis. Mol. Psychiatr. 2014;19:1156–1162. doi: 10.1038/mp.2014.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zahr N.M., Kaufman K.L., Harper C.G. Clinical and pathological features of alcohol-related brain damage. Nat. Rev. Neurol. 2011;7:284–294. doi: 10.1038/nrneurol.2011.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rajabian A., Rameshrad M., Hosseinzadeh H. Therapeutic potential of Panax ginseng and its constituents, ginsenosides and gintonin, in neurological and neurodegenerative disorders: a patent review. Expert Opin. Ther. Pat. 2019;29:55–72. doi: 10.1080/13543776.2019.1556258. [DOI] [PubMed] [Google Scholar]
  • 12.Guan L., Mao Z., Yang S., Wu G., Chen Y., Yin L., Qi Y., Han L., Xu L. Dioscin alleviates Alzheimer's disease through regulating RAGE/NOX4 mediated oxidative stress and inflammation. Biomed. Pharmacother. 2022;152 doi: 10.1016/j.biopha.2022.113248. [DOI] [PubMed] [Google Scholar]
  • 13.Kim S., Kang I.-H., Nam J.-B., Cho Y., Chung D.-Y., Kim S.-H., Kim J.-S., Cho Y.-D., Hong E.-K., Sohn N.-W., Shin J.-W. Ameliorating the effect of astragaloside IV on learning and memory deficit after chronic cerebral hypoperfusion in rats. Molecules. 2015;20:1904–1921. doi: 10.3390/molecules20021904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cao Y., Yang Y., Wu H., Lu Y., Wu S., Liu L., Wang C., Huang F., Shi H., Zhang B., Wu X., Wang Z. Stem-leaf saponins from Panax notoginseng counteract aberrant autophagy and apoptosis in hippocampal neurons of mice with cognitive impairment induced by sleep deprivation. J Ginseng Res. 2020;44:442–452. doi: 10.1016/j.jgr.2019.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhang H.-S., Wang S.-Q. Notoginsenoside R1 from Panax notoginseng inhibits TNF-alpha-induced PAI-1 production in human aortic smooth muscle cells. Vasc. Pharmacol. 2006;44:224–230. doi: 10.1016/j.vph.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 16.Gu B., Nakamichi N., Zhang W.-S., Nakamura Y., Kambe Y., Fukumori R., Takuma K., Yamada K., Takarada T., Taniura H., Yoneda Y. Possible protection by notoginsenoside R1 against glutamate neurotoxicity mediated by N-methyl-D-aspartate receptors composed of an NR1/NR2B subunit assembly. J. Neurosci. Res. 2009;87:2145–2156. doi: 10.1002/jnr.22021. [DOI] [PubMed] [Google Scholar]
  • 17.Wang Y.-H., Du G.-H. Ginsenoside Rg1 inhibits beta-secretase activity in vitro and protects against Abeta-induced cytotoxicity in PC12 cells. J. Asian Nat. Prod. Res. 2009;11:604–612. doi: 10.1080/10286020902843152. [DOI] [PubMed] [Google Scholar]
  • 18.Zhou X., Chen L.-L., Xie R.F., Lam W., Zhang Z.-J., Jiang Z.-L., Cheng Y.-C. Chemosynthesis pathway and bioactivities comparison of saponins in radix and flower of Panax notoginseng (Burk. F.H. Chen, J Ethnopharmacol. 2017;201:56–72. doi: 10.1016/j.jep.2016.11.008. [DOI] [PubMed] [Google Scholar]
  • 19.Xie Y., Wang C. Herb–drug interactions between Panax notoginseng or its biologically active compounds and therapeutic drugs: a comprehensive pharmacodynamic and pharmacokinetic review. J. Ethnopharmacol. 2023;307 doi: 10.1016/j.jep.2023.116156. [DOI] [PubMed] [Google Scholar]
  • 20.Kim D.-H. Chemical diversity of Panax ginseng, Panax quinquifolium, and Panax notoginseng. J Ginseng Res. 2012;36:1–15. doi: 10.5142/jgr.2012.36.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Liu H., Lu X., Hu Y., Fan X. Chemical constituents of Panax ginseng and Panax notoginseng explain why they differ in therapeutic efficacy. Pharmacol. Res. 2020;161 doi: 10.1016/j.phrs.2020.105263. [DOI] [PubMed] [Google Scholar]
  • 22.Smith I., Williamson E.M., Putnam S., Farrimond J., Whalley B.J. Effects and mechanisms of ginseng and ginsenosides on cognition. Nutr. Rev. 2014;72:319–333. doi: 10.1111/nure.12099. [DOI] [PubMed] [Google Scholar]
  • 23.Nt R., Ts K., Kc C., J R., Nh K., A. M, M. A, A. H, A. A, I. A A role of ginseng and its constituents in the treatment of central nervous system disorders. Evid. base Compl. Alternative Med. : eCAM. 2016;2016 doi: 10.1155/2016/2614742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rasch B., Born J. About sleep's role in memory. Physiol. Rev. 2013;93:681–766. doi: 10.1152/physrev.00032.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Deng W., Aimone J.B., Gage F.H. New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat. Rev. Neurosci. 2010;11:339–350. doi: 10.1038/nrn2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Yau S., Li A., So K.-F. Involvement of adult hippocampal neurogenesis in learning and forgetting. Neural Plast. 2015;2015 doi: 10.1155/2015/717958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lazarov O., Hollands C. Hippocampal neurogenesis: learning to remember. Prog. Neurobiol. 2016;138–140:1–18. doi: 10.1016/j.pneurobio.2015.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Livingston G., Huntley J., Sommerlad A., Ames D., Ballard C., Banerjee S., Brayne C., Burns A., Cohen-Mansfield J., Cooper C., Costafreda S.G., Dias A., Fox N., Gitlin L.N., Howard R., Kales H.C., Kivimäki M., Larson E.B., Ogunniyi A., Orgeta V., Ritchie K., Rockwood K., Sampson E.L., Samus Q., Schneider L.S., Selbæk G., Teri L., Mukadam N. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet. 2020;396:413–446. doi: 10.1016/S0140-6736(20)30367-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chen G., Xu T., Yan Y., Zhou Y., Jiang Y., Melcher K., Xu H.E. Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol. Sin. 2017;38:1205–1235. doi: 10.1038/aps.2017.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Armstrong R.A. The molecular biology of senile plaques and neurofibrillary tangles in Alzheimer's disease. Folia Neuropathol. 2009;47:289–299. [PubMed] [Google Scholar]
  • 31.Thal D.R., Fändrich M. Protein aggregation in Alzheimer's disease: aβ and τ and their potential roles in the pathogenesis of AD. Acta Neuropathol. 2015;129:163–165. doi: 10.1007/s00401-015-1387-2. [DOI] [PubMed] [Google Scholar]
  • 32.Hurtado D.E., Molina-Porcel L., Iba M., Aboagye A.K., Paul S.M., Trojanowski J.Q., Lee V.M.-Y. A{beta} accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am. J. Pathol. 2010;177:1977–1988. doi: 10.2353/ajpath.2010.100346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lauckner J., Frey P., Geula C. Comparative distribution of tau phosphorylated at Ser 262 in pre-tangles and tangles. Neurobiol. Aging. 2003;24:767–776. doi: 10.1016/s0197-4580(02)00228-2. [DOI] [PubMed] [Google Scholar]
  • 34.Metaxas A., Kempf S.J. Neurofibrillary tangles in Alzheimer's disease: elucidation of the molecular mechanism by immunohistochemistry and tau protein phospho-proteomics. Neural Regen Res. 2016;11:1579–1581. doi: 10.4103/1673-5374.193234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Vanden Dries V., Stygelbout V., Pierrot N., Yilmaz Z., Suain V., De Decker R., Buée L., Octave J.-N., Brion J.-P., Leroy K. Amyloid precursor protein reduction enhances the formation of neurofibrillary tangles in a mutant tau transgenic mouse model. Neurobiol. Aging. 2017;55:202–212. doi: 10.1016/j.neurobiolaging.2017.03.031. [DOI] [PubMed] [Google Scholar]
  • 36.Yang T., Zhang F. Targeting transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) for the intervention of vascular cognitive impairment and dementia. Arterioscler. Thromb. Vasc. Biol. 2021;41:97–116. doi: 10.1161/ATVBAHA.120.314804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Fulop G.A., Tarantini S., Yabluchanskiy A., Molnar A., Prodan C.I., Kiss T., Csipo T., Lipecz A., Balasubramanian P., Farkas E., Toth P., Sorond F., Csiszar A., Ungvari Z. Role of age-related alterations of the cerebral venous circulation in the pathogenesis of vascular cognitive impairment. Am. J. Physiol. Heart Circ. Physiol. 2019;316:H1124–H1140. doi: 10.1152/ajpheart.00776.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Volkert D., Chourdakis M., Faxen-Irving G., Frühwald T., Landi F., Suominen M.H., Vandewoude M., Wirth R., Schneider S.M. ESPEN guidelines on nutrition in dementia. Clin. Nutr. 2015;34:1052–1073. doi: 10.1016/j.clnu.2015.09.004. [DOI] [PubMed] [Google Scholar]
  • 39.Fávaro-Moreira N.C., Krausch-Hofmann S., Matthys C., Vereecken C., Vanhauwaert E., Declercq A., Bekkering G.E., Duyck J. Risk factors for malnutrition in older adults: a systematic review of the literature based on longitudinal data. Adv. Nutr. 2016;7:507–522. doi: 10.3945/an.115.011254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gibson G.E., Hirsch J.A., Fonzetti P., Jordan B.D., Cirio R.T., Elder J. Vitamin B1 (thiamine) and dementia. Ann. N. Y. Acad. Sci. 2016;1367:21–30. doi: 10.1111/nyas.13031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Doorduijn A.S., Visser M., van de Rest O., Kester M.I., de Leeuw F.A., Boesveldt S., Fieldhouse J.L.P., van den Heuvel E.G.H.M., Teunissen C.E., Scheltens P., van der Flier W.M., de van der Schueren M.A.E. Associations of AD biomarkers and cognitive performance with nutritional status: the NUDAD Project. Nutrients. 2019;11:1161. doi: 10.3390/nu11051161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Swaab D.F., Bao A.-M., Lucassen P.J. The stress system in the human brain in depression and neurodegeneration. Ageing Res. Rev. 2005;4:141–194. doi: 10.1016/j.arr.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 43.Otte C., Gold S.M., Penninx B.W., Pariante C.M., Etkin A., Fava M., Mohr D.C., Schatzberg A.F. Major depressive disorder. Nat. Rev. Dis. Prim. 2016;2 doi: 10.1038/nrdp.2016.65. [DOI] [PubMed] [Google Scholar]
  • 44.Campbell S., Macqueen G. The role of the hippocampus in the pathophysiology of major depression. J. Psychiatry Neurosci. 2004;29:417–426. [PMC free article] [PubMed] [Google Scholar]
  • 45.Goeldner C., Ballard T.M., Knoflach F., Wichmann J., Gatti S., Umbricht D. Cognitive impairment in major depression and the mGlu 2 receptor as a therapeutic target. Neuropharmacology. 2013;64:337–346. doi: 10.1016/j.neuropharm.2012.08.001. [DOI] [PubMed] [Google Scholar]
  • 46.Washington P.M., Villapol S., Burns M.P. Polypathology and dementia after brain trauma: does brain injury trigger distinct neurodegenerative diseases, or should they be classified together as traumatic encephalopathy? Exp. Neurol. 2016;275(Pt 3):381–388. doi: 10.1016/j.expneurol.2015.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Collins-Praino L.E., Corrigan F. Does neuroinflammation drive the relationship between tau hyperphosphorylation and dementia development following traumatic brain injury? Brain Behav. Immun. 2017;60:369–382. doi: 10.1016/j.bbi.2016.09.027. [DOI] [PubMed] [Google Scholar]
  • 48.VanItallie T.B. Traumatic brain injury (TBI) in collision sports: possible mechanisms of transformation into chronic traumatic encephalopathy (CTE) Metabolism. 2019;100S doi: 10.1016/j.metabol.2019.07.007. [DOI] [PubMed] [Google Scholar]
  • 49.Wu Z., Wang Z.-H., Liu X., Zhang Z., Gu X., Yu S.P., Keene C.D., Cheng L., Ye K. Traumatic brain injury triggers APP and Tau cleavage by delta-secretase, mediating Alzheimer's disease pathology. Prog. Neurobiol. 2020;185 doi: 10.1016/j.pneurobio.2019.101730. [DOI] [PubMed] [Google Scholar]
  • 50.Brett B.L., Gardner R.C., Godbout J., Dams-O’Connor K., Keene C.D. Traumatic brain injury and risk of neurodegenerative disorder. Biol. Psychiatr. 2022;91:498–507. doi: 10.1016/j.biopsych.2021.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Deslauriers J., van Wijngaarde M., Geyer M.A., Powell S., Risbrough V.B. Effects of LPS-induced immune activation prior to trauma exposure on PTSD-like symptoms in mice. Behav. Brain Res. 2017;323:117–123. doi: 10.1016/j.bbr.2017.01.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Miller M.W., Lin A.P., Wolf E.J., Miller D.R. Oxidative stress, inflammation, and neuroprogression in chronic PTSD. Harv. Rev. Psychiatr. 2018;26:57–69. doi: 10.1097/HRP.0000000000000167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sumner J.A., Cleveland S., Chen T., Gradus J.L. Psychological and biological mechanisms linking trauma with cardiovascular disease risk. Transl. Psychiatry. 2023;13:25. doi: 10.1038/s41398-023-02330-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sergei Sergeivich Korsakov 1853-1900). Korsakov's psychosis. JAMA. 1970;212:1700. [PubMed] [Google Scholar]
  • 55.Learning Disability disease: Malacards - Research Articles, Drugs, Genes, Clinical Trials, (n.d.) https://www.malacards.org/card/learning_disability?search=Learning%20memory%20disorder (accessed August 11, 2023)..
  • 56.Dementia disease: Malacards - Research Articles, Drugs, Genes, Clin. Trials, (n.d.). https://www.malacards.org/card/dementia?search=Learning%20memory%20disorder (accessed August 11, 2023).
  • 57.Zhu T., Xie W.-J., Wang L., Jin X.-B., Meng X.-B., Sun G.-B., Sun X.-B. Notoginsenoside R1 activates the NAMPT-NAD+-SIRT1 cascade to promote postischemic angiogenesis by modulating Notch signaling. Biomed. Pharmacother. 2021;140 doi: 10.1016/j.biopha.2021.111693. [DOI] [PubMed] [Google Scholar]
  • 58.Liu J., Wang Y., Qiu L., Yu Y., Wang C. Saponins of Panax notoginseng: chemistry, cellular targets and therapeutic opportunities in cardiovascular diseases. Expet Opin. Invest. Drugs. 2014;23:523–539. doi: 10.1517/13543784.2014.892582. [DOI] [PubMed] [Google Scholar]
  • 59.Yan S., Li Z., Li H., Arancio O., Zhang W. Notoginsenoside R1 increases neuronal excitability and ameliorates synaptic and memory dysfunction following amyloid elevation. Sci. Rep. 2014;4:6352. doi: 10.1038/srep06352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hodgkin A.L., Huxley A.F. A quantitative description of membrane current and its application to conduction and excitation in nerve. J. Physiol. 1952;117:500–544. doi: 10.1113/jphysiol.1952.sp004764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hu T., Li S., Liang W.-Q., Li S.-S., Lu M.-N., Chen B., Zhang L., Mao R., Ding W.-H., Gao W.-W., Chen S.-W., XiYang Y.-B., Zhang J., Wang X.-Y. Notoginsenoside R1-induced neuronal repair in models of alzheimer disease is associated with an alteration in neuronal hyperexcitability, which is regulated by Nav. Front. Cell. Neurosci. 2020;14:280. doi: 10.3389/fncel.2020.00280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cao Y., Li Q., Zhou A., Ke Z., Chen S., Li M., Gong Z., Wang Z., Wu X. Notoginsenoside R1 reverses abnormal autophagy in hippocampal neurons of mice with sleep deprivation through Melatonin receptor 1A. Front. Pharmacol. 2021;12 doi: 10.3389/fphar.2021.719313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wang M., Liu H., Xu L., Li M., Zhao M. The protective effect of notoginsenoside R1 on isoflurane-induced neurological impairment in the rats via regulating miR-29a expression and neuroinflammation. Neuroimmunomodulation. 2022;29:70–76. doi: 10.1159/000518215. [DOI] [PubMed] [Google Scholar]
  • 64.Rudakewich M., Ba F., Benishin C.G. Neurotrophic and neuroprotective actions of ginsenosides Rb(1) and Rg(1) Planta Med. 2001;67:533–537. doi: 10.1055/s-2001-16488. [DOI] [PubMed] [Google Scholar]
  • 65.Shi Y.-Q., Huang T.-W., Chen L.-M., Pan X.-D., Zhang J., Zhu Y.-G., Chen X.-C. Ginsenoside Rg1 attenuates amyloid-beta content, regulates PKA/CREB activity, and improves cognitive performance in SAMP8 mice. J Alzheimers Dis. 2010;19:977–989. doi: 10.3233/JAD-2010-1296. [DOI] [PubMed] [Google Scholar]
  • 66.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. 2012;1822:286–292. doi: 10.1016/j.bbadis.2011.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Song X.-Y., Hu J.-F., Chu S.-F., Zhang Z., Xu S., Yuan Y.-H., Han N., Liu Y., Niu F., He X., Chen N.-H. Ginsenoside Rg1 attenuates okadaic acid induced spatial memory impairment by the GSK3β/tau signaling pathway and the Aβ formation prevention in rats. Eur. J. Pharmacol. 2013;710:29–38. doi: 10.1016/j.ejphar.2013.03.051. [DOI] [PubMed] [Google Scholar]
  • 68.Quan Q., Wang J., Li X., Wang Y. Ginsenoside Rg1 decreases Aβ(1-42) level by upregulating PPARγ and IDE expression in the hippocampus of a rat model of Alzheimer's disease. PLoS One. 2013;8 doi: 10.1371/journal.pone.0059155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Li F., Wu X., Li J., Niu Q. Ginsenoside Rg1 ameliorates hippocampal long-term potentiation and memory in an Alzheimer's disease model. Mol. Med. Rep. 2016;13:4904–4910. doi: 10.3892/mmr.2016.5103. [DOI] [PubMed] [Google Scholar]
  • 70.Chen X., Zhang J., Fang Y., Zhao C., Zhu Y. Ginsenoside Rg1 delays tert-butyl hydroperoxide-induced premature senescence in human WI-38 diploid fibroblast cells. J Gerontol A Biol Sci Med Sci. 2008;63:253–264. doi: 10.1093/gerona/63.3.253. [DOI] [PubMed] [Google Scholar]
  • 71.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 A Biol Sci Med Sci. 2014;69:282–294. doi: 10.1093/gerona/glt091. [DOI] [PubMed] [Google Scholar]
  • 72.Zhu G., Wang Y., Li J., Wang J. Chronic treatment with ginsenoside Rg1 promotes memory and hippocampal long-term potentiation in middle-aged mice. Neuroscience. 2015;292:81–89. doi: 10.1016/j.neuroscience.2015.02.031. [DOI] [PubMed] [Google Scholar]
  • 73.Chen L., Yao H., Chen X., Wang Z., Xiang Y., Xia J., Liu Y., Wang Y. Ginsenoside Rg1 decreases oxidative stress and down-regulates akt/mTOR signalling to attenuate cognitive impairment in mice and senescence of neural stem cells induced by D-galactose. Neurochem. Res. 2018;43:430–440. doi: 10.1007/s11064-017-2438-y. [DOI] [PubMed] [Google Scholar]
  • 74.Zhong S.-J., Wang L., Gu R.-Z., Zhang W.-H., Lan R., Qin X.-Y. Ginsenoside Rg1 ameliorates the cognitive deficits in D-galactose and AlCl3-induced aging mice by restoring FGF2-Akt and BDNF-TrkB signaling axis to inhibit apoptosis. Int. J. Med. Sci. 2020;17:1048–1055. doi: 10.7150/ijms.43979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhang X., Wang J., Xing Y., Gong L., Li H., Wu Z., Li Y., Wang J., Wang Y., Dong L., Li S. Effects of ginsenoside Rg1 or 17β-estradiol on a cognitively impaired, ovariectomized rat model of Alzheimer's disease. Neuroscience. 2012;220:191–200. doi: 10.1016/j.neuroscience.2012.06.027. [DOI] [PubMed] [Google Scholar]
  • 76.Shi C., Zheng D., Fang L., Wu F., Kwong W.H., Xu J. Ginsenoside Rg1 promotes nonamyloidgenic cleavage of APP via estrogen receptor signaling to MAPK/ERK and PI3K/Akt. Biochim. Biophys. Acta. 2012;1820:453–460. doi: 10.1016/j.bbagen.2011.12.005. [DOI] [PubMed] [Google Scholar]
  • 77.F S., J W., F G., J W., G Z. Ginsenoside Rg1 prevents cognitive impairment and hippocampal neuronal apoptosis in experimental vascular dementia mice by promoting GPR30 expression. Neural Plast. 2021;2021 doi: 10.1155/2021/2412220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Benishin C.G., Lee R., Wang L.C., Liu H.J. Effects of ginsenoside Rb1 on central cholinergic metabolism. Pharmacology. 1991;42:223–229. doi: 10.1159/000138801. [DOI] [PubMed] [Google Scholar]
  • 79.Wang Z., Zhu K., Chen L., Ou Yang L., Huang Y., Zhao Y. Preventive effects of ginsenoside Rg1 on post-traumatic stress disorder (PTSD)-like behavior in male C57/B6 mice. Neurosci. Lett. 2015;605:24–28. doi: 10.1016/j.neulet.2015.08.017. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang Z., Song Z., Shen F., Xie P., Wang J., Zhu A.-S., Zhu G. Ginsenoside Rg1 prevents PTSD-like behaviors in mice through promoting synaptic proteins, reducing Kir4.1 and TNF-α in the Hippocampus. Mol. Neurobiol. 2021;58:1550–1563. doi: 10.1007/s12035-020-02213-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Chen F., Eckman E.A., Eckman C.B. Reductions in levels of the Alzheimer's amyloid beta peptide after oral administration of ginsenosides. Faseb. J. 2006;20:1269–1271. doi: 10.1096/fj.05-5530fje. [DOI] [PubMed] [Google Scholar]
  • 82.Chen L.-M., Lin Z.-Y., Zhu Y.-G., Lin N., Zhang J., Pan X.-D., Chen X.-C. Ginsenoside Rg1 attenuates β-amyloid generation via suppressing PPARγ-regulated BACE1 activity in N2a-APP695 cells. Eur. J. Pharmacol. 2012;675:15–21. doi: 10.1016/j.ejphar.2011.11.039. [DOI] [PubMed] [Google Scholar]
  • 83.Nie L., Xia J., Li H., Zhang Z., Yang Y., Huang X., He Z., Liu J., Yang X. Ginsenoside Rg1 ameliorates behavioral abnormalities and modulates the hippocampal proteomic change in triple transgenic mice of Alzheimer's disease. Oxid. Med. Cell. Longev. 2017;2017 doi: 10.1155/2017/6473506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Qi D., Zhu Y., Wen L., Liu Q., Qiao H. Ginsenoside Rg1 restores the impairment of learning induced by chronic morphine administration in rats. J. Psychopharmacol. 2009;23:74–83. doi: 10.1177/0269881107082950. [DOI] [PubMed] [Google Scholar]
  • 85.Wang Q., Sun L.-H., Jia W., Liu X.-M., Dang H.-X., Mai W.-L., Wang N., Steinmetz A., Wang Y.-Q., Xu C.-J. Comparison of ginsenosides Rg1 and Rb1 for their effects on improving scopolamine-induced learning and memory impairment in mice. Phytother Res. 2010;24:1748–1754. doi: 10.1002/ptr.3130. [DOI] [PubMed] [Google Scholar]
  • 86.Miao H.H., Zhen Y., Ding G.N., Hong F.X., Xie Z.C., Tian M. Ginsenoside Rg1 attenuates isoflurane-induced caspase-3 activation via inhibiting mitochondrial dysfunction. Biomed. Environ. Sci. 2015;28:116–126. doi: 10.3967/bes2015.014. [DOI] [PubMed] [Google Scholar]
  • 87.Miao H.-H., Wang M., Wang H.-X., Tian M., Xue F.-S. Ginsenoside Rg1 attenuates isoflurane/surgery-induced cognitive disorders and sirtuin 3 dysfunction. Biosci. Rep. 2019;39 doi: 10.1042/BSR20190069. BSR20190069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhang Y., Zhang Z., Wang H., Cai N., Zhou S., Zhao Y., Chen X., Zheng S., Si Q., Zhang W. Neuroprotective effect of ginsenoside Rg1 prevents cognitive impairment induced by isoflurane anesthesia in aged rats via antioxidant, anti-inflammatory and anti-apoptotic effects mediated by the PI3K/AKT/GSK-3β pathway. Mol. Med. Rep. 2016;14:2778–2784. doi: 10.3892/mmr.2016.5556. [DOI] [PubMed] [Google Scholar]
  • 89.J Y., P J., W X.Z., D W.T. Ameliorative effect of ginsenoside Rg1 on lipopolysaccharide-induced cognitive impairment: role of cholinergic system. Neurochem. Res. 2017;42 doi: 10.1007/s11064-016-2171-y. [DOI] [PubMed] [Google Scholar]
  • 90.Zhang Y., Hu W., Zhang B., Yin Y., Zhang J., Huang D., Huang R., Li W., Li W. Ginsenoside Rg1 protects against neuronal degeneration induced by chronic dexamethasone treatment by inhibiting NLRP-1 inflammasomes in mice. Int. J. Mol. Med. 2017;40:1134–1142. doi: 10.3892/ijmm.2017.3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Huang L., Peng Z., Lu C., Chen Y., Lv J.-W., Qin M., Liao D.-F., Liu X.-M., Shi Z. Ginsenoside Rg1 alleviates repeated alcohol exposure-induced psychomotor and cognitive deficits. Chin. Med. 2020;15:44. doi: 10.1186/s13020-020-00325-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wang Y., Kan H., Yin Y., Wu W., Hu W., Wang M., 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. doi: 10.1016/j.pbb.2014.02.012. [DOI] [PubMed] [Google Scholar]
  • 93.Kezhu W., Pan X., Cong L., Liming D., Beiyue Z., Jingwei L., Yanyan Y., Xinmin L. Effects of ginsenoside Rg1 on learning and memory in a reward-directed instrumental conditioning task in chronic restraint stressed rats. Phytother Res. 2017;31:81–89. doi: 10.1002/ptr.5733. [DOI] [PubMed] [Google Scholar]
  • 94.Ghaeminia M., Rajkumar R., Koh H.-L., Dawe G.S., Tan C.H. Ginsenoside Rg1 modulates medial prefrontal cortical firing and suppresses the hippocampo-medial prefrontal cortical long-term potentiation. J Ginseng Res. 2018;42:298–303. doi: 10.1016/j.jgr.2017.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Gao J., Wan F., Tian M., Li Y., Li Y., Li Q., Zhang J., Wang Y., Huang X., Zhang L., Si Y. Effects of ginsenoside-Rg1 on the proliferation and glial-like directed differentiation of embryonic rat cortical neural stem cells in vitro. Mol. Med. Rep. 2017;16:8875–8881. doi: 10.3892/mmr.2017.7737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Wu Z., Huang J., Bai X., Wang Q., Wang F., Xu J., Tang H., Yin C., Wang Y., Yu F., Zhang H. Ginsenoside-Rg1 mitigates cardiac arrest-induced cognitive damage by modulating neuroinflammation and hippocampal plasticity. Eur. J. Pharmacol. 2022 doi: 10.1016/j.ejphar.2022.175431. [DOI] [PubMed] [Google Scholar]
  • 97.Liu L., Du X., Yang Q., Li M., Ran Q., Liu Q., Yang L., Sun L., Guo Y., Li Y., Chen Y., Zhu X., Li Q. Ginsenoside Rg1 promotes remyelination and functional recovery in demyelinating disease by enhancing oligodendrocyte precursor cells-mediated myelin repair. Phytomedicine. 2022;106 doi: 10.1016/j.phymed.2022.154309. [DOI] [PubMed] [Google Scholar]
  • 98.Liu Z.-J., Zhao M., Zhang Y., Xue J.-F., Chen N.-H. Ginsenoside Rg1 promotes glutamate release via a calcium/calmodulin-dependent protein kinase II-dependent signaling pathway. Brain Res. 2010;1333:1–8. doi: 10.1016/j.brainres.2010.03.096. [DOI] [PubMed] [Google Scholar]
  • 99.Sun W., Zhang F., Wang H., Wang C., Zhou Z., Zhou Y. Ginsenoside Rg1 fails to rescue PTSD-like behaviors in a mice model of single-prolonged stress. Biochem. Biophys. Res. Commun. 2020;528:243–248. doi: 10.1016/j.bbrc.2020.05.159. [DOI] [PubMed] [Google Scholar]
  • 100.Guo Y., Wang L.-P., Li C., Xiong Y.-X., Yan Y.-T., Zhao L.-Q., Li S.-D., Sun J., Luo H.-Y., Xian C.J. Effects of ginsenoside Rb1 on expressions of phosphorylation akt/phosphorylation mTOR/phosphorylation PTEN in artificial abnormal hippocampal microenvironment in rats. Neurochem. Res. 2018;43:1927–1937. doi: 10.1007/s11064-018-2612-x. [DOI] [PubMed] [Google Scholar]
  • 101.Zhang B., Hata R., Zhu P., Sato K., Wen T.-C., Yang L., Fujita H., Mitsuda N., Tanaka J., Samukawa K., Maeda N., Sakanaka M. Prevention of ischemic neuronal death by intravenous infusion of a ginseng saponin, ginsenoside Rb(1), that upregulates Bcl-x(L) expression. J. Cerebr. Blood Flow Metabol. 2006;26:708–721. doi: 10.1038/sj.jcbfm.9600225. [DOI] [PubMed] [Google Scholar]
  • 102.Tohda C., Matsumoto N., Zou K., Meselhy M.R., Komatsu K. Abeta(25-35)-induced memory impairment, axonal atrophy, and synaptic loss are ameliorated by M1, A metabolite of protopanaxadiol-type saponins. Neuropsychopharmacology. 2004;29:860–868. doi: 10.1038/sj.npp.1300388. [DOI] [PubMed] [Google Scholar]
  • 103.Bae M.-Y., Cho J.-H., Choi I.-S., Park H.-M., Lee M.-G., Kim D.-H., Jang I.-S. Compound K, a metabolite of ginsenosides, facilitates spontaneous GABA release onto CA3 pyramidal neurons. J. Neurochem. 2010;114:1085–1096. doi: 10.1111/j.1471-4159.2010.06833.x. [DOI] [PubMed] [Google Scholar]
  • 104.Hou J.-G., Xue J.-J., Lee M.-R., Sun M.-Q., Zhao X.-H., Zheng Y.-N., Sung C.-K. Compound K is able to ameliorate the impaired cognitive function and hippocampal neurogenesis following chemotherapy treatment. Biochem. Biophys. Res. Commun. 2013;436:104–109. doi: 10.1016/j.bbrc.2013.05.087. [DOI] [PubMed] [Google Scholar]
  • 105.Yang Q., Lin J., Zhang H., Liu Y., Kan M., Xiu Z., Chen X., Lan X., Li X., Shi X., Li N., Qu X. Ginsenoside compound K regulates amyloid β via the nrf2/keap 1 signaling pathway in mice with scopolamine hydrobromide-induced memory impairments. J. Mol. Neurosci. 2019;67:62–71. doi: 10.1007/s12031-018-1210-3. [DOI] [PubMed] [Google Scholar]
  • 106.Zong W., Zeng X., Chen S., Chen L., Zhou L., Wang X., Gao Q., Zeng G., Hu K., Ouyang D. Ginsenoside compound K attenuates cognitive deficits in vascular dementia rats by reducing the Aβ deposition. J. Pharmacol. Sci. 2019;139:223–230. doi: 10.1016/j.jphs.2019.01.013. [DOI] [PubMed] [Google Scholar]
  • 107.Liu Q., Liu L., Liu H., Jiang J., Guo S., Wang C., Jia Y., Tian Y. Compound K attenuated hepatectomy-induced post-operative cognitive dysfunction in aged mice via LXRα activation. Biomed. Pharmacother. 2019;119 doi: 10.1016/j.biopha.2019.109400. [DOI] [PubMed] [Google Scholar]
  • 108.Jiao H., Jia J. Ginsenoside compound K acts via LRP1 to alleviate Amyloid β42-induced neuroinflammation in microglia by suppressing NF-κB. Biochem. Biophys. Res. Commun. 2022;590:14–19. doi: 10.1016/j.bbrc.2021.12.071. [DOI] [PubMed] [Google Scholar]
  • 109.Lin T., Liu Y., Shi M., Liu X., Li L., Liu Y., Zhao G. Promotive effect of ginsenoside Rd on proliferation of neural stem cells in vivo and in vitro. J. Ethnopharmacol. 2012;142:754–761. doi: 10.1016/j.jep.2012.05.057. [DOI] [PubMed] [Google Scholar]
  • 110.Liu J., Yan X., Li L., Zhu Y., Qin K., Zhou L., Sun D., Zhang X., Ye R., Zhao G. Ginsennoside rd attenuates cognitive dysfunction in a rat model of Alzheimer's disease. Neurochem. Res. 2012;37:2738–2747. doi: 10.1007/s11064-012-0866-2. [DOI] [PubMed] [Google Scholar]
  • 111.Zhang X., Shi M., Ye R., Wang W., Liu X., Zhang G., Han J., Zhang Y., Wang B., Zhao J., Hui J., Xiong L., Zhao G. Ginsenoside Rd attenuates tau protein phosphorylation via the PI3K/AKT/GSK-3β pathway after transient forebrain ischemia. Neurochem. Res. 2014;39:1363–1373. doi: 10.1007/s11064-014-1321-3. [DOI] [PubMed] [Google Scholar]
  • 112.Yan X., Hu G., Yan W., Chen T., Yang F., Zhang X., Zhao G., Liu J. Ginsenoside Rd promotes non-amyloidogenic pathway of amyloid precursor protein processing by regulating phosphorylation of estrogen receptor alpha. Life Sci. 2017;168:16–23. doi: 10.1016/j.lfs.2016.11.002. [DOI] [PubMed] [Google Scholar]
  • 113.Liu J., Yan X., Li L., Li Y., Zhou L., Zhang X., Hu X., Zhao G. Ginsenoside rd improves learning and memory ability in APP transgenic mice. J. Mol. Neurosci. 2015;57:522–528. doi: 10.1007/s12031-015-0632-4. [DOI] [PubMed] [Google Scholar]
  • 114.Wan Q., Ma X., Zhang Z.-J., Sun T., Xia F., Zhao G., Wu Y.-M. Ginsenoside reduces cognitive impairment during chronic cerebral hypoperfusion through brain-derived neurotrophic factor regulated by epigenetic modulation. Mol. Neurobiol. 2017;54:2889–2900. doi: 10.1007/s12035-016-9868-4. [DOI] [PubMed] [Google Scholar]
  • 115.Wang H., Jiang N., Lv J., Huang H., Liu X. Ginsenoside Rd reverses cognitive deficits by modulating BDNF-dependent CREB pathway in chronic restraint stress mice. Life Sci. 2020;258 doi: 10.1016/j.lfs.2020.118107. [DOI] [PubMed] [Google Scholar]
  • 116.Han S.-K., Joo M.-K., Kim J.-K., Jeung W., Kang H., Kim D.-H. Bifidobacteria-fermented red ginseng and its constituents ginsenoside rd and protopanaxatriol alleviate anxiety/depression in mice by the amelioration of gut dysbiosis. Nutrients. 2020;12:901. doi: 10.3390/nu12040901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.B L., I S., H L., Dh H. Effect of ginsenoside Re on depression- and anxiety-like behaviors and cognition memory deficit induced by repeated immobilization in rats. J. Microbiol. Biotechnol. 2012;22 doi: 10.4014/jmb.1112.12046. [DOI] [PubMed] [Google Scholar]
  • 118.Wang H., Lv J., Jiang N., Huang H., Wang Q., Liu X. Ginsenoside Re protects against chronic restraint stress-induced cognitive deficits through regulation of NLRP3 and Nrf2 pathways in mice. Phytother Res. 2021 doi: 10.1002/ptr.6947. [DOI] [PubMed] [Google Scholar]
  • 119.Liu Y.-W., Zhu X., Li W., Lu Q., Wang J.-Y., Wei Y.-Q., Yin X.-X. Ginsenoside Re attenuates diabetes-associated cognitive deficits in rats. Pharmacol. Biochem. Behav. 2012;101:93–98. doi: 10.1016/j.pbb.2011.12.003. [DOI] [PubMed] [Google Scholar]
  • 120.Shi J., Xue W., Zhao W., Li K. Pharmacokinetics and dopamine/acetylcholine releasing effects of ginsenoside Re in hippocampus and mPFC of freely moving rats. Acta Pharmacol. Sin. 2013;34:214–220. doi: 10.1038/aps.2012.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Tran T.-V., Shin E.-J., Dang D.-K., Ko S.K., Jeong J.H., Nah S.-Y., Jang C.-G., Lee Y.J., Toriumi K., Nabeshima T., Kim H.-C. Ginsenoside Re protects against phencyclidine-induced behavioral changes and mitochondrial dysfunction via interactive modulation of glutathione peroxidase-1 and NADPH oxidase in the dorsolateral cortex of mice. Food Chem. Toxicol. 2017;110:300–315. doi: 10.1016/j.fct.2017.10.019. [DOI] [PubMed] [Google Scholar]
  • 122.Bt N., Ej S., Jh J., N S., Sy N., Sk K., Jk B., Y L., Xg L., Dj K., T N., Hc K. Ginsenoside Re attenuates memory impairments in aged Klotho deficient mice via interactive modulations of angiotensin II AT1 receptor, Nrf2 and GPx-1 gene. Free Radical Biol. Med. 2022;189 doi: 10.1016/j.freeradbiomed.2022.07.003. [DOI] [PubMed] [Google Scholar]
  • 123.Shin E.-J., Nguyen B.T., Sharma N., Tran N.K.C., Nguyen Y.N.D., Hwang Y., Park J.H., Nah S.-Y., Ko S.K., Byun J.K., Lee Y., Kim D.-J., Jeong J.H., Kim H.-C. Ginsenoside Re mitigates memory impairments in aged GPx-1 KO mice by inhibiting the interplay between PAFR, NFκB, and microgliosis in the hippocampus. Food Chem. Toxicol. 2023 doi: 10.1016/j.fct.2023.113627. [DOI] [PubMed] [Google Scholar]
  • 124.Cui J., Jiang L., Xiang H. Ginsenoside Rb3 exerts antidepressant-like effects in several animal models. J. Psychopharmacol. 2012;26:697–713. doi: 10.1177/0269881111415735. [DOI] [PubMed] [Google Scholar]
  • 125.Du Y., Fu M., Wang Y.T., Dong Z. Neuroprotective effects of ginsenoside Rf on amyloid-β-induced neurotoxicity in vitro and in vivo. J Alzheimers Dis. 2018;64:309–322. doi: 10.3233/JAD-180251. [DOI] [PubMed] [Google Scholar]
  • 126.Fan Y., Wang N., Rocchi A., Zhang W., Vassar R., Zhou Y., He C. Identification of natural products with neuronal and metabolic benefits through autophagy induction. Autophagy. 2017;13:41–56. doi: 10.1080/15548627.2016.1240855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Cui J., Shan R., Cao Y., Zhou Y., Liu C., Fan Y. Protective effects of ginsenoside Rg2 against memory impairment and neuronal death induced by Aβ25-35 in rats. J. Ethnopharmacol. 2021;266 doi: 10.1016/j.jep.2020.113466. [DOI] [PubMed] [Google Scholar]
  • 128.Gao Z.-W., Ju R.-L., Luo M., Wu S.-L., Zhang W.-T. The anxiolytic-like effects of ginsenoside Rg2 on an animal model of PTSD. Psychiatr. Res. 2019;279:130–137. doi: 10.1016/j.psychres.2018.12.034. [DOI] [PubMed] [Google Scholar]
  • 129.Zhang J.-J., Chen K.-C., Zhou Y., Wei H., Qi M.-H., Wang Z., Zheng Y.-N., Chen R.-X., Liu S., Li W. Evaluating the effects of mitochondrial autophagy flux on ginsenoside Rg2 for delaying D-galactose induced brain aging in mice. Phytomedicine. 2022;104 doi: 10.1016/j.phymed.2022.154341. [DOI] [PubMed] [Google Scholar]
  • 130.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. doi: 10.1016/j.jep.2007.10.026. [DOI] [PubMed] [Google Scholar]
  • 131.Lee B., Sur B., Park J., Kim S.-H., Kwon S., Yeom M., Shim I., Lee H., Hahm D.-H. Ginsenoside rg3 alleviates lipopolysaccharide-induced learning and memory impairments by anti-inflammatory activity in rats. Biomol Ther (Seoul) 2013;21:381–390. doi: 10.4062/biomolther.2013.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Jang S.K., Yu J.M., Kim S.T., Kim G.H., Park D.W., Lee D.I., Joo S.S. An Aβ42 uptake and degradation via Rg3 requires an activation of caveolin, clathrin and Aβ-degrading enzymes in microglia. Eur. J. Pharmacol. 2015;758:1–10. doi: 10.1016/j.ejphar.2015.03.071. [DOI] [PubMed] [Google Scholar]
  • 133.Kim J., Shim J., Lee S., Cho W.-H., Hong E., Lee J.H., Han J.-S., Lee H.J., Lee K.W. Rg3-enriched ginseng extract ameliorates scopolamine-induced learning deficits in mice. BMC Compl. Alternative Med. 2016;16:66. doi: 10.1186/s12906-016-1050-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Sur B., Lee B. Ginsenoside Rg3 modulates spatial memory and fear memory extinction by the HPA axis and BDNF-TrkB pathway in a rat post-traumatic stress disorder. J. Nat. Med. 2022;76:821–831. doi: 10.1007/s11418-022-01636-z. [DOI] [PubMed] [Google Scholar]
  • 135.Chu S., Gu J., Feng L., Liu J., Zhang M., Jia X., Liu M., Yao D. Ginsenoside Rg5 improves cognitive dysfunction and beta-amyloid deposition in STZ-induced memory impaired rats via attenuating neuroinflammatory responses. Int. Immunopharm. 2014;19:317–326. doi: 10.1016/j.intimp.2014.01.018. [DOI] [PubMed] [Google Scholar]
  • 136.Choi S.-Y., Kim K.-J., Song J.-H., Lee B.-Y. Ginsenoside Rg5 prevents apoptosis by modulating heme-oxygenase-1/nuclear factor E2-related factor 2 signaling and alters the expression of cognitive impairment-associated genes in thermal stress-exposed HT22 cells. J Ginseng Res. 2018;42:225–228. doi: 10.1016/j.jgr.2017.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wang Y.-Z., Chen J., Chu S.-F., Wang Y.-S., Wang X.-Y., Chen N.-H., Zhang J.-T. Improvement of memory in mice and increase of hippocampal excitability in rats by ginsenoside Rg1's metabolites ginsenoside Rh1 and protopanaxatriol. J. Pharmacol. Sci. 2009;109:504–510. doi: 10.1254/jphs.08060fp. [DOI] [PubMed] [Google Scholar]
  • 138.Hou J., Xue J., Lee M., Yu J., Sung C. Long-term administration of ginsenoside Rh1 enhances learning and memory by promoting cell survival in the mouse hippocampus. Int. J. Mol. Med. 2014;33:234–240. doi: 10.3892/ijmm.2013.1552. [DOI] [PubMed] [Google Scholar]
  • 139.Lu C., Wang Y., Lv J., Jiang N., Fan B., Qu L., Li Y., Chen S., Wang F., Liu X. Ginsenoside Rh2 reverses sleep deprivation-induced cognitive deficit in mice. Behav. Brain Res. 2018;349:109–115. doi: 10.1016/j.bbr.2018.03.005. [DOI] [PubMed] [Google Scholar]
  • 140.Hou J., Xue J., Lee M., Liu L., Zhang D., Sun M., Zheng Y., Sung C. Ginsenoside Rh2 improves learning and memory in mice. J. Med. Food. 2013;16:772–776. doi: 10.1089/jmf.2012.2564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Lv J., Lu C., Jiang N., Wang H., Huang H., Chen Y., Li Y., Liu X. Protective effect of ginsenoside Rh2 on scopolamine-induced memory deficits through regulation of cholinergic transmission, oxidative stress and the ERK-CREB-BDNF signaling pathway. Phytother Res. 2021;35:337–345. doi: 10.1002/ptr.6804. [DOI] [PubMed] [Google Scholar]
  • 142.Shi L.-S., Ji C.-H., Liu Y., Gu J.-H., Tang W.-Q., Zhang W., Guan W. Ginsenoside Rh2 administration produces crucial antidepressant-like effects in a CUMS-induced mice model of depression. Brain Behav. 2022;12 doi: 10.1002/brb3.2705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.She L., Xiong L., Li L., Zhang J., Sun J., Wu H., Ren J., Wang W., Zhao X., Liang G. Ginsenoside Rk3 ameliorates Aβ-induced neurotoxicity in APP/PS1 model mice via AMPK signaling pathway. Biomed. Pharmacother. 2023;158 doi: 10.1016/j.biopha.2022.114192. [DOI] [PubMed] [Google Scholar]
  • 144.Lee K.-W., Jung S.Y., Choi S.-M., Yang E.J. Effects of ginsenoside Re on LPS-induced inflammatory mediators in BV2 microglial cells. BMC Compl. Alternative Med. 2012;12:196. doi: 10.1186/1472-6882-12-196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Li Y., Wang F., Luo Y. Ginsenoside Rg1 protects against sepsis-associated encephalopathy through beclin 1-independent autophagy in mice. J. Surg. Res. 2017;207:181–189. doi: 10.1016/j.jss.2016.08.080. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data availability is not applicable to this article as no new data were created or analyzed in this study.


Articles from Heliyon are provided here courtesy of Elsevier

RESOURCES