ArticlePDF AvailableLiterature Review

Beneficial effects of Panax ginseng for the treatment and prevention of neurodegenerative diseases: Past findings and future directions

Authors:
  • Gachon University College of Korean Medicine

Abstract

In recent years, several therapeutical drugs have been rationally designed and synthesized based on the novel knowledge gained from investigating the actions of biologically active chemicals derived from foods, plants, and medicinal herbs. One of the major advantages of these naturalistic chemicals is their ability to interact with multiple targets in the body resulting in a combined beneficial effect. Ginseng is a perennial herb (Araliaceae family), a species within the genus Panax, and a highly valued and popular medicinal plant. Evidence for the medicinal and health benefits of Panax ginseng and its components in preventing neurodegeneration has increased significantly in the past decade. The beneficial effects of Panax ginseng on neurodegenerative diseases have been primarily attributed to the antioxidative and immunomodulatory activities of its ginsenosides components. Mechanistic studies on the neuroprotective effects of ginsenosides revealed that they act not only as antioxidants but also as modulators of intracellular neuronal signaling and metabolism, cell survival/death genes, and mitochondrial function. The goal of the present paper is to provide a brief review of recent knowledge and developments concerning the beneficial effects as well as the mechanism of action of Panax ginseng and its components in the treatment and prevention of neurodegenerative diseases.
Accepted Manuscript
Beneficial effects of Panax ginseng for the treatment and prevention of
neurodegenerative diseases: past findings and future directions
Ki Hyun Kim, Dahae Lee, Hye Lim Lee, Chang-Eop Kim, Kiwon Jung, Ki Sung Kang
PII: S1226-8453(16)30269-X
DOI: 10.1016/j.jgr.2017.03.011
Reference: JGR 270
To appear in: Journal of Ginseng Research
Received Date: 16 November 2016
Accepted Date: 15 March 2017
Please cite this article as: Kim KH, Lee D, Lee HL, Kim C-E, Jung K, Kang KS, Beneficial effects of
Panax ginseng for the treatment and prevention of neurodegenerative diseases: past findings and future
directions, Journal of Ginseng Research (2017), doi: 10.1016/j.jgr.2017.03.011.
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
1
Running title: Neuroprotective evidence of ginseng
1
2
3
Beneficial effects of Panax ginseng for the treatment and prevention of neurodegenerative
4
diseases: past findings and future directions
5
6
Ki Hyun Kim
a,
, Dahae Lee
a,
, Hye Lim Lee
b
, Chang-Eop Kim
b
, Kiwon Jung
c,*
, Ki Sung Kang
b,*
7
8
9
10
a
School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Korea
11
b
College of Korean Medicine, Gachon University, Seongnam 13120, Korea
12
c
Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam 13844,
13
Korea
14
15
16
These authors contributed equally to the work described in this study.
17
*
Corresponding authors:
18
Tel.:82-31-881-7173; Fax: 82-31-850-9315: E-mail: pharmj@cha.ac.kr (Jung KW)
19
Tel.: 82-31-750-5402; Fax: 82-31-750-5416; E-mail: kkang@gahon.ac.kr (Kang KS)
20
21
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
2
ABSTRACT
22
In recent years, several therapeutical drugs have been rationally designed and synthesized
23
based on the novel knowledge gained from investigating the actions of biologically active chemicals
24
derived from foods, plants, and medicinal herbs. One of the major advantages of these naturalistic
25
chemicals is their ability to interact with multiple targets in the body resulting in a combined
26
beneficial effect. Ginseng is a perennial herb (Araliaceae family), a species within the genus Panax,
27
and a highly valued and popular medicinal plant. Evidence for the medicinal and health benefits of
28
Panax ginseng and its components in preventing neurodegeneration has increased significantly in the
29
past decade. The beneficial effects of Panax ginseng on neurodegenerative diseases have been
30
primarily attributed to the antioxidative and immunomodulatory activities of its ginsenosides
31
components. Mechanistic studies on the neuroprotective effects of ginsenosides revealed that they act
32
not only as antioxidants but also as modulators of intracellular neuronal signaling and metabolism,
33
cell survival/death genes, and mitochondrial function. The goal of the present paper is to provide a
34
brief review of recent knowledge and developments concerning the beneficial effects as well as the
35
mechanism of action of Panax ginseng and its components in the treatment and prevention of
36
neurodegenerative diseases.
37
38
KEYWORDS: Panax ginseng; Gginsenosides; Antioxidant; Neurodegenerative diseases
39
40
41
42
43
44
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
3
1. Introduction
45
1. 1. Classification, structures, and chemical properties of ginseng components
46
Ginseng is a perennial herb (Araliaceae family), a species within the genus Panax, and a
47
highly valued and popular medicinal plant [1]. The name Ginseng originates from the Chinese
48
words Jen Sheng and meansman-herb because of the human-like shape of the root or rhizome
49
of the plant. The word Panax means cure all and describes the traditional belief that ginseng has
50
properties that heal all bodily diseases. So far, 14 plants, including 12 species and two infraspecific
51
taxa, have been classified under the genus Panax [2]. The three major commercial ginseng sorts are
52
the Korean ginseng (Panax ginseng Meyer), the Chinese ginseng (Panax notoginseng (Burk.) F. H.
53
Chen), and the American ginseng (Panax quinquefolius L.), and they have been used worldwide as
54
herbal medicines for thousands of years [3].
55
Korean ginseng (Panax ginseng Meyer) is a well-known medicinal herb cultivated in eastern
56
Asian countries [4]. Panax ginseng is native to China and Korea but is now widely cultivated in other
57
countries like Japan, Russia, the United States, and Canada. The root of the Korean ginseng has been
58
traditionally used to treat various diseases, particularly as an adaptogen since it is suggested to
59
normalize body functions and increase physical strength [5,6]. Because fresh ginseng tends to be
60
easily degraded at room temperature, it has been traditionally processed into white ginseng through
61
air-drying of the root, or into red ginseng through root steaming followed by drying [7-9]. In Korea,
62
red ginseng and various processed ginseng products are used popularly as functional foods or
63
nutritional supplements. Based on recent studies, red ginseng has been reported to have biological
64
benefits while inducing fewer side effects compared to fresh and white ginseng [7,10-14]. In addition,
65
Korean red ginseng is known to possess various biological activities including boosting the immune
66
system, improving the blood circulation, enhancing memory, anti-fatigue effects, antioxidant effects,
67
and positive effects on menopausal disorder [10-14].
68
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
4
Korean ginseng is known to have various therapeutic benefits mediated by its well studied active
69
components [15-21]. Indeed, Korean ginseng is reported to contain various functional constituents,
70
including most notably ginseng saponins (also called ginsenosides), polyacetylenes, phenolic
71
compounds, sesquiterpenes, alkaloids, polysaccharides, and oligopeptides [22].
72
73
1.2. Ginsenosides
74
First attempts to isolate ginsenosides happened in the 1960s [23,24] and most were identified
75
from the Panax species. Ginsenosides are biosynthesized from 2,3-oxidosqualene, which leads to the
76
formation of cycloartenol, dammarenediol-II, and β-amyrin by the action of three different enzymes.
77
Dammarenediol-II and β-amyrin are eventually biotransformed into ginsenosides [2]. Based on their
78
chemical structures, ginsenosides are typically divided into two groups: a four-ring dammarane-type
79
and a five-ring oleanane-type. Dammarenediol-II is the precursor of the dammarane-type, including
80
ginsenosides Rb1, Rb2, Re, and Rg1, which account for a significant portion of ginsenosides found in
81
ginseng species. Oleanane-type, on the other hand, are biosynthesized from β-amyrin. However,
82
oleanane-type ginsenosides such as Ro are rare and often undetectable
i
n Panax ginseng.
83
Dammarane-type ginsenosides are further classified into two groups: protopanaxadiols (PPD) and
84
protopanaxatriols (PPT). Dammarenediol-II is hydroxylated into PPD, 3β,12β,20-trihydroxydammar-
85
24-ene. Consequently, a number of ginsenosides are biosynthesized by the O-glycosylation of PPD,
86
which involves the attachment of saccharides to carbon (C)-3 and/or C-20. PPD-type ginsenosides
87
include Rb1, Rb2, Rc, Rd, Rg3, Rh2, and Rh3 (Fig. 1). Dammarenediol-II is further hydroxylated
88
into PPT, 3β,6α,12β,20-tetrahydroxydammar-24-ene. A variety of ginsenosides are biosynthesized
89
by O-glycosylation of PPT, which involves the linkage of saccharides to C-6 and/or C-20. Typically,
90
the hydroxyl group at C-3 remains free in PPT-type ginsenosides. Typical PPT-type ginsenosides in
91
Panax ginseng are Re, Rf, Rg1, Rg2, and Rh1 (Fig. 1). While most naturally occurring ginsenosides
92
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
5
are of the (S)-configuration at C-20, some artifactual ginsenosides exist in two epimeric forms at the
93
carbon. The pseudoginsenoside F11 belongs to the PPT group although the carbon chain at C-20 is
94
replaced by a tetrahydrofuran ring (Fig. 1). Several new ginsenosides such as 25-OH-PPD and 25-
95
OHPPT were recently isolated from ginseng berries [25]. Four malonyl derivatives of ginsenosides
96
Rb1, Rb2, Rc and Rd have also been reported [25]. The malonyl derivatives and ginsenosides Ro are
97
also called acidicginsenosides while the others are calledneutralginsenosides [26].
98
Heat treatment induces deglycosylation of ginsenosides. As a result, red ginseng has relatively
99
high concentrations of the less polar ginsenosides transformed from fresh ginseng ginsenosides. Red
100
ginseng contains ginsenosides Rg2, Rg6, F4, 20(E)-F4, Rh1, Rh4, Rk3, Rg3, Rg5, Rz1, Rk1, Rg9,
101
and Rg10, which are converted from the major ginsenosides Rb1, Rb2, Rc, Rd, Rg1, and Re [7].
102
Generally, ginsenosides deglycosylation during the process of red ginseng production results in these
103
conversions: [Rg1 Rh1 (Rh4, Rk3)], [Re Rg2 (F4, Rg6)], [Rf (Rg9, 20Z-Rg9, Rg10)],
104
and [(Rb1, Rc, Rb2, Rd) Rg3 (Rg5, Rk1, Rz1)] [7]. These results are consistent with the
105
experimental evidence that the levels of the less polar ginsenosides such as Rg2, Rh1, and Rg3
106
progressively increase, whereas the levels of the natural ginsenosides such as Rg1, Re, Rb1, Rc, and
107
Rd progressively decrease during the heat-processed red ginseng production [27].
108
109
1.3. Polyacetylenes
110
Polyacetylenes are representative non-saponin components of ginseng. The first polyacetylene
111
identified and extracted from Panax ginseng was panaxynol [28]. Since then, many polyacetylenic
112
substances, including panaxydol and ginsenoynes A-E, have been identified and extracted from
113
Panax ginseng (Fig. 2) [22]. Panaxytriol is a hydrated compound with an epoxy ring derived from
114
panaxydol by heat and acid treatment (Fig. 2). These Panax ginseng polyacetylenes are believed to
115
possess anti-cancer properties. However, their in vivo efficacy has not been determined due to their
116
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
6
chemical instability.
117
118
1.4. Phenolic compounds
119
Phenolic compounds generally possess antioxidative and anti-cancer biological properties.
120
However, phenolic compounds found in ginseng are relatively less investigated. More than 10
121
phenolic compounds have previously been reported in fresh and/or processed ginseng (Fig. 2). These
122
include salicylic acid, vanillic acid, ascorbic acid, p-coumaric acid, ferulic acid, caffeic acid, gentisic
123
acid, p-hydroxybenzoic acid, maltol, cinnamic acid, protocatechuic acid, syringic acid, and quercetin
124
[29]. A recent study revealed that chlorogenic acid, gentisic acid, p- and m-coumaric acid, and rutin
125
are the major phenolic compounds in 36 year old ginseng fruit, leaves, and roots [30]. Korean
126
ginseng, which is suggested to provide more health benefits than other ginseng species, usually
127
contains more phenolic compounds than Chinese ginseng [31].
128
129
1.5. Sesquiterpenes
130
A number of sesquiterpene hydrocarbons as well as oxygenated sesquiterpenes have been
131
identified as volatile constituents of the Panax ginseng. More than 15 sesquiterpenes have been
132
identified as volatile constituents of the P. ginseng. These include sesquiterpene hydrocarbons like β-
133
panasinsene, african-2-ene, β-elemene, calarene, (E)-β-farnesene, α-humulene, α-neoclovene, 2-epi-
134
(E)-β-caryophyllene, β-neoclovene, β-selinene, and bicyclogermacrene and oxygenated
135
sesquiterpenes like spathulenol, humulene epoxide II, ginsenol, hexadecanoic acid, and falcarinol
136
(Fig. 2) [32,33].
137
138
1.6. Alkaloids
139
Alkaloids are another non-saponin component of Korean ginseng and include 1-
140
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
7
carbomethoxy-β-carboline, N
9
-formylharman, harman, norharman, perlolyrine, 4-methyl-5-
141
thiazoleethanol, and spinacine (Fig. 2) [22]. Recently, a new indole alkaloid, ginsenine, with a seven-
142
membered lactam unit, was isolated from Panax ginseng berries [34]. These alkaloids are minor
143
components of Panax ginseng and their biological activities are also limited.
144
145
1.7. Polysaccharides
146
Korean ginseng contains various polysaccharides. The hypoglycemic glycans, panasans A-E,
147
panaxans I-L, panaxans M-P, and panaxans Q-U have been isolated from the roots of Panax ginseng
148
[3,35-37]. It has been recognized that the composition of the polysaccharides vary depending on
149
strains and/or places of production [35,37]; however, acid hydrolysis, reduction, acetylation followed
150
by gas-liquid chromatography of these glycans showed that they consist of diverse combination of
151
neutral sugars including rhamnose, mannose, galactose, arabinose, galactose and glucose. In addition,
152
the immunomodulating g1lycans, ginsenan PA and ginsenan PB, were identified in the Panax
153
ginseng root [38]. These immunomodulating glycans are composed of L-arabinose, D-galactose, L-
154
rhamnose, D-galacturonic acid, and D-glucuronic acid, but their exact structure is still unknown.
155
Other immunomodulating glycans, such as acidic polysaccharide ginsenan S-IA and ginsenan S-II A,
156
have also been identified. On the other hand, ginseng polysaccharides are mainly composed of
157
neutral polysaccharides (starch-like glucans) and acidic substances (ginseng pectin) [39]. Ginseng
158
pectins have been reported to show a wider range of pharmacological activities compared to neutral
159
polysaccharides [40,41], and they are known to be composed of galacturonic acid, galactose, glucose,
160
arabinose, rhamnose, glucuronic acid, and mannose [41], however their exact structure is also
161
unknown.
162
163
164
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
8
2. Basic and clinical evidences on the beneficial effects of Panax ginseng on neurodegenerative
165
diseases
166
167
2.1. Alzheimer's disease
168
Alzheimer’s disease is a neurodegenerative disorder that affects the central nervous system
169
(CNS) and results in a loss of memory and basic motor functions. It is one of the most common
170
causes of mental deterioration in elderly people, and accounts for around 50–60% of the overall cases
171
of dementia [42,43]. The major brain areas affected in Alzheimer’s disease include the cerebral
172
cortex, locus coeruleus, nucleus basalis of Meynert, and hippocampus [44]. The pathological
173
characteristics include extracellular deposits of amyloid β (derived from amyloid precursor protein)
174
in senile plaques, intracellular formation of neurofibrillary tangles (containing an abnormally
175
phosphorylated form of tau, a microtubule associated protein), and loss of neuronal synapses and
176
pyramidal neurons [42,45].
177
Since the exact mechanism underlying Alzheimer’s disease is not fully understood, current
178
therapies are largely based on a number of theories/hypotheses regarding the pathogenesis of the
179
disease. The cholinergic hypothesis of Alzheimer’s disease is based on the reported presynaptic
180
deficits observed Alzheimer’s disease affected brains and on the role of the cholinergic system in
181
animal and human behavior [42,46]. The most common treatment strategy in Alzheimer’s disease
182
involves acetylcholine (ACh), an important neurotransmitter in cognition and memory processes, that
183
is known to be decreased in Alzheimer’s disease. Treatment options include the use of ACh
184
precursors, ACh-releasing agents, and acetylcholinesterase (AChE) inhibitors [47,48]. Other
185
therapeutical interventions, although with fewer proven beneficial effects, also involve antioxidative
186
agents that scavenge free radicals and anti-inflammatory agents that treat the amyloid β cascade [48].
187
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
9
Basic and clinical evidence of the beneficial effects of Panax ginseng on Alzheimer’s disease
188
are summarized in Table 1. In males Sprague-Dawley rats, Panax ginseng extracts have been
189
reported to exert neuroprotective effects by ameliorating the advanced glycation end product-induced
190
memory impairment and mitigating the Alzheimer-like pathophysiological changes through
191
downregulation of the RAGE/NF-κB pathway [49]. Moreover, ginsenosides attenuated d-galactose-
192
and AlCl
3
-induced spatial memory impairment and Alzheimer-like pathophysiological changes in
193
male Wistar rats through restoration of amyloid β formation, tau phosphorylation, and the function of
194
various neurotransmitters including Glu, Asp, GABA, ACh, DA, Gly and 5-HT [50]. Ginsenosides
195
are reported to improve memory loss in C57BL/6J mice with severe hippocampal damage and in
196
aged SAMP8 mice (senescence-accelerated mouse) by upregulating plasticity-related proteins such as
197
PSD-95, PKCγ and BDNF [51].
198
Ginsenoside Rb1 protected against amyloid β-induced neurotoxicity in SH-SY5Y cells by
199
regulating the CAP1, CAPZB, TOMM40, and DSTN proteins related to the actin cytoskeleton
200
organization and by decreasing the levels of apoptotic proteins like PARP-1 and Bax [52]. The
201
expression of BDNF, a key modulator of neuronal survival, activity, and synaptic transmission, and a
202
key player in hippocampal-dependent learning and memory, was increased in male ICR mice treated
203
with ginsenoside Rh1, resulting in enhanced survival of dentate gyrus cells. Ginsenoside Rh1 was
204
also reported to protect newborn neurons from death during the neuronal differentiation process
205
[53,54].
206
Ginsenoside Rg5 improved cognition and amyloid β deposition in a Wistar rat model by
207
increasing IGF-1 and BDNF levels and decreasing TNF-α and IL-1β as well as COX-2 and iNOS
208
levels [55]. Ginsenoside Rg5 and Rh3 reversed scopolamine-induced memory deficits in male ICR
209
mice by inhibiting AChE activity and increasing BDNF expression and CREB activation [56].
210
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
10
Ginsenoside Rg1 reduced the mRNA and protein expressions of TLR3, TLR4, NF-κB, and TRAF-6
211
and down-regulated the levels of TNF-α and IFN-β in a NG108-15 neuroglial cell line stimulated by
212
amyloid β peptide 2535 (Aβ2535) [57]. Moreover, ginsenoside Rg1 attenuated okadaic acid-
213
induced memory impairment in male Sprague-Dawley rats through the GSK3β/tau signaling pathway
214
and the
prevention of amyloid β formation [58].
215
The potential efficacy of a heat-processed form of ginseng on cognitive function and
216
behavioral symptoms has been recently reported in a clinical study in patients with moderately severe
217
Alzheimer’s disease. Indeed, ginseng-treated patients showed a significant improvement on the Mini-
218
Mental State Examination (MMSE) and Alzheimer's Disease Assessment Scale (ADAS). Moreover,
219
patients treated with higher ginseng doses (4.5 g/day) showed further improvements in their ADAS
220
cognitive, ADAS non-cognitive, and MMSE scores as early as 12 weeks following treatment. This
221
improvement was sustained over a follow-up period of 24 weeks [59]. However, the effects of
222
ginseng on Alzheimer’s disease remain inconclusive as reported in a recent meta-analysis study
223
including seven main databases for randomized clinical trials [60]. The main limitations of the
224
available studies are small sample size, poor methodological qualities, and the absence of placebo
225
controls [60].
226
227
2.2. Parkinsons Disease
228
Parkinson's disease is a neurodegenerative disorder commonly affecting the elderly. It is
229
characterized by degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc)
230
as well as other regions of the CNS [61-65]. Dopaminergic degeneration is typically associated with
231
the
presence of protein deposits, called Lewy bodies, in the neuronal cytoplasm and thread-like
232
proteinaceous inclusions, called Lewy neurites, within neuronal neurites [66-68]. The main clinical
233
symptoms include resting tremor, bradykinesia, rigidity, and
postural instability, which result in
234
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
11
impaired movement and other neurological dysfunctions. The current understanding of the
235
pathophysiology of Parkinson's disease largely stemmed from elegant neurochemical investigations
236
in the 1950s-60s that demonstrated over 80% reduction in striatal dopamine along with the loss of
237
SNpc dopaminergic neurons in most Parkinson's disease patients.
238
While the pathogenic mechanism of human Parkinson's disease is still not fully understood,
239
oxidative stress and cytotoxicity are thought to play an important role in the degeneration of
240
dopaminergic neurons [63,64,69-74]. Mechanistically, it is known that dopamine neurotransmitter is
241
chemically labile and its oxidation products, such as dopamine quinones and semi-quinones, are
242
highly cytotoxic to neurons in general and dopaminergic neurons in particular [63,64,69]. Elevated
243
formation of these neurotoxic intermediates contributes to neuronal damage and degeneration. This
244
mechanistic hypothesis is supported by many in vitro as well as in vivo studies.
245
Basic and clinical evidence of the beneficial effects of Panax ginseng on Parkinson's disease
246
are summarized in Table 2. Extracts of Panax ginseng had neuroprotective effects on 1-methyl-4-
247
phenylpyridinium ion (MPP+)-induced apoptosis in SH-SY5Y cells through decreasing the levels of
248
apoptotic proteins like Bax, Bcl-2, cytochrome c, and cleaved caspase-3 [75]. In both in vivo
249
(C57BL/6J mice) and in vitro (PC12 cells) models of Parkinson's disease, ginsenoside Rg1 exerted
250
neuroprotective effects through the Wnt/β-catenin signaling pathway including Wnt-1, β-catenin,
251
GSK-3β, and p-GSK-3β. Neuroprotective effects of ginsenoside Rg1 on MPP+-induced apoptosis in
252
PC12 cells were also mediated through the decrease in apoptotic proteins levels including Bcl-xL and
253
cleaved caspase-3 [76]. Ginsenoside Rd was also shown to exert neuroprotective effects on MPP+-
254
induced apoptosis in SH-SY5Y cells by decreasing the levels of apoptotic proteins including P-Akt,
255
Bax, and Bcl-2 [77].
256
257
2.3. Brain Ischemia and Stroke
258
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
12
Stroke is the third leading cause of death in the industrialized world and the leading cause of
259
disability [78]. There are two mechanistically distinct modes of cerebral ischemia, namely global and
260
focal ischemia. Global ischemia commonly develops after transient cardiac arrest. The typical
261
histological picture following global ischemic insults is described by delayed neuronal death sparing
262
glial cells. Under normothermic conditions, 10 minutes of global brain ischemia is lethal in humans.
263
Focal ischemia occurs after transient or permanent flow reduction in the territory of a cerebral artery
264
resulting from embolic or thrombotic vessel occlusion. The typical histological picture following
265
focal ischemia is a pan-necrosis that includes all brain cell types [78].
266
Basic and clinical evidence of the beneficial effects of Panax ginseng on stroke are
267
summarized in Table 3. In male Sprague–Dawley rats, ginsenoside Rd has been reported to protect
268
against ischemic cerebral damage by promoting clearance of extracellular glutamate through the up-
269
regulation of GLT-1 expression via PI3K/AKT and ERK1/2 pathways [79]. Moreover, administration
270
of ginsenoside Rd increased the expression of non-selective cation channels including TRPM7,
271
ASIC1a, and ASIC2a [80], and decreased the levels of apoptotic proteins like CytoC, AIF, and
272
caspase-3 [81]. Post-ischemic synthesis of two damaging enzymes, COX-2 and iNOS, were also
273
significantly decreased by ginsenoside Rd [82]. Ginsenoside Rb1 was also reported to promote
274
extracellular glutamate clearance by up-regulating GLT-1 expression via PI3K/AKT and ERK1/2
275
pathways in male Sprague-Dawley rats [83].
276
Although no prospective clinical trials are available for Panax ginseng, a multi-center,
277
double-blinded and randomized controlled clinical trial of 140 Chinese patients demonstrated that a
278
low dose of aspirin (50 mg per day) combined with notogiseng capsules (200 mg, three times a day)
279
significantly ameliorated neurological deficits and improved daily life activities compared to patients
280
treated with aspirin alone [84].
281
282
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
13
2.4. Huntington's Disease
283
Huntington's disease is a neurodegenerative disorder caused by a CAG repeat expansion in
284
the gene encoding for the huntingtin protein [85,86]. Clinical symptoms of
Huntington's disease
285
comprise adult-onset personality changes, generalized motor dysfunctions, and cognitive decline. The
286
peak age of adult-onset Huntington's disease is between 35 and 50 years [85,87]. Commonly reported
287
symptoms include progressive weight loss, alterations in sexual behavior, and disturbances in the
288
wake-sleep cycle that occur very early during the course of the disease possibly due to hypothalamic
289
dysfunction [88]. At later disease stages, characteristic symptoms include motor impairments,
290
progressive dementia, and gradual impairment of mental processes involved in comprehension,
291
reasoning, judgment, and memory [89,90]. Most affected patients eventually succumb to the disease
292
due to aspiration pneumonia caused by swallowing difficulties [89].
293
Basic evidence of the beneficial effects of Panax ginseng on Parkinson's disease are
294
summarized in Table 4. In a cellular model of Huntington's disease with primary medium spiny
295
striatal neuronal cultures, ginsenosides Rb1, Rc and Rg5 exerted protective effects on glutamate-
296
induced apoptosis and were suggested as a potential treatment choice [91]. In a Sprague-Dawley rat
297
model of Huntington's disease, PPT were reported to have neuroprotective effects on 3-nitropropionic
298
acid-induced oxidative stress in males. Oral administration of PPT resulted in marked improvements
299
in body weight and locomotor activity. Beneficial effects of PPT were mediated by increasing the
300
Nrf2 entry into the nucleus while enhancing the expression of HO-1 and NAD(P)H quinone oxidase 1
301
in the striatum [92].
302
303
3. Concluding Remarks and Future Perspectives
304
Oxidative stress and dysregulation of the inflammatory network are being recognized as
305
important components in the pathogenesis of neurodegenerative diseases [93-95]. Oxidative stress
306
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
14
has been linked to neuronal cell death associated with certain neurodegenerative conditions [96,97].
307
Because of its high metabolic rate and relatively reduced capacity for cellular regeneration compared
308
with other organs, the brain is believed to be particularly susceptible to the damaging effects of
309
reactive oxygen species (ROS). An acute oxidative insult to brain tissue can amplify ROS generation,
310
increase the accumulation of oxidized biomolecules, and promote oxidative stress [98]. Accumulation
311
of ROS in the brain stimulates the oxidation of lipids [99], protein [100], and DNA [101], which are
312
characteristic changes of many neuronal pathologies.
313
In the case of Parkinsons and Alzheimers diseases, various indices of ROS damage have
314
been reported within specific brain regions that undergo selective neurodegeneration [102-104].
315
Many researchers in the neurodegenerative field are seeking ways to modulate or emulate the
316
protective effects of key enzymatic components that regulate oxidative stress, with the aim of
317
developing rational drugs or genetic therapies [105,106].
318
A growing number of studies have demonstrated the efficacy of ginseng components
319
extracted from its fruit, roots, and leaves in reducing or blocking neuronal death in various
320
experimental neurodegeneration models [56,60]. Ginseng components, particularly ginsenosides, are
321
capable of protecting neurons both in vitro and in vivo by modulating biological processes including
322
oxidative stress, excitotoxicity, apoptotic neuronal death, and the kinase and ubiquitin-proteasome
323
signaling pathways [20,107]. Indeed, ginsenosides are receiving increasing interest from consumers
324
as well as researchers because of their unique ability to prevent neurodegeneration [108,109].
325
Extensive research over the last 10 years has indicated that components derived from Panax ginseng
326
target ROS and therefore may prevent neurodegenerative diseases [20,110].
327
Evidence for the medicinal and health benefits of Panax ginseng and its components in
328
preventing neurodegenerative diseases is increasing [20,56,111-113]. The current clinical results did
329
not report any serious adverse effects of ginseng [60], but it may alter the blood haemostasis and
330
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
15
anticoagulation with warfarin [108]. The beneficial effects of ginseng have been attributed to the
331
presence of ginsenosides that are powerful antioxidants and free iron scavengers. Mechanistic studies
332
on the neuroprotective effects of ginsenosides revealed that they act not only as antioxidant metal
333
chelators but also as modulators of intracellular neuronal signaling and metabolism, cell
334
survival/death genes, and mitochondrial function. It has been shown that ginsenosides modulate
335
caspase-dependent and caspase-independent programmed cell death. Indeed, several ginsenosides
336
significantly inhibit the activation of caspase-3, key apoptotic player, and are able to modulate
337
mitogen-activated protein kinases known to play an important role in neuronal apoptosis. However,
338
findings from clinical studies on ginseng for neurodegenerative diseases showed that the effects of
339
ginseng were still inconclusive. The main limitations of the available studies were small sample size,
340
poor methodological qualities, and absence placebo controls. Larger, well-designed clinical studies
341
are a prerequisite to successfully elucidate the effect of ginseng on neurodegenerative diseases.
342
343
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
16
REFERENCES
344
[1] Helms S. Cancer prevention and therapeutics: Panax ginseng. Altern Med Rev 2004; 9: 259-
345
274.
346
[2] Shin BK, Kwon SW, Park JH. Chemical diversity of ginseng saponins from Panax ginseng. J
347
Ginseng Res 2015; 39: 287-298.
348
[3] Kim DH. Chemical diversity of Panax ginseng, Panax quinquifolium, and Panax notoginseng.
349
J Ginseng Res 2012; 36: 1-15.
350
[4] Park JD, Rhee DK, Lee YH. Biological activities and chemistry of saponins from Panax
351
ginseng CA Meyer. Phytochem Rev 2005; 4: 159-175.
352
[5] Liu CX, Xiao PG. Recent advances on ginseng research in China. J Ethnopharmacol 1992;
353
36: 27-38
354
[6] Wang J, Li S, Fan Y, Chen Y, Liu D, Cheng H, Gao X, Zhou Y. Anti-fatigue activity of the
355
water-soluble polysaccharides isolated from Panax ginseng CA Meyer. J Ethnopharmacol
356
2010; 130: 421-423.
357
[7] Lee SM, Bae BS, Park HW, Ahn NG, Cho BG, Cho YL, Kwak YS. Characterization of
358
Korean Red Ginseng (Panax ginseng Meyer): History, preparation method, and chemical
359
composition. J Ginseng Res 2015; 39: 384-391.
360
[8] Han MS, Han IH, Lee D, An JM, Kim SN, Shin MS, Yamabe N, Hwang GS, Yoo HH, Choi
361
SJ. Beneficial effects of fermented black ginseng and its ginsenoside 20 (S)-Rg3 against
362
cisplatin-induced nephrotoxicity in LLC-PK1 cells. J Ginseng Res 2016; 40: 135-140.
363
[9] Park JY, Choi P, Kim HK, Kang KS, Ham J. Increase in apoptotic effect of Panax ginseng by
364
microwave processing in human prostate cancer cells: in vitro and in vivo studies. J Ginseng
365
Res 2016; 40: 62-67.
366
[10] Babiker LB, Gadkariem EA, Alashban RM, Aljohar HI. Investigation of stability of Korean
367
ginseng in herbal drug product. Am J Appl Sci 2014; 11: 160.
368
[11] Zhang D, Yasuda T, Yu Y, Zheng P, Kawabata T, Ma Y, Okada S. Ginseng extract scavenges
369
hydroxyl radical and protects unsaturated fatty acids from decomposition caused by iron-
370
mediated lipid peroxidation. Free Radic Biol Med 1996; 20: 145-150.
371
[12] Yun TK, Choi SY, Yun HY. Epidemiological study on cancer prevention by ginseng: are all
372
kinds of cancers preventable by ginseng? J Korean Med Sci 2001; 16: S19-S27.
373
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
17
[13] Joo SS, Won TJ, Lee DI. Reciprocal activity of ginsenosides in the production of
374
proinflammatory repertoire, and their potential roles in neuroprotection in vitro. Planta Med
375
2005; 71: 476-481.
376
[14] Jung CH, Seog HM, Choi IW, Choi HD, Cho HY. Effects of wild ginseng (Panax ginseng CA
377
Meyer) leaves on lipid peroxidation levels and antioxidant enzyme activities in streptozotocin
378
diabetic rats. J Ethnopharmacol 2005; 98: 245-250.
379
[15] Fang Y, Shen N, Chen X. Beneficial changes in prostacyclin and thromboxane A2 induced by
380
ginsenosides in myocardial infarction and reperfusion injury in dogs. Zhongguo Yao Li Xue
381
Bao 1986; 7: 226-230.
382
[16] Sakata T, Etou H, Fujimoto K, Ockuma K, Hayashi T, Arichi S. Central effects of
383
ginsenosides on the feeding behavior and response to stress in rats. In, Korea-Japan Panax
384
Ginseng Symposium; 1987: 20-28.
385
[17] Fujimoto K, Sakata T, Ishimaru T, Etou H, Ookuma K, Kurokawa M, Machidori H.
386
Attenuation of anorexia induced by heat or surgery during sustained administration of
387
ginsenoside Rg1 into rat third ventricle. Psychopharmacol 1989; 99: 257-260.
388
[18] Xie JT, McHendale S, Yuan CS. Ginseng and diabetes. Am J Chin Med 2005; 33: 397-404.
389
[19] Yang G, Park D, Lee J, Song BS, Jeon TH, Kang SJ, Jeon JH, Shin S, Jeong HS, Lee HJ.
390
Suppressive effects of red ginseng preparations on SW480 colon cancer xenografts in mice.
391
Food Sci Biotechnol 2011; 20: 1649-1653.
392
[20] Cho IH. Effects of Panax ginseng in neurodegenerative diseases. J Ginseng Res 2012; 36: 342.
393
[21] Kang SW, Min HY. Ginseng, the'immunity boost': the effects of Panax ginseng on immune
394
system. J Ginseng Res 2012; 36: 354-368.
395
[22] Park J. Recent studies on the chemical constituents of Korean ginseng (Panax ginseng CA
396
Meyer). Korean J Ginseng Sci 1996; 20: 389-415.
397
[23] Elyakov G, Strigina L, Uvarova N, Vaskovsky V, Dzizenko A, Kochetkov N. Glycosides
398
from ginseng roots. Tetrahedron Lett 1964; 5: 3591-3597.
399
[24] Shibata S, Fujita M, Itokawa H, Tanaka O, Ishii T. Studies on the constituents of Japanese and
400
Chinese crude drugs. XI. Panaxadiol, a sapogenin of ginseng roots. Chem Pharm Bull 1963;
401
11: 759-761.
402
[25] Lu JM, Yao Q, Chen C. Ginseng compounds: an update on their molecular mechanisms and
403
medical applications. Curr Vasc Pharmacol 2009; 7: 293-302.
404
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
18
[26] Fuzzati N. Analysis methods of ginsenosides. J Chromatogr B Analyt Technol Biomed Life
405
Sci 2004; 812: 119-133.
406
[27] Lee SM. Thermal conversion pathways of ginsenoside in red ginseng processing. Nat Prod
407
Sci 2014; 20(2): 119-125.
408
[28] Takahashi M, Yoshikura M. Studies on the components of Panax ginseng CA Meyer. V. On
409
the structure of a new acetylene derivative" Panaxynol"(3). Synthesis of 1, 9-(cis)-
410
heptadecadiene-4, 6-diyn-3-ol. Yakugaku Zasshi 1966; 86: 1053.
411
[29] Kong YH, Lee YC, Choi SY. Neuroprotective and anti-inflammatory effects of phenolic
412
compounds in Panax ginseng CA Meyer. J Ginseng Res 2009; 33: 111-114.
413
[30] Chung IM, Lim JJ, Ahn MS, Jeong HN, An TJ, Kim SH. Comparative phenolic compound
414
profiles and antioxidative activity of the fruit, leaves, and roots of Korean ginseng (Panax
415
ginseng Meyer) according to cultivation years. J Gnseng Res 2016; 40: 68-75.
416
[31] Lee H, Yoo B, Byun S. Differences in phenolic acids between Korean ginsengs and mountain
417
ginsengs. Korean J Biotechnol Bioeng 2000; 15: 323-328.
418
[32] Iwabuchi H, Yoshikura M, Kamisako W. Studies on the sesquiterpenoids of Panax ginseng C.
419
A. Meyer. II. Isolation and structure determination of ginsenol, a novel sesquiterpene alcohol.
420
Chem Pharm Bull (Tokyo) 1988; 36: 2447-2451.
421
[33] Richter R, Basar S, Koch A, König WA. Three sesquiterpene hydrocarbons from the roots of
422
Panax ginseng CA Meyer (Araliaceae). Phytochemistry 2005; 66: 2708-2713.
423
[34] Wang JY, Li XG, Yang XW. Ginsenine, a new alkaloid from the berry of Panax ginseng CA
424
Meyer. J Asian Nat Prod Res 2006; 8: 605-608.
425
[35] Konno C, Murakami M, Oshima Y, Hikino H. Isolation and hypoglycemic activity of
426
panaxans Q, R, S, T and U, glycans of Panax ginseng roots. J Ethnopharmacol 1985; 14: 69-
427
74.
428
[36] Konno C, Sugiyama K, Kano M, Takahashi M, Hikino H. Isolation and hypoglycaemic
429
activity of panaxans A, B, C, D and E, glycans of Panax ginseng roots. Planta Med 1984; 50:
430
434-436.
431
[37] Oshima Y, Konno C, Hikino H. Isolation and hypoglycemic activity of panaxans I, J, K and L,
432
glycans of Panax ginseng roots. J Ethnopharmacol 1985; 14: 255-259.
433
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
19
[38] Tomoda M, Takeda K, Shimizu N, Gonda R, Ohara N, Takada K, Hirabayashi K.
434
Characterization of two acidic polysaccharides having immunological activities from the root
435
of Panax ginseng. Biol Pharm Bull 1993; 16: 22-25.
436
[39] Zhang X, Yu L, Bi HT, Li XH, Ni WH, Han H, Li N, Wang BQ, Zhou YF, Tai GH. Total
437
fractionation and characterization of the water-soluble polysaccharides isolated from Panax
438
ginseng C. A. Meyer. Carbohyd Polym 2009; 77: 544-552.
439
[40] Fan YY, Sun CX, Gao XG, Wang F, Li XZ, Kassim RM, Tai GH, Zhou YF. Neuroprotective
440
effects of ginseng pectin through the activation of ERK/MAPK and Akt survival signaling
441
pathways. Mol Med Rep 2012; 5: 1185-1190.
442
[41] Fan YY, Cheng HR, Liu D, Zhang X, Wang B, Sun L, Tai GH, Zhou YF. The Inhibitory
443
Effect of Ginseng Pectin on L-929 Cell Migration. Arch Pharm Res 2010; 33: 681-689.
444
[42] Francis PT, Palmer AM, Snape M, Wilcock GK. The cholinergic hypothesis of Alzheimer’s
445
disease: a review of progress. J Neurol Neurosurg Psychiatry 1999; 66: 137-147.
446
[43] Tong LM, Fong H, Huang Y. Stem cell therapy for Alzheimer’s disease and related disorders:
447
current status and future perspectives. Exp Mol Med 2015; 47: e151.
448
[44] Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in
449
Alzheimer disease. Cold Spring Harb Perspect Med 2011; 1: a006189.
450
[45] Musiek ES, Xiong DD, Holtzman DM. Sleep, circadian rhythms, and the pathogenesis of
451
Alzheimer disease. Exp Mol Med 2015; 47: e148.
452
[46] Ciechanover A, Kwon YT. Degradation of misfolded proteins in neurodegenerative diseases:
453
therapeutic targets and strategies. Exp Mol Med 2015; 47: e147.
454
[47] Bajda M, Guzior N, Ignasik M, Malawska B. Multi-target-directed ligands in Alzheimer's
455
disease treatment. Curr Med Chem 2011; 18: 4949-4975.
456
[48] Mancuso C, Bates TE, Butterfield DA, Calafato S, Cornelius C, Lorenzo AD, Dinkova
457
Kostova AT, Calabrese V. Natural antioxidants in Alzheimer's disease. Expert Opin Investig
458
Drugs 2007; 16: 1921-1931.
459
[49] Tan X, Gu J, Zhao B, Wang S, Yuan J, Wang C, Chen J, Liu J, Feng L, Jia X. Ginseng
460
improves cognitive deficit via the RAGE/NF-κB pathway in advanced glycation end product-
461
induced rats. Journal of ginseng research 2015; 39: 116-124.
462
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
20
[50] Zhang Y, Pi Z, Song F, Liu Z. Ginsenosides attenuate d-galactose-and AlCl 3-inducedspatial
463
memory impairment by restoring the dysfunction of the neurotransmitter systems in the rat
464
model of Alzheimer's disease. J Ethnopharmacol 2016; 194: 188-195.
465
[51] Zhao H, Li Q, Zhang Z, Pei X, Wang J, Li Y. Long-term ginsenoside consumption prevents
466
memory loss in aged SAMP8 mice by decreasing oxidative stress and up-regulating the
467
plasticity-related proteins in hippocampus. Brain Res 2009; 1256: 111-122.
468
[52] Hwang JY, Shim JS, Song MY, Yim SV, Lee SE, Park KS. Proteomic analysis reveals that
469
the protective effects of ginsenoside Rb1 are associated with the actin cytoskeleton in β-
470
amyloid-treated neuronal cells. J Ginseng Res 2015.
471
[53] Hou J, Xue J, Lee M, Yu J, Sung C. Long-term administration of ginsenoside Rh1 enhances
472
learning and memory by promoting cell survival in the mouse hippocampus. Int J Mol Med
473
2014; 33: 234-240.
474
[54] Zhao H, Li Q, Li Y. Long-term ginsenoside administration prevents memory loss in aged
475
female C57BL/6J mice by modulating the redox status and up-regulating the plasticity-related
476
proteins in hippocampus. Neurosci 2011; 183: 189-202.
477
[55] Chu S, Gu J, Feng L, Liu J, Zhang M, Jia X, Liu M, Yao D. Ginsenoside Rg5 improves
478
cognitive dysfunction and beta-amyloid deposition in STZ-induced memory impaired rats via
479
attenuating neuroinflammatory responses. Int Immunopharmacol 2014; 19: 317-326.
480
[56] Kim EJ, Jung IH, Van Le TK, Jeong JJ, Kim NJ, Kim DH. Ginsenosides Rg5 and Rh3 protect
481
scopolamine-induced memory deficits in mice. J Ethnopharmacol 2013; 146: 294-299.
482
[57] Zhao BS, Liu Y, Gao XY, Zhai HQ, Guo JY, Wang XY. Effects of ginsenoside Rg1 on the
483
expression of toll-like receptor 3, 4 and their signalling transduction factors in the NG108-15
484
murine neuroglial cell line. Molecules 2014; 19: 16925-16936.
485
[58] Song XY, Hu JF, Chu SF, Zhang Z, Xu S, Yuan YH, Han N, Liu Y, Niu F, He X.
486
Ginsenoside Rg1 attenuates okadaic acid induced spatial memory impairment by the
487
GSK3β/tau signaling pathway and the Aβ formation prevention in rats. Eur J Pharmacol 2013;
488
710: 29-38.
489
[59] Heo JH, Lee ST, Chu K, Oh MJ, Park HJ, Shim JY, Kim M. Heat-processed ginseng
490
enhances the cognitive function in patients with moderately severe Alzheimer's disease. Nutr
491
Neurosci 2012; 15: 278-282.
492
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
21
[60] Wang Y, Yang G, Gong J, Lu F, Diao Q, Sun J, Zhang K, Tian J, Liu J. Ginseng for
493
Alzheimer's Disease: A Systematic Review and Meta-Analysis of Randomized Controlled
494
Trials. Curr Top Med Chem 2016; 16: 529-536.
495
[61] Hornykiewicz O. Dopamine in the basal ganglia: its role and therapeutic implications
496
(including the clinical use of L-DOPA). Brit Med Bull 1973; 29: 172-178.
497
[62] Hornykiewicz O. How L-DOPA was discovered as a drug for Parkinson's disease 40 years
498
ago. Wien Klin Wochenschr 2001; 113: 855-862.
499
[63] Mizuno Y, Mori H, Kondo T. Parkinson''s disease: From etiology to treatment. Int Med 1995;
500
34: 1045-1054.
501
[64] Sherer T, Betarbet R, Greenamyre J. Pathogenesis of Parkinson's disease. Curr Opin Investig
502
Drugs 2001; 2: 657-662.
503
[65] Bae JR, Lee BD. Function and dysfunction of leucine-rich repeat kinase 2 (LRRK2):
504
Parkinson’s disease and beyond. BMB Reports 2015; 48: 243.
505
[66] Pollanen MS, Dickson DW, Bergeron C. Pathology and biology of the Lewy body. J
506
Neuropathol Exp Neurol 1993; 52: 183-191.
507
[67] Kuzuhara S, Mori H, Izumiyama N, Yoshimura M, Ihara Y. Lewy bodies are ubiquitinated.
508
Acta Neuropathol 1988; 75: 345-353.
509
[68] Chung YC, Shin WH, Baek JY, Cho EJ, Baik HH, Kim SR, Won SY, Jin BK. CB2 receptor
510
activation prevents glial-derived neurotoxic mediator production, BBB leakage and peripheral
511
immune cell infiltration and rescues dopamine neurons in the MPTP model of Parkinson's
512
disease. Exp Mol Med 2016; 48: e205.
513
[69] Zhu BT. CNS dopamine oxidation and catechol-O-methyltransferase: importance in the
514
etiology, pharmacotherapy, and dietary prevention of Parkinson's disease. Int J Mol Med
515
2004; 13: 343-354.
516
[70] Fahn S, Cohen G. The oxidant stress hypothesis in Parkinson's disease: evidence supporting it.
517
Ann Neurol 1992; 32: 804-812.
518
[71] Fahn S, Bressman SB. Should levodopa therapy for parkinsonism be started early or late?
519
Evidence against early treatment. Can J Neurol Sci 1984; 11: 200-205.
520
[72] Alam ZI, Daniel SE, Lees AJ, Marsden DC, Jenner P, Halliwell B. A generalised increase in
521
protein carbonyls in the brain in Parkinson's but not incidental Lewy body disease. J
522
Neurochem 1997; 69: 1326-1329.
523
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
22
[73] Diamond SG, Marchkham CH, Hoehn MM, McDowell FH, Muenter MD. Multicenter study
524
of Parkinson mortality with early versus later dopa treatment. Ann Neurol 1987; 22: 8-12.
525
[74] Parodi J, Ormeño D, Ochoa-de la Paz LD. Amyloid pore-channel hypothesis: effect of ethanol
526
on aggregation state using frog oocytes for an Alzheimer’s disease study. BMB Reports 2015;
527
48: 13.
528
[75] Hu S, Han R, Mak S, Han Y. Protection against 1-methyl-4-phenylpyridinium ion (MPP+)-
529
induced apoptosis by water extract of ginseng (Panax ginseng CA Meyer) in SH-SY5Y cells.
530
J Ethnopharmacol 2011; 135: 34-42.
531
[76] Zhou T, Zu G, Zhang X, Wang X, Li S, Gong X, Liang Z, Zhao J. Neuroprotective effects of
532
ginsenoside Rg1 through the Wnt/β-catenin signaling pathway in both in vivo and in vitro
533
models of Parkinson's disease. Neuropharmacol 2016; 101: 480-489.
534
[77] Liu Y, Zhang RY, Zhao J, Dong Z, Feng DY, Wu R, Shi M, Zhao G. Ginsenoside Rd Protects
535
SH-SY5Y Cells against 1-Methyl-4-phenylpyridinium Induced Injury. Int J Mol Sci 2015; 16:
536
14395-14408.
537
[78] Woodruff TM, Thundyil J, Tang SC, Sobey CG, Taylor SM, Arumugam TV.
538
Pathophysiology, treatment, and animal and cellular models of human ischemic stroke. Mol
539
Neurodegener 2011; 6: 11.
540
[79] Zhang X, Shi M, Bjoras M, Wang W, Zhang G, Han J, Liu Z, Zhang Y, Wang B, Chen J, et al.
541
Ginsenoside Rd promotes glutamate clearance by up-regulating glial glutamate transporter
542
GLT-1 via PI3K/AKT and ERK1/2 pathways. Front Pharmacol 2013; 4: 152.
543
[80] Zhang Y, Zhou L, Zhang X, Bai J, Shi M, Zhao G. Ginsenoside-Rd attenuates TRPM7 and
544
ASIC1a but promotes ASIC2a expression in rats after focal cerebral ischemia. Neurol Sci
545
2012; 33: 1125-1131.
546
[81] Ye R, Zhang X, Kong X, Han J, Yang Q, Zhang Y, Chen Y, Li P, Liu J, Shi M. Ginsenoside
547
Rd attenuates mitochondrial dysfunction and sequential apoptosis after transient focal
548
ischemia. Neurosci 2011; 178: 169-180.
549
[82] Ye R, Yang Q, Kong X, Han J, Zhang X, Zhang Y, Li P, Liu J, Shi M, Xiong L. Ginsenoside
550
Rd attenuates early oxidative damage and sequential inflammatory response after transient
551
focal ischemia in rats. Neurochem Int 2011; 58: 391-398.
552
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
23
[83] Dong X, Zheng L, Lu S, Yang Y. Neuroprotective effects of pretreatment of ginsenoside Rb1
553
on severe cerebral ischemiainduced injuries in aged mice: Involvement of antioxidant
554
signaling. Geriatr Gerontol Int 2016; doi: 10.1111/ggi.12699.
555
[84] He L, Chen X, Zhou M, Zhang D, Yang J, Yang M, Zhou D. Radix/rhizoma notoginseng
556
extract (sanchitongtshu) for ischemic stroke: a randomized controlled study. Phytomedicine
557
2011; 18: 437-442.
558
[85] Rub U, Vonsattel JPG, Heinsen H, Korf HW. The Neuropathology of Huntington's Disease:
559
Classical Findings, Recent Developments and Correlation to Functional Neuroanatomy
560
Conclusions and Outlook. Adv Anat Embryol Cel 2015; 217: 125-131.
561
[86] Saudou F, Humbert S. The biology of Huntingtin. Neuron 2016; 89: 910-926.
562
[87] Zuccato C, Valenza M, Cattaneo E. Molecular mechanisms and potential therapeutical targets
563
in Huntington's disease. Physiol Rev 2010; 90: 905-981.
564
[88] Politis M, Pavese N, Tai YF, Tabrizi SJ, Barker RA, Piccini P. Hypothalamic involvement in
565
Huntington's disease: an in vivo PET study. Brain 2008; 131: 2860-2869.
566
[89] Bates G, Jones L. Huntington’s disease Oxford: Oxford Universtity Press. CIT0001 2002.
567
[90] Rosenblatt A. Neuropsychiatry of Huntington's disease. Dialogues Clin Neurosci 2007; 9: 191.
568
[91] Wu J, Jeong HK, Bulin SE, Kwon SW, Park JH, Bezprozvanny I. Ginsenosides protect
569
striatal neurons in a cellular model of Huntington's disease. J Neurosci Res 2009; 87: 1904-
570
1912.
571
[92] Gao Y, Chu SF, Li JP, Zhang Z, Yan JQ, Wen ZL, Xia CY, Mou Z, Wang ZZ, He WB.
572
Protopanaxtriol protects against 3-nitropropionic acid-induced oxidative stress in a rat model
573
of Huntington's disease. Acta Pharmacol Sin 2015; 36: 311-322.
574
[93] Amor S, Puentes F, Baker D, Van Der Valk P. Inflammation in neurodegenerative diseases.
575
Immunol 2010; 129: 154-169.
576
[94] Kannappan R, Gupta SC, Kim JH, Reuter S, Aggarwal BB. Neuroprotection by spice-derived
577
nutraceuticals: you are what you eat! Mol Neurobiol 2011; 44: 142-159.
578
[95] Marchetti B, Abbracchio MP. To be or not to be (inflamed)–is that the question in anti-
579
inflammatory drug therapy of neurodegenerative disorders? Trends Pharmacol Sci 2005; 26:
580
517-525.
581
[96] Andersen JK. Oxidative stress in neurodegeneration: cause or consequence? Nat Med 2004;
582
10: 18-25.
583
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
24
[97] Son JH, Shim JH, Kim KH, Ha JY, Han JY. Neuronal autophagy and neurodegenerative
584
diseases. Exp Mol Med 2012; 44: 89-98.
585
[98] Ferrante RJ, Shinobu LA, Schulz JB, Matthews RT, Thomas CE, Kowall NW, Gurney ME,
586
Beal MF. Increased 3nitrotyrosine and oxidative damage in mice with a human copper/zinc
587
superoxide dismutase mutation. Ann Neurol 1997; 42: 326-334.
588
[99] Adibhatla RM, Hatcher JF, Dempsey RJ. Phospholipase A2, hydroxyl radicals, and lipid
589
peroxidation in transient cerebral ischemia. Antioxid Redox Signal 2003; 5: 647-654.
590
[100] Hall N, Carney J, Cheng M, Butterfield D. Ischemia/reperfusion-induced changes in
591
membrane proteins and lipids of gerbil cortical synaptosomes. Neurosci 1995; 64: 81-89.
592
[101] Won MH, Kang TC, Jeon GS, Lee JC, Kim DY, Choi EM, Lee KH, Do Choi C, Chung MH,
593
Cho SS. Immunohistochemical detection of oxidative DNA damage induced by ischemia–
594
reperfusion insults in gerbil hippocampus in vivo. Brain Res 1999; 836: 70-78.
595
[102] Dexter D, Carter C, Wells F, JavoyAgid F, Agid Y, Lees A, Jenner P, Marsden CD. Basal
596
lipid peroxidation in substantia nigra is increased in Parkinson's disease. J Neurochem 1989;
597
52: 381-389.
598
[103] Hensley K, Maidt ML, Yu Z, Sang H, Markesbery WR, Floyd RA. Electrochemical analysis
599
of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific
600
accumulation. J Neurosci 1998; 18: 8126-8132.
601
[104] Butterfield DA, Castegna A, Lauderback CM, Drake J. Evidence that amyloid beta-peptide-
602
induced lipid peroxidation and its sequelae in Alzheimer’s disease brain contribute to
603
neuronal death. Neurobiol Aging 2002; 23: 655-664.
604
[105] Yang HY, Lee TH. Antioxidant enzymes as redox-based biomarkers: a brief review. BMB
605
Reports 2015; 48: 200.
606
[106] Ye M, Chung HS, Lee C, Song JH, Shim I, Kim YS, Bae H. Bee venom phospholipase A2
607
ameliorates motor dysfunction and modulates microglia activation in Parkinson's disease
608
alpha-synuclein transgenic mice. Exp Mol Med 2016; 48: e244.
609
[107] Radad K, Moldzio R, Rausch WD. Ginsenosides and their CNS targets. CNS Neurosci Ther
610
2011; 17: 761-768.
611
[108] Nguyen CT, Luong TT, Kim GL, Pyo S, Rhee DK. Korean Red Ginseng inhibits apoptosis in
612
neuroblastoma cells via estrogen receptor β-mediated phosphatidylinositol-3 kinase/Akt
613
signaling. J Ginseng Res 2015; 39: 69-75.
614
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
25
[109] Kim S, Kim MS, Park K, Kim HJ, Jung SW, Nah SY, Han JS, Chung C. Hippocampus-
615
dependent cognitive enhancement induced by systemic gintonin administration. J Ginseng
616
Res 2016; 40: 55-61.
617
[110] Kang KS, Ham J, Kim YJ, Park JH, Cho EJ, Yamabe N. Heat-processed Panax ginseng and
618
diabetic renal damage. J Ginseng Res 2013; 37: 379-388.
619
[111] González-Burgos E, Fernandez-Moriano C, Gómez-Serranillos MP. Potential neuroprotective
620
activity of Ginseng in Parkinson’s disease: a review. J Neuroimmune Pharmacol 2015; 10:
621
14-29.
622
[112] Ong WY, Farooqui T, Koh HL, Farooqui AA, Ling EA. Protective effects of ginseng on
623
neurological disorders. Front Aging Neurosci 2015; 7.
624
[113] Li N, Liu Y, Li W, Zhou L, Li Q, Wang X, He P. A UPLC/MS-based metabolomics
625
investigation of the protective effect of ginsenosides Rg1 and Rg2 in mice with Alzheimer's
626
disease. J Ginseng Res 2016; 40: 9-17.
627
628
629
630
631
632
633
634
635
636
637
638
639
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
26
FIGURE LEGENDS
640
641
FIGURE 1. Structure of selected ginsenosides. (A) protopanaxadiols (PPD). (B) protopanaxatriols
642
(PPT). (C) derivatives of PPD and PPT. (D) other ginsenosides. Glc, β-D-glucose; Rha, α-L-
643
rhamnose; Ara(p), α-L-arabinose(pyranose); Ara(f), α-L-arabinose(furanose); Xyl, β-D-xylose;
644
GlcUA, β-D-glucuronic acid; mal, malonyl; Ac, acetyl.
645
646
FIGURE 2. Structure of selected non-saponin constituents. (A) polyacetylenes. (B) phenolic
647
compounds. (C) sesquiterpenes. (D) alkaloids.
648
649
650
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
27
Table 1. Effects of Panax ginseng and its active ingredient on Alzheimer’s disease.
651
Active
ingredient
Taget
molecules
Cell lines
or animal strain (toxicants)
Effective doses
(treatment time)
References
Rg1 TNF-α, IFN-β,
iNOS, TLR3,
TLR4, NF-κB and
TRAF-6
NG108-15 cells (amyloid β
peptide 25-35, Aβ25-35)
8, 16 and 32
µg/mL (24h)
[57]
Rb1 CAP1, CAPZB,
TOMM40, DSTN,
PARP-1 and Bax
SH-SY5Y cells (amyloid β) 100 µM (24 h)
[52]
Panax
ginseng
extract
RAGE and NF-κB
Male Sprague-Dawley rats
(advanced glycation end
product)
0.25, 0.5 and 1
g/kg/day (30
days)
[49]
Ginseng total
saponin
Aβ, tau, Glu, Asp,
GABA, Ach, DA,
Gly and 5-HT
Male Wistar rats (d-
galactose with AlCl
3
)
2 g/kg/day (30
days)
[50]
Ginseng total
saponin
PSD-95, PKCγ
and BDNF
Female C57BL/6J mice
(aged mice: 12 months old)
0.056 and 0.112%
(w/v) (8 months)
[54]
Ginseng total
saponin
PSD-95, p-
NMDAR1, p-
CaMKII, p-PKA
Cβ, PKCγ, p-
CREB and BDNF
Male SAMP8 and SAMR1
mice (aged mice: 4 months
old)
100 and 200
mg/kg/day (7
months)
[51]
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
28
Rh1 BDNF Male ICR mice (aged mice:
6 months old)
10 mg/kg/day (3
months)
[53]
Rg5 TNF-α, IL-1β,
IGF-1, BDNF,
COX-2, iNOS
and Aβ
Wistar rats (streptozotocin)
10 and 20
mg/kg/day (28
days)
[55]
Rg5 and Rh3 BDNF and CREB Male ICR mice
(scopolamine) 10 mg/kg (1h) [56]
Rg1 GSK3β and tau Male Sprague-Dawley rats
(okadaic acid)
20 mg/kg/day (25
days)
[58]
652
653
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
29
Table 2. Effects of Panax ginseng and its active ingredient on Parkinson’s disease.
654
Active
ingredient
Taget
molecules
Cell lines
or animal strain (toxicants)
Effective doses
(treatment time) References
Panax
ginseng
extract
Bax, Bcl-2, cytochrome c
and cleaved caspase-3
SH-SY5Y cells (1-methyl-
4-phenylpyridinium ion,
MPP
+
)
0.2 mg/mL (60
h) [75]
Rg1 Wnt-1, β-catenin, GSK-
3β and p-GSK-3β,
cleaved caspase-3 and
Bcl-xL
PC12 cells (MPP
+
) 20 µM (24 h) [76]
Rd Bax, Bcl-2 and P-Akt SH-SY5Y cells (MPP
+
) 1 and 10 µM
(72 h) [77]
Rg1 Wnt-1, β-catenin, GSK-
3β and p-GSK-3β
Male C57BL/6J mice (1-
methyl-4-phenyl-1,2,3,6-
tetrahydropyridine, MPTP)
5, 10 and 20
mg/kg/day (15
days)
[76]
655
656
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
30
Table 3. Effects of Panax ginseng and its active ingredient on brain ischemia and stroke.
657
Active
ingredient
Taget
molecules
Cell lines
or animal strain (toxicants)
Effective doses
(treatment time) References
Rb1 ERK1/2 C57BL/6J mice (aged
mice: 12 months old)
5 mg/kg/every 3
days (1 year) [83]
Rd GLT-1, PI3K/AKT
and ERK1/2
Male Sprague–Dawley rats
(middle cerebral artery
occlusion, MCAO)
30 mg/kg (1h) [79]
Rd TRPM-1, -2, -3, -4, -
5, -6, -7, ASIC1a,
ASIC2a, NR1,
NR2A and NR2B
Male Sprague–Dawley rats
(MCAO) 10 mg/kg (15 min) [80]
Rd CytoC, AIF, and
Caspase-3
Male Sprague–Dawley rats
(MCAO) 50 mg/kg (30 min) [81]
Rd COX-2 and iNOS Male Sprague–Dawley rats
(MCAO) 50 mg/kg (30 min) [82]
658
659
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
31
Table 4. Effects of Panax ginseng and its active ingredient on Huntington's disease.
660
Active
ingredient
Taget
molecules
Cell lines
or animal strain (toxicants)
Effective doses
(treatment time) References
Protopanaxtriol
Nrf2, HO-1, NQO1
and PCNA
Male Sprague-Dawley
rats (3-nitropropionic
acid)
20 mg/kg (30 min) [92]
661
662
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
33
664
665
666
667
668
669
670
671
672
673
674
Figure 1.
675
676
R
1
O
OH
H
H
H
R
2
O
H
3
12
20
A
HO
OH
H
H
H
R
2
O
H
3
12
20
B
OR
1
6
20(S)-protopanaxadiol (PPD)
R
1
= R
2
= H
20(S)-protopanaxatriol (PPT)
R
1
= R
2
= H
Ginsenoside Rb1: R
1
= Glc
2
-Glc, R
2
= Glc
6
-Glc
Ginsenoside Rb2: R
1
= Glc
2
-Glc, R
2
= Glc
6
-Ara(p)
Ginsenoside Rc: R
1
= Glc
2
-Glc, R
2
= Glc
6
-Ara(f)
Ginsenoside Rd: R
1
= Glc
2
-Glc, R
2
= Glc
Ginsenoside Rg3: R
1
= Glc
2
-Glc, R
2
= H
Ginsenoside Rh2: R
1
= Glc, R
2
= H
Ginsenoside Rb3: R
1
= Glc
2
-Glc, R
2
= Glc
6
-Xyl
Ginsenoside Ra1: R
1
= Glc
2
-Glc, R
2
= Glc
6
-Ara(p)
4
-Xyl
Ginsenoside Ra2: R
1
= Glc
2
-Glc, R
2
= Glc
6
-Ara(f)
2
-Xyl
Ginsenoside Ra3: R
1
= Glc
2
-Glc, R
2
= Glc
6
-Glc
3
-Xyl
Ginsen oside Rs3: R
1
= Glc
2
-Glc-Ac, R
2
= H
Malonyl-ginsenoside Rb1 : R
1
= Glc
2
-Glc
6
-mal, R
2
= Glc
6
-Glc
Malonyl-ginsenoside Rb2 : R
1
= Glc
2
-Glc
6
-mal, R
2
= Glc
6
-Ara(p)
Malonyl-ginsenoside Rc: R
1
= Glc
2
-Glc
6
-mal, R
2
= Glc
6
-Ara(f)
Malonyl-ginsenoside Rd: R
1
= Glc
2
-Glc
6
-mal, R
2
= Glc
Ginsen oside Rh1: R
1
= Glc, R
2
= H
Ginsen oside Rg1: R
1
= Glc, R
2
= Glc
Ginsen oside Rf: R
1
= Glc
2
-Glc, R
2
= H
Ginsen oside Re: R
1
= Glc
2
-Rha, R
2
= Glc
Ginsen oside F1: R
1
= H, R
2
= Glc
Ginsen oside R1: R
1
= Glc
2
-Xyl, R
2
= Glc
Ginsen oside Rg2: R
1
= Glc
2
-Rha, R
2
= H
R
1
O
OH
H
H
HH
OH
C
R
2
20(R)-ginsenoside Rh1: R
1
= H, R
2
= O-Glc
20(R)-ginsenoside Rg2: R
1
= H, R
2
= O-Glc
2
-Rha
20(R)-ginsenoside Rg3: R
1
= Glc
2
-Glc, R
2
= H
20(R)-ginsenoside Rs3 : R
1
= Glc
2
-Glc
6
-Ac, R
2
= H
R
1
O
OH
H
H
HH
R
2
Ginsen oside F4: R
1
= H, R
2
= O-Glc
2
-Rha
Ginsen oside Rh4: R
1
= H, R
2
= O-Glc
Ginsen oside Rg5: R
1
= Glc
2
-Glc, R
2
= H
Ginsen oside Rs4: R
1
= Glc
2
-Glc
6
-Ac, R
2
= H
R
1
O
OH
H
H
HH
R
2
Ginsen oside Rg6: R
1
= H, R
2
= O-Glc
2
-Rha
Ginsen oside Rk3: R
1
= H, R
2
= O-Glc
Ginsen oside Rk1: R
1
= Glc
2
-Glc, R
2
= H
Ginsen oside Rs5: R
1
= Glc
2
-Glc
6
-Ac, R
2
= H
RO H
H
D
Ginsen oside Ro: R = Glc UA
2
-Glc
COOGlc
HO
OH
H
H
HH
R
OH
OH
25-OH -PPT: R = OH
25-OH -PPD: R = H
HO
OH
H
H
HH
OGlc
2
-Rha
O
HO
20(R)-pseudoginsenoside F11
MANUS CRIP T
ACCEP TED
ACCEPTED MANUSCRIPT
34
677
678
679
680
681
682
683
684
685
686
687
Figure 2.
688
689
Panaxynol
Panaxydol
HO
O
HO
O
HO
Ginseno yne A
OH
Cl
HO
OH
OH
HO
O
HO
O
O
Ginsenoyne B
Ginsenoyne C
Ginsenoyne D
Ginsenoyne E
OH
OH
Panaxytriol
HO
A
OH
O
OH
Salicylic acid
OH
O
HO OCH
3
Vanillic acid
OH
OH
O
m-Coumaric acid
O
OOH
Maltol
B
Beta-panasinsene African-2-ene Beta-elemene Calarene
C
HH
H
H
Beta-selinene
H
HO
H
Spathulenol
O
Humulene epoxide II
OH
H
Ginsenol
N
HN
OCH
3
O
1-Carbomethoxy-beta-carboline
N
HN
Harman
N
S
OH
4-Methyl-5-thiazoleethanol
NH
H
N
N
COOH
Spinacine
D
... P. ginseng has been used in traditional medicine since ancient times. It has a positive effect on cardiovascular and neurodegenerative diseases, cancer, and diabetes mellitus [42][43][44][45][46]. Extensive research has associated the biological activities of Korean P. ginseng and its products with various functional components, including ginsenosides, polyacetylenes, phenolic compounds, alkaloids, polysaccharides, oligopeptides, and essential oils [47]. ...
Article
Flavonoids are plant polyphenols that exhibit biological activity with antibacterial, antiviral, antioxidant, anti-inflammatory, antimutagenic, and anticarcinogenic effects. The medicinal plants of Kuzbass have high contents of flavonoids and other polyphenolic compounds. Therefore, they can be used in medicinal preparations to prevent or treat serious diseases. We studied the following plants collected in Kuzbass: common thyme (Thymus vulgaris Linn., leaves and stems), woolly burdock (Arctium tomentosum Mill., roots), alfalfa (Medicago sativa L., leaves and stems), common lungwort (Pulmonaria officinalis L., leaves and stems), common yarrow (Achillea millefolium L., leaves and stems), red clover (Trifolium pratense L., leaves and stems), common ginseng (Panax ginseng, roots), sweetvetch (Hedysarum neglectum Ledeb., roots), and cow parsnip (Heracleum sibiricum L., inflorescences, leaves, and stems). To extract flavonoids, we used ethanol at concentrations of 40, 55, 60, 70, and 75%. Spectrophotometry was used to determine total flavonoids, while high-performance liquid chromatography was employed to study the qualitative and quantitative composition of the extracts. The highest yield of flavonoids was found in H. sibiricum leaves (at all concentrations except 70%), followed by the 55% and 70% ethanol extracts of T. vulgaris leaves and stems, as well as the 75% ethanol extract of A. millefolium leaves and stems. Thus, these plants have the greatest potential in being used in medicines. High-performance liquid chromatography showed the highest contents of polyphenolic compounds in the samples of P. officinalis, A. millefolium, T. vulgaris, and T. pratense. Our results can be used in further research to produce new medicinal preparations based on the medicinal plants of Kuzbass.
... A common medicinal plant used to cure a variety of diseases is ginseng. Ginseng has been used for its antioxidant properties, immune system stimulation, stress relief, and central nervous system (CNS) function [67], [68]. Its pharmacological effects have been shown in cancer, diabetes, and heart disease. ...
Article
The gut microbiota is a varied population of microorganisms that live in the human gastrointestinal system. Emerging research emphasizes the importance of this microbial ecology in general health and its influence on a variety of disorders. The review explores the synergy between herbal treatment and traditional medicine, emphasizing their cultural significance and therapeutic benefits. It delves into the intricate relationship between herbal remedies, traditional healing practices, and their sustained usage over centuries. The review highlights the pivotal role of the gut microbiota in herbal medicine, elucidating how treatments influence the gastrointestinal microorganisms, impacting overall health. Dietary phytochemicals are underscored for their significance in herbal medicine and nutritional well-being, along with the interdependence of plant extracts and botanicals. The investigation explores the molecular connections between phytoconstituents and gut microbiota, aiming to deepen the understanding of herbal medicine's tailored approach to specific health challenges. The summary concludes by emphasizing herbal treatments' unique ability to regulate gut flora, contributing to overall gastrointestinal wellbeing. In closing, the review provides a concise overview, serving as a valuable resource for integrative medicine research, with recommendations for future exploration of herbal medicine's potential in healthcare.
... G-Rh2, first isolated from red ginseng, is a steroidal saponin belonging to the protopanaxadiol type, and has various potent biological functions, including antitumor, antiobesity, anti-inflammatory and antioxidant activities, preventing neurodegenerative diseases and so on [10][11][12][13][14]. In this study, we aimed to explore the anti-inflammatory effect of G-Rh2 by constructing an LPS-induced RAW 264.7 cell inflammatory model in vitro. ...
Article
Full-text available
Lipopolysaccharide (LPS) triggers a severe systemic inflammatory reaction in mammals, with the dimerization of TLR4/MD-2 upon LPS stimulation serving as the pivotal mechanism in the transmission of inflammatory signals. Ginsenoside Rh2 (G-Rh2), one of the active constituents of red ginseng, exerts potent anti-inflammatory activity. However, whether G-Rh2 can block the TLR4 dimerization to exert anti-inflammatory effects remains unclear. Here, we first investigated the non-cytotoxic concentration of G-Rh2 on RAW 264.7 cells, and detected the releases of pro-inflammatory cytokines in LPS-treated RAW 264.7 cells, and then uncovered the mechanisms involved in the anti-inflammatory activity of G-Rh2 through flow cytometry, fluorescent membrane localization, Western blotting, co-immunoprecipitation (Co-IP), molecular docking and surface plasmon resonance (SPR) analysis in LPS-stimulated macrophages. Our results show that G-Rh2 stimulation markedly inhibited the secretion of LPS-induced interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and nitric oxide (NO). Additionally, G-Rh2 blocked the binding of LPS with the membrane of RAW264.7 cells through direct interaction with TLR4 and MD-2 proteins, leading to the disruption of the dimerization of TLR4 and MD-2, followed by suppression of the TLR4/NF-κB signaling pathway. Our results suggest that G-Rh2 acts as a new inhibitor of TLR4 dimerization and may serve as a promising therapeutic agent against inflammation.
... Although the content of ginsenosides was lower in cultivated plants that were aggregated from field soil, it is reported that ginsenosides treat many chronic diseases and modulate various physiological activities [24]. One of the various advantages of ginsenosides is their ability to interact with target molecules in the cells resulting in combined pharmacologically beneficial effects [13,45]. WSG naturally grown in China and Korea showed greater health benefits [29]. ...
Article
Full-text available
Wild-simulated ginseng (WSG, Panax ginseng C.A. Meyer) is grown in mountainous forests, without the chemical treatment or installation of artificial facilities. This study aimed to investigate monthly changes in growth characteristics and ginsenoside contents in WSG to suggest the optimal harvest time. Four-year-old WSG were collected in the same area every month, and their growth characteristics and ginsenoside contents were measured. The growth characteristics of aerial and root parts were measured from May to July and from March to December, respectively. For the aerial part, most growth characteristics of WSG decreased over time, except for stem length. For the root part, rhizome length increased over time except for September, while the root diameter and weight of root part were mostly consistent. The root length increased by September, while the number of rootlets was the highest at May. At July, the total ginsenoside content of WSG was significantly the highest, while the total ginsenoside content at October was the lowest. This result was believed to be due to the F2, Rd, and Rg1 contents of the aerial part, rather than the root part. Also, based on these growths and the ginsenoside contents of WSG, the optimal harvest time for WSG is considered to be late spring–summer (May–July) when the aerial part can be identified.
... Ginseng (Panax ginseng Mey.), a perennial herbaceous medicinal plant in the Araliaceae family, is renowned as the king of herbs [1][2][3][4]. Due to its high medicinal value, it is widely utilized in clinical treatments and the production of health foods worldwide [5][6][7]. Ginsenosides, a class of important secondary metabolites in the Panax genus, are the primary active ingredients of ginseng, belonging to the triterpene compound family. ...
Article
Full-text available
Ginseng, an important medicinal plant, is characterized by its main active component, ginsenosides. Among more than 40 ginsenosides, Rg1 is one of the ginsenosides used for measuring the quality of ginseng. Therefore, the identification and characterization of genes for Rg1 biosynthesis are important to elucidate the molecular basis of Rg1 biosynthesis. In this study, we utilized 39,327 SNPs and the corresponding Rg1 content from 344 core ginseng cultivars from Jilin Province. We conducted a genome-wide association study (GWAS) combining weighted gene co-expression network analysis (WGCNA), SNP-Rg1 content association analysis, and gene co-expression network analysis; three candidate Rg1 genes (PgRg1-1, PgRg1-2, and PgRg1-3) and one crucial candidate gene (PgRg1-3) were identified. Functional validation of PgRg1-3 was performed using methyl jasmonate (MeJA) regulation and RNAi, confirming that this gene regulates Rg1 biosynthesis. The spatial–temporal expression patterns of the PgRg1-3 gene and known key enzyme genes involved in ginsenoside biosynthesis differ. Furthermore, variations in their networks have a significant impact on Rg1 biosynthesis. This study established an accurate and efficient method for identifying candidate genes, cloned a novel gene controlling Rg1 biosynthesis, and identified 73 SNPs significantly associated with Rg1 content. This provides genetic resources and effective tools for further exploring the molecular mechanisms of Rg1 biosynthesis and molecular breeding.
Article
Full-text available
Background: Plants have long been recognized for their potential to influence neurological health, with both neuroprotective and neurotoxic properties. This review explores the dual nature of plant-derived compounds and their impact on the human brain. Discussion: Numerous studies have highlighted the neuroprotective effects of various phytoconstituents, such as those found in Ginkgo biloba, Centella asiatica, Panax ginseng, Withania somnifera, and Curcuma longa. The neuroprotective compounds have demonstrated antioxidant, anti-inflammatory, and cognitive-enhancing properties, making them promising candidates for combating neurodegenerative diseases and improving brain function. Polyphenolic compounds, triterpenic acids, and specific phytocompounds like the ones from EGb 761 extract have shown interactions with key enzymes and receptors in the brain, leading to neuroprotective outcomes. However, this review also acknowledges the neurotoxic potential of certain plants, such as the Veratrum species, which contains steroidal alkaloids that can cause DNA damage and disrupt neurological function, or Atropa belladonna, which interfere with the normal functioning of the cholinergic system in the body, leading to a range of symptoms associated with anticholinergic toxicity. Conslusions: This review also emphasizes the need for further research to elucidate the complex mechanisms underlying the neuroprotective and neurotoxic effects of plant-derived compounds, as well as to identify novel phytoconstituents with therapeutic potential. Understanding the complex relationship between plants and the human brain is crucial for harnessing the benefits of neuroprotective compounds while mitigating the risks associated with neurotoxic substances. This review provides a comprehensive overview of the knowledge on the neurological properties of plants and highlights the importance of continued research in this field for the development of novel therapeutic strategies targeting brain health and neurological disorders.
Article
Full-text available
Areas of the body accessible to gastric secretions, such as the stomach and duodenum, are most commonly damaged by circumscribed lesions of the upper gastrointestinal tract mucosa. Peptic ulcer disease is the term for this illness (PUD). About 80% of peptic ulcers are duodenal ulcers, with stomach ulcers accounting for the remaining 20%. Duodenal ulcers are linked to the two primary results about Helicobacter pylori infection and COX inhibitor users. Additional causes might include drinking, smoking, stress, and coffee consumption. The indications and symptoms of a duodenal ulcer depend on the patient's age and the lesion's location. For duodenal ulcers, proton pump inhibitors (PPIs) are the usual course of treatment. This comprehensive study included an in-depth literature search in the literature and methods section using electronic databases such as PubMed, ScienceDirect, and Google Scholar. The search method included publications published from the inception of the relevant database to the present. Inclusion criteria included studies investigating different treatment options for duodenal ulcer disease, including traditional pharmacotherapy and naturopathic treatments. Data mining includes information on treatment techniques, treatment outcomes, and possible synergies between conventional and herbal treatments. In addition, this review critically examines the available information on the effectiveness, safety, and possible side effects of different treatments. The inclusion of conventional and herbal treatments is intended to provide a comprehensive overview of the many treatment options available for duodenal ulcer disease. A more comprehensive and personalized treatment plan can be achieved by incorporating dietary changes, lifestyle modifications, and, if necessary, herbal therapies to complement other treatments normally. Graphical Abstract
Article
Full-text available
α-Synuclein (α-Syn) has a critical role in microglia-mediated neuroinflammation, which leads to the development of Parkinson's disease (PD). Recent studies have shown that bee venom (BV) has beneficial effects on PD symptoms in human patients or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxin-induced PD mice. This study investigated whether treatment with BV-derived phospholipase A2 (bvPLA2) would improve the motor dysfunction and pathological features of PD in human A53T α-Syn mutant transgenic (A53T Tg) mice. The motor dysfunction of A53T Tg mice was assessed using the pole test. The levels of α-Syn, microglia and the M1/M2 phenotype in the spinal cord were evaluated by immunofluorescence. bvPLA2 treatment significantly ameliorated motor dysfunction in A53T Tg mice. In addition, bvPLA2 significantly reduced the expression of α-Syn, the activation and numbers of microglia, and the ratio of M1/M2 in A53T Tg mice. These results suggest that bvPLA2 could be a promising treatment option for PD.
Article
Full-text available
The cannabinoid (CB2) receptor type 2 has been proposed to prevent the degeneration of dopamine neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. However, the mechanisms underlying CB2 receptor-mediated neuroprotection in MPTP mice have not been elucidated. The mechanisms underlying CB2 receptor-mediated neuroprotection of dopamine neurons in the substantia nigra (SN) were evaluated in the MPTP mouse model of Parkinson’s disease (PD) by immunohistochemical staining (tyrosine hydroxylase, macrophage Ag complex-1, glial fibrillary acidic protein, myeloperoxidase (MPO), and CD3 and CD68), real-time PCR and a fluorescein isothiocyanate-labeled albumin assay. Treatment with the selective CB2 receptor agonist JWH-133 (10 μg kg−1, intraperitoneal (i.p.)) prevented MPTP-induced degeneration of dopamine neurons in the SN and of their fibers in the striatum. This JWH-133-mediated neuroprotection was associated with the suppression of blood–brain barrier (BBB) damage, astroglial MPO expression, infiltration of peripheral immune cells and production of inducible nitric oxide synthase, proinflammatory cytokines and chemokines by activated microglia. The effects of JWH-133 were mimicked by the non-selective cannabinoid receptor WIN55,212 (10 μg kg−1, i.p.). The observed neuroprotection and inhibition of glial-mediated neurotoxic events were reversed upon treatment with the selective CB2 receptor antagonist AM630, confirming the involvement of the CB2 receptor. Our results suggest that targeting the cannabinoid system may be beneficial for the treatment of neurodegenerative diseases, such as PD, that are associated with glial activation, BBB disruption and peripheral immune cell infiltration.
Article
Full-text available
Oxidative stress plays a crucial role in the pathology of neurodegenerative diseases such as Alzheimer's Parkinson's, Huntington and amyotrophic lateral sclerosis as well. Increased markers of oxidative damage are present in such conditions. They involve protein, lipids, DNA and even RNA oxidation. Several evidences indicate that increase in reactive oxygen species, an antioxidant systems deficit, decreased repair DNA mechanisms, proteolysis and the loss of immune system regulation are contributing factors to an increased oxidative stress. All those processes ultimately lead to a progressive neuronal death and damage. Alzheimer's a neurofibrillary tangles disorder, Parkinson's disease, a decrease in dopaminergic neurons, Huntington's, a loss of neostriatal neurons and Amyotrophic lateral sclerosis, a motor neuron disease, are common neurodegenerative diseases. Although these disorders differ from important pathways, they share some pathogenic features including inflammation, genetic mutations and inappropriate protein aggregates (Lewy bodies, amyloid plaques). Glial activation and dysfunction in dopaminergic, cholinergic and gabaergic systems are also common. Oxidative stress and mitochondrial dysfunction that eventually lead to neuronal death play a fundamental in the progressive impairment that patients with that kind of neurodegenerative diseases show. In current review, aspects that closely associate an oxidative stress state to neurodegeneration processes occurring in some neuropathologic conditions.
Book
1. Introduction.- 2. The neuropathological grading of Huntington disease.- 3. The cerebral cortex in Huntington's disease.- 4. Degeneration of select motor and limbic nuclei of the thalamus in Huntington's disease.- 5. Consistent and widespread degeneration of the cerebellum in Huntington's disease.- 6. Elucidation of the role of the premotor oculomotor brainstem nuclei in the pathogenesis of oculomotor dysfunctions in Huntington's disease.- 7. Widespread brainstem neurodegeneration in Huntington's disease.- 8. Intraneuronal transport and defense mechanisms with possible pathogenetic relevance in Huntington's disease.- 9. The disease protein huntingtin and neuronal protein aggregations in Huntington's disease.- 10. Pathological nerve cell alterations in Huntington's disease (HD) and their possible role for the demise of nerve cells.- 11. Conclusions and outlook.
Article
Ethnopharmacological relevance: Panax ginseng C.A.Mey. is a traditional Chinese herbal medicine, which has been used to treat Alzheimer's disease (AD) for thousands of years. Ginsenoside is one of the major compounds found in P. ginseng. This study aimed to explore the attenuation of spatial memory impairment by ginsenosides and its correlation with restoring the dysfunction of the neurotransmitter systems in AD model rats to understand the mechanism underlying the anti-AD effect of P. ginseng. Materials and methods: In this study, the AD model was established by combining d-galactose (d-gal) with AlCl3 (Al) for 60 days. From day 30, the ginsenosides group was intragastrically administered with ginsenosides for 30 days. The ethology of rats was tested through the Morris water maze test(MWM). Histopathological changes in the hippocampus of rats were observed through hematoxylin and eosin staining. The expressions of amyloid β peptide (Aβ) and phospho-tau (p-tau) in the hippocampus and cortex of rats were detected by immunohistochemistry. A liquid chromatography-mass spectrometry assay was used to measure neurotransmitter concentrations in the hippocampus, cortex, and blood. Results: Ginsenosides could significantly decrease the escape latency time and the average latency time in the place navigation test and increase the times of crossing the platform area, the percentage of residence time, and the distance in the original platform quadrant in the spatial probe test. Ginsenosides could repair the damage of the hippocampus and reduce the expressions of Aβ and p-tau. Ginsenosides could also increase γ-aminobutyric acid, acetylcholine, and dopamine levels and decrease glutamate and aspartic acid levels in the hippocampus and cortex and increase glycine and serotonin levels in the blood. Conclusions: After effectively administrated, ginsenosides attenuate d-gal- and Al-induced spatial memory impairment. The possible mechanism of the beneficial effect is restoring the dysfunction of various neurotransmitters.
Article
Huntingtin (HTT) is now a famous protein because an abnormal expansion of a glutamine stretch (polyQ) in its N-terminal sequence leads to the devastating neurodegenerative disorder Huntington's disease (HD). The gene encoding huntingtin, HTT, and its dominantly inherited mutation were identified more than 20 years ago. Subsequently, in the hope of finding a cure for HD, there has been intense research aimed at understanding the molecular mechanisms underlying the deleterious effects of the presence of the abnormal polyQ expansion in HTT. Notwithstanding with the value of this approach, evidence has been emerging of a potential role of context and function of the HTT protein in the specificity and severity of the pathogenicity. HTT is ubiquitous both at the tissue and subcellular levels. It interacts with many partners and has long been considered having no clearly defined cellular function. Based on research over the past 20 years, specifically focused on the function of wild-type HTT, we reconsider the literature describing HTT-regulated molecular and cellular mechanisms that could be dysfunctional in HD and their possible physiological consequences for patients.
Article
This monograph describes the progress in neuropathological HD research made during the last century, the neuropathological hallmarks of HD and their pathogenic relevance. Starting with the initial descriptions of the progressive degeneration of the striatum as one of the key events in HD, the worldwide practiced Vonsattel HD grading system of striatal neurodegeneration will be outlined. Correlating neuropathological data with results on the functional neuroanatomy of the human brain, subsequent chapters will highlight recent HD findings: the neuronal loss in the cerebral neo-and allocortex, the neurodegeneration of select thalamic nuclei, the affection of the cerebellar cortex and nuclei, the involvement of select brainstem nuclei, as well as the pathophysiological relevance of these pathologies for the clinical picture of HD. Finally, the potential pathophysiological role of neuronal huntingtin aggregations and the most important and enduring challenges of neuropathological HD research are discussed. © Springer International Publishing Switzerland 2015. All rights reserved.
Article
Aim: Ginsenosides, a class of ginseng compounds of herbal medicine, have been shown to have therapeutic potential for the neuroprotection of brain damage after cerebral ischemia because of their activities including anti-oxidant and anti-inflammation. In the elderly population, aging-induced oxidative stress has been implicated in exacerbating brain injury, which might also be ameliorated by anti-oxidants, such as ginsenosides. However, this hypothesis has yet to be explored. Methods: Here we present in vivo studies highlighting a protective function of ginsenoside Rb1, a natural steroid glycoside derivative purified from saponin of Panax ginseng, in neurological injury during aging. Results: Compared with young mice, the recovery of brain damage after middle cerebral artery occlusion is significantly impaired in aged mice, whereas the long-term pretreatment with ginsenoside Rb1 through oral administration can greatly prevent the injury in a dose-dependent manner. In addition, we further explored the involvement of oxidative stress and extracellular signal-regulated kinase activation in aged mice stimulated by cerebral ischemia, both of which were found to be blocked by ginsenoside Rb1. Conclusions: These observations suggest that ginsenoside Rb1 could represent promising applications as anti-oxidants for the anti-aging treatment of neurological disorders, such as stroke, in elderly patients. Geriatr Gerontol Int 2015; ●●: ●●-●●.
Article
According to the results of my study on the chromatographic analysis of fresh ginseng (Panax ginseng C. A. Meyer) roots, most of the contents of protopanxadiol ginsenosides Rb1, Rc, Rb2, and Rd are derived from the corresponding malonyl ginsenosides in fresh ginseng by a heat process. Also, I confirmed that acetyl ginsenosides are naturally occurring constituents in fresh ginseng, not decarboxylates from malonyl ginsenosides. Seven neutral ginsenosides Rg1, Re, Rf, Rc, Rb1, Rb2, and Rd were transformed to specific conversions in red ginseng preparation conditions. The conversion paths progress by three rules concluded from my study. These conversion rules are I: the ether bond is stable at positions 3 and 6 in the dammarane skeleton, II: the ether bond between sugars is stable in glycosides, and III: the ether bond to glycosides is unstable at position 20 in the dammarane skeleton.