Zum Hauptinhalt springen

Antioxidant Activity of Panax ginseng to Regulate ROS in Various Chronic Diseases

Md Niaj Morshed ; Jong Chan Ahn ; et al.
In: Applied Sciences, Jg. 13 (2023-02-23), S. 2893-2893
Online unknown

Antioxidant Activity of Panax ginseng to Regulate ROS in Various Chronic Diseases 

Reactive oxygen species (ROS)-the byproduct of regular cell activity formed by various cellular components—play a significant role in pathological and physiological conditions. Alternatively, antioxidants are compounds that reduce or scavenge reactive species in cells. An asymmetry between the antioxidant defense system and ROS from intracellular and extracellular sources cause chronic diseases such as cancer, inflammation, tumorigenesis, cardiovascular and neurogenerative diseases. However, Panax ginseng and its secondary metabolites (known as ginsenosides, phenolic compounds, peptides, acid polysaccharides, polyacetylene, and alkaloids) are well-recognized as antioxidants in many in vitro and in vivo experiments which show beneficial activity in regulating ROS in these diseases. There are extensive evidences that P. ginseng can destroy cancer cells specifically by increasing oxidative stress through ROS generation without significantly harming normal cells. Additionally, numerous studies have examined the antioxidant activity of ginseng and its derivatives on ROS-mediated signaling pathways which are discussed herein. This review summarizes the potential antioxidant activity of P. ginseng in several chronic diseases, and gives updated research evidence with related mechanisms and the future possibilities of nano-formulated compounds of P. ginseng and other polyphenols.

Keywords: ROS; antioxidant; oxidative stress; Panax ginseng; ginsenosides; secondary metabolites; chronic disease

1. Introduction

Ginseng comes from the genus Panax of the Araliaceae family with nine different species such as Panax ginseng (Korean ginseng), Panax notoginseng (Chinese ginseng), Panax japonicum (Japanese ginseng), and Panax quinquefolius (American ginseng) [[1]]. Among all ginseng, four types of P. ginseng can be categorized according to how they are processed, for example, fresh ginseng, white ginseng (air-dried), red ginseng (steamed), and sun ginseng [[2]]. The word "ginseng" is derived from the Chinese word "rénshēn" which means "human" as the roots of ginseng are shaped like the human leg [[3]]. Ginseng has long been recognized as 'the king of herbs' due to its ability to improve fitness and relax the mind [[4]]. P. ginseng is endemic to Korea and China and has been used in traditional treatment [[5]]. However, consumers from Korea and others prefer Korean ginseng due to its current medical research findings of being useful in boosting blood circulation and better cognition, acting as a mind booster, and it being supposed to bolster one's soul, increase the body's immune system, and control diabetes, along with possessing anti-aging and anticancer properties [[6]]. P. ginseng contains a vast amount of secondary metabolites such as phenolic acids (gallic acid, caffeic acid, coumaric acid, salicylic acid, cinnamic acid, maltol, etc.), flavonoids, acid polysaccharides, amino acids, phytosterol, carbohydrates, minerals, ginseng oil, and certain vitamins [[4], [8]]. P. ginseng has attracted the interest of researchers worldwide due to its pharmacological efficacy and potent medical applications.

Instead of entire ginseng and other components, a majority of researches have been conducted on specific ginsenosides to treat a variety of medical problems [[9]]. Ginsenosides are mainly triterpene saponins of ginseng. To date, more than 218 ginsenosides (major types: Rc, Rb1, Rb2, Rg1, Rd, and Re; minor types: Rh1, Rh2, and Rg3) have been identified from different parts of ginseng (leaves, roots, berries, and flower buds) and these metabolites have become popular for research. Ginsenosides are the therapeutically active components obtained from ginseng and are widely recognized for their oxidative stress [[10]], apoptosis [[11]], inflammation [[12]], angiogenesis [[13]], anticancer [[14]], and cancer metastatic properties associated with cell proliferation. Ginsenosides are divided into two groups based on the glycon structure: oleanane and dammarane [[15]]. According to the chemical structure, dammarane-type ginsenosides can be further classified into two categories: protopanaxadiol (PPD) and protopanaxatriol (PPT) [[16]], whereas the minor categories depending on aglycone moieties include ocotillol and oleanane [[17]]. Ginsenosides Rc, Rb2, Rh2, Rg3, Rh4, Ck, Rk1, Rk3, and Rd are strong bioactive components that have been shown to greatly inhibit the proliferation of cancer cells by regulating ROS in mitochondria [[18]]. The following are listed in decreasing order of how well ginsenosides scavenge intracellular ROS: Rb2 > Rc > Rg2 > Rh2 > Rh1 > Rf > Rg3 > Rg1 > Rb1 > Re > Rd [[19]]. Different studies have shown that the transformation of ginseng referred to as ginsenosides have stronger activity than crude ginseng [[20]].

P. ginseng and ginsenosides have excellent ROS-regulating activity in various disease families such as sensor impairment, cardiovascular diseases, neurogenerative diseases, cancer, diabetes, inflammation, and vice-versa. This review was designed to investigate the antioxidant properties of P. ginseng and ginsenosides regulating ROS in different chronic diseases.

2. Research Methodology

Existing knowledge was incorporated to prepare a review on the ROS-regulating activity of P. ginseng and its secondary metabolites in different chronic diseases. The relevant information was gathered by looking through articles published in the MDPI, Elsevier, Taylor and Francis, Wiley, Springer, Google Scholar, PubMed, and NCBI databases between 1996 and January 2023.

3. ROS, Oxidative Stress, and Antioxidants

Reactive oxygen species (ROS) including hydrogen peroxide (H2O2), superoxide (O2∙), and hydroxyl (HO∙) radicals were initially recognized as potentially hazardous by-products; they are now acknowledged to serve significant roles as secondary messengers in numerous intercellular pathways [[21]]. ROS are produced during ATP production by the electron transport chain and NADPH (nicotinamide adenine dinucleotide phosphate) oxidase system [[22]]. Moreover, our bodies usually produce huge amounts of ROS due to our daily lifestyles including extended working circumstances, sitting for a long time, wearing restrictive clothing, using illicit substances often, eating unhealthily, and smoking or drinking too much alcohol [[23]]. ROS positively impact on immunological activity and intracellular signaling at mild to moderate levels [[24]]. A higher concentration of ROS can cause oxidative stress, DNA damage, redox homeostasis, tumor progression, and drug resistance which are related to the development of various diseases. ROS play a crucial role in cell proliferation, differentiation, and the control of signal transduction at certain levels (Figure 1) [[22]]. According to Sies (1985), "Oxidative stress is defined as the imbalance of pro-oxidant and the antioxidant protective capacity that promotes ROS or RNS which might cause potential damage" [[25]]. Undeniably, oxidative stress is associated with more than 100 diseases as a source or outcome [[26]]. It is well known that oxidative stress leads to cell death by damaging important bio-compounds such as proteins, DNA, and lipids [[28]]. Oxidative stress acts as a contributor to many chronic diseases such as cancer, neuro-generative disease, inflammation, cardiovascular disease, etc. [[29]].

On the other hand, an antioxidant is a compound that reduces or scavenges reactive species or blocks the oxidation in cells [[30]]. In other words, antioxidants have the power to stop or delay the oxidation reaction to regulate the excessive production of oxidants [[31]]. Thiols and polyphenols are common examples of antioxidants for their reducing behavior [[32]]. Plants and animals consist of two types of antioxidants: non-enzymatic (vitamin E, C, carotenoids, lipoic acid, and others) and enzymatic (catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), Glutathione-S-Transferase (GST), Glutathione reductase (GR), etc.). These enzymatic antioxidants have important functions in regulating cell homeostasis [[33]]. In a nutshell, the antioxidant mechanisms are (a) inhibiting the production of reactive species, (b) scavenging oxidants, (c) restoring the damaged molecule, (d) blocking the formation of harmful secondary metabolites and inflammation mediators, and (e) developing and boosting the natural antioxidant defense system. These defensive mechanisms work together to prevent oxidative stress in the body [[34]]. However, in redox biology, superoxide dismutase quickly turns into H2O2 and O2 via the SOD enzyme. The Fenton reaction can decrease metal ions to produce OH. from H2O2, which is issued in systemic inflammation [[35]]. OH. are reactive and so damages macromolecules. Antioxidant enzymes (catalase, glutathione peroxidase) can detoxify H2O2 to avoid the production of OH. (Figure 2).

Natural antioxidants have numerous biological effects including preventing ROS production and inhibiting free radicals [[37]]. The administration of natural sources of an antioxidant such as ginseng, pomegranate, curcumin, sesame, garlic, peppermint, and olive leaves demonstrated beneficial effects on ROS-mediated diseases in both animal and human studies [[38]].

4. Pathways Related to Oxidative Stress in Cells

There are multiple mechanisms involved in the normal cellular process. These mechanisms produce antioxidant enzymes such as SOD, CAT, GPx, HO-1, and G-S-T to inhibit the production of ROS which protect the cell from damage by maintaining antioxidant/oxidation balance [[20]]. Several pathways including NF-κB, Keap1/Nrf2/ARE, Wnt/β catenin, and PI3k/Akt are the most important for regulating the redox balance.

NF-κB is an essential nuclear transcription factor that regulates innate immunity, tumor progression, and inflammatory responses [[40]]. Excessive ROS production activates NF-κB by activating the MMP [[41]]. NF-κB is an important factor in the expression of COX-2, iNOS, and cytokines [[42]]. Cox-2 generates prostaglandin E2 in the cell which causes skin cancer and iNOS produces nitric oxide that leads to inflammation [[43]]. However, the suppression of NF-κB activity may reduce inflammation and oxidative stress [[31]].

The Keap1/Nrf2/ARE (kelch-like epoxychloropropqane-related protein 1/nuclear factor erythroid2-related factor 2/antioxidant response element) signaling pathway plays a vital role in the improvement in oxidative stress [[44]]. Nrf2 is an important transcription factor that protects the cell from oxidative stress [[45]]. In extreme antioxidant stress conditions, Nrf2 dissociates from keap1 and binds with the ARE which down-regulates antioxidant enzyme production, resulting in DNA damage, apoptosis, autophagy, etc. [[46]].

Furthermore, the Wnt signaling pathway is crucial for numerous basic processes of embryonic development and the homeostasis of normal cells [[47]]. The Wnt/β-catenin pathway is generally composed of β catenin, glycogen synthase kinase-3 (gsk-3), and casein kinase-1 (CK-1) proteins. In normal physiological conditions, gsk-3β and CK-1 phosphorylate β-catenin, the ligase complex of E3-ubiquitin, specifically targets β-catenin for destruction and ubiquitination. During oxidative stress, β-catenin is not decomposed, leading to the activation of the Wnt/β-catenin pathway [[48]].

In addition, the PI3k/Akt signaling pathway is also essential in the mechanism of oxidative stress which controls cell proliferation and cell apoptosis [[49]]. The stimulation of Akt activity helps to regulate apoptosis by enhancing Bcl-2 and reducing Bax and caspase-3 expression. Many researchers have shown that the PI3K/Akt signaling pathway offers antioxidative activity which inhibits apoptosis and mitigates ischemia-reperfusion injury [[50]]. However, excessive ROS production blocks the PI3k/Akt, resulting in tumorigenesis [[51]] (Figure 3).

5. Roles of P. ginseng in the Oxidative-Stress-Related Signaling Pathways

It has been reported that P. ginseng and its derivatives are some of the antioxidant-rich sources involved in the regulation of many oxidative-stress-related pathways [[20]]. Ginseng and ginsenosides have antioxidant activity, inhibiting oxidative stress, and their preventive effects are explained by their ability to scavenge ROS. Several in vitro and in vivo experiments have depicted that P. ginseng and ginsenosides increase antioxidant enzymes activity to inhibit ROS. P. ginseng showed a barrier role against oxidative stress in several cell lines. Furthermore, the supplementation of P. ginseng increases antioxidant enzymes and reduces the generation of ROS and MDA in various tissues such as heart, lung, kidney, and liver in animal models.

Several studies have shown that ginsenoside Rg1 can stimulate the Nrf2/ARE pathway and mitigate neuronal injury prompted by ischemia reperfusion (I/R) [[52]]. Rg1 prevents the inflammation and oxidative stress of diabetic rats by inducing the Keap1/Nrf2 pathway, increases the survival rate of cells, and also minimizes the excessive ROS and cell apoptosis by increasing the Nrf1/HO-1 pathway [[53]]. Similarly, the administration of ginsenoside Rd reduces serum CK and LDH and induces HO-1 and Nrf2 expression to protect against myocardial I/R injury [[54]]. In addition, by activating the Nrf2 pathway, ginsenoside Rb1 increases GSH levels and decreases MDA content. This antioxidant regulatory activity of Rb1 relieves diabetic retinopathy [[55]]. Moreover, Rb1 enhances antioxidant enzymes activity that reduce the MDA level in spinal-cord-injured (SCI) rats by stimulating the Nrf2/HO-1 signaling pathway [[57]]. In addition, ginsenoside Rg1 remarkably improves SOD and GSH contents, inhibits MDA production and ROS levels, and exerts antioxidant and anti-inflammatory activity via the activation of the Nrf2/HO-1 signaling pathway [[58]]. 20(S)-Rg3 and 20(R) compounds induce the expression of antioxidant enzymes and protect H2O2-mediated myocardial cell injury through the activation of the Nrf2/HO-1 pathway [[59]]. Along with these reports, ginsenoside Rg3 inhibits oxidative damage and apoptosis via activating Nrf2/ARE through Akt activation [[60]]. Ginsenoside compound K (CK) inhibits diabetic nephropathy via inhibiting ROS generation and inflammatory cytokines IL-1β through the upregulation of the NF-κB/p38 signaling pathway [[61]]. Both ginsenoside Rb1 and Rg1 increase the activity of catalase and reduce ROS formulation in case of inhibiting myocardial oxidation [[62]]. Ginsenoside Rg1 induces SOD and GSH-x enzyme activity and reduces the ROS and MDA level to attenuate oxidative-stress-mediated aging and the Wnt/β-catenin pathway.

Similarly, red ginseng (RG) also provides significant antioxidant activity in several disease cases such as diabetes, skin cancer, hepatic and nerve disorder, etc. RG enhances the antioxidant activity and reduces the oxidative stress that is exposed in Table 1.

6. P. ginseng in ROS-Mediated Diseases

In mammalian cells, mitochondria are significant for pathophysiological processes including oxidative phosphorylation (OXPHOS), the formation of cell development, and mediating key events that determine cell functions and states. According to some reviews, many diseases such as I/R injury, cardiovascular diseases, neurogenerative diseases, cancer, and metabolic disorders have been linked to mitochondrial dysfunction [[73]]. Several studies have demonstrated that P. ginseng regulates mitochondrial ROS, apoptosis, dynamics, biogenesis, and mitophagy to have a pharmaceutical impact. The overexpression of mitochondrial ROS leads to a wide variety of disorders and many of them lead to causes of death [[74]] which have been summarized below.

6.1. Antioxidant Activities of P. ginseng in Sensor Impairment

6.1.1. Ototoxicity

Age-related hearing loss is considered to be an ROS-mediated disorder. Other causes of hearing loss are noise, antibiotics (Aminoglycoside, cisplatin) consumption, and immune-mediated hearing loss [[75]]. Many studies have exhibited that ginseng is helpful to prevent ototoxicity caused by different sources. Aminoglycosides including gentamicin react with iron in the inner ear and produce ROS with damage to hair cells and neurons. Choung et al. showed that ginsenoside Rb1 and Rb2 are effective against aminoglycoside-induced hearing loss by attenuating ROS generation and IL-6 inhibition [[76]]. The organ of Corti reaches its maximum intensities of ROS and RNS generation after seven to ten days of noise insult [[77]]. Additionally, ginsenoside Ck and Rg2 have therapeutic effects against noise-induced hearing loss in mice by reducing the levels of ROS and RNS [[78]]. Ginseng extract protects against cisplatin-induced ototoxicity of the auditory cell line (HEI-OC1) due to its anti-apoptotic and anti-oxidative stress effects [[79]].

6.1.2. Ocular Disease

It is believed that oxidative stress is involved in many age-related eye illnesses including retinal degeneration and cataract, glaucoma, and diabetes retinopathy, etc.; cataract is an age-related loss of transparency of the eye lens because of the formation of protein complexes in the lens [[80], [82]]. ROS and ultraviolet radiations damage crystalline proteins during aging, resulting in the insoluble protein clumps of the lens being opaque which interferes with vision. Park et al. recognized that ginsenoside CK blocked ROS production via Nrf2/HO-1 activation in the H2O2-stimulated ARPE-19 cell line to prevent cataracts [[82]]. After cataracts, glaucoma is the second leading reason for blindness which is associated with intra-ocular pressure and a loss of vision [[83]]. An overproduction of ROS leads to apoptosis in retinal ganglion cells that cause glaucoma [[84]]. Several studies showed evidence that ginseng supplements and ginsenosides are very useful against glaucoma. Ginsenoside Rb1 protects retinal ganglion cells against apoptosis caused by H2O2-induced oxidative stress [[85]]. Moreover, patients with glaucoma who consumed 3 g of Korean red ginseng daily for 4 weeks observed an improvement in their daytime visual acuity and ocular pain [[86]]. Eight weeks of consumption of KRG reduces the symptoms of dry eye in glaucoma patients by improving the tear film stability [[87]].

6.2. Antioxidant Activities of P. ginseng in Neurogenerative Diseases

The central nervous system (CNS) is extremely vulnerable to oxidative injury due to its utilization of a high pace of oxygen [[88]]. An overproduction of ROS and inadequate antioxidant defense systems have been connected to the pathophysiology of numerous neurogenerative disorders such as Huntington's disease (HD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Alzheimer's disease (AD) [[89]]. Neuroprotection inhibits or delays the neurogenerative process to minimize neuronal death [[90]]. In this case, secondary metabolites of plants which are rich in antioxidant content can protect the nerve cells from free-radical-induced oxidative stress to prevent neurogenerative diseases [[91]]. The research into and recognition of the potential impact of Panax ginseng on ROS-mediated neurogenerative diseases are growing day by day.

6.2.1. Parkinson's Disease

Parkinson's disease (PD) is a chronic neurodegenerative condition that affects approximately 2% of people over 60 years old worldwide. PD depends on the interplay between various genetic and environmental factors and is marked by the development and accumulation of misfolded-α-synuclein [[90]]. The hallmark symptoms of PD include motor disorders (rigidity, tremor, and bradykinesia) and non-motor disorders (depression, sleep disturbance, and autonomic dysfunction) resulting from the gradual deterioration of the dopaminergic pathway [[92]]. Several studies have depicted that ginseng and its bioactive components, ginsenosides, have therapeutic actions on PD. P. ginseng extracts can inhibit ROS generation, eliminate Bax/Bcl2, increase the cytochrome C release, and stimulate caspase-3 expression to alleviate cell death [[93]]. Ginsenoside Rg1 suppressed the oxidative stress to mediate the neuroprotective action in MPTP (1-methyl-4-phenyl-1,2,3,6-twtrahydropyridine)-induced substantia nigra [[94]]. Rg1 activates total superoxide dismutase (SOD) and inhibits glutathione reduction, reducing c-Jun and N-terminal kinase (JNK) in the substantia nigra of C57BL/6 mice [[94]]. Furthermore, Rg1 decreases ROS production and mitochondrial cytochrome C and blocks the activation of caspase 3 and the formulation of the iNOS protein and NO in PC12 cells [[96]]. Again, Rg1 attenuates ROS generation and NF-ĸB translocation in MPP+-induced MES23.5 cells for reducing the expression of DMT1-IRE [[97]]. Ginsenoside Re shows neuroprotective action against the neurotoxicity of substantia nigra. Ginsenoside Re increases the Bcl-2 mRNA and Bcl-2 protein expression decreases the iNOS, Bax, and Bax mRNA and inhibits the cleavage of caspase-3 to protect the SN neuron from MPTP-induced apoptosis [[98]].

6.2.2. Alzheimer's Disease

Alzheimer's disease (AD) is a cognitive condition defined by the accumulation of senile plaques, the development of neurofibrillary tangle, and finally the death of neurons. The improper degradation of the amyloid precursor protein (APP) is the primary mechanism causing AD progression [[99]]. Several changes in molecular and cellular pathways including mitochondrial dysfunction, antioxidant decreases, oxidative stress increases, synaptic impairment, and amyloid Aβ clearance capacity are present in the AD brain [[101], [103]]. Many studies have distinguished that P. ginseng extract, powder, and ginsenosides were applied to AD in in vivo and in vitro studies. The total saponins of ginseng consumption for seven months revealed a remarkable reduction in memory loss by inhibiting oxidative stress and increasing the proteins associated with plasticity in aged mice [[104]]. Ginsenoside Rb1 shields neurons from Aβ1-42 neurotoxicity via an antioxidant mechanism [[105]]. Rb1 pre-treatment in PC12 cells for 1 day inhibits the overproduction of ROS and lipid peroxidation enhances the activation of caspase-3 and Bcl-2/bax for promoting cell survival [[106]]. In H2O2-induced PC12 cells, Rg1 prevents NF-κB/P65, ERK1/2, and Akt stimulation [[107]]. Rg1 can protect PC12 cells from cytotoxicity caused by Aβ25-35 by preventing β-secretase activities [[108]]. However, the ref. [[109]] experiment found for the first time that ginsenoside Rk3 can trigger the intracellular ROS level and Aβ-induced neuronal injury by stimulating the AMPK pathway and the upregulation of Nrf2. Interestingly, this study also confirmed that the pharmacological activity of ginsenoside Rk3 is better than the control drug donepezil in case of the treatment of AD. Recently, a new ginseng component gintonin has been discovered that is effective in reducing the severity of AD-related neuropathies [[110]].

6.2.3. Others

P. ginseng and its active components are also effective in Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), depression, neuroprotection, and improvement in cognition [[111]]. Wang et al. recognized that ginseng sesquiterpenoids down-regulate the NF-κB and BDNF/TrkB signaling pathways, and increase SOD production in the hippocampus of the ICR mice model. The result exhibited that the SP of ginseng shows antidepressant activity via Sirt1/NF-κB and BDNF/TrkB pathways [[112]]. Water extract of Korean red ginseng acts as a neuroprotector through the regulation of the Nrf2 signaling pathway [[113]]. Ginsenoside Rb1 exhibits strong antioxidant activity in the treatment of many neurological diseases including strokes [[114]]. Ginsenosides Rg3 and Rg1 are useful in cognition improvement via regulating the NF-κB and PI3K/Akt signaling pathways in mice models [[115]]. It has been reported that P. ginseng fibrous root (GFR) shows good antioxidant activity to scavenge free radicals. GFR enhanced the expression of antioxidant enzymes to trigger the intracellular ROS which ultimately accelerated Alzheimer's and other neurogenerative diseases [[117]].

6.3. Antioxidant Activities of P. ginseng in Cardiovascular Diseases

Cardiovascular disease (CVD) is a critical challenge among 50% of the world's population [[118]]. CVD includes hypertension, coronary artery disease, massive heart failure, and peripheral vascular disease, etc., which are common and affect newborns, children, and adults of both sexes [[119]]. Research shows that the enhancement of reactive oxygen species and oxygen consumption is one of the crucial factors in CVD outbreak. For example, heart failure can be caused by ROS-inducing cardiac apoptosis and necrosis [[120]], as well as increased oxidant generation via the NADH/NADPH oxidant, and superoxide-generated endothelium breakdown results in hypertension and coronary artery disease [[122]]; furthermore, an overproduction of ROS and oxidant-mediated myocyte apoptosis and necrosis causes myocardial infarction [[124]]. An excessive amount of free oxygen from heart ischemia causes myocardial damage; however, ginseng consumption increases blood flow by inhibiting free oxygen and myocardial damage [[126]]. Ginsenoside Rb1 can inhibit the production of ROS to reduce homocysteine which causes endothelial dysfunction [[127]]. Ginsenoside Re protects the myocardial cell from oxidative damage and increases myocardial cell viability in heart ischemia [[128]]. Total ginsenosides enhance coronary artery perfusion flow by activating the PI3K/Akt-eNOS signaling pathway that ultimately produces NO levels [[129]]. Ginsenoside-Rb1 administration increases eNOS expression which also increases NO levels and decreases super oxides in the porcine coronary artery via the vasodilating mechanism [[127]]. In addition, the saponin fractions of Korean red ginseng decrease blood pressure and prompt reflex tachycardia due to their hypotensive effect and the mechanism of NO donation [[130]]. In another study, total ginsenosides were effective against right ventricular hypertrophy, promoted systolic pressure, and reduced pulmonary pressure by controlling the ERK-1/MAPK signaling pathway [[131]]. Hong et al. demonstrated that the consumption of PPT-rich ginseng enhances the activation of eNOS, stimulates NO formulation, and improves the thickness of the vessel walls to attenuate hypertension [[132]]. Previous studies have depicted that ginsenoside Rg1 inhibited Bcl-2 and caspase-3 expression during myocardial infarction via ischemia to lower myocardial cell death and decreased left ventricular hypertrophy [[133]]. In another study, ginsenoside CK reduced the burden of myocardial infraction by increasing the protein kinase B(Akt) and nitrogen oxide synthetase (eNOS) followed by ischemia via the Akt/PI3K pathway [[134]]. Although it has also been reported that ginsenoside Rg3 mitigates myocardial ischemia-reperfusion injury (MIRI) via the AKT/eNOS and Bcl-2/Bax signaling pathways [[135]], ginsenoside Rd reduces MIRI via the Nrf-2/HO-1 pathway [[136]], and ginsenoside Rb1 inhibits cardiomyocyte autophagy via the PI3K/Akt/mTOR pathway, thus controlling MIRI [[137]]. Recently, ref. [[138]] confirmed that ginsenoside Rh2 provides anti-inflammatory and antioxidant activity on MIRI via the regulation of the Nrf2/HO-1/NLRP3 signaling pathway.

6.4. Antioxidant Activities of P. ginseng in Cancer

According to growing data, ROS are implicated in multiple steps of tumorigenesis from the initiation of the tumor to metastasis [[139]]. Excessive amounts of ROS in the cell or the defective antioxidant defense mechanisms quicken the cellular damage and initiate carcinogenesis [[140]]. It has been reported that the cancer cell initiates more ROS than their counterpart [[141]]. ROS play a dual role in cancer. First, the overabundance of ROS instigates autophagy, apoptosis, and cell cycle arrest signals [[142]]. Second, ROS can influence the initiation, growth, and transmission of cancer via the activation of the signaling pathways which affect cell proliferation, survivability, angiogenesis, and metastasis [[139]]. In general, when ROS are low to moderate, they may contribute to the initiation of a tumor, and a high amount of ROS causes massive cell damage and death, typically at the early stages of tumor formation [[144]]. So, it may be possible to destroy cancer cells specifically by raising oxidative stress through exogenous ROS production without significantly harming normal cells [[145]].

P. ginseng and its derivatives, ginsenosides, might be a promising supplemental therapy for cancer patients. According to growing researches, ginsenosides have a lot of potential to offer multiple strategies for treating different cancers (Table 2). Therefore, it might be difficult for cancer cells to develop resistance against ginsenosides. Additionally, ginsenosides are desirable candidates for new therapeutics due to their ability to destroy tumor cells. In general, ginsenosides can produce ROS in a variety of cancer cells which can enhance autophagy, apoptosis, paraptosis, and decrease cell proliferation in vivo and in vitro [[139]]. However, according to current research, by comparing between healthy and cancer cell lines (CCD841 and HT-29), ref. [[146]] confirmed that P. ginseng berry extract exhibited prebiotic activity against colorectal cancer by increasing ROS production without affecting the normal cells. It has also been newly proposed that ginsenoside Rh1 from P. ginseng is a potential compound to prevent and treat lung cancer through the regulation of metastasis and apoptosis [[147]]. P. ginseng extract suppresses ROS production by increasing the antioxidant biomarker GSH, and reduces the inflammatory biomarker TNF-α and apoptotic biomarker caspase-3 to prevent doxorubicin-induced cardiovascular disease [[148]].

6.5. Antioxidant Activities of P. ginseng in Other Diseases

ROS play a crucial role in the development of diabetes, kidney diseases, aging, etc. [[173]]. Mitochondrial DNA damaged by ROS is the primary cause of aging. Oxidative damage causes mitochondrial dysfunction and the translation and multiplication of mitochondrial DNA that promotes ROS production which damages mtDNA [[174]]. However, ginsenoside Rd consumption for one month quickens the cellular senescence to increase the antioxidant enzyme GPx and GR in mitochondria [[175]]. Moreover, ginsenoside Rb2 increases SOD, CAT activities, and also blood albumin which reduces oxidative stress from the skin cell [[176]]. According to Ramesh et al., Korean red ginseng reduces MDA levels, creatinine, AST, ALT, and urine nitrogen at higher levels Furthermore, KRG induces SOD, GPx, GST, GR, and CAT activity in the lungs and heart [[70]]. Additionally, P. ginseng derivative syringaresinol (SYR) shows antioxidant activity and stimulated autophagy in H2O2-induced Hacat cells, therefore inhibiting the mRNA expression of MMP-2 and MMP-9 related to skin aging [[177]]. SYR may have therapeutic potential to treat diabetic cardiomyopathy by reducing oxidative stress, fibrosis, and inflammation [[178]].

Furthermore, Korean red ginseng prevented the blood glucose levels in STZ-induced diabetic rats [[179]]. In addition, it was recognized that ginsenoside Rd consumption via acute renal failure in rats increases SOD and catalase in renal tissue and serum [[180]]. Recent studies have shown that ginseng triggers pro-inflammatory cytokine (IL-6, IL-1β, and TNF-α) expression as well as activates ROS-mediated pathways to show antifatigue activity through anti-oxidation and anti-inflammatory activity [[181]]. Research has shown that P. ginseng plays as anti-inflammatory, immunostimulatory, neuroprotective, hepatoprotective, antiplatelet, antidiabetic, and anti-angiogenesis roles. Korean ginseng and its ginsenosides are effective in various anti-inflammatory diseases including colitis, gastritis, and hepatitis. Han et al. depicted that ginseng shows anti-inflammatory activity by preventing Akt [[182]]. Moreover, Rg1 could be a useful approach for preventing acute liver damage by stimulating the Nrf2 signaling pathway [[183]]. Additionally, ginseng can regulate streptozotocin-induced diabetes by increasing antioxidant enzymes [[69]]. Moreover, ref. [[184]] depicted that fermented black ginseng (P. ginseng) can reduce ROS levels in H2O2-induced Hacat cells via its antioxidant activity compared to black and white ginseng. Furthermore, FBG shows higher anti-wrinkle and anti-melanogenic activity than BG and WG [[185]].

7. Nano-Formulation Forms of Phenolic Compounds as a Solid Alternative in Current Treatment M...

P. ginseng consists of approximately 80–90% organic and about 10–20% inorganic compounds. It has already been shown that the active ingredients of P. ginseng comprise a vast number of beneficial activities [[186]]. Several studies have discussed the pharmacological activities of P ginseng such as antioxidant, anti-inflammatory, anticancer, neuroprotective, cardioprotective, antidiabetic, anti-allergic, anti-stress, hypolipidemic, antifatigue, anti-depressive, antitumor, anti-adhesive, anti-aging, etc., which express the potentials of ginseng as a complementary and alternative medicine (CAM) [[187]]. Additionally, Acero et al. [[189]] demonstrated that P. ginseng is an ayurvedic medicine (AM), traditional medicine (TM), and also a traditional Chinese medicine (TCM). P. ginseng is most frequently employed in therapeutic applications due to its significant pharmacological activities [[190]]. Moreover, ginsenosides Rg, Re, and Rb are still exploited in Chinese pharmacopeia as molecular markers for quality assessment [[191]]. Along with ginsenosides, some essential phenolic compounds from P. ginseng have been reported such as syringic, ferulic, cinnamic, gentistic, and p-hydrobenzoic acid [[188]]. Researchers from a variety of disciplines have been interested in these active compounds [[192]]. However, these natural by-products have limited therapeutic efficacy due to high hydrophobicity, poor bioavailability, low in vivo stability, and short half-life. In fact, modification and conjugation (nano-formulation) have been hastening the study and the therapeutic use of these components [[193]]. A significant step forward in the effort to boost therapeutic efficacy by lowering toxicity, raising bioavailability, enhancing stability, and optimizing pharmacokinetics is the use of natural ingredients in nano-formulations [[194]]. In recent years, a wide range of nanomaterial-based delivery systems have been subjected to ginsenosides, curcumin, quercetin, resveratrol, ferulic acid, gamboxylic acid, and other polyphenols. These nano-formulations exhibit improved solubility, slower release, more precise targeting, greater bioavailability, and suitability for conventional drug administration methods including injections and oral capsules in comparison to free forms (Table 3) [[195]]. Furthermore, nanocarriers also reduce the side effects of drugs as it only takes small doses for them to have biological impact [[196]].

8. Discussion

Mitochondria are collectively known as the power houses of eukaryotic cells. The major roles of mitochondria are cell metabolism and regulating pathways related to energy generation. Furthermore, mitochondria are the main source of ROS which play a significant role in the cellular redox biology and are influenced by their generation and accumulation [[207], [209]]. ROS are highly reactive due to their unpaired electron that can interact with several macromolecules in the cell such as proteins, nucleic acids, carbohydrates, and lipids, and change their molecular functions. This redox state control is essential for maintaining the functions of the organ, cell viability, cell activity, and cell proliferation. An oversupply of ROS from various sources (both internal and external) has been shown cellular biomolecular damages and mitochondrial dysfunction and trigger mitochondrial biogenesis, which is the main reason for several chronic diseases such as neurogenerative, cardiovascular, metabolic diseases, cancer, etc.

However, antioxidants act as an essential role in the scavenging and inducing of free radicals. Natural antioxidants, for example, polyphenols, flavonoids, acid polysaccharides, amino acids, phytosterol, and carbohydrates, found in food and plants have remarkable efficacy to protect the healthcare system from oxidative stress [[208]]. These compounds diminish ROS production and increase mitochondrial biogenesis as well as improve the function of mitochondria [[207]]. Among the thousands of antioxidant-rich sources, P. ginseng is the most lexical source of antioxidants that is traditionally used to prevent and treat diseases, which has been discussed in this review in Section 6. P. ginseng increases the activity of antioxidant enzymes such as SOD, CAT, GPx, GST, and the Nr-f2-related enzyme Ho-1 that inhibits ROS production to protect cells from apoptosis, autophagy, and tumor progression. However, they promote the generation of ROS to kill the cancer cell. As shown above, P. ginseng and ginsenosides regulate ROS in ROS-mediated disease states. Among all of the secondary metabolites of ginseng, the ginsenosides Rh2, Rg1, Rg3, Ro, F2, and compound K have the higher efficacy to kill cancer cells by regulating ROS generation.

Despite the fact that P. ginseng has a reputation for being safe, several studies have raised concerns regarding the side effects of ginseng such as allergies and toxicity to the lung, heart, liver, kidney, and reproductive organs [[209]]. As a result, the use of ginseng in humans need to go through a standard consuming system as well as a number of additional controlled conditions including optimum regular dose durations and precise assessment of the patient's overall health. Several studies have shown that to minimize the side effects from plant extracts and their components, nanotechnology has become an essential tool. By developing a nanocarrier system, the stability and solubility of these compounds has been shown to be improved [[196]].

9. Conclusion and Future Perspectives

In this study, we discussed that P. ginseng and its constituents have physiological processes that reduce the symptoms of certain diseases through the antioxidant mechanisms in cells and animals. P. ginseng is well-known for its antioxidant activities that alleviate several chronic diseases such as sensor impairment, cardiovascular diseases, neurogenerative disease, cancer, aging, and metabolic diseases. Although P. ginseng has been proven to increase antioxidant activity in humans as measured by several oxidative stress indications, the limitations of P. ginseng are not deniable. Furthermore, it will be essential to establish the precise efficacy, cytotoxicity, and systemic mechanisms for these chronic diseases via antioxidant action in clinical trials in the future.

Figures and Tables

Graph: Figure 1 In normal cells, the balance between antioxidants and ROS remain at equilibrium due to the chemical reactions of antioxidant enzymes (SOD, CAT, GPx, and GST). However, the overproduction or scavenging of ROS breaks this equilibrium system. At the moderate or basal state, ROS perform as secondary messengers in several intracellular pathways that are essential for healthy cells. However, higher concentrations of ROS can cause oxidative stress, DNA damage, redox homeostasis, autophagy, apoptosis, tumor progression, and drug resistance which are related to the development of various diseases.

Graph: Figure 2 Scavenge and production of reactive oxygen species (ROS). Organic and inorganic constituents can produce, convert, and scavenge ROS. Antioxidant enzymes (SOD, CAT, GPx, and GR) intercept ROS through the chemical reactions. Oxidases convert oxygen to O2·−, which is then dismutated to H2O2 via SOD. H2O2 can be converted to H2O via CAT or GPx or to hydroxyl radical (·OH) after reaction with Fe2+. Abbreviations: SOD-Superoxide dismutase, CAT-catalase, GPx-glutathione Peroxidase, GR-glutathione reductase. GSH-glutathione. GSSG-glutathione disulfide. O2•−-superoxide anion. H2O2-hydrogen peroxide, (HO∙)-hydroxyl, ROS-reactive oxygen species.

Graph: Figure 3 The overproduction of ROS causes oxidative damage which dysregulates several signaling pathways. (a) Oxidative stress increases NF-κB, thus increasing the expression of COX-2, iNOS, and pro-inflammatory cytokines that may cause skin cancer and inflammation. However, ginsenosides Rg1, Rg3, Rb1, and Rh3 increase the antioxidant enzymes (SOD, CAT, GST, and GPx) activity, which suppresses the activation of NF-κB and oxidative stress totrigger skin cancer and inflammation. (b) In extreme antioxidant stress conditions, Nrf2 dissociates from keap1 and binds with the ARE which down-regulates the antioxidant enzyme production, resulting in DNA damage, apoptosis, autophagy, etc., whereas, Rg3, Rb1, and Rh3 increase the antioxidant enzymes by activating the Nrf2 that suppresses the disease conditions. (c) During oxidative stress, β-catenin is not decomposed, leading to the activation of the Wnt/β-catenin pathway. Ginsenoside Rg1 inhibits Wnt activation and increases the activation of β catenin, glycogen synthase kinase-3 (gsk-3), and casein kinase-1 (CK-1) proteins that reduce cell proliferation; (d) similarly, ginsenosides Rg3, Rb1, and Rk1 increase antioxidant enzyme activity and PI3K/Akt to decrease oxidative stress and tumorigenesis. Abbreviations: (ROS-reactive oxygen species, MMP = matrix metalloproteinases, NF-κB-nuclear factor-kappa B, COX-2-cyclooxygenase-2, iNOS-inducible nitric oxide synthase, Keap-1-Kelch-like epoxy chloropropane-related protein-1, Nrf-2-nuclear factor erythroid 2-related factor 2, ARE-antioxidant response element, HO-1-Heme oxygenase-1, NQO-1-NAD(P)H: quinone oxidoreductase 1, SOD-superoxide dismutase, GSH-Px = GSH and glutathione peroxidase, CK-1-casein kinase-1, gsk-3-glycogen synthase kinase-3, PI3k-Phosphatidylinositol-3-kinase, and Akt-protein kinase B).

Table 1 Antioxidant activity of ginseng/ginsenosides in oxidative-stress-mediated diseases.

DiseasesGinseng/GinsenosidesActionModelReferences
Intestinal I/R injuryRg1↓ROS, ↓apoptosis↓ROS, ↓tunnel positive cellIn vitroIn vivo[52]
Adriamycin-induced cardiotoxicityRg3↓ROS, ↓MDA,↑SOD, ↑eNOSIn vitro[60]
Inflammation in asthmaRg3↑Nrf2, ↑HO-1, ↑SOD, ↑GSH,↓MDAIn vivo/Mice[63]
Diabetic nephropathyCK↓ROS, ↓IL-1βIn vivo/Mice[61]
Myocardial cell injury20(S)-Rg3 and 20(R)↓ROS, ↓MDA↑SOD, ↑GSH-Px, ↑CATIn vitro[59]
Myocardial ischemia-reperfusionRd↓CK, ↓LDH,↑HO-1In vivo/Rats[54]
Spinal cord injuryRb1↓MDA, ↑SOD, ↑CAT, ↑GSHIn vivo/Rats[57]
Rat SCI modelRg1↓MDA, ↑SOD, ↑GSHIn vivo/Rats[58]
Cerebral ischemia/reperfusion damageRe↑SOD, ↑GSH-Px, ↓MDA,In vivo/Rats[64]
Mitochondrial oxidationRb1, Rg1↑CAT, ↑complexes I-V, ↑ATP↓ROSIn vivo/Mice[62]
Cerebral hypoxic-ischemic damageRed ginseng↑NQO1, ↑HO1, ↑SOD2, ↑Gpx1,↓IL-1β, ↓iNOSIn vivo[65]
Hepatic diseaseRed ginseng↓MDA, ↓DCF, ↑GPx, ↑GR, ↑CAT, ↑SODIn vitro[66]
PheochromocytomaRed ginseng↓DCF, ↑GCLC, ↑SOD, ↑CAT↑Nrf2, ↑HO-1In vitro[67]
Skin cancerRed ginseng↑GSH, ↑SOD, ↑CAT, ↑Vit C, ↓TBARSIn vivo/Mice[68]
DiabetesRed ginseng↑GSH, ↑SOD, ↑CAT, ↑GPx, ↑GR, ↓MDAIn vivo/Mice[69]
AgingRed ginseng↓MDA, ↑SOD, ↑CAT, ↑GPx, ↑GR, ↑GST[70]
Nerve disorderRed ginseng↑GPx, ↓MDA, ↓ROSIn vivo/Mice[71]
Kidney diseaseRd↑SOD, ↑CAT, ↑GPx,↓MDA, ↓urea nitrogen, ↓creatinineIn vivo/Rats[72]
Notes: ↑-Increases, ↓-Decrease, abbreviations: ROS-reactive oxygen species, MDA-malondialdehyde, SOD-superoxide dismutase, Enos-endothelial nitric oxide synthase, Nrf2-nuclear factor erythroid-2-related factor 2, HO-1-Heme oxygenase-1, GSH-glutathione, IL-1β-interleukin-1β, GSH-Px-glutathione peroxidase, CAT-catalase, Ck-compound K, LDH-lactate dehydrogenase, ATP-adenosine triphosphate, NQO1-NAD(P)H quinone oxidoreductase 1, iNOS-inducible nitric oxide synthase, DCF-Deoxycoformycin, GR-glutathione reductase, GCLC-Glutamate-cysteine ligase catalytic, Vit-c-Vitamin C, TBARS-Thiobarbituric acid reactive substances, GST-glutathione-S-Transferase.

Table 2 ROS-associated effects of ginseng and ginsenosides on cancer cells.

Cancer CellGinseng/GinsenosidesMechanismsEffectReferences
Leukemia JurkatcellsRh2 and Rg3ROS↑, MTP↓, caspase-3/9↑, Bax/Bcl-2↑, Cyt C↑Stimulated mitochondrial ROS that inhibited cell proliferation and induced apoptosis.[149]
AcuteLymphoblasticleukemia cells20(S)-Ginsenoside Rh2ROS↑, MTP↓, Cyt C↑, caspase-3/9↑, LC3-I↑, LC3-II↑, Atg5↓, Beclin-1↑Generated mitochondrial ROS that inhibited autophagy and induced apoptosis.[150]
Lung cancerNCI-H460 cellCompound KROS↑, MMP↓, caspase-3↑Increased γ-ray-induced apoptosis by enhancing intracellular ROS production that reduced MMP and increased caspase-3.[151]
Colorectalcancer HCT-116Rh2ROS↓, Bax/Bcl-2↑, capase-3↑, G1↓, S ↓Reduced the formation of ROS which activated autophagy and antiproliferative effects.[152]
ColorectalcancerProtopanaxadiolROS↑, NF-κB↑ROS production activated the NF-ĸB pathway that induced paraptosis.[153]
Colon cancerHCT-116 cellsCompound KROS↑, Mcl-1↓, Bcl-2↓, survivin↓, XIAP↓, cFLIP↓, Bax↑, tBid↑, Cyt C↑, LC3-II↑, Atg7↑, JNK↑, ERK↓, p38↓, p53↑, DR5↑, CHOP↑Stimulated TRAIL-induced apoptosis by upregulating DR5 in both autophagy in a dependent and dependent manner.[154]
HepatomaHepG2 cellsRh2Caspase-3/9↑, cytosol Cat B↓, leupeptin (Leu) ↑, MTP↓, Bid↑, tBid↑, Cyt C↓Mitochondrial apoptotic and ROS-accumulation-pathway-induced apoptosis.[155]
HepatomaHepG2 cellsRh2PARP↑, ROS↑, p-p38↑, p-AMPK↑Activation of AMPK-mediated ROS generation that induced apoptosis.[156]
HepatomaHep3B cellsRg3 and Rh2ROS↑, caspase-3↑, Bcl-2↓, Bax↑, Cyt C↑, MTP↓Mitochondria-mediated apoptosis pathway and ROS generation caused apoptosis.[157]
Breast cancerMCF-7 cellsCompound KMTP↓, AMPK↑, COX-2↓, PGE2↓Modulation of the AMPK signaling pathway and ROS production induced apoptosis.[158]
Breast cancerMDA-MB-231 cellsRg3ROS↑, Bax/Bcl-2↓, MTP↓, caspase-3↑, PARP↑Apoptosis was caused by the activation of the mitochondrial death pathway.[159]
Breast cancerMCF-7 cellsBG-AuNps and BG-AgNpsROS↑ROS production caused oxidative cell damage and apoptosis.[160]
Cervicalcarcinoma HeLa cellsRh2ROS↑, MTP↓, Caspase-3↓, JNK1↑, SEK1↑, JNK2↑, c-Jun↑, Smac↑, Bax↑, Ca2+Initiated apoptosis by generating ROS and Ca2+which activated SEK1 and JNK1.[161]
Bladder cancerT24 cellsCompound KCyt C↑, Bax↑, Bcl- 2↓, p-p38MAPK↑, procaspase-3/9↑, p38↑, ROS↑, glutathione↓Induced apoptosis through ROS-mediated P38 MAPK pathway.[162]
Gastric carcinoma SGC7901 cellsF2PARP↓, ASK-1↑, JNK↑, Bcl-2↓, Cyt C↑, Caspase-3/9↑Induced apoptosis via modulating the ASK-1/JNK signaling cascade and ROS–mitochondria pathway.[10]
Esophagealcancer cellsRoROS↑, CYBB/Nox2↑, LC3B-II↑, ATG7↑, ESR2↓, NCF1↑, SQSTM1/p62↑, CSTB↓ CSTD↓, p-CHEK1↑, EGFR↑, DDIT3↑, ATM↑, ATR↑, BRCA1↑, GFP-LC3B puncta↑, Lysosomal pH↑, autophagic vacuoles↑ERSR2-NCF1-ROS signaling pathway suppressed autophagy and decreased CHEK1 degradation which sensitized 5-fluorouracil inducing cell death.[163]
NeuroblastomaSH-SY5Y cellRg1ROS↓, JNK↑, caspase-3↑Inhibited the generation of ROS and activated the JNK pathway to inhibit MPP+-induced apoptosis.[164]
AstrocytomaCRT-MG cellsCompound K, ginsenoside Rh2ROS↑, caspase-3↑, Cyt C↑, p-p38↑, Fas↑, MIP↓Increased Fas-dependent apoptosis in ROS/caspase/ mitochondrial pathways.[165]
NeuroblastomaSH-SY5Y cellGinseng water extractROS↑, Bcl-2↓, Bax↑, Cyt C↑, caspase-3↑Alleviated oxidative stress in the mitochondrial apoptotic pathway to prevent MPP+-induced apoptosis.[166]
GliomaU87cells20(S)-ginsenoside Rg3ROS↑, p21↑, p16↑, p53↑, Akt↑ Enhanced ROS generation via Akt- and p53/p21-dependent pathways that induced senescence-like growth arrest.[167]
GlioblastomaU87MG cellsRg3Bcl-2↓, Bax↑, pro-caspase3↓, MEK1/2↑, ROS↑Induced apoptosis via ROS and MEK pathway.[168]
Lung cancerA549 cellsRh1↑P53, ↑Bax, ↑caspase 3, ↑caspase 9, ↓BclIntrinsic pathway.[147]
Colorectal carcinoma SW-480 and HCT-116Rh2Bax↑, Bad ↑, ROS ↑, NF-ĸB↑, Bad↓, Bcl-xL↓, P53↓, Cytosolic vacuolization↓ROS/NF-ĸB signaling pathway/P53 and NF-ĸB signaling pathway. (Upregulation of ROS induced apoptosis by activating NF-ĸB and P53.)[169]
HepatomaHepG2 cellsKorean white ginseng extract Cyt C↑, c-Jun↑, SAPK↑, MDA↓, Caspase-3↑, Iĸ-b↓,Apoptosis induction through JNK/NF- ĸB/Cyt C apoptotic pathway and antioxidant activities.[170]
Breast cancerMDA-MB-231cellsRg3NF-ĸB↓, Bcl-2↓, Akt ↓, IĸB↓, p-ERK↓NF-ĸB inhibition via Akt and ERK inactivation.[171]
Lung cancerA549, H1299, andH358 cellsRg3EMT↓, EGFR ↓, MAPK↓, NF-κB↓MAPK and NF-κB blocking to induce apoptosis.[172]
Notes: ↑ = upregulation, ↓ = down regulation, abbreviations: ROS = reactive oxygen species, MTP-microsomal triglyceride transfer protein, Bax-Bcl-associated-2 protein, Cyt C-cytochrome C, LC-3I-light chain-3I, Atg-5-autophagy-related 5, MMP-matrix metalloproteinase, NF-κB-nuclear factor kappa B, XIAP-X linked inhibitor of apoptosis protein, cFLIP-cellular FLICE-like inhibitory protein, tBid-truncated Bid, JNK$-c-Jun N-terminal kinase, ERK-extracellular signal-regulated kinase, CHOP-C/EBP homogenous protein, PARP-Poly (ADP ribose) polymerase-1, AMPK-AMP-activated protein kinase, Cox-2-Cyclooxygenase-2, PGE-2-Prostaglandin E2, Smac-second mitochondria-derived activator of caspase, MAPK-mitogen-activated protein kinase, ASK-1-apoptosis signal-regulating kinase, NCF-1-neutrophil cytosolic factor-1, CSTB-Cystatin B, CSTB-Cystatin D, EGFR-epidermal growth factor receptor, DDIT3-DNA damage-inducible transcript 3 protein, ATM-Ataxia-telangiectasia-mutated, ATR-Ataxia-telangiectasia and Rad-3 related, BRCA-1-breast cancer gene-1, EMT-epithelial mesenchymal transition, and EGFR-epidermal growth factor receptor.

Table 3 Nano-formulations of active components.

CompoundsNano-FormulationsBenefitsReferences
Ginsenoside Rg3Mixed micelles containing baicalin copolymer P123.G-Rg3 conjugated with nanoparticle.G-Rg3 with CNCnanocomposite.Liposomes of G-Rg3Increases drug absorption in cancer cells and boosts baicalin solubility.Inhibits the growth ofhepatocellular carcinoma (HCC) and prevents lung metastasis.Enhances antioxidant activity.Antitumor activity is greater than free formulation.Increases uptake, sustains release, bioavailability, and enhances cytotoxicity in cancer cells.[194, 197-198]
Ginsenoside Rg5Liposomes of G-Rg5Inhibits tumor growth, tumor targeting.[199]
Ginsenoside Rh2MicellesEnhances water solubility and drug uptake.Elongates drug retention.
Mesoporous silicasExcellent biocompatibility to normal cell (Hacat cell).
CurcuminPLA polymer,chitosan, gum arabicEnhances cellular uptake, and antioxidant and anticancer activities.[200-201]
Increases antioxidant activity.[202]
QuercetinPLAInduces self-life.[203]
Chitosan-CMCEnhances stability, solubility, and antioxidant activity.[204]
Chitosan-pluronic F-127-STPPWound healing.[205]
ResveratrolPLGA-PEGImproves nonvascular effects.[206]
Lecithin/chitosanPromotes anti-inflammatory effects.[207]
Ferulic acidFerulic acid polymer nanoparticlesReduces ROS activity.[208]
Nanoparticles of poly ferulic acidImproved colon cancer treatment.[208]
Abbreviation: PLA-poly lactic acid, PGMD-poly-glycerol-malic acid–dodecanedioic acid, CMC-carboxymethyl chitosan, PEG-polyethylene glycol.

Author Contributions

Writing the original manuscript, M.N.M. and J.C.A.; review and editing, R.M., E.J.R. and D.U.Y.; editing, M.R.K., R.A. and D.H.J.; review, editing, and supervision, D.C.Y. and S.K.J. All authors have read and agreed to the published version of the manuscript.

Data Availability Statement

The data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

Acknowledgments

We would like to thank the Ministry of Agriculture, Food and Rural Affairs (MAFRA), and of Agriculture and Forestry (IPET.) for support via the relevant resources and technology.

Footnotes 1 Disclaimer/Publisher's Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. References Angelova N., Kong H.W., Van Der Heijden R., Yang S.Y., Choi Y.H., Kim H.K., Wang M., Hankemeier T., Van Der Greef J., Xu G. Recent methodology in the phytochemical analysis of ginseng. Phytochem. Anal. Int. J. Plant Chem. Biochem. Tech. 2008; 19: 2-16. 10.1002/pca.1049. 18058794 2 Bae J.-S., Park H.-S., Park J.-W., Li S.-H., Chun Y.-S. Red ginseng and 20(S)-Rg3 control testosterone-induced prostate hy-perplasia by deregulating androgen receptor signaling. J. Nat. Med. 2012; 66: 476-485. 10.1007/s11418-011-0609-8. 22101440 3 Nocerino E., Amato M., Izzo A.A. The aphrodisiac and adaptogenic properties of ginseng. Fitoterapia. 2000; 71: S1-S5. 10.1016/S0367-326X(00)00170-2 4 Metwaly A.M., Lianlian Z., Luqi H., Deqiang D. Black ginseng and its saponins: Preparation, phytochemistry and pharmacological effects. Molecules. 2019; 241856. 10.3390/molecules24101856 5 Abbas M., Haq M., Anwar F. Pharmaceutical and Medicinal Applications of Panax Ginseng and Ginsenosides and Their Theuropatic Role in Different. Nat. Volatiles Essent. Oils. 2022; 9: 745-765 6 Kwak G.-Y., Ying Z., Awais M., Akter R., Xu F., Baik S., Jung D., Yang D.C., Wenying Y. Discrimination of Panax ginseng from counterfeits using single nucleotide polymorphism: A focused review. Front. Plant Sci. 2022; 2116: 903306 7 Adil M., Jeong B.R. In vitro cultivation of Panax ginseng CA Meyer. Ind. Crops Prod. 2018; 122: 239-251. 10.1016/j.indcrop.2018.05.076 8 Chung I.-M., Lim J.-J., Ahn M.-S., Jeong H.-N., An T.-J., Kim S.-H. Comparative phenolic compound profiles and antiox-idative activity of the fruit, leaves, and roots of Korean ginseng (Panax ginseng Meyer) according to cultivation years. J. Ginseng Res. 2016; 40: 68-75 9 Buettner C., Yeh G.Y., Phillips R.S., Mittleman M.A., Kaptchuk T.J. Systematic review of the effects of ginseng on cardio-vascular risk factors. Ann. Pharmacother. 2006; 40: 83-95. 16332943 Mao Q., Zhang P.-H., Wang Q., Li S.-L. Ginsenoside F2 induces apoptosis in humor gastric carcinoma cells through reactive oxygen species-mitochondria pathway and modulation of ASK-1/JNK signaling cascade in vitro and in vivo. Phytomedicine. 2014; 21: 515-522. 10.1016/j.phymed.2013.10.013 Li L., Wang Y., Guo R., Li S., Ni J., Gao S., Gao X., Mao J., Zhu Y., Wu P. Ginsenoside Rg3-loaded, reactive oxygen species-responsive polymeric nanoparticles for alleviating myocardial ischemia-reperfusion injury. J. Control. Release. 2020; 317: 259-272. 10.1016/j.jconrel.2019.11.032. 31783047 Shin M.-S., Song J.H., Choi P., Lee J.H., Kim S.-Y., Shin K.-S., Ham J., Kang K.S. Stimulation of innate immune function by Panax ginseng after heat processing. J. Agric. Food Chem. 2018; 66: 4652-4659. 10.1021/acs.jafc.8b00152 Zeng D., Wang J., Kong P., Chang C., Li J., Li J. Ginsenoside Rg3 inhibits HIF-1α and VEGF expression in patient with acute leukemia via inhibiting the activation of PI3K/Akt and ERK1/2 pathways. Int. J. Clin. Exp. Pathol. 2014; 7: 2172-2178. 24966925 Chung K.-S., Cho S.-H., Shin J.-S., Kim D.-H., Choi J.-H., Choi S.Y., Rhee Y.K., Hong H.-D., Lee K.-T. Ginsenoside Rh2 induces cell cycle arrest and differentiation in human leukemia cells by upregulating TGF-β expression. Carcinogenesis. 2012; 34: 331-340. 10.1093/carcin/bgs341 He Y., Hu Z., Li A., Zhu Z., Yang N., Ying Z., He J., Wang C., Yin S., Cheng S. Recent advances in biotransformation of saponins. Molecules. 2019; 242365. 10.3390/molecules24132365. 31248032 Qi L.-W., Wang H.-Y., Zhang H., Wang C.-Z., Li P., Yuan C.-S. Diagnostic ion filtering to characterize ginseng saponins by rapid liquid chromatography with time-of-flight mass spectrometry. J. Chromatogr. A. 2012; 1230: 93-99. 10.1016/j.chroma.2012.01.079. 22349142 Ahuja A., Kim J.H., Kim J.-H., Yi Y.-S., Cho J.Y. Functional role of ginseng-derived compounds in cancer. J. Ginseng Res. 2018; 42: 248-254. 10.1016/j.jgr.2017.04.009 Jin Y., Huynh D.T.N., Nguyen T.L.L., Jeon H., Heo K.-S. Therapeutic effects of ginsenosides on breast cancer growth and metastasis. Arch. Pharmacal Res. 2020; 43: 773-787. 10.1007/s12272-020-01265-8 Chae S., Kang K.A., Youn U., Park J.S., Hyun J.W. A comparative study of the potential antioxidant activities of ginsenosides. J. Food Biochem. 2010; 34: 31-43. 10.1111/j.1745-4514.2009.00287.x He B., Chen D., Zhang X., Yang R., Yang Y., Chen P., Shen Z. Oxidative Stress and Ginsenosides: An Update on the Molecular Mechanisms. Oxidative Med. Cell. Longev. 2022; 2022: 9299574. 10.1155/2022/9299574. 35498130 Dröge W. The plasma redox state and ageing. Ageing Res. Rev. 2002; 1: 257-278. 10.1016/S1568-1637(01)00008-3 Trachootham D., Alexandre J., Huang P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic ap-proach?. Nat. Rev. Drug Discov. 2009; 8: 579-591. 10.1038/nrd2803. 19478820 Bindu M., Annamalai P. Combined Effect of Alcohol and Cigarette Smoke on Lipid Peroxidation and Antioxidant Status in Rats; CSIR: New Delhi, India. 2004: 40-44 Chan L.Y., Kwok H.H., Chan R.W.Y., Peiris M.J.S., Mak N.K., Wong R.N.S., Chan M.C.W., Yue P.Y.K. Dual functions of ginsenosides in protecting human endothelial cells against influenza H9N2-induced inflammation and apoptosis. J. Ethnopharmacol. 2011; 137: 1542-1546. 10.1016/j.jep.2011.08.022 Khansari N., Shakiba Y., Mahmoudi M. Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer. Recent Pat. Inflamm. Allergy Drug Discov. 2009; 3: 73-80. 10.2174/187221309787158371 Luo Z., Xu X., Sho T., Zhang J., Xu W., Yao J., Xu J. ROS-induced autophagy regulates porcine trophectoderm cell apoptosis, proliferation, and differentiation. Am. J. Physiol. Cell Physiol. 2019; 316: C198-C209. 10.1152/ajpcell.00256.2018. 30485137 Devasagayam T.P., Steenken S., Obendorf M.S., Schulz W.A., Sies H. Formation of 8-hydroxy (deoxy) guanosine and gen-eration of strand breaks at guanine residues in DNA by singlet oxygen. Biochemistry. 1991; 30: 6283-6289. 10.1021/bi00239a029. 2059635 Gutteridge J.M., Halliwell B. Invited review free radicals in disease processes: A compilation of cause and consequence. Free. Radic. Res. Commun. 1993; 19: 141-158. 10.3109/10715769309111598. 8244084 Halliwell B. Reactive oxygen species and the central nervous system. J. Neurochem. 1992; 59: 1609-1623. 10.1111/j.1471-4159.1992.tb10990.x Bhatia V., Tandon R.K. Stress and the gastrointestinal tract. J. Gastroenterol. Hepatol. 2005; 20: 332-339. 10.1111/j.1440-1746.2004.03508.x Pisoschi A.M., Pop A. The role of antioxidants in the chemistry of oxidative stress: A review. Eur. J. Med. Chem. 2015; 97: 55-74. 10.1016/j.ejmech.2015.04.040 Alzoghaibi M.A. Concepts of oxidative stress and antioxidant defense in Crohn's disease. World J. Gastroenterol. WJG. 2013; 19: 6540. 10.3748/wjg.v19.i39.6540 Pammi S.S.S., Suresh B., Giri A. Antioxidant potential of medicinal plants. J. Crop Sci. Biotechnol. 2023; 26: 13-26. 10.1007/s12892-022-00159-z Duarte T.L., Lunec J. Review: When is an antioxidant not an antioxidant? A review of novel actions and reactions of vitamin C. Free Radic Res. 2005; 39: 671-686. 10.1080/10715760500104025. 16036346 Cheung C.C., Zheng G.J., Li A.M., Richardson B.J., Lam P.K. Relationships between tissue concentrations of polycyclic aromatic hydrocarbons and antioxidative responses of marine mussels, Perna viridis. Aquat. Toxicol. 2001; 52: 189-203. 10.1016/S0166-445X(00)00145-4. 11239681 Halliwell B. Biochemistry of oxidative stress. Biochem. Soc. Trans. 2007; 35: 1147-1150. 10.1042/BST0351147 Kehrer J.P., Klotz L.-O. Free radicals and related reactive species as mediators of tissue injury and disease: Implications for health. Crit. Rev. Toxicol. 2015; 45: 765-798. 10.3109/10408444.2015.1074159 Halliwell B. Free radicals and antioxidants: Updating a personal view. Nutr. Rev. 2012; 70: 257-265. 10.1111/j.1753-4887.2012.00476.x Adwas A.A., Elsayed A., Azab A., Quwaydir F. Oxidative stress and antioxidant mechanisms in human body. J. Appl. Bio-Technol. Bioeng. 2019; 6: 43-47 Ji X., Hou C., Shi M., Yan Y., Liu Y. An insight into the research concerning Panax ginseng CA Meyer polysaccharides: A review. Food Rev. Int. 2022; 38: 1149-1165. 10.1080/87559129.2020.1771363 Azab A.E., Albasha M.O., Elsayed A.S.I. Prevention of nephropathy by some natural sources of antioxidants. Yangtze Med. 2017; 1: 235. 10.4236/ym.2017.14023 Bassères D.S., Baldwin A.S. Nuclear factor-κB and inhibitor of κB kinase pathways in oncogenic initiation and progression. Oncogene. 2006; 25: 6817-6830. 10.1038/sj.onc.1209942. 17072330 Jung Y.R., Kim D.H., Kim S.R., An H.J., Lee E.K., Tanaka T., Kim N.D., Yokozawa T., Park J.N., Chung H.Y. Anti-wrinkle effect of magnesium lithospermate B from Salvia miltiorrhiza BUNGE: Inhibition of MMPs via NF-κB signaling. PLoS ONE. 2014; 9e102689. 10.1371/journal.pone.0102689. 25099178 Kim J.-K., Kim Y., Na K.-M., Surh Y.-J., Kim T.-Y. [6]-Gingerol prevents UVB-induced ROS production and COX-2 expression in vitro and in vivo. Free. Radic. Res. 2007; 41: 603-614. 10.1080/10715760701209896 Wang Y., Wang L., Wen X., Hao D., Zhang N., He G., Jiang X. NF-κB signaling in skin aging. Mech. Ageing Dev. 2019; 184: 111160. 10.1016/j.mad.2019.111160. 31634486 Loboda A., Damulewicz M., Pyza E., Jozkowicz A., Dulak J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell. Mol. Life Sci. 2016; 73: 3221-3247. 10.1007/s00018-016-2223-0 Dai X., Yan X., Wintergerst K.A., Cai L., Keller B.B., Tan Y. Nrf2: Redox and Metabolic Regulator of Stem Cell State and Function. Trends Mol. Med. 2020; 26: 185-200. 10.1016/j.molmed.2019.09.007. 31679988 Yamamoto M., Kensler T.W., Motohashi H. The KEAP1-NRF2 system: A thiol-based sensor-effector apparatus for main-taining redox homeostasis. Physiol. Rev. 2018; 98: 1169-1203. 10.1152/physrev.00023.2017 Katoh M., Katoh M. WNT signaling pathway and stem cell signaling network. Clin. Cancer Res. 2007; 13: 4042-4045. 10.1158/1078-0432.CCR-06-2316 Kahn M. Can we safely target the WNT pathway?. Nat. Rev. Drug Discov. 2014; 13: 513-532. 10.1038/nrd4233 Xie Y., Shi X., Sheng K., Han G., Li W., Zhao Q., Jiang B., Feng J., Li J., Gu Y. PI3K/Akt signaling transduction pathway, erythropoiesis and glycolysis in hypoxia. Mol. Med. Rep. 2019; 19: 783-791. 10.3892/mmr.2018.9713 Wen Z., Hou W., Wu W., Zhao Y., Dong X., Bai X., Peng L., Song L. 6-O-galloylpaeoniflorin attenuates cerebral ischemia reperfusion-induced neuroinflammation and oxidative stress via PI3K/Akt/Nrf2 activation. Oxidative Med. Cell. Longev. 2018; 2018: 8678267. 10.1155/2018/8678267 Zu G., Guo J., Che N., Zhou T., Zhang X. Protective effects of ginsenoside Rg1 on intestinal ischemia/reperfusion inju-ry-induced oxidative stress and apoptosis via activation of the Wnt/β-catenin pathway. Sci. Rep. 2016; 6: 38480. 10.1038/srep38480. 27910952 Gao L., Wang X., Tang Y., Huang S., Hu C.-A.A., Teng Y. FGF19/FGFR4 signaling contributes to the resistance of hepato-cellular carcinoma to sorafenib. J. Exp. Clin. Cancer Res. 2017; 36: 8. 10.1186/s13046-016-0478-9 Zeng X., Li J., Li Z. Ginsenoside Rd mitigates myocardial ischemia-reperfusion injury via Nrf2/HO-1 signaling pathway. Int. J. Clin. Exp. Med. 2015; 8: 14497 Dong C., Liu P., Wang H., Dong M., Li G., Li Y. Ginsenoside Rb1 attenuates diabetic retinopathy in streptozotocin-induced diabetic rats. Acta Cirúrgica Bras. 2019; 34. 10.1590/s0102-8650201900201 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. 10.3892/etm.2018.6286. 30116359 Zhang Z., Yang K., Mao R., Zhong D., Xu Z., Xu J., Xiong M. Ginsenoside Rg1 inhibits oxidative stress and inflammation in rats with spinal cord injury via Nrf2/HO-1 signaling pathway. Neuroreport. 2021; 33: 81-89. 10.1097/WNR.0000000000001757 Zhao Y., Wang Y., Zhang M., Gao Y., Yan Z. Protective Effects of Ginsenosides (20R)-Rg3 on H2O2-Induced Myocardial Cell Injury by Activating Keap-1/Nrf2/HO-1 Signaling Pathway. Chem. Biodivers. 2021; 18: e2001007. 10.1002/cbdv.202001007 Wang X., Chen L., Wang T., Jiang X., Zhang H., Li P., Lv B., Gao X. Ginsenoside Rg3 antagonizes adriamycin-induced cardiotoxicity by improving endothelial dysfunction from oxidative stress via upregulating the Nrf2-ARE pathway through the activation of akt. Phytomedicine. 2015; 22: 875-884. 10.1016/j.phymed.2015.06.010 Song W., Wei L., Du Y., Wang Y., Jiang S. Protective effect of ginsenoside metabolite compound K against diabetic nephropathy by inhibiting NLRP3 inflammasome activation and NF-κB/p38 signaling pathway in high-fat di-et/streptozotocin-induced diabetic mice. Int. Immunopharmacol. 2018; 63: 227-238. 10.1016/j.intimp.2018.07.027. 30107367 Xu M., Ma Q., Fan C., Chen X., Zhang H., Tang M. Ginsenosides Rb1 and Rg1 protect primary cultured astrocytes against oxygen-glucose deprivation/reoxygenation-induced injury via improving mitochondrial function. Int. J. Mol. Sci. 2019; 206086. 10.3390/ijms20236086 Huang W.-C., Huang T.-H., Yeh K.-W., Chen Y.-L., Shen S.-C., Liou C.-J. Ginsenoside Rg3 ameliorates allergic airway inflammation and oxidative stress in mice. J. Ginseng Res. 2021; 45: 654-664. 10.1016/j.jgr.2021.03.002. 34764720 Zhou X.-M., Cao Y.-L., Dou D.-Q. Protective Effect of Ginsenoside-Re against Cerebral Ischemia/Reperfusion Damage in Rats. Biol. Pharm. Bull. 2006; 29: 2502-2505. 10.1248/bpb.29.2502 Liu L., Vollmer M.K., Ahmad A.S., Fernandez V.M., Kim H., Doré S. Pretreatment with Korean red ginseng or dimethyl fumarate attenuates reactive gliosis and confers sustained neuroprotection against cerebral hypoxic-ischemic damage by an Nrf2-dependent mechanism. Free Radic. Biol. Med. 2019; 131: 98-114. 10.1016/j.freeradbiomed.2018.11.017 Sohn S.-H., Kim S.-K., Kim Y.-O., Kim H.-D., Shin Y.-S., Yang S.-O., Kim S.-Y., Lee S.-W. A comparison of antioxidant activity of Korean White and Red Ginsengs on H2O2-induced oxidative stress in HepG2 hepatoma cells. J. Ginseng Res. 2013; 37: 442-450. 10.5142/jgr.2013.37.442. 24233437 Park S.-H., Jang J.-H., Chen C.-Y., Na H.-K., Surh Y.-J. A formulated red ginseng extract rescues PC12 cells from PCB-induced oxidative cell death through Nrf2-mediated upregulation of heme oxygenase-1 and glutamate cysteine ligase. Toxicology. 2010; 278: 131-139. 10.1016/j.tox.2010.04.003 Sharma J., Goyal P.K. Chemoprevention of chemical-induced skin cancer by Panax ginseng root extract. J. Ginseng Res. 2015; 39: 265-273. 10.1016/j.jgr.2015.01.005 Kim H.-J., Lee S.-G., Chae I.-G., Kim M.-J., Im N.-K., Yu M.-H., Lee E.-J., Lee I.-S. Antioxidant effects of fermented red ginseng extracts in streptozotocin-induced diabetic rats. J. Ginseng Res. 2011; 35: 129. 10.5142/jgr.2011.35.2.129 Ramesh T., Kim S.-W., Hwang S.-Y., Sohn S.-H., Yoo S.-K., Kim S.-K. Panax ginseng reduces oxidative stress and restores antioxidant capacity in aged rats. Nutr. Res. 2012; 32: 718-726. 10.1016/j.nutres.2012.08.005. 23084645 Lee J. Antioxidant effects of Korean red ginseng extracts on the glutathione and lipid peroxidation in the liver of mouse treated with paraquat. Korean J. Biomed. Lab. Sci. 2000; 6: 45-53 Yokozawa T., Liu Z.W., Dong E. A study of ginsenoside-Rd in a renal ischemia-reperfusion model. Nephron. 1998; 78: 201-206. 10.1159/000044911 Murphy M.P., Hartley R.C. Mitochondria as a therapeutic target for common pathologies. Nat. Rev. Drug Discov. 2018; 17: 865-886. 10.1038/nrd.2018.174 Huang Q., Gao S., Zhao D., Li X. Review of ginsenosides targeting mitochondrial function to treat multiple disorders: Current status and perspectives. J. Ginseng Res. 2021; 45: 371-379. 10.1016/j.jgr.2020.12.004. 34025130 Pak J.H., Kim Y., Yi J., Chung J.W. Antioxidant therapy against oxidative damage of the inner ear: Protection and precon-ditioning. Antioxidants. 2020; 91076. 10.3390/antiox9111076. 33147893 Choung Y.H., Kim S.W., Tian C., Min J.Y., Lee H.K., Park S.N., Lee J.B., Park K. Korean red ginseng prevents gentamicin-induced hearing loss in rats. Laryngoscope. 2011; 121: 1294-1302. 10.1002/lary.21756. 21541943 Yamashita D., Jiang H.-Y., Schacht J., Miller J.M. Delayed production of free radicals following noise exposure. Brain Res. 2004; 1019: 201-209. 10.1016/j.brainres.2004.05.104 Hong B.N., Kim S.Y., Yi T.H., Kang T.H. Post-exposure treatment with ginsenoside compound K ameliorates auditory functional injury associated with noise-induced hearing loss in mice. Neurosci. Lett. 2011; 487: 217-222. 10.1016/j.neulet.2010.10.026 Im G.J., Chang J.W., Choi J., Chae S.W., Ko E.J., Jung H.H. Protective effect of Korean red ginseng extract on cisplatin ototoxicity in HEI-OC1 auditory cells. Phytother. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2010; 24: 614-621. 10.1002/ptr.3082 Moreau K.L., King J.A. Protein misfolding and aggregation in cataract disease and prospects for prevention. Trends Mol. Med. 2012; 18: 273-282. 10.1016/j.molmed.2012.03.005 Truscott R.J.W., Friedrich M.G. The etiology of human age-related cataract. Proteins don't last forever. Biochim. Biophys. Acta BBA Gen. Subj. 2016; 1860: 192-198. 10.1016/j.bbagen.2015.08.016 Park C., Cha H.-J., Song K.-S., Kim H.-S., Bang E., Lee H., Jin C.-Y., Kim G.-Y., Choi Y.H. Nrf2-mediated activation of HO-1 is required in the blocking effect of compound K, a ginseng saponin metabolite, against oxidative stress damage in ARPE-19 human retinal pigment epithelial cells. J. Ginseng Res. 2022in press. 10.1016/j.jgr.2022.09.007 Quigley H.A., Broman A.T. The number of people with glaucoma worldwide in 2010 and 2020. Br. J. Ophthalmol. 2006; 90: 262-267. 10.1136/bjo.2005.081224 Sim R.H., Sirasanagandla S.R., Das S., Teoh S.L. Treatment of Glaucoma with Natural Products and Their Mechanism of Action: An Update. Nutrients. 2022; 14534. 10.3390/nu14030534 Liu Z., Chen J., Huang W., Zeng Z., Yang Y., Zhu B. Ginsenoside Rb1 protects rat retinal ganglion cells against hypoxia and oxidative stress. Mol. Med. Rep. 2013; 8: 1397-1403. 10.3892/mmr.2013.1658. 24002069 Lee K., Yang H., Kim J.Y., Choi W., Seong G.J., Kim C.Y., Lee J.M., Bae H.W. Effect of red ginseng on visual function and vision-related quality of life in patients with glaucoma. J. Ginseng Res. 2021; 45: 676-682. 10.1016/j.jgr.2021.03.004 Bae H.W., Kim J.H., Kim S., Kim M., Lee N., Hong S., Seong G.J., Kim C.Y. Effect of Korean Red Ginseng supplementation on dry eye syndrome in glaucoma patients–A randomized, double-blind, placebo-controlled study. J. Ginseng Res. 2015; 39: 7-13. 10.1016/j.jgr.2014.07.002 Sayre L.M., Perry G., Smith M.A. Oxidative Stress and Neurotoxicity. Chem. Res. Toxicol. 2008; 21: 172-188. 10.1021/tx700210j. 18052107 Li J., O W., Li W., Jiang Z.G., Ghanbari H.A. Oxidative stress and neurodegenerative disorders. Int. J. Mol. Sci. 2013; 14: 24438-24475. 10.3390/ijms141224438 Braak H., Del Tredici K., Rüb U., de Vos R.A., Jansen Steur E.N., Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol. Aging. 2003; 24: 197-211. 10.1016/S0197-4580(02)00065-9 Kang M.H., Ji Y.-J., Han Y.M., Jang G.Y., Kim D.H., Lee J.H., Kim G.-S., Choi S.J., Kim H.D. Sasa borealis Ethanol Extract Protects PC12 Neuronal Cells against Oxidative Stress. Appl. Sci. 2022; 126155. 10.3390/app12126155 Schapira A.H. Molecular and clinical pathways to neuroprotection of dopaminergic drugs in Parkinson disease. Neurology. 2009; 72: S44-S50. 10.1212/WNL.0b013e3181990438. 19221314 Hu S., Han R., Mak S., Han Y. Protection against 1-methyl-4-phenylpyridinium ion (MPP+)-induced apoptosis by water extract of ginseng (Panax ginseng C.A. Meyer) in SH-SY5Y cells. J. Ethnopharmacol. 2011; 135: 34-42. 10.1016/j.jep.2011.02.017 Chen X.C., Zhou Y.C., Chen Y., Zhu Y.G., Fang F., Chen L.M. Ginsenoside Rg1 reduces MPTP-induced substantia nigra neuron loss by suppressing oxidative stress. Acta Pharmacol. Sin. 2005; 26: 56-62. 10.1111/j.1745-7254.2005.00019.x. 15659115 Shi C., Zhang Y.X., Zhang Z.F. Effect of phosphorylated-ERK1/2 on inducible nitric oxide synthase expression in the sub-stantia nigra of mice with MPTP-induced Parkinson disease. Nan Fang Yi Ke Da Xue Xue Bao. 2009; 29: 60-63. 19218114 Chen X.C., Zhu Y.G., Zhu L.A., Huang C., Chen Y., Chen L.M., Fang F., Zhou Y.C., Zhao C.H. Ginsenoside Rg1 attenuates dopamine-induced apoptosis in PC12 cells by suppressing oxidative stress. Eur. J. Pharmacol. 2003; 473: 1-7. 10.1016/S0014-2999(03)01945-9 Xu H., Jiang H., Wang J., Xie J. Rg1 protects the MPP+-treated MES23. 5 cells via attenuating DMT1 up-regulation and cellular iron uptake. Neuropharmacology. 2010; 58: 488-494. 10.1016/j.neuropharm.2009.09.002. 19744503 Xu B.-B., Liu C.-Q., Gao X., Zhang W.-Q., Wang S.-W., Cao Y.-L. Possible mechanisms of the protection of ginsenoside Re against MPTP-induced apoptosis in substantia nigra neurons of Parkinson's disease mouse model. J. Asian Nat. Prod. Res. 2005; 7: 215-224. 10.1080/10286020410001690172 Selkoe D.J., Schenk D. Alzheimer's disease: Molecular understanding predicts amyloid-based therapeutics. Annu. Rev. Pharmacol. Toxicol. 2003; 43: 545. 10.1146/annurev.pharmtox.43.100901.140248 Tanzi R.E., Bertram L. The latest suspect. Nature. 2008; 454: 707-708. 10.1038/454706a Bazan N.G., Palacios-Pelaez R., Lukiw W.J. Hypoxia signaling to genes. Mol. Neurobiol. 2002; 26: 283-298. 10.1385/MN:26:2-3:283 Frank B., Gupta S. A review of antioxidants and Alzheimer's disease. Ann. Clin. Psychiatry. 2005; 17: 269-286. 10.1080/10401230500296428. 16402761 Finch C.E., Morgan T.E. Systemic inflammation, infection, ApoE alleles, and Alzheimer disease: A position paper. Curr. Alzheimer Res. 2007; 4: 185-189. 10.2174/156720507780362254 Zhao H., Li Q., Zhang Z., Pei X., Wang J., Li Y. Long-term ginsenoside consumption prevents memory loss in aged SAMP8 mice by decreasing oxidative stress and up-regulating the plasticity-related proteins in hippocampus. Brain Res. 2009; 1256: 111-122. 10.1016/j.brainres.2008.12.031 Qian Y.-H., Han H., Hu X.-D., Shi L.-L. Protective effect of ginsenoside Rb1 on β-amyloid protein (1-42)-induced neuro-toxicity in cortical neurons. Neurol. Res. 2009; 31: 663-667. 10.1179/174313209X385572. 19138476 Xie X., Wang H.-T., Li C.-L., Gao X.-H., Ding J.-L., Zhao H.-H., Lu Y.-L. Ginsenoside Rb1 protects PC12 cells against β-amyloid-induced cell injury. Mol. Med. Rep. 2010; 3: 635-639. 21472290 Liu Q., Kou J.-P., Yu B.-Y. Ginsenoside Rg1 protects against hydrogen peroxide-induced cell death in PC12 cells via inhibiting NF-κB activation. Neurochem. Int. 2011; 58: 119-125. 10.1016/j.neuint.2010.11.004. 21078355 Wang Y.-H., Du G.-H. Ginsenoside Rg1 inhibits β-secretase activity in vitro and protects against Aβ-induced cytotoxicity in PC12 cells. J. Asian Nat. Prod. Res. 2009; 11: 604-612. 10.1080/10286020902843152. 20183297 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: 114192. 10.1016/j.biopha.2022.114192. 36587558 Bouzroud S., El Maaiden E., Sobeh M., Merghoub N., Boukcim H., Kouisni L., El Kharassi Y. Biotechnological Approaches to Producing Natural Antioxidants: Anti-Ageing and Skin Longevity Prospects. Int. J. Mol. Sci. 2023; 241397. 10.3390/ijms24021397. 36674916 Hou W., Wang Y., Zheng P., Cui R. Effects of ginseng on neurological disorders. Front. Cell. Neurosci. 2020; 14: 55. 10.3389/fncel.2020.00055. 32265659 Wang W., Liu X., Liu J., Cai E., Zhao Y., Li H., Zhang L., Li P., Gao Y. Sesquiterpenoids from the root of Panax ginseng attenuates lipopolysaccharide-induced depressive-like behavior through the brain-derived neurotrophic fac-tor/tropomyosin-related kinase B and sirtuin type 1/nuclear factor-κB signaling pathways. J. Agric. Food Chem. 2018; 66: 265-271. 10.1021/acs.jafc.7b04835. 29237268 Liu L., Kelly M.G., Wierzbicki E.L., Escober-Nario I.C., Vollmer M.K., Doré S. Nrf2 plays an essential role in long-term brain damage and neuroprotection of Korean red ginseng in a permanent cerebral ischemia model. Antioxidants. 2019; 8273. 10.3390/antiox8080273. 31382635 Dong X., Zheng L., Lu S., Yang Y. Neuroprotective effects of pretreatment of ginsenoside R b1 on severe cerebral ischemia-induced injuries in aged mice: Involvement of anti-oxidant signaling. Geriatr. Gerontol. Int. 2017; 17: 338-345. 10.1111/ggi.12699 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 Complement. Altern. Med. 2016; 16316. 10.1186/s12906-016-1050-z. 26887326 Garbelli R., De Bock F., Medici V., Rousset M., Villani F., Boussadia B., Arango-Lievano M., Jeanneteau F., Daneman R., Bartolomei F. PDGFRβ+ cells in human and experimental neuro-vascular dysplasia and seizures. Neuroscience. 2015; 306: 18-27. 10.1016/j.neuroscience.2015.07.090 Zhu Y., Wang Z., Yu S., Zhao C., Xu B., Liu R., Xu L., Guo Y. Neuroprotective Effect of Ginseng Fibrous Root Enzymatic Hydrolysate against Oxidative Stress. Molecules. 2022; 277824. 10.3390/molecules27227824 Kritharides L., Brown A., Brieger D., Ridell T., Zeitz C., Jeremy R., Tonkin A., Walsh W., White H. Overview and de-terminants of cardiovascular disease in indigenous populations. Heart Lung Circ. 2010; 19: 337-343. 10.1016/j.hlc.2010.02.017 Pratt C. Alternative prevention and treatment of cardiovascular disease, part 2. Prim. Care Clin. Off. Pract. 2010; 37: 339-366. 10.1016/j.pop.2010.02.010. 20493340 Ing D.J., Zang J., Dzau V.J., Webster K.A., Bishopric N.H. Modulation of cytokine-induced cardiac myocyte apoptosis by nitric oxide, Bak, and Bcl-x. Circ. Res. 1999; 84: 21-33. 10.1161/01.RES.84.1.21. 9915771 Von Harsdorf R.d., Li P.-F., Dietz R. Signaling pathways in reactive oxygen species–induced cardiomyocyte apoptosis. Circulation. 1999; 99: 2934-2941. 10.1161/01.CIR.99.22.2934 Kanani P.M., Sinkey C.A., Browning R.L., Allaman M., Knapp H.R., Haynes W.G. Role of oxidant stress in endothelial dysfunction produced by experimental hyperhomocyst (e) inemia in humans. Circulation. 1999; 100: 1161-1168. 10.1161/01.CIR.100.11.1161 B Britten M., M Zeiher A., Schächinger V. Clinical importance of coronary endothelial vasodilator dysfunction and thera-peutic options. J. Intern. Med. 1999; 245: 315-327. 10.1046/j.1365-2796.1999.00449.x Ferrari R., Agnoletti L., Comini L., Gaia G., Bachetti T., Cargnoni A., Ceconi C., Curello S., Visioli O. Oxidative stress during myocardial ischaemia and heart failure. Eur. Heart J. 1998; 19: B2-B11. 10.2174/1381612043384718. 15134567 Anversa P., Cheng W., Liu Y., Leri A., Redaelli G., Kajstura J. Apoptosis and myocardial infarction. Basic Res. Cardiol. 1998; 93: s008-s012. 10.1007/s003950050195. 9879436 Lim K.H., Ko D., Kim J.-H. Cardioprotective potential of Korean Red Ginseng extract on isoproterenol-induced cardiac injury in rats. J. Ginseng Res. 2013; 37: 273. 10.5142/jgr.2013.37.273. 24198652 Zhou W., Chai H., Lin P.H., Lumsden A.B., Yao Q., Chen C. Ginsenoside Rb1 blocks homocysteine-induced endothelial dysfunction in porcine coronary arteries. J. Vasc. Surg. 2005; 41: 861-868. 10.1016/j.jvs.2005.01.054 Xie J.-T., Shao Z.-H., Vanden Hoek T.L., Chang W.-T., Li J., Mehendale S., Wang C.-Z., Hsu C.-W., Becker L.B., Yin J.-J. Antioxidant effects of ginsenoside Re in cardiomyocytes. Eur. J. Pharmacol. 2006; 532: 201-207. 10.1016/j.ejphar.2006.01.001 Yi X.Q., Li T., Wang J.R., Wong V.K.W., Luo P., Wong I.Y.F., Jiang Z.H., Liu L., Zhou H. Total ginsenosides increase coronary perfusion flow in isolated rat hearts through activation of PI3K/Akt-eNOS signaling. Phytomedicine. 2010; 17: 1006-1015. 10.1016/j.phymed.2010.06.012 Jeon B.H., Kim C.S., Park K.S., Lee J.W., Park J.B., Kim K.-J., Kim S.H., Chang S.J., Nam K.Y. Effect of Korea red ginseng on the blood pressure in conscious hypertensive rats. Gen. Pharmacol. Vasc. Syst. 2000; 35: 135-141. 10.1016/S0306-3623(01)00096-9 Qin N., Gong Q.-h., Wei L.-w., Wu Q., Huang X.-n. Total Ginsenosides Inhibit the Right Ventricular Hypertrophy Induced by Monocrotaline in Rats. Biol. Pharm. Bull. 2008; 31: 1530-1535. 10.1248/bpb.31.1530 Hong S.Y., Kim J.Y., Ahn H.Y., Shin J.-H., Kwon O. Panax ginseng Extract Rich in Ginsenoside Protopanaxatriol Attenuates Blood Pressure Elevation in Spontaneously Hypertensive Rats by Affecting the Akt-Dependent Phosphorylation of Endo-thelial Nitric Oxide Synthase. J. Agric. Food Chem. 2012; 60: 3086-3091. 10.1021/jf204447y. 22380784 Zhu D., Wu L., Li C.R., Wang X.W., Ma Y.J., Zhong Z.y., Zhao H.B., Cui J., Xun S.F., Huang X.L. Ginsenoside Rg1 protects rat cardiomyocyte from hypoxia/reoxygenation oxidative injury via antioxidant and intracellular calcium homeostasis. J. Cell. Biochem. 2009; 108: 117-124. 10.1002/jcb.22233. 19530220 Tsutsumi Y.M., Tsutsumi R., Mawatari K., Nakaya Y., Kinoshita M., Tanaka K., Oshita S. Compound K, a metabolite of ginsenosides, induces cardiac protection mediated nitric oxide via Akt/PI3K pathway. Life Sci. 2011; 88: 725-729. 10.1016/j.lfs.2011.02.011. 21338613 Wang Y., Hu Z., Sun B., Xu J., Jiang J., Luo M. Ginsenoside Rg3 attenuates myocardial ischemia/reperfusion injury via Akt/endothelial nitric oxide synthase signaling and the B-cell lymphoma/B-cell lymphoma-associated X protein pathway. Mol. Med. Rep. 2015; 11: 4518-4524. 10.3892/mmr.2015.3336 Qin G.-W., Lu P., Peng L., Jiang W. Ginsenoside Rb1 inhibits cardiomyocyte autophagy via PI3K/Akt/mTOR signaling pathway and reduces myocardial ischemia/reperfusion injury. Am. J. Chin. Med. 2021; 49: 1913-1927. 10.1142/S0192415X21500907 Fan Z.-X., Yang C.-J., Li Y.-H., Yang J., Huang C.-X. Ginsenoside Rh2 attenuates myocardial ischaemia-reperfusion injury by regulating the Nrf2/HO-1/NLRP3 signalling pathway. Exp. Ther. Med. 2023; 25: 35. 10.3892/etm.2022.11734 Sodrul I.M., Wang C., Chen X., Du J., Sun H. Role of ginsenosides in reactive oxygen species-mediated anticancer therapy. Oncotarget. 2018; 9: 2931. 10.18632/oncotarget.23407 Klaunig J.E., Xu Y., Isenberg J.S., Bachowski S., Kolaja K.L., Jiang J., Stevenson D.E., Walborg Jr E.F. The role of oxidative stress in chemical carcinogenesis. Environ. Health Perspect. 1998; 106: 289-295 Cairns R.A., Harris I.S., Mak T.W. Regulation of cancer cell metabolism. Nat. Rev. Cancer. 2011; 11: 85-95. 10.1038/nrc2981 Ichijo H., Nishida E., Irie K., Dijke P.t., Saitoh M., Moriguchi T., Takagi M., Matsumoto K., Miyazono K., Gotoh Y. In-duction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science. 1997; 275: 90-94. 10.1126/science.275.5296.90 Moon D.-O., Kim M.-O., Choi Y.H., Hyun J.W., Chang W.Y., Kim G.-Y. Butein induces G2/M phase arrest and apoptosis in human hepatoma cancer cells through ROS generation. Cancer Lett. 2010; 288: 204-213. 10.1016/j.canlet.2009.07.002 Gorrini C., Harris I.S., Mak T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013; 12: 931-947. 10.1038/nrd4002 Liu J., Wang Z. Increased oxidative stress as a selective anticancer therapy. Oxidative Med. Cell. Longev. 2015; 2015: 294303. 10.1155/2015/294303 De Giani A., Oldani M., Forcella M., Lasagni M., Fusi P., Di Gennaro P. Synergistic Antioxidant Effect of Prebiotic Ginseng Berries Extract and Probiotic Strains on Healthy and Tumoral Colorectal Cell Lines. Int. J. Mol. Sci. 2023; 24373. 10.3390/ijms24010373. 36613815 Nahar J., Boopathi V., Murugesan M., Rupa E.J., Yang D.C., Kang S.C., Mathiyalagan R. Investigating the Anticancer Activity of G-Rh1 Using In Silico and In Vitro Studies (A549 Lung Cancer Cells). Molecules. 2022; 278311. 10.3390/molecules27238311 Al-Hussaniy H.A., Al-Kuraishy H.M., Abdulameer A.-G.A. The Use of Panax Ginseng to Reduce the Cardiotoxicity of Doxorubicin and Study its Effect on Modulating Oxidative Stress, Inflammatory, and Apoptosis Pathways. Open Access Maced. J. Med. Sci. 2022; 10: 715-719. 10.3889/oamjms.2022.9479 Xia T., Wang Y.N., Zhou C.X., Wu L.M., Liu Y., Zeng Q.H., Zhang X.L., Yao J.H., Wang M., Fang J.P. Ginsenoside Rh2 and Rg3 inhibit cell proliferation and induce apoptosis by increasing mitochondrial reactive oxygen species in human leukemia Jurkat cells. Mol. Med. Rep. 2017; 15: 3591-3598. 10.3892/mmr.2017.6459 Xia T., Wang J., Wang Y., Wang Y., Cai J., Wang M., Chen Q., Song J., Yu Z., Huang W. Inhibition of autophagy potentiates anticancer property of 20 (S)-ginsenoside Rh2 by promoting mitochondria-dependent apoptosis in human acute lympho-blastic leukaemia cells. Oncotarget. 2016; 7: 27336. 10.18632/oncotarget.8285. 27027340 Chae S., Kang K.A., Chang W.Y., Kim M.J., Lee S.J., Lee Y.S., Kim H.S., Kim D.H., Hyun J.W. Effect of compound K, a metabolite of ginseng saponin, combined with γ-ray radiation in human lung cancer cells in vitro and in vivo. J. Ag-Ricultural Food Chem. 2009; 57: 5777-5782. 10.1021/jf900331g Zhu C., Liu F., Qian W., Zhang T., Li F. Combined effect of sodium selenite and ginsenoside Rh2 on HCT116 human col-orectal carcinoma cells. Arch. Iran. Med. 2016; 19: 23 Wang C.-Z., Li B., Wen X.-D., Zhang Z., Yu C., Calway T.D., He T.-C., Du W., Yuan C.-S. Paraptosis and NF-κB activation are associated with protopanaxadiol-induced cancer chemoprevention. BMC Complement. Altern. Med. 2013; 132. 10.1186/1472-6882-13-2. 23281928 Chen L., Meng Y., Sun Q., Zhang Z., Guo X., Sheng X., Tai G., Cheng H., Zhou Y. Ginsenoside compound K sensitizes human colon cancer cells to TRAIL-induced apoptosis via autophagy-dependent and-independent DR5 upregulation. Cell Death Dis. 2016; 7: e2334. 10.1038/cddis.2016.234. 27512955 Chen F., Deng Z., Xiong Z., Zhang B., Yang J., Hu J. A ROS-mediated lysosomal–mitochondrial pathway is induced by ginsenoside Rh2 in hepatoma HepG2 cells. Food Funct. 2015; 6: 3828-3837. 10.1039/C5FO00518C. 26449932 Kim M.-J., Yun H., Kim D.-H., Kang I., Choe W., Kim S.-S., Ha J. AMP-activated protein kinase determines apoptotic sensitivity of cancer cells to ginsenoside-Rh2. J. Ginseng Res. 2014; 38: 16-21. 10.1016/j.jgr.2013.11.010. 24558305 Park H.-M., Kim S.-J., Kim J.-S., Kang H.-S. Reactive oxygen species mediated ginsenoside Rg3-and Rh2-induced apoptosis in hepatoma cells through mitochondrial signaling pathways. Food Chem. Toxicol. 2012; 50: 2736-2741. 10.1016/j.fct.2012.05.027. 22634290 Kim A.D., Kang K.A., Zhang R., Lim C.M., Kim H.S., Kim D.H., Jeon Y.J., Lee C.H., Park J., Chang W.Y. Ginseng saponin metabolite induces apoptosis in MCF-7 breast cancer cells through the modulation of AMP-activated protein kinase. Environ. Toxicol. Pharmacol. 2010; 30: 134-140. 10.1016/j.etap.2010.04.008. 21787643 Kim B.-M., Kim D.-H., Park J.-H., Na H.-K., Surh Y.-J. Ginsenoside Rg3 induces apoptosis of human breast cancer (MDA-MB-231) cells. J. Cancer Prev. 2013; 18: 177. 10.15430/JCP.2013.18.2.177 Wang D., Markus J., Kim Y.-J., Wang C., Pérez Z.E.J., Ahn S., Aceituno V.C., Mathiyalagan R., Yang D.C. Coalescence of functional gold and monodisperse silver nanoparticles mediated by black Panax ginseng Meyer root extract. Int. J. Nanomed. 2016; 11: 6621. 10.2147/IJN.S113692 Ham Y.-M., Lim J.-H., Na H.-K., Choi J.-S., Park B.-D., Yim H., Lee S.-K. Ginsenoside-Rh2-induced mitochondrial depo-larization and apoptosis are associated with reactive oxygen species-and Ca2+-mediated c-Jun NH2-terminal kinase 1 activation in HeLa cells. J. Pharmacol. Exp. Ther. 2006; 319: 1276-1285. 10.1124/jpet.106.109926 Wang H., Jiang D., Liu J., Ye S., Xiao S., Wang W., Sun Z., Xie Y., Wang J. Compound K induces apoptosis of bladder cancer T24 cells via reactive oxygen species-mediated p38 MAPK pathway. Cancer Biother. Radiopharm. 2013; 28: 607-614. 10.1089/cbr.2012.1468 Zheng K., Li Y., Wang S., Wang X., Liao C., Hu X., Fan L., Kang Q., Zeng Y., Wu X. Inhibition of autophagosome-lysosome fusion by ginsenoside Ro via the ESR2-NCF1-ROS pathway sensitizes esophageal cancer cells to 5-fluorouracil-induced cell death via the CHEK1-mediated DNA damage checkpoint. Autophagy. 2016; 12: 1593-1613. 10.1080/15548627.2016.1192751 Chen X.-C., Fang F., Zhu Y.-G., Chen L.-M., Zhou Y.-C., Chen Y. Protective effect of ginsenoside Rg1 on MPP+-induced apoptosis in SHSY5Y cells. J. Neural Transm. 2003; 110: 835-845. 10.1007/s00702-003-0005-y Choi K., Choi C. Proapoptotic Ginsenosides Compound K and Rh2 Enhance Fas-induced Cell Death of Human Astrocytoma Cells Through Distinct Apoptotic Signaling Pathways. Cancer Res. Treat. 2009; 41: 36-44. 10.4143/crt.2009.41.1.36 Sin S., Kim S.Y., Kim S.S. Chronic treatment with ginsenoside Rg3 induces Akt-dependent senescence in human glioma cells. Int. J. Oncol. 2012; 41: 1669-1674. 10.3892/ijo.2012.1604 Choi Y.J., Lee H.J., Kang D.W., Han I.H., Choi B.K., Cho W.H. Ginsenoside Rg3 induces apoptosis in the U87MG human glioblastoma cell line through the MEK signaling pathway and reactive oxygen species. Oncol. Rep. 2013; 30: 1362-1370. 10.3892/or.2013.2555. 23783960 Li B., Zhao J., Wang C.-Z., Searle J., He T.-C., Yuan C.-S., Du W. Ginsenoside Rh2 induces apoptosis and paraptosis-like cell death in colorectal cancer cells through activation of p53. Cancer Lett. 2011; 301: 185-192. 10.1016/j.canlet.2010.11.015 Lee M., Sorn S., Baek S., Jang S., Kim S. Antioxidant and apoptotic effects of Korean white ginseng extracted with the same ratio of protopanaxadiol and protopanaxatriol saponins in human hepatoma HepG2 cells. Ann. N. Y. Acad. Sci. 2009; 1171: 217-227. 10.1111/j.1749-6632.2009.04918.x Ghafouri-Fard S., Balaei N., Shoorei H., Hasan S.M.F., Hussen B.M., Talebi S.F., Taheri M., Ayatollahi S.A. The effects of Ginsenosides on PI3K/AKT signaling pathway. Mol. Biol. Rep. 2022; 49: 6701-6716. 10.1007/s11033-022-07270-y. 35220526 Jung D.-H., Nahar J., Mathiyalagan R., Rupa E.J., Ramadhania Z.M., Han Y., Yang D.-C., Kang S.C. A Focused Review on Molecular Signalling Mechanisms of Ginsenosides Anti-Lung Cancer and Anti-inflammatory Activities. Anti Cancer Agents Med. Chem. Former. Curr. Med. Chem. Anti Cancer Agents. 2023; 23: 3-14 Krata N., Zagożdżon R., Foroncewicz B., Mucha K. Oxidative Stress in Kidney Diseases: The Cause or the Consequence?. Arch. Immunol. Ther. Exp. 2018; 66: 211-220. 10.1007/s00005-017-0496-0. 29214330 Cui H., Kong Y., Zhang H. Oxidative stress, mitochondrial dysfunction, and aging. J. Signal Transduct. 2012; 2012: 646354. 10.1155/2012/646354. 21977319 Yokozawa T., Satoh A., Cho E.J. Ginsenoside-Rd attenuates oxidative damage related to aging in senescence-accelerated mice. J. Pharm. Pharmacol. 2004; 56: 107-113. 10.1211/0022357022449. 14980007 Oh M.H., Chung H.Y., Yong H.S., Kim K.W., Oura H., Yokozawa T. Effects of ginsenoside Rb2 on the antioxidants in SAM-R/1 mice. Korean Biochem. J. 1992; 25: 492-497 Choi W., Kim H.S., Park S.H., Kim D., Hong Y.D., Kim J.H., Cho J.Y. Syringaresinol derived from Panax ginseng berry attenuates oxidative stress-induced skin aging via autophagy. J. Ginseng Res. 2022; 46: 536-542. 10.1016/j.jgr.2021.08.003 Li G., Yang L., Feng L., Yang J., Li Y., An J., Li D., Xu Y., Gao Y., Li J. Syringaresinol protects against type 1 diabetic cardiomyopathy by alleviating inflammation responses, cardiac fibrosis, and oxidative stress. Mol. Nutr. Food Res. 2020; 64: 2000231. 10.1002/mnfr.202000231 Jung C.-H., Seog H.-M., Choi I.-W., Choi H.-D., Cho H.-Y. Effects of wild ginseng (Panax ginseng C.A. Meyer) leaves on lipid peroxidation levels and antioxidant enzyme activities in streptozotocin diabetic rats. J. Ethnopharmacol. 2005; 98: 245-250. 10.1016/j.jep.2004.12.030. 15814255 Yokozawa T., Wu Liu Z. The role of ginsenoside-Rd in cisplatin-induced acute renal failure. Ren. Fail. 2000; 22: 115-127. 10.1081/JDI-100100858. 10803758 Lu G., Liu Z., Wang X., Wang C. Recent advances in panax ginseng CA Meyer as a herb for anti-fatigue: An effects and mechanisms review. Foods. 2021; 101030. 10.3390/foods10051030. 34068545 Han S.Y., Kim J., Kim E., Kim S.H., Seo D.B., Kim J.-H., Shin S.S., Cho J.Y. AKT-targeted anti-inflammatory activity of Panax ginseng calyx ethanolic extract. J. Ginseng Res. 2018; 42: 496-503. 10.1016/j.jgr.2017.06.003 Ning C., Gao X., Wang C., Huo X., Liu Z., Sun H., Yang X., Sun P., Ma X., Meng Q. Hepatoprotective effect of ginsenoside Rg1 from Panax ginseng on carbon tetrachloride-induced acute liver injury by activating Nrf2 signaling pathway in mice. Environ. Toxicol. 2018; 33: 1050-1060. 10.1002/tox.22616 Ramadhania Z.M., Yang D.U., Moektiwardojo M., Han Y., Park J.K., Rupa E.J., Yang D.C., Lee S.J., Kang S.C. Enhanced Anti-Skin Aging Effects of Fermented Black Ginseng (Panax ginseng CA Meyer) by Aspergillus niger KHNT-1. Appl. Sci. 2023; 13550. 10.3390/app13010550 Lakshmi T., Anitha R., Geetha R. Panax ginseng–a universal panacea in the herbal medicine with diverse pharmacological spectrum–a review. Asian J. Pharm. Clin. Res. 2011; 4: 14-18 Patel S., Rauf A. Adaptogenic herb ginseng (Panax) as medical food: Status quo and future prospects. Biomed. Pharma Cotherapy. 2017; 85: 120-127. 10.1016/j.biopha.2016.11.112. 27930975 Shahrajabian M.H., Sun W., Cheng Q. A review of ginseng species in different regions as a multipurpose herb in traditional Chinese medicine, modern herbology and pharmacological science. J. Med. Plants Res. 2019; 13: 213-226 Acero N., Ortega T., Villagrasa V., Leon G., Muñoz-Mingarro D., Castillo E., González-Rosende M.E., Borrás S., Rios J.L., Bosch-Morell F. Phytotherapeutic alternatives for neurodegenerative dementias: Scientific review, discussion and therapeutic proposal. Phytother. Res. 2023. 10.1002/ptr.7727 Xu X.-F., Gao Y., Xu S.-Y., Liu H., Xue X., Zhang Y., Zhang H., Liu M.-N., Xiong H., Lin R.-C. Remarkable impact of steam temperature on ginsenosides transformation from fresh ginseng to red ginseng. J. Ginseng Res. 2018; 42: 277-287. 10.1016/j.jgr.2017.02.003 Zhao Q., Zhao N., Ye X., He M., Yang Y., Gao H., Zhang X. Rapid discrimination between red and white ginseng based on unique mass-spectrometric features. J. Pharm. Biomed. Anal. 2019; 164: 202-210. 10.1016/j.jpba.2018.10.007. 30391809 Tian X., Wang P., Li T., Huang X., Guo W., Yang Y., Yan M., Zhang H., Cai D., Jia X. Self-assembled natural phytochemicals for synergistically antibacterial application from the enlightenment of traditional Chinese medicine combination. Acta Pharm. Sin. B. 2020; 10: 1784-1795. 10.1016/j.apsb.2019.12.014 Watkins R., Wu L., Zhang C., Davis R.M., Xu B. Natural product-based nanomedicine: Recent advances and issues. Interna-Tional J. Nanomed. 2015; 10: 6055 Qiao L., Han M., Gao S., Shao X., Wang X., Sun L., Fu X., Wei Q. Research progress on nanotechnology for delivery of active ingredients from traditional Chinese medicines. J. Mater. Chem. B. 2020; 8: 6333-6351. 10.1039/D0TB01260B Kong W., Wei J., Abidi P., Lin M., Inaba S., Li C., Wang Y., Wang Z., Si S., Pan H. Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat. Med. 2004; 10: 1344-1351. 10.1038/nm1135. 15531889 Deng Y., Zhang X., Shen H., He Q., Wu Z., Liao W., Yuan M. Application of the nano-drug delivery system in treatment of cardiovascular diseases. Front. Bioeng. Biotechnol. 2020; 7: 489. 10.3389/fbioe.2019.00489 Yu H., Teng L., Meng Q., Li Y., Sun X., Lu J., Lee R.J., Teng L. Development of liposomal Ginsenoside Rg3: Formulation optimization and evaluation of its anticancer effects. Int. J. Pharm. 2013; 450: 250-258. 10.1016/j.ijpharm.2013.04.065. 23628402 Ke Y., Huang L., Song Y., Liu Z., Liang L., Wang L., Wang T. Preparation and pharmacological effects of minor ginsenoside nanoparticles: A review. Front. Pharmacol. 2022; 13: 974274. 10.3389/fphar.2022.974274 Wang X., Zheng W., Shen Q., Wang Y., Tseng Y., Luo Z., Wang X., Shi L., Li C., Liu J. Identification and construction of a novel biomimetic delivery system of paclitaxel and its targeting therapy for cancer. Signal Transduct. Target. Ther. 2021; 6: 33. 10.1038/s41392-020-00390-6 Alippilakkotte S., Sreejith L. Pectin mediated synthesis of curcumin loaded poly(lactic acid) nanocapsules for cancer treat-ment. J. Drug Deliv. Sci. Technol. 2018; 48: 66-74. 10.1016/j.jddst.2018.09.001 Kumari M., Sharma N., Manchanda R., Gupta N., Syed A., Bahkali A.H., Nimesh S. PGMD/curcumin nanoparticles for the treatment of breast cancer. Sci. Rep. 2021; 11: 3824. 10.1038/s41598-021-81701-x. 33589661 Tan C., Xie J., Zhang X., Cai J., Xia S. Polysaccharide-based nanoparticles by chitosan and gum arabic polyelectrolyte complexation as carriers for curcumin. Food Hydrocoll. 2016; 57: 236-245. 10.1016/j.foodhyd.2016.01.021 Martínez-Ávila M., Guajardo-Flores D., Antunes-Ricardo M. Nanoformulations applied to the delivery of phenolic com-pound. Phytochemical Nanodelivery Systems as Potential Biopharmaceuticals; Elsevier: Amsterdam, The Netherlands. 2023: 283-325 Yan L., Wang R., Wang H., Sheng K., Liu C., Qu H., Ma A., Zheng L. Formulation and characterization of chitosan hy-drochloride and carboxymethyl chitosan encapsulated quercetin nanoparticles for controlled applications in foods system and simulated gastrointestinal condition. Food Hydrocoll. 2018; 84: 450-457. 10.1016/j.foodhyd.2018.06.025 Choudhary A., Kant V., Jangir B.L., Joshi V.G. Quercetin loaded chitosan tripolyphosphate nanoparticles accelerated cu-taneous wound healing in Wistar rats. Eur. J. Pharmacol. 2020; 880: 173172. 10.1016/j.ejphar.2020.173172 Bhatt P., Fnu G., Bhatia D., Shahid A., Sutariya V. Nanodelivery of Resveratrol-Loaded PLGA Nanoparticles for Age-Related Macular Degeneration. AAPS PharmSciTech. 2020; 21: 291. 10.1208/s12249-020-01836-4 Saha M., Saha D.R., Ulhosna T., Sharker S.M., Shohag M.H., Islam M.S., Ray S.K., Rahman G.M.S., Reza H.M. QbD based development of resveratrol-loaded mucoadhesive lecithin/chitosan nanoparticles for prolonged ocular drug delivery. J. Drug Deliv. Sci. Technol. 2021; 63: 102480. 10.1016/j.jddst.2021.102480 Chmielowski R.A., Abdelhamid D.S., Faig J.J., Petersen L.K., Gardner C.R., Uhrich K.E., Joseph L.B., Moghe P.V. Athero-inflammatory nanotherapeutics: Ferulic acid-based poly (anhydride-ester) nanoparticles attenuate foam cell for-mation by regulating macrophage lipogenesis and reactive oxygen species generation. Acta Biomater. 2017; 57: 85-94. 10.1016/j.actbio.2017.05.029. 28522412 Khan S.U., Fatima K., Aisha S., Hamza B., Malik F. Redox balance and autophagy regulation in cancer progression and their therapeutic perspective. Med. Oncol. 2022; 40: 12. 10.1007/s12032-022-01871-0 Chodari L., Dilsiz Aytemir M., Vahedi P., Alipour M., Vahed S.Z., Khatibi S.M.H., Ahmadian E., Ardalan M., Eftekhari A. Targeting mitochondrial biogenesis with polyphenol compounds. Oxidative Med. Cell. Longev. 2021; 2021: 4946711. 10.1155/2021/4946711. 34336094 Zhao B., Wang X., Liu H., Lv C., Lu J. Structural characterization and antioxidant activity of oligosaccharides from Panax ginseng C. A. Meyer. Int. J. Biol. Macromol. 2020; 150: 737-745. 10.1016/j.ijbiomac.2020.02.016 Paik D.J., Lee C.H. Review of cases of patient risk associated with ginseng abuse and misuse. J. Ginseng Res. 2015; 39: 89-93. 10.1016/j.jgr.2014.11.005

By Md Niaj Morshed; Jong Chan Ahn; Ramya Mathiyalagan; Esrat Jahan Rupa; Reshmi Akter; Md. Rezaul Karim; Dae Hyo Jung; Dong Uk Yang; Deok Chun Yang and Seok Kyu Jung

Reported by Author; Author; Author; Author; Author; Author; Author; Author; Author; Author

Titel:
Antioxidant Activity of Panax ginseng to Regulate ROS in Various Chronic Diseases
Autor/in / Beteiligte Person: Md Niaj Morshed ; Jong Chan Ahn ; Mathiyalagan, Ramya ; Esrat Jahan Rupa ; Akter, Reshmi ; Md. Rezaul Karim ; Dae Hyo Jung ; Dong Uk Yang ; Deok Chun Yang ; Seok Kyu Jung
Link:
Zeitschrift: Applied Sciences, Jg. 13 (2023-02-23), S. 2893-2893
Veröffentlichung: MDPI AG, 2023
Medientyp: unknown
ISSN: 2076-3417 (print)
DOI: 10.3390/app13052893
Schlagwort:
  • Fluid Flow and Transfer Processes
  • Process Chemistry and Technology
  • General Engineering
  • General Materials Science
  • Instrumentation
  • Computer Science Applications
Sonstiges:
  • Nachgewiesen in: OpenAIRE
  • Rights: OPEN

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -