Excellent Paper Polyphenols For Different Disease PDF

You might also like

You are on page 1of 17

Ageing Research Reviews 11 (2012) 329–345

Contents lists available at SciVerse ScienceDirect

Ageing Research Reviews


journal homepage: www.elsevier.com/locate/arr

Review

Natural polyphenols against neurodegenerative disorders: Potentials and pitfalls


Azadeh Ebrahimi ∗ , Hermann Schluesener
Division of Immunopathology of the Nervous System, Department of Neuropathology, Institute of Pathology and Neuropathology, University of Tuebingen, Calwer Str. 3, D-72076
Tuebingen, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Within the last years, a rapidly growing number of polyphenolic compounds with neuroprotective effects
Received 1 November 2011 have been described. Many efforts have been made to explore the mechanisms behind the neuroprotec-
Received in revised form tive action of polyphenols. However, many pathways and mechanisms considered for mediating these
23 December 2011
effects are rather general than specific. Moreover, despite the beneficial effects of polyphenols in exper-
Accepted 31 January 2012
imental treatment of neurodegeneration, little has been achieved in bringing them into routine clinical
Available online 9 February 2012
applications. In this review, we have summarized the protective effects of polyphenols against neu-
rodegeneration, and we have also discussed some of the barricades in translating these biochemical
Keywords:
Polyphenol
compounds, into relevant therapeutics for neurodegenerative diseases.
Neurodegenerative disease © 2012 Elsevier B.V. All rights reserved.
Neuroprotection
Alzheimer’s disease
Parkinson’s disease

1. Why polyphenols? success in bringing these compounds into routine clinical applica-
tion has been limited.
Natural polyphenols are the most commonly found chemical Studies on neuroprotective effects of polyphenols can be divided
compounds in consumed herbal beverage and food worldwide into the following categories: (1) neuroprotective action through
(Joseph et al., 2007; Ramos, 2007). They constitute a large group antioxidant pathways, (2) interaction with signaling pathways,
of phytochemicals with more than 8000 identified compounds (3) neuroprotection through modulation of neural mediators and
(Table 1). enzymes like acetylcholine (Ach) and acetylcholinesterase (AChE),
The primary function of these compounds is protection of plants (4) inhibition of NMDA neurotoxicity and (5) anti-amyloidogenic
against reactive oxygen species (ROS), produced during photosyn- effects.
thesis, and consumption by herbivores (Ramos, 2007). Within the In this review, we have focused on neuroprotective effects of
previous decades, most of the studies on polyphenols have been natural polyphenols and their interaction with specific molecular
focused on anti-oxidant properties of these chemical compounds as targets and pathways of neurodegeneration. We have discussed
their most prominent effect (Halliwell, 2001). Along with introduc- potentials and pitfalls of using these chemical compounds as neu-
ing resveratrol, as a potential anti-aging agent, much focus has been roprotective agents in in vitro and in vivo experiments as well as
placed on protective effects of various polyphenols against aging in clinical setting.
and related neurodegenerative diseases (Kay, 2010). Increase in life
span by polyphenols can be associated with increased or improved 2. Polyphenols as antioxidants
brain function. For instance, epigallocatechin gallate (EGCG) post-
poned the onset of neurological symptoms and prolonged life span The most prominently discussed effect of polyphenols is their
in a mice model of amyotrophic lateral sclerosis (ALS) (Koh et al., antioxidant activity. It is now established that oxidative/nitrosative
2006; Xu et al., 2006). Long term treatment with epigallocatechin stress (OS/NS) has a pivotal role in pathophysiology of neurodegen-
gallate increased the life span and enhanced movement abilities in erative diseases and many other types of human maladies (Aluise
a transgenic Drosophila melanogaster model of PD (Ortega-Arellano et al., 2010; Bjelakovic et al., 2011; Gotz et al., 1994; Halliwell, 2001;
et al., 2011). Despite the prominent evidence of neuroprotective Tesch and Lim, 2011). Oxidative damage to neuronal molecules,
effects of polyphenols from in vitro and preclinical models, overall accumulation of iron ion species in the brain, and decreased cellular
reserve antioxidant pool are major pathological aspects of neu-
rodegenerative disorders, like Parkinson’s disease (PD), Alzheimer’s
∗ Corresponding author. Fax: +49 7071 294846. disease (AD) or Amyotrophic lateral sclerosis (Gotz et al., 1990;
E-mail address: azadehebr@yahoo.com (A. Ebrahimi). Halliwell, 1992; Mandel et al., 2004b, 2005; Riederer et al., 1989;

1568-1637/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
doi:10.1016/j.arr.2012.01.006
330 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

Table 1
A simplified classification of natural polyphenols.

Polyphenols Falvonoids Anthocyanins E.g. aurantinidin, cyanidin, delphinidin, europinidin, luteolinidin,


pelargonidin, malvidin, peonidin, petunidin, rosinidin, etc.
Flavonols E.g. 3-hydroxyflavone, azaleatin, fisetin, galangin, gossypetin,
kaempferide, kaempferol, isorhamnetin, morin, myricetin,
natsudaidain, pachypodol, quercetin, rhamnazin, rhamnetin, etc.
Flavones E.g. apigenin, luteolin, tangeritin, chrysin, 6-hydroxyflavone, baicalein,
scutellarein, wogonin, diosmin, flavoxate, etc.
Flavanones E.g. butin, eriodictyol, hesperetin, hesperidin, homoeriodictyol,
isosakuranetin, naringenin, naringin, pinocembrin, poncirin,
sakuranetin, sakuranin, sterubin, etc.
Isoflavonoids Isoflavones E.g. genistein, daidzein, lonchocarpane, laxiflorane, etc.
Isoflavanes E.g. equol, etc.
Flavanols Monomers E.g. catechin, epicatechin (EC), epigallocatechin (EGC), epicatechin
gallate (ECG), epigallocatechin gallate (EGCG), epiafzelechin,
fisetinidol, guibourtinidol, mesquitol, robinetinidol, etc.
Oligomers and polymers E.g. theaflavins, thearubigins, condensed tannins, proanthocyanidins
etc.
Non-flavonoids Phenolic acids Derivatives of cinnamic acid E.g. p-coumaric acid, caffeic acid, chlorogenic acid, ferulic acid, sinapic
acid, etc.
Derivatives of benzoic acid E.g. gallic acid, gentisic acid, protocatechuic acid, syringic acid, vanillic
acid, etc.
Lignans E.g. pinoresinol, podophyllotoxin, steganacin, etc.
Stilbenes E.g. resveratrol analogs, etc.

Weinreb et al., 2004). It has been shown, that severe hypoxia or Leibold, 1999; Hofer et al., 2002; Thomas and Kim, 2005; Wang
ischemia episodes increase the susceptibility to development of AD and Pantopoulos, 2005; Zhou et al., 2004). Catechins, like EGCG, are
(Desmond et al., 2002). Hypoxia, in fact, can induce amyloid pre- a group of polyphenols which exert their neuroprotective effects
cursor protein (APP) up-regulation at both the mRNA and protein through induction of the HIF-1␣ pathway (Weinreb et al., 2007;
level and subsequently leads to amyloid beta (A␤) accumulation Zhou et al., 2004). Resveratrol is another non-flavonoid polyphenol
(Chen et al., 2003; Jendroska et al., 1997; Shi et al., 2000). NS can with significant antioxidant activity, activating the HIF-1␣ pathway
damage bio-molecules through reactive species such as peroxyni- (Harikumar and Aggarwal, 2008).
trite and plays also a crucial role in PD by triggering mitochondrial However, there is also evidence of definite links between
dysfunction (Lin and Beal, 2006; Mattson, 2004; Mythri et al., 2011; hypoxia, HIF-1 activation and APP/A␤ production (Wang et al.,
Reddy, 2006). 2006b; Zhang et al., 2007a,b). The precise role of HIF-1 alpha path-
Polyphenols exert their antioxidant effects through differ- way in neurodegeneration, albeit positive or negative, is in fact a
ent mechanisms like interaction with the HIF-1 alpha pathway, matter of debate (Ogunshola and Antoniou, 2009). HIF-1␣ acts like
inducing expression of protective genes against OS, regulation of a double-edged sword that can be both beneficial and detrimental
reactive oxygen species by interacting with oxidative pathways to neural cell survival (Ogunshola and Antoniou, 2009).
and scavenging metal ions as pathogenic free radicals (Kelsey et al., On the other hand, there are contradictory reports on effects
2010). Table 2 summarizes recent studies on antioxidant effects of of the same polyphenol on HIF-1␣ expression and activity. For
polyphenols in neurodegenerative processes. example, in one study EGCG has been shown to exert antioxidant
effects through HIF-1 pathway activation, while in another study an
inhibitory effect on the same pathway has been reported (Domingo
2.1. Hypoxia inducible factor 1 (HIF-1) alpha pathway
et al., 2010; Weinreb et al., 2007; Zhou et al., 2004). Similarly,
other polyphenols have been reported to exert antioxidant effects
One strategy of neuroprotection is activation of hypoxia sig-
through inhibition rather than activation of the HIF-1 pathway (Lu
nal transduction pathways through which the hypoxic condition is
et al., 2009; Terzuoli et al., 2010).
sensed and appropriate genes are activated and expressed in order
Therefore, claiming neuroprotective effects for polyphenols
to mediate compensatory survival conditions for the cells. A high
based on HIF-1 pathway modulation in vitro is still a matter of
percentage of hypoxic responses in cells, are controlled by hypoxia-
debate.
inducible transcription factor-1 (Ogunshola and Antoniou, 2009).
The HIF-1 complex is a heterodimer, consisting of two subunits:
The HIF-1␣ oxygen-regulated subunit and the HIF-1␤ subunit, also
known as aryl hydrocarbon receptor nuclear translocation (ARNT) 2.2. ROS regulation
(Ratcliffe et al., 1998; Semenza, 1998; Soucek et al., 2003; Wang
and Semenza, 1993; Wenger and Gassmann, 1997). Alterations Free oxygen radicals can damage cellular micro-organelles
in oxygen levels regulate HIF-1␣ activity. Under normoxic condi- directly and in this regard mitochondrial damage is of great impor-
tions, HIF-1␣ is degraded by HIF prolyl-4-hydroxylases (Jaakkola tance to neurodegenerative diseases (Higuchi et al., 2003; Lin and
et al., 2001). These enzymes require iron as co-factor and oxy- Beal, 2006). Oxygen radicals can also reduce free metal ions to active
gen as co-substrate (Ivan et al., 2001; Jaakkola et al., 2001). In radicals, like super oxide anions, which are responsible for reduc-
contrast, under hypoxic conditions, HIF-1␣ is translocated to the tion of ferric (Fe3+ ) to ferrous (Fe2+ ) through the Fenton reaction
nucleus where it dimerises with ARNT and subsequently binds to (Higuchi et al., 2003).
hypoxic binding sites of target genes involved in cell survival, gly- Polyphenols scavenge superoxide and hydroxyl radicals, as
colysis, angiogenesis, erythropoiesis, and iron metabolism (Hofer well as the 1,1-diphenyl-3-picrylhydrazyl radical, peroxyl radicals,
et al., 2002). Consequently, stabilizing HIF-1␣ would be a strategy nitric oxide, carbon-center free radicals, singlet oxygen and lipid
to promote further cytoprotective events. Some natural polyphe- free radicals, and peroxynitrite (Guo et al., 1996; Lin et al., 2003;
nols induce HIF-1␣ protein and lead to a further increase in mRNA Morel et al., 1993; Nanjo et al., 1996; Pannala et al., 1997; Salah
levels of HIF-1␣ target genes (Hanson et al., 1999; Hanson and et al., 1995; Spencer et al., 2001b; Zhao et al., 2001).
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 331

Table 2
Antioxidant effects of polyphenols in in vivo and in vitro models of neurotoxicity and neurodegeneration.

Substance Studied cell line/animal model Effect References

Aloe-emodin N-methyl-d-aspartate • Elevates levels of RNA and protein Lin et al. (2007)
(NMDA)-induced toxicity in retinal expression of superoxide dismutase
ganglion cells (RGCs) (SOD)
• Attenuates NMDA-induced apoptosis
of RGCs
Curcumin N27 dopaminergic neurons • Protects against mitochondrial Mythri et al. (2011)
complex I inhibition (leading to
mitochondrial dysfunction) and NS
Curcumin Homocysteine-induced neurotoxicity • Reduces Malondialdehyde (MDA)a Ataie et al. (2010)
in rats and Superoxide anion levels
• Reduces lipid peroxidation
Improves learning and memory in rats
Curcumin N27 dopaminergic neuronal cell line • Increases glutathioneb (GSH) levels Harish et al. (2010)
Curcumin 3-Nitropropionic acid (3-NP)-induced • Improves the 3-NP-induced motor Kumar et al. (2007)
neurotoxicity in rats and cognitive impairment
• Attenuates 3-NP-induced OS
(including lipid peroxidation, reduced
GSH and nitrite activity)
• Restores the decreased succinate
dehydrogenasec activity
Epigallocatechingallat (EGCG) Glucose oxidase-induced neurotoxicity • Enhances cellular resistance to Romeo et al. (2009)
in H 19-7 (a rat neuronal cell line) glucose oxidase-mediated oxidative
damage
• Elevates heme oxygenase-1d (HO-1)
mRNA and protein expression
• Activates transcription factor Nrf2e
EGCG Glutamate-induced toxicity in HT22 • Reduces glutamate-induced Kang et al. (2010)
mouse hippocampus neuronal cells, oxidative cytotoxicity
Kainic acid-induced neurotoxicity in • Inactivates the NF-␬B signaling
rats pathwayf
• Reduces ROS accumulation and
NF-␬B transcriptional activity
EGCG Transient global cerebral ischemia • Reduces the development of delayed Park et al. (2009)
C57BL/6 in mice neuronal death after transient global
cerebral ischemia in mouse brain
EGCG Age-associated oxidative damage in rat • Amplifies the activities of enzymic Srividhya et al. (2008)
brain antioxidants like SOD, catalase,
glutathione peroxidase, glutathione
reductase and glucose-6-phosphate
dehydrogenase
• Improves the activity of non-enzymic
antioxidants like tocopherol, ascorbic
acid and glutathione
• Ameliorates the MDA and protein
carbonyl levels
EGCG Progressive neurotoxic model of • Decreases protein levels and mRNA Weinreb et al. (2007)
long-term serum deprivation in human expression of the beta subunit of the
SH-SY5Y neuroblastoma cells enzyme prolyl 4-hydroxylase
• Decreases protein levels of two
molecular chaperones that are
associated with HIF regulation, the
immunoglobulin-heavy-chain binding
protein and the heat shock protein 90
beta
EGCG SOD1-G93A transgenic mice (a model • Maintains the normal expression of Koh et al. (2006), Xu et al.
of ALS) p85a PI3-K, pAkt, and pGSK-3 (2006)
(molecular signals of survival)
• Reduces activation of NF-kB and the
cleaved form of caspase-3
• Reduces microglial activation
Prolongs the life span
• Delays the onset of symptoms
Mangiferin Glutamate-induced neurotoxicity in • Prevents neuronal death, oxidative Lemus-Molina et al. (2009)
rat cerebral cortex neurons stress and mitochondrial
depolarization
Mangiferin 1-Methyl-4-phenyl pyridinium • Restores the GSH content (to 60% of Amazzal et al. (2007)
(MPP(+))-induced oxidative stress in control levels), and down-regulates
the murine neuroblastoma cell line both SOD and catalase mRNA
N2A expression
• Quenches reactive oxygen
intermediates
Mangiferin Glutamate-induced neurotoxicity in • Reduces ROS formation Campos-Esparza et al.
Morin rat primary culture of neurons • Activates enzymatic antioxidant (2009)
system
• Restores mitochondrial membrane
potential
332 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

Table 2 (Continued )

Substance Studied cell line/animal model Effect References

Polyphenol-rich Hedeoma Biochemical assay on rat brain • Inhibits lipid peroxidation Dade et al. (2011)
multiflorum extract homogenates Scavenges
2,2 -diphenyl-1-picrylhydrazyl (DPPH)
radicals
Polyphenol-rich osmanthus Glutamate, arachidonic acid, and • Scavenges DPPH and hydroxyl anions Lee et al. (2007)
fragrans extract 6-hydroxydopamine-induced Inhibits lipid peroxidation
neurotoxicity in rat primary cortical
neurons
Red wine polyphenol Rat model of ischemic cerebral stroke • Prevent the burst of excitatory amino Ritz et al. (2008)
compounds acids in response to ischemia
• Reduce brain infarct volumes
• Enhance the residual cerebral blood
flow during occlusion and reperfusion
• Modulate expression of proteins
involved in the maintenance of
neuronal caliber and axon formation
Resveratrol Lipopolysaccaride (LPS)-induced • Reduces NADPHg oxidase-mediated Zhang et al. (2010)
dopaminergic neurodegeneration in rat generation of ROS
• Inhibits microglia activation
• Attenuates the activation of MAPK
and NF-␬B signaling pathways
• Implies neuroprotection against
LPS-induced dopaminergic
neurodegeneration
Resveratrol Glutamate-induced toxicity in mice • Induces heme oxygenase 1d (HO-1) in Sakata et al. (2010)
primary culture of neurons a dose- and time-dependent manner
Optimized ischemic-reperfusion stroke • Protects mouse neurons, subjected to
model in mice an optimized ischemic-reperfusion
stroke model
• Protects neurons against ecitotoxicity
Resveratrol 1-methyl-4-phenyl-1,2,3,6- • Protects from MPTP-induced motor Lu et al. (2008)
tetrahydropyridine (MPTP)- induced coordination impairment, hydroxyl
parkinson in mice radical overloading, and neuronal loss
• Scavenges free radicals
Resveratrol MPP(+)-induced neurotoxicity in • Prevents accumulation of ROS, Okawara et al. (2007)
dopaminergic neurons of midbrain depletion of cellular glutathione, and
slice culture cellular oxidative damage induced by
MPP(+)
• Activates sirtuin family of
NAD-dependent histone deacetylases
Resveratrol A-beta induced toxicity in neurons • Maintains normal expression of Manczak et al. (2010)
from a mouse model (Tg2576 line) and peroxiredoxins and mitochondrial
mouse neuroblastoma (N2a) cells structural genes
• Maintains normal mitochondrial
function
Tea polyphenol NMDA-induced neurotoxicity in mice • Attenuates the increased production Chen et al. (2008)
of synaptosomal ROS
• Reduces the deteriorative
ROS-sensitive Na(+), K(+)-ATPase and
Mg(2+)-ATPase activity
a
Malondialdehyde (MDA) is a highly reactive electrophile species that occurs naturally form degradation of polyunsaturated lipids and is a biomarker for measuring the
levels of OS.
b
Gluthation (GSH) is an antioxidant, preventing damage to important cellular components caused by ROS such as free radicals and peroxides.
c
Succinate dehydrogenase is a Complex II mitochondrial enzyme.
d
Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme.
e
Transcription factor Nrf2 is a master regulator of the antioxidant response.
f
NF␬B signaling pathway is a pathway which is activated in response to cell stresses including OS.
g
NADPH is the reduced form of NADP+ , a coenzyme used in anabolic reactions, such as lipid and nucleic acid synthesis.

Amongst different polyphenols, EGCG has shown to be the most in many cell degeneration pathways and act as a non-specific rather
efficient radical scavenger, even among its other counterparts like than specific neural damage mechanism. Although ROS regulation
ECG, EC and EGC (Guo et al., 1999; Nanjo et al., 1996, 1999). Strong can be mentioned as an additive mechanism of neuroprotection of
scavenging properties of EGCG are due to several hydroxyl groups polyphenols, claiming a therapeutic effect for polyphenols via ROS
at the side rings of the chemical core (Guo et al., 1999; Nanjo et al., regulation as the only mechanism seems to be overdrawn.
1996, 1999). Hydroxyl groups are especially important in biological
chemistry because of their tendency to form hydrogen bonds both 2.3. Metal ion chelating
as donor and acceptor. In fact, polyphenols with hydroxyl groups
can act as strong reducing agents and vice versa. Polyphenols are also able to chelate metal ions, like copper (II)
Another important feature of some polyphenols is modulating and iron (III), to prevent free radical damage (Guo et al., 1999).
the activity of enzymes involved in OS. In this regard, previous stud- In this regard, some polyphenols have shown to be more efficient
ies have shown that EGCG can increase the activity of SOD and than traditional antioxidants like Vitamin E and C (Nanjo et al.,
catalase, two important antioxidant enzymes in the mouse stria- 1996; Pannala et al., 1997). Studies have shown that some polyphe-
tum (Levites et al., 2001). However, ROS generation is the final step nols can inhibit lipid peroxidation in the brain (Guo et al., 1996;
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 333

Table 3
Polyphenols with neuroprotective properties and their target cellular signaling pathways.

Substance Pathway References

Curcumin PI-3 K/MAPK signaling pathways Lin et al. (2011)


EGCG NF-␬B signaling pathway Kang et al. (2010), Kim et al. (2007)
EGCG Akt signaling pathways Chao et al. (2010)
EGCG HIF-1␣ pathway Weinreb et al. (2007)
EGCG Erk1/2 pathway Levites et al. (2002a), Weinreb et al. (2007)
EGCG PKC pathway Kalfon et al. (2007), Levites et al. (2003)
Fisetin ERK pathway Maher et al. (2011)
Fisetin NF-␬B signaling pathway Zheng et al. (2008)
GSE Erk1/2 pathway Kim et al. (2011), Min et al. (2010)
Huperzine A PKC pathway, MAPK pathway Peng et al. (2006), Peng et al. (2007)
Naringenin MAPK pathway Yang et al. (2011)
Resveratrol ERK pathway Maher et al. (2011)
Resveratrol NF-␬B signaling pathway Chen et al. (2005), Jang and Surh (2003), Zhang et al. (2010)
Resveratrol SIRT1- uncoupling protein 2 pathway Anekonda (2006), Anekonda and Reddy (2006), Della-Morte et al. (2009)
Resveratrol AMPK signaling pathway Dasgupta and Milbrandt (2007)
Resveratrol Nrf2/ARE antioxidant pathway Chen et al. (2005), Ghanim et al. (2011)
Resveratrol PKC pathway Han et al. (2004)
Resveratrol PI3K/AKT pathway Fukui et al. (2010)
Mangiferin, morin Erk1/2 pathway, Akt signaling pathway, NF-␬B signaling pathway Campos-Esparza et al. (2009)

Levites et al., 2002b). Polyphenols seem to exert this effect, through PKC over-expression has been shown to reduce amyloid
chelating ferrous ions. For instance, EGCG attenuates paraquat- plaque formation and A␤ levels in human APP transgenic mice
induced microsomal lipid peroxidation and increases the survival (Etcheberrigaray et al., 2004). There is strong evidence, that PKC
of paraquat-poisoned mice, a PD model, through this mechanism signaling pathways regulate important molecular events involved
(Higuchi et al., 2003; Ossowska et al., 2006). in associative memory storage, and signaling deficits of PKC sig-
The ability of polyphenols to chelate metal ions contributes to naling pathways play an important role in the pathophysiology of
their neuroprotective activity via inhibition of transition metal cat- neurodegenerative disorders like AD (Alkon et al., 2007; Mattson,
alyzed free radical formation. Two attachment sites for metal ions 1991). EGCG, huperzine A and resveratrol have shown to interact
have been suggested within the molecular structure of flavonols, with PKC signaling pathway (Table 3).
a subclass of polyphenols: The o-diphenolic groups in the 30, 40- The MAPK signaling pathway is a general target of flavonoids,
dihydroxy positions in the B ring, and the keto structure 4-keto, notably in the nervous system in the context of oxidative insults
3-hydroxy or 4-keto and 5-hydroxy in the C ring (Thompson et al., (Williams et al., 2004). ERK1/2 and c-jun amino-terminal kinase
1976; van Acker et al., 1996). These functional groups bind tran- (JNK), two important components of MAPK signaling pathway, are
sition metal ions, such as iron or copper (Rice-Evans et al., 1996). involved in various forms of cellular plasticity, such as differentia-
With a closer look at the molecular structures, the same chemical tion and apoptosis (Mielke and Herdegen, 2000; Yuan and Yankner,
and molecular features can also be found in other polyphenol sub- 2000).
classes (Bors et al., 2001). Therefore, we can expect a broad range The phosphatidylinositol 3 kinase (PI3K)-Akt/PKB pathway has
of polyphenols with the ability to chelate metal ions. a pivotal role in neuronal survival (Rodgers and Theibert, 2002).
Activation of Akt/PKB in neurons leads to the inhibition of central
proteins of the cell death machinery, such as the proapoptotic Bcl-2
2.4. Modulating cell signaling pathways family member BAD and members of the caspase family (Bonni
et al., 1999; Datta et al., 1999; Zha et al., 1996). Akt1, an effec-
Several polyphenols have been shown to interact with cellu- tor molecule of (PI3K)-Akt/PKB pathway, has also been shown to
lar signaling pathways which are directly or indirectly involved attenuate the apoptotic effect of Abeta (25–35) (Martin et al., 2001).
in neurodegeneration. Most of these pathways are, in fact, sig- Quercetin, has been shown to exert inhibitory activity on (PI3K)-
naling pathways involved in cell survival and programmed cell Akt/PKB pathway through inhibiting PI 3K (Spencer et al., 2003).
death. In this respect, polyphenols have been reported to act at However, a large number of protein kinases, important medi-
phosphoinositide 3-kinase (PI 3K), Akt/protein kinase B (Akt/PKB), ators of different cell signaling pathways, have been reported as
tyrosine kinases, protein kinase C (PKC), and mitogen activated pro- being potential targets of different polyphenols (Williams et al.,
tein kinase (MAPK) signaling cascades (Table 3) (Williams et al., 2004). Therefore, we have an apparent lack of selectivity of action
2004). They affect cellular function by altering the phosphoryla- for polyphenols in this respect. This might arise from the fact that
tion mode and expression level of targeted molecules within the many studies reporting the interactions of polyphenols with cell
mentioned pathways. signaling pathways have not defined the primary cellular site of
The flavonoid superfamily of polyphenols has the potential to action of the studied polyphenol.
bind to the ATP-binding sites of a large number of proteins of cel-
lular signaling pathways, including mitochondrial ATPase, calcium
plasma membrane ATPase, protein kinase A, protein kinase C and 3. Polyphenol’s anti-acetycholinesterase activity
topoisomerase (Barzilai and Rahamimoff, 1983; Boege et al., 1996;
Conseil et al., 1998; Di Pietro et al., 1975; Kantengwa and Polla, The concept of cholinergic system deficit in neurodegeneration
1991; Revuelta et al., 1997; Rosenblat et al., 1999; Ursini et al., and its important role in cognition was proposed nearly 30 years
1994), as well as to benzodiazapine binding sites of GABA-A recep- ago (Perry et al., 1978). Loss of cholinergic activity, atrophy of the
tors and adenosine receptors (Dekermendjian et al., 1999; Medina nucleus basalis of Meynert as the major source of acetylcholine,
et al., 1997). Resveratrol and the citrus flavanones, hesperetin and and loss of cortically projecting cholinergic neurons, along with
naringenin inhibit the activity of a number of protein kinases (Pallas increasing cognitive deficits, are some of notable findings in differ-
et al., 2009; Sabarinathan et al., 2011; Vauzour et al., 2007). ent neurodegenerative diseases, like Alzheimer’s and Parkinson’s
334 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

diseases (Bohnen et al., 2005; Davis et al., 1999; Hanyu et al., 2002; and patients with vascular dementia (Xu et al., 1999, 1995; Zhang
Klein et al., 2010; Lahiri et al., 2004; Rosengarten et al., 2010; et al., 2002). Several meta-analyses have shown that administra-
Teipel et al., 2005; Whitehouse, 1998). Cholinergic dysfunction tion of huperzine A for at least 8 weeks might lead to a significant
in neurodegenerative diseases can be the result of reduction in improvement in cognitive function, mood, behavior and daily activ-
Ach synthesis due to reduced choline acetyltransferase (ChAT) or ity of patients with AD. Most of its side effects are cholinergic in
choline uptake, cholinergic neuronal and axonal abnormalities, and nature and are generally mild and of brief duration (Li et al., 2008;
degeneration of cholinergic neurons (Fisher, 2008). Accordingly, Man et al., 2008; Wang et al., 2009a). Combinatorial regimens with
using acetylcholinesterase inhibitors, which exert their efficacy other selective AChE inhibitors have shown even more promising
through stimulation of both muscarinic and nicotinic acetylcholine results (Camps and Munoz-Torrero, 2001; Wang et al., 2003; Zhang
receptors (mAChR and nAChR), has been a proper therapeutic et al., 2007c). Huperzine A has also butyrylcholinesterase inhibitory
approach to alleviate the cognitive symptoms of neurodegenerative activity which is not as promising as the anti-AChE activity (Ogura
diseases (Lahiri et al., 2004; Larner, 2010; Rosengarten et al., 2010). et al., 2000).
There are two distinct receptor subtypes in the brain for Ach: nico- Not all polyphenols have an anti-cholinesterase activity. Some
tinic and muscarinic. nAChRs are mainly ligand-gated ion channels, of them have a reverse effect. For example, caffeic acid increases
while mAChRs are metabotropic receptors. Muscarinic receptors AchE activity and expression (Rezg et al., 2008).
include five distinct receptor subtypes (M1–M5). M1 mAChR is the
most abundant subtype in cerebral cortex and hippocampus, the
4. Polyphenols and protective effects against NMDA
most sensitive brain areas to the development of amyloid plaques
neurotoxicity
and neurofibrilary tangles (Levey et al., 1991; Wei et al., 1994).
Several natural polyphenols have shown cholinesterase
The role of NMDA neurotoxicity and glutamate excitotoxicity
inhibitory effect (Zhang et al., 2009). In most in vivo studies,
in neurodegenerative diseases like Huntington’s disease (HD), AD
the anticholinergic activity of polyphenol was accompanied by
and even in cognitive impairment associated with aging has been
improvement of cognitive functions, like learning and memory
confirmed many years ago (Choi et al., 1990; Fan and Raymond,
(Coma et al., 2010; Huang and Zhang, 2010; Kim et al., 2004;
2007; Rosi et al., 2004; Waxman and Lynch, 2005). Excessive acti-
Liu et al., 2010; Papandreou et al., 2009; Tota et al., 2010). How-
vation of NMDA receptors induces the production of damaging
ever, the exact mechanism of interaction of polyphenols with
free radicals (e.g., NO and ROS) and other enzymatic processes
the cholinergic system is still not clear. EGCG has shown strong
that contribute to neuronal damage and cell death (Bonfoco et al.,
anti-acetylcholinesterase activity (Zhang et al., 2009). In another
1995; Budd et al., 2000; Dawson et al., 1991; Lafon-Cazal et al.,
study, EGCG has been reported to modulate nAChR signaling
1993; Lipton et al., 1993; Lipton and Rosenberg, 1994). Therefore,
pathway through down regulation of ␣9-nAChR expression as
blocking the NMDA pathway has been a therapeutic strategy for
well as inhibition of (3H)-Nic/␣9-nAChR binding activity (Tu et al.,
cognitive impairment not only in neurodegenerative diseases but
2011). Resveratrol has shown in a study to block acetylcholine
also in psychiatric disorders with cognitive dysfunction (Lipton,
release from adrenal chromaffin cells (Chang-Mu et al., 2010).
2007; Shim et al., 2008). There is strong evidence of protective
Some polyphenols such as huperzine A, quercetin, kuwanon U,
effects of several natural polyphenols against NMDA neurotoxic-
E, and C, kaempferol, tri- and tetrahydroxyflavone, etc. have
ity (Table 4) (Chang-Mu et al., 2010; Davis et al., 1999; Shim et al.,
shown anti-butyrylcholinesterase effects in addition to their
2008; Yazawa et al., 2006; Zhang et al., 2008). Polyphenols, act at
anti-cholinesterase activity (Kim et al., 2011; Min et al., 2010; Tota
different locations within the NMDA pathway. (Fig. 1).
et al., 2010; Wang et al., 2011, 2010c, 2001; Zhang et al., 2009).
In most of these studies, a protective effect against NMDA exci-
Huperzine A has shown the most promising effects in this
totoxicity has been reported. However, the mechanism of such
respect (Wang et al., 2011, 2010c, 2001; Zhang et al., 2009). Studies
protection has not been clearly addressed. A few in vitro studies
have shown that huperzine A is highly specific for AChE (Zangara,
reported a reduction in frequency and amplitude of AMPA/NMDA
2003). The richest natural source of huperzine A is the plant
receptor mediated spontaneous excitatory postsynaptic currents
Huperzia serrata, a fascinating fungal reservoir of other AChEIs as
(sEPSCs) in pyramidal neurons. But it remains unclear, whether
well (Wang et al., 2010d; Zhu et al., 2010). The drug is a potent,
this is the result of polyphenols’ antioxidant activity or their direct
reversible and selective inhibitor of acetylcholinesterase and its
NMDA receptor blocking effect (Gao et al., 2006; Yazawa et al.,
potency is similar or superior to other AChEIs (Wang and Tang,
2006; Zhang et al., 2008).
2005; Zangara, 2003). Huperzine A has shown a rival potency in
AChE inhibition over drugs, like tacrine, galanthamine, and rivastig-
mine, which are currently being used in the treatment of AD 5. Polyphenols and amyloidopathies
patients (Liang and Tang, 2004; Ogura et al., 2000; Wang and
Tang, 1998a,b; Wang et al., 2006a, 1986; Zhao and Tang, 2002). Tau hyperphosphorylation and beta amyloid accumulation are
Huperzine A has the highest AChE inhibitory (AChEI) activity (IC50) believed to be the core pathologies of tauopathies and amy-
after donepezil, while tacrine, physostigmine, galantamine, and loidopathies (Iqbal et al., 2010; Kawahara and Kuroda, 2000;
rivastigmine were less potent (Ma et al., 2007). Compared to the Kawahara et al., 2000). Excessive A␤ accumulation can be the result
other AChE inhibitors, huperzine A has also shown better penetra- of either increased production or decreased clearance, which in
tion through the blood–brain barrier, higher oral bioavailability, both situations is toxic to the cells (Selkoe, 2001). A␤ aggregation
and longer duration of AChEI activity (Wang et al., 2006a). The leads to the formation of senile plaques (SP) and stimulates a series
mechanism of action of this drug has still not been clearly defined. of biological signaling pathways which leads to an impairment
In silico studies have shown a direct binding of huperzine A to of neuronal synapses and dendrites through oxidative stress and
acetylcholinesterase (Kitisripanya et al., 2011). Clinical trials with inflammatory responses (Heneka and O‘Banion, 2007; Roberson
huperzine A, for treatment of cognitive and functional impairments and Mucke, 2006). Beside SP and A␤ aggregation, neurofibrilary
of AD and schizophrenia and the increase in memory performance tangles (NFTs), consisting of abnormally hyperphosphorylated tau
of normal individuals, have been promising (Sun et al., 1999; Xu protein, are another pathologic hallmark of Alzheimer’s disease
et al., 1999, 1995; Zhang et al., 2002, 2007c). In China, huperzine A (Iqbal et al., 2009; Wallin et al., 2006). For a long time, these two
has been studied in phase IV clinical trials and revealed a signifi- pathological features have been the major therapeutic targets for
cant improvement of memory of elderly people, patients with AD drug development in tau-amyloidopathies. Polyphenols exert their
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 335

Table 4
Recent studies on protective effects of polyphenols against NMDA neurotoxicity.

Substance Cell line/animal model Effect References

Catechin, curcumin, tannic Rat NMDA neurotoxicity model, primary • Inhibits glutamate-induced excitotoxicity Yazawa et al.
acid culture of neurons • Inhibits PKC activity, and subsequent phosphorylation of NR1 of (2006)
the NMDA receptor
• Reduces glutamate-mediated Ca2+ influx
Inhibits glutamate-induced caspase-3 activation
• Reduces glutamate-induced ROS generation
EGCG Unilateral cerebral ischemia in gerbils • Reduces excitotoxin-induced MDA production and neuronal Lee et al. (2004)
damage
• Attenuates the increase in MDA level caused by cerebral ischemia
• Reduces the formation of postischemic brain edema and infarct
volume
Honokiol Mice NMDA toxicity model • Ameliorates behavioral and neurotoxic effects of NMDA Chang-Mu
• Reduces seizure occurrence, score, and latency et al. (2010)
• Decreases ROS production
Resveratrol Acute oxygen-glucose deprivation (OGD) • Reduces the frequency and amplitude of Zhang et al.
model in rat hippocampus slices 2-amino-3-(5-methyl-3-oxo-1,2- oxazol-4-yl) propanoic acid (2008)
(AMPA)-mediated sEPSCs in pyramidal neurons
• Attenuates OGD-induced neuronal impairment
• Reduces the OGD-enhanced AMPA/NMDA receptor mediated
neuronal EPSCs
Tea polyphenol Mice NMDA toxicity model • Ameliorates behavioral and neurotoxic effects of NMDA Chang-Mu
• Reduces seizure occurrence, score, and latency et al. (2010)
• Decreases ROS production
Trans-resveratrol CA1 region of rat hippocampus slices with • Suppresses glutamate-induced currents in postsynaptic CA1 Gao et al.
NMDA toxicity model pyramidal neurons (2006)
• Inhibits postsynaptic glutamate receptors
Morin, mangiferin Glutamate-induced neurotoxicity in rat • Protects cortical neurons from excitotoxic death Campos-
primary culture of neurons • Reduces ROS levels and maintains the homeostasis of the Esparza et al.
enzymatic antioxidant system after excitotoxic event (2009)
• Inhibits glutamate-induced calpain activity
• Regulates the release of pro-apoptotic proteins implicated in
caspase-dependent and -independent apoptotic neuronal death
• Modulates the activity of the Akt and Erk1/2 kinases after
excitotoxic events
• Prevents the activation of NF-␬B and its subsequent translocation
to the nucleus

effect through, modulation of ␣-, ␤- and ␥-secretases, inhibition of


A␤ oligomer formation, inhibition of A␤-induced neurotoxicity and
inhibition of A␤-induced neuroinflammation (Fig. 2).
Several natural polyphenols, have effectively reduced A␤ depo-
sition and A␤ protein concentrations in brain and serum, among
which EGCG has shown the most promising anti-amyloidogenic
effects (Table 5) (Ray et al., 2011; Vislocky and Fernandez, 2010).
Many studies have shown a direct binding of polyphenols to beta
sheet structures (Benaki et al., 2009; Bieschke et al., 2010; Hafner-
Bratkovic et al., 2008; Huang and Zhang, 2010). ECGC induces
␣-secretase cleavage activity and inhibit ␤- and ␥-secretases (Lee
et al., 2009; Obregon et al., 2006). Several other polyphenols reduce
A␤ levels through direct or indirect modulatory effects on ␣, ␤, and
␥-secretases (Table 5). Myricetin, quercetin, kaempherol, morin,
and apigenin directly inhibit ␤-secretase activity in a concentration
dependent manner (Shimmyo et al., 2008a). Some of these com-
pounds, like Dihydroguaiaretic acid (NDGA), GSE, tannic acid and
wine related polyphenols, affect beta aggregates and destabilize
Fig. 1. A schematic illustration of NMDA pathway and polyphenol targets within preformed A␤, while some others affect A␤ (1–40) and A␤ (1–42)
pathway. Polyphenols interact with the NMDA pathway through inhibitory activ- and inhibit polymerization and fA␤ formation (Table 5) (Fig. 2).
ity on various molecular targets. Catechin has shown to be an effective inhibitor
of caspase 3 (Yazawa et al., 2006). NMDA-induced intracellular Ca2+ accumulation
can activate caspase 3, contributing to ROS generation and neural damage (Zeng 6. Barrels and barricades in polyphenol research
et al., 2010). Polyphenols, such as morin, tannic acid, magniferin and resveratrol
affect glutamate-mediated Ca2+ influx (Yazawa et al., 2006). Catechin can eliminate
glutamate-induced toxicity by reducing ROS generation and further neural degen- The broad accessibility of polyphenols as a component of daily
eration. Catechin is also able to inhibit PKC activity (Yazawa et al., 2006). Curcumin dietary intake has always raised the question regarding the preva-
and tannic acid are two polyphenols reducing glutamate induced excitotoxicity by lence and incidence of neurodegenerative diseases in populations
inhibiting PKC activity and subsequent phosphorylation of NR1 of the NMDA recep- with high polyphenol daily dietary intake, like Chinese populations
tor, thereby reducing glutamate induced Ca2+ influx (Yazawa et al., 2006). EGCG
has been shown to interact with the NMDA pathway in unilateral cerebral ischemic
(Checkoway et al., 2002; Ng et al., 2008; Commenges et al., 2000;
gerbils by attenuating ischemia-induced MDA elevation (Lee et al., 2004). Morin Dai et al., 2006). Although some epidemiological studies suggest a
and magniferin are able to inhibit calpain activity and further cell degeneration negative correlation of the amount of daily polyphenol consump-
(Campos-Esparza et al., 2009). tion and the incidence of neurodegenrative disorders, the number
336 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

Fig. 2. A schematic illustration of amyloidogenic and non-amyloidogenic pathways. Polyphenols exert their effect at different sites within the amyloidogenic pathway.
Basically, there are two pathways through which APP can be cleaved. Depending on the enzyme which cleaves the APP, the final product can be amyloidogenic or non-
amyloidogenic (Nunan and Small, 2000). In the non-amyloidogenic pathway, APP is cleaved by the membrane-bound enzyme ␣-secretase within its A␤ domain, resulting
in secretion of extracellular soluble sAPP␣ fragments and a short membrane-bound COOH-terminal fragment, ␣-CTF or C83, which is cleaved by ␥-secretase to p3, a 3-kDa
peptide, whereas APP intracellular domain (AICD) is secreted into the cytoplasm (Cao and Sudhof, 2004; Esch et al., 1990). Within the amyloidogenic pathway, APP cleavage
by beta-site APP-cleaving enzyme1 (BACE-1, ␤-secretase) and ␥-secretase leads to extracellular sAPP␤ fragments and the membrane bound ␤-C-Terminal Fragment (␤-CTF)
or C99 fragment, which is subsequently cleaved by ␥-secretase at the C-terminal part of the A␤ domain. This leads to the release of APP into the intracellular domain (AICD)
and secretion of A␤ into the extracellular spaces (Selkoe, 1998, 2001; Tanzi and Bertram, 2005). Excessive production of A␤ or decreased A␤ clearance in the later pathway
leads to the accumulation and aggregation of A␤, which is toxic to cells. Therefore, inhibition of either ␤ or ␥-secretases or stimulation of ␣-secretase has been a therapeutic
target in amyloidopathies (Nunan and Small, 2000). The polyphenol targets within pathway are indicated by * and a representative compound has been mentioned for each
target. For more polyphenols that can affect the pathway, please refer to Table 5. N: nucleus, G: Golgi apparatus, ER: endoplasmic reticulum.

of such studies are still too small and other confounding variables body (Scalbert and Williamson, 2000; Singh et al., 2008; Thompson
need to be considered to make a realistic conclusion (Weinreb et al., et al., 1976). Proanthocyanidins are virtually not absorbed in the gut
2009). The accurate and exact polyphenol intake has also never because of their high molecular weight and hydrosolubility (Deprez
been investigated or reported in these epidemiological studies. In et al., 2001; Donovan et al., 2002; Holt et al., 2002; Manach et al.,
fact, it is extremely difficult to quantify the health effects of nat- 2005). Except catechins and proanthocyanidins, polyphenols are
ural polyphenols as part of dietary intake, regarding their great mostly glycosylated in food, and glycosylation influences absorp-
diversity in foods, the limited information about their quantity and tion of some of these polyphenols through the gut barrier (Hollman
metabolism, the interacting effect of foods’ chemicals on each other et al., 1995; Morand et al., 2000; Scalbert and Williamson, 2000).
and the presence of too many potential other bioactive compounds For catechins, intestinal absorption can be influenced by esterifica-
(Spencer et al., 2008). tion with gallic acid. The recovery of galloylated catechins in human
However, there are also many of in vivo and in vitro studies on urine after black tea consumption has been reported 10-fold lower
neuroprotective effects of polyphenols. Within the last decade, sig- than that of non-galloylated catechins (Warden et al., 2001).
nificant short course human interventions have been performed In addition, polyphenols that are absorbed via GI are extensively
in order to confirm the bio-efficacy of different polyphenol sub- metabolized through the first-pass effect (Scalbert et al., 2002).
classes (Kennedy et al., 2010; Krikorian et al., 2010). However, they Most dietary polyphenols are quickly eliminated in both urine and
mostly failed to show a validated neuroprotective effect for the bile after ingestion and they undergo phase I and II liver metabolism
studied polyphenols. The poor outcome of these studied might be (Lin et al., 2003; Possemiers et al., 2011). In human subjects, a
due to several factors including the lack of standardized biomark- post-prandial peak 1–2 h after ingestion of various flavonols and
ers of intake, using non-purified compounds, lack of long term flavanols has been reported but this plasma concentration peak
efficacy investigations, lack of standards for an effective dose and is achieved after a longer time course for isoflavones and other
blood level, and lack of knowledge of pharmacokinetics (Kay, 2010). polyphenols (Scalbert and Williamson, 2000). For some polyphe-
Moreover, there is still a lack of a complete understanding of intake, nols, the plasma concentrations decline rapidly within 1–2 h, which
absorption, metabolism and excretion of polyphenols. Therefore shows a rapid metabolism and elimination. To maintain a high con-
the validity of many in vivo and in vitro experiments performed centration of these polyphenols in plasma, a repeated ingestion of
on neuroprotective effects of polyphenolic compounds are still a them is required (Scalbert and Williamson, 2000; van het Hof et al.,
matter of debate. 1999).
The discussed health effects of natural polyphenols depend on In the context of anti-oxidant activity, some polyphenols, like
their bioavailability. EGCG and grape seed polyphenol extract (GSE), exert their effect
Not all natural polyphenols are absorbable through ingestion. locally in the intestine by blocking iron transport across the basolat-
Glycon forms of polyphenols can be absorbed from the small intes- eral membrane of the enterocyte (Kim et al., 2008; Ma et al., 2010),
tine. Most of the natural polyphenols are in the form of ester, glyco- but polyphenols mainly exert their anti-oxidant effect in the body
sides or polymers which are poorly absorbed via the gastrointesti- through their metabolites. Metabolic modifications of polyphenols
nal tract (GI), highly metabolized, and rapidly eliminated from the have been reported in many studies to alter their antioxidant nature
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 337

Table 5
Polyphenols with protective effect against beta amyloidopathy and protein aggregation.

Substance Studied cell line/animal model Effect References

Alvidin Biochemical assay (UV–visible • Implies anti-amyloidogenic effects against whole A␤ Riviere et al. (2008)
measurements) peptides (1–40 and 1–42)
Blueberry polyphenolic Primary hippocampal neurons, LPS, • Antagonizes LPS, DA- or A␤ (42)-induced deficits in CAR Joseph et al. (2010)
fractions dopamine (DA) or A␤ (42)-induced in primary hippocampus neuronal cells
deficits in Ca2+ recovery (CAR)
Curcumin Brain sections of variant • Inhibits in vitro conversion of prion protein (PrP)a and Hafner-Bratkovic
Creutzfeldt–Jakob disease cases formation of protease resistant PrP et al. (2008)
• Binds to the alpha-helical intermediate of PrP
• Binds to the native form of PrP
Curcumin APP/PS1 transgenic mice model of • Reduces A␤ levels in the brain and serum of mice Wang et al. (2009b)
AD) • Reduces amyloid plaques and microgliosis in the brain of
transgenic mice
• Reduces brain A␤ burden and microglia activation
Dihydroguaiaretic acid Biochemical assay (fluorescence • Inhibits A␤ fibril (fA␤)b formation and extension from A␤ Ono et al. (2004)
(NDGA) spectroscopic analysis) (1–40) and A␤ (1–42)
• Destabilizes preformed fA␤ (1–40) and fA␤ (1–42)
EGCG Yeast prion protein Sup35 • Inhibits the formation of aggregating amyloid forms Duennwald and
Biochemical assay (prions) of Sup35 Shorter (2010)
• Remodels preassembled prions
EGCG Biochemical assay • Binds directly to beta-sheet-rich aggregates and Bieschke et al.
mediates the conformational change without their (2010)
disassembly into monomers or small diffusible oligomers
• Converts large, mature alpha-synuclein and A␤ fibrils
into smaller, amorphous protein aggregates that are
nontoxic to mammalian cells
EGCG Biochemical assay (a generic model • Inhibits in vitro fibril formation by RCM kappa-CN Hudson et al.
of fibril formation by protein, • Inhibits amyloid-fibril formation (2009)
reduced and carboxymethylated
kappa-casein (RCM kappa-CN))
EGCG Biochemical assay • Inhibits the fibrillogenesis of both alpha-synuclein and Ehrnhoefer et al.
A␤ (2008)
• Binds directly to unfolded polypeptides and preventing
their conversion into toxic aggregation intermediates
• Promotes the formation of unstructured, nontoxic
alpha-synuclein and A␤ oligomers of a new type
EGCG Inflammatory response induced by • Inhibits IL-6, IL-8, VEGF and PGE2 Kim et al. (2007)
interleukin (IL)-1␤ and A␤ (25–35) • Attenuates cyclooxygenase-2 expression and activation
in human astrocytoma, U373MG of NF-␬B induced by IL-1␤ and A␤ (25–35)
cells • Suppresses IL-1␤ and A␤ (25–35)-induced
phosphorylation of the mitogen-activated protein kinase
p38 and the c-Jun N-terminal kinase, two protein
mediators of MAPK signaling pathway
• Induces mitogen-activated protein kinase phosphatase-1
expression
EGCG Wild type N2 (Bristol), transgenic • Attenuates hydrogen peroxide levels Brown et al. (2006)
mutant strain daf-16 (mgDf50), • Attenuates age-related behavioral decline
and transgenic A␤ muscle • Alleviates an A␤-induced pathological behavior
expressing strain of C. elegans
(behavioral and biochemical
analysis)
EGCG N2a cells stably transfected with • Elevates active ADAM10 protein Obregon et al.
“Swedish” mutant human APP • Increases APP␣ cleavage (2006)
(SweAPP N2a cells) • Increases ␣-secretase cleavage activity
• Produces no significant alteration in ␤-or ␥-secretase
activities
• Elevates soluble sAPP␣
EGCG Human SH-SY5Y neuroblastoma • Shows potent iron-chelating activity Reznichenko et al.
cells, Chinese hamster ovary cells • Increases transferrin receptor TfR protein and mRNA (2006)
(CHO) overexpressing “Swedish” levels
mutant human APP • Reduces immature and full-length cellular holo-APP
• Reduces toxic A␤ peptide generation in Chinese hamster
ovary cells overexpressing the “Swedish” mutant human
APP
EGCG Human SH-SY5Y neuroblastoma, • Increases PKC␣ and PKC in the membrane and the Levites et al. (2003)
rat pheochromocytoma PC12 cells, cytosolic fractions of mice hippocampus
C57/BL mice • Enhances the release of non-amyloidogenic sAPP␣
• Decreases membrane-bound holo-APP levels, with a
concomitant increase in sAPP␣ levels in the hippocampus
EGCG Cultured hippocampal neurons • Increases cell survival Choi et al. (2001)
exposed to A␤ • Decreases caspase activity
• Employs neuroprotection through scavenging ROS
EGCG MPTP- and DA-induced • Prevents the accumulation of iron and ␣-synuclein in the Mandel et al.
neurodegeneration in mice and substantia nigra (2004a)
rats
338 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

Table 5 (Continued )

Substance Studied cell line/animal model Effect References

EGCG Yeast model of HD, Transgenic HD • Inhibits mutant htt exon 1 protein aggregation Ehrnhoefer et al.
flies overexpressing a pathogenic • Modulates misfolding and oligomerization of mutant htt (2006)
Huntingtin gene (htt) exon 1 exon 1 protein
protein • Reduces polyQ-mediated htt-protein aggregation and
cytotoxicity in yeast model of HD
• Ameliorates photoreceptor degeneration and motor
function in flies
EGCG Biochemical assay • Binds to beta-sheet-rich aggregates and mediates the Bieschke et al.
conformational change without their disassembly into (2010)
monomers or small diffusible oligomers
• Converts large, mature alpha-synuclein and A␤ fibrils into
smaller, nontoxic amorphous aggregates
EGCG Human SH-SY5Y neuroblastoma • Reduces the levels of cellular holo- APP in SH-SY5Y cells Avramovich-Tirosh
cells (serum deprivation model), • Reduces levels of toxic A␤ peptides in CHO cells et al. (2007)
CHO cells overexpressing over-expressing the APP “Swedish” mutation
“Swedish” mutant human APP • Reduces the pro-apoptotic proteins, Bad and Bax
• Inhibits the cleavage and activation of caspase-3
• Improves neuronal differentiation
EGCG N2a cells stably transfected with • Promotes non-amyloidogenic processing of APP Smith et al. (2010)
“Swedish” mutant human APP • Upregulates ␣-secretase
(SweAPP N2a cells)
EGCG PS2 transgenic mice model of AD • Enhances memory function Lee et al. (2009)
• Induces brain alpha-secretase activity
• Reduces brain beta- and gamma-secretase activities
Ellagic acid Biochemical assay • Inhibits beta-secretase (BACE1) activity Kwak et al. (2005)
Exifone Biochemical assay • Inhibits heparin-induced tau filament formation Taniguchi et al.
• Inhibits the formation of A␤ fibrils (2005)
GSE Tg2576 transgenic mice model of • Inhibits A␤ protein aggregation Guo et al. (2010)
AD • Attenuates AD-type cognitive deterioration along with
reducing HMW soluble oligomeric A␤ in the brain
GSE Transgenic HD PC-12 cells, • Inhibits polyQ aggregation Wang et al. (2010a)
transgenic HD model drosophila, • Improves life span
R6/2 rodent model of HD • Attenuates motor skill decay
GSE TMHT transgenic mice model of • Induces unfolding of tau and diminishes structural stability Ksiezak-Reding
tauopathy • Neutralizes phospho-epitopes and disrupts fibrillary et al. (2010), Wang
conformation leading to disintegration of paired helical et al. (2010b)
filaments (PHFs)c
• Attenuates AD type tau neuropathology development in
brain
• Attenuates extracellular signal-receptor kinase 1/2
signaling in the brain.
GSE APP/PS1 transgenic mice model of • Reduces A␤ levels in the brain and serum of the mice Wang et al. (2009b)
AD • Reduces amyloid plaques and microgliosis in the brain of
transgenic mice
• Reduces brain A␤ burden and microglia activation
Green tea polyphenol Mice model of AD induced by • Ameliorates deleterious effects of d-galactose and A␤ Lu et al. (2006)
d-galactose and A␤ (25–35) (25–35)
(behavioral study) • Improves animal’s learning and memory
• Reduces prolonged latency time and the error numbers
• Increases the autonomic activities
Myricetin Biochemical assay • Inhibits fA␤ formation from A␤ Ono et al. (2003)
Destabilizes preformed fA␤
Myricetin A␤-induced neurotoxicity in rat • Reduces apoptotic changes and caspase-3 activation Shimmyo et al.
primary neuronal cells • Decreases A beta 1–40 and A beta 1–42 levels in culture (2008b)
media
• Activates and up-regulates alpha-secretase (ADAM10)
protein levels
• Directly binds and inhibits beta-secretase (BACE-1)
Myricetin In silico study • Directly binds and inhibits beta-secretase (BACE-1) Chakraborty et al.
(2011)
Oligonol A␤-induced oxidative cell death on • Attenuates A␤-induced cytotoxicity, apoptotic features, Li et al. (2004)
rat pheochromocytoma cells intracellular ROS accumulation, and lipid peroxidation
(PC12) • Increases cellular glutathione pool
• Suppresses A␤-induced activation of NF-␬B
Piceid Biochemical assay • Destabilizes A␤ fibrils and oligomers back to monomers Riviere et al.
• Inhibits A␤ polymerization (2009), Riviere
et al. (2007)
Phenolsulfon-phthalein Biochemical assay • Inhibits amyloid fibril formation by islet amyloid Levy et al. (2008)
polypeptide (IAPP)
Resveratrol HEK293 and N2a cells stably • Increases cytosolic calcium levels and promotes AMPK Vingtdeux et al.
transfected with human APP695, activation by the calcium/calmodulin-dependent protein (2010)
APP/PS1 transgenic mice model of kinase kinase-beta
AD • Loweres extracellular A␤ accumulation
• Inhibits AMPK target, mTOR, to trigger autophagy and
lysosomal degradation of A␤
• Activates AMPK and reduces cerebral A␤ levels and
deposition in cortex
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 339

Table 5 (Continued )

Substance Studied cell line/animal model Effect References

Resveratrol Tg19959 transgenic mice model of • Diminishes plaque formation in a region specific manner Karuppagounder
AD • Reduces brain glutathione et al. (2009)
• Increases brain cysteine
Resveratrol Biochemical assay • Inhibits A␤ polymerization Riviere et al. (2007)
Resveratrol HEK293 cells stably transfected • Shows no inhibition of A␤ production Marambaud et al.
with human APP695 (a cell model Shows no effect on ␤- and -secretases (2005)
of AD) • Promotes intracellular degradation of A␤ via proteasome
system
• Reduces secreted and intracellular A␤ levels in different
cell lines
Resveratrol A␤-induced neurotoxicity in • Restores GSH content Savaskan et al.
SH-SY5Y neuroblastoma cells, • Employs neuroprotective effects through antioxidant (2003)
biochemical assay activity
• Inhibits A␤ (42) fibril formation
• Employs no effect on A␤ (42) oligomerization
Resveratrol Biochemical assay • Inhibits BACE1 (beta-secretase) activity Choi et al. (2011),
Jeon et al. (2007)
Salvianolic acid B A␤ (25–35) mediated injury of • Antagonizes A␤ (25–35)-induced cytotoxicity Zhou et al. (2009)
PC-12 cells
Tannic acid Biochemical assay • Inhibits fA␤ formation and extension from A␤ (1–40) and Ono et al. (2004)
A␤ (1–42)
• Destabilizes preformed fA␤ (1–40) and fA␤ (1–42)
Wine-related polyphenols Biochemical assay • Inhibits fA␤formation and extension from A␤ (1–40) and Ono et al. (2004)
A␤ (1–42)
• Destabilizes preformed fA␤ (1–40) and fA␤ (1–42)
a
Prion protein (PrP), in misfolded form, produces transmissible spongiform encephalopathies including bovine spongiform encephalopathy (BSE, also known as “mad cow
disease”) in cattle and Creutzfeldt–Jakob disease (CJD) in humans.
b
Poly Q or polyglutamine is a polypeptide induced by trinucleotide repeat expansion important in trinucleotide repeat disorders like HD.
c
PHFs or paired helical filaments are aggregations of hyperphosphorylated tau protein.

so that their metabolites are less effective scavengers and antioxi- plasma concentration is needed for EGCG to reach a reasonable
dants than the original substances (Miyake et al., 2000; Shirai et al., therapeutic level in brain, and in this case, such a high concentration
2001; Terao et al., 2001; Yamamoto et al., 1999). Glucuronides and will raise questions regarding the safety and adverse effects of the
O-methylated forms and intracellular metabolites of flavonoids for drug (Suganuma et al., 1998). Catechins are able to cross BBB after
example, have shown a reduced ability to donate hydrogen and less oral administration, but they have been found in glucuronide and
effective scavenging capacity (Spencer et al., 2001a). Moreover, the 30-O-methyl glucuronide forms in the brain tissue (Abd El Mohsen
capacity of polyphenol molecules for metal ion scavenging is lim- et al., 2002; Mandel et al., 2006). Production of conjugates has
ited and usually high concentrations of a polyphenol are needed to been a good experimental strategy to promote polyphenol absorp-
produce an effective metal ion scavenging effect (Halliwell et al., tion and activity. For example, the glutamoyl diester of curcumin
2000; Ma et al., 2010). Considering the very low bioavailability of is a better neuroprotective agent than curcumin alone (Mythri
these natural compounds, notably in the brain, it is unlikely that et al., 2011). For increasing the bioavailability of EGCG in vivo,
metal ion scavenging property of polyphenols produces a signifi- using fully acetylated EGCG has been suggested, which functions
cant therapeutic effect. as a pro-drug. The acetyl group can be hydrolyzed intracellular by
It should be also noted that polyphenols are extensively metabo- esterases to release EGCG inside the cell. This strategy minimizes
lized by gut microflora (Gardana et al., 2009; Possemiers et al., 2011; auto-oxidation and allows better cellular uptake (Chao et al., 2010;
Rastmanesh, 2011). Acid hydrolysis of polyphenol conjugates by Huo et al., 2008; Lambert et al., 2006). Free radical grafting of gallic
microflora in the gut is actually required for producing bioavailable acid on multi-walled carbon nanotubes has been shown to improve
bioactive polyphenolic compounds from primary phenolic struc- biological properties of gallic acid (Cirillo et al., 2011). However, the
tures. While oxidative and conjugative metabolism of polyphenols application of such conjugates is still not common and the outcome
in liver generates hydrophilic high molecular weight biotransfor- of using them has not been sufficiently addressed.
mation products, their mainly anaerobic, reductive and hydrolytic More importantly, hazards, risks, and safety of consuming
metabolism in the gut microbial community generates non-polar polyphenols as life-long therapeutics should always be considered.
low molecular weight products (Sousa et al., 2008). There are few reports about adverse effects of polyphenols (Cordier
Another important question to answer is whether the inves- and Steenkamp, 2011; Mennen et al., 2005). Feeding rats with high
tigated poylphenols reach the brain in sufficient concentrations doses of quercetin (2% or 4%) caused chronic nephropathy in one
and in biologically active forms. Generally there is little infor- study (Dunnick and Hailey, 1992). Adding quercetin (0.1%) to the
mation regarding the interaction of polyphenols with the BBB. diet of mice in a study significantly reduced mice life expectancy
There are two main obstacles for polyphenols to cross the BBB: (Jones and Hughes, 1982). High-dose green tea polyphenols in
(1) endothelium of the brain microvessles (BBB) and (2) multidrug the diet (1%) disrupted kidney function through the reduction of
resistance-associated proteins (MRPs) (Singh et al., 2008). Cur- antioxidant enzymes and heat-shock protein expressions in mice
cumin is highly lipophilic and can cross the BBB (Wang et al., 2005). (Inoue et al., 2011). Even carcinogenic effects have been reported
However, the drug is metabolized very rapidly before reaching the for some polyphenols. For example, caffeic acid induced gastroin-
BBB by conjugation, and as a result, the bioavailability for brain is testinal (GI) and kidney tumors in rats and mice when presented in
very low (Kelloff et al., 1996). Increasing levels of EGCG in brain 2% and more in their diet (Hagiwara et al., 1991). Similarly, green
tissue after direct gastric lavage of EGCG has also been shown tea catechins (1% of the diet) enhanced tumor development in the
(Suganuma et al., 1998). However even after continuous admin- colon of male rats (Hirose et al., 2001). Some polyphenols, such as
istration of EGCG until 24 h, levels of the compound in brain tissue naringenin, have interaction with liver cytochrome P450 system,
reaches 5–10% of its blood levels. This suggests that a very high which is a main enzymatic system involved in drug metabolism and
340 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

bioactivation (Lilja et al., 2000; Veronese et al., 2003). Such interac- Bors, W., Michel, C., Stettmaier, K., 2001. Structure–activity relationships governing
tions might be problematic when the polyphenol is used with drugs antioxidant capacities of plant polyphenols. Methods Enzymol. 335, 166–180.
Brown, M.K., Evans, J.L., Luo, Y., 2006. Beneficial effects of natural antioxidants EGCG
of narrow therapeutic range like phenytoin. Other reported hazards and alpha-lipoic acid on life span and age-dependent behavioral declines in
related to polyphenol consumption include thyroid toxicity (Chang Caenorhabditis elegans. Pharmacol. Biochem. Behav. 85, 620–628.
and Doerge, 2000; Doerge and Chang, 2002; Doerge and Sheehan, Budd, S.L., Tenneti, L., Lishnak, T., Lipton, S.A., 2000. Mitochondrial and extramito-
chondrial apoptotic signaling pathways in cerebrocortical neurons. Proc. Natl.
2002; Ferreira et al., 2002), malnutrition (Arts and Hollman, 2005; Acad. Sci. U.S.A. 97, 6161–6166.
Temme and Van Hoydonck, 2002; Zijp et al., 2000), estrogenic activ- Campos-Esparza, M.R., Sanchez-Gomez, M.V., Matute, C., 2009. Molecular mecha-
ity and infertilization (Delclos et al., 2001; Hughes, 1988; Mendez nisms of neuroprotection by two natural antioxidant polyphenols. Cell Calcium
45, 358–368.
et al., 2002; Sharpe and Franks, 2002; Wisniewski et al., 2003). Stud-
Camps, P., Munoz-Torrero, D., 2001. Tacrine-huperzine A hybrids (huprines): a new
ies on safety assessment of polyphenols are needed before human class of highly potent and selective acetylcholinesterase inhibitors of interest
intervention trials are designed. Guidelines for such assessments for the treatment of Alzheimer’s disease. Mini Rev. Med. Chem. 1, 163–174.
Cao, X., Sudhof, T.C., 2004. Dissection of amyloid-beta precursor protein-dependent
in natural products have been previously presented and can be
transcriptional transactivation. J. Biol. Chem. 279, 24601–24611.
adapted to polyphenol research (Schilter et al., 2003). Chakraborty, S., Kumar, S., Basu, S., 2011. Conformational transition in the sub-
The way to bring natural polyphenols into routine clinical appli- strate binding domain of beta-secretase exploited by NMA and its implication
cation against neurodegeneration seems to be long. Comprehensive in inhibitor recognition: BACE1-myricetin a case study. Neurochem. Int. 58,
914–923.
investigation models have been previously proposed to cover all Chang-Mu, C., Jen-Kun, L., Shing-Hwa, L., Shoei-Yn, L.S., 2010. Characterization of
aspects of polyphenol studies (Kay, 2010). Such models require a neurotoxic effects of NMDA and the novel neuroprotection by phytopolyphenols
multidisciplinary approach, including epidemiological, clinical and in mice. Behav. Neurosci. 124, 541–553.
Chang, H.C., Doerge, D.R., 2000. Dietary genistein inactivates rat thyroid peroxidase
cellular-molecular studies. Structurally modified bioactive com- in vivo without an apparent hypothyroid effect. Toxicol. Appl. Pharmacol. 168,
pound might be used and systemic metabolism, pharmacokinetics, 244–252.
brain bioavailability, local metabolism and modification in CNS and Chao, J., Lau, W.K., Huie, M.J., Ho, Y.S., Yu, M.S., Lai, C.S., Wang, M., Yuen,
W.H., Lam, W.H., Chan, T.H., Chang, R.C., 2010. A pro-drug of the green tea
bioactive by-products need to be considered. polyphenol (−)-epigallocatechin-3-gallate (EGCG) prevents differentiated SH-
SY5Y cells from toxicity induced by 6-hydroxydopamine. Neurosci. Lett. 469,
360–364.
Checkoway, H., Powers, K., Smith-Weller, T., Franklin, G.M., Longstreth Jr., W.T.,
References Swanson, P.D., 2002. Parkinson’s disease risks associated with cigarette
smoking, alcohol consumption, and caffeine intake. Am. J. Epidemiol. 155,
Abd El Mohsen, M.M., Kuhnle, G., Rechner, A.R., Schroeter, H., Rose, S., Jenner, P., 732–738.
Rice-Evans, C.A., 2002. Uptake and metabolism of epicatechin and its access to Chen, C.M., Lin, J.K., Liu, S.H., Lin-Shiau, S.Y., 2008. Novel regimen through com-
the brain after oral ingestion. Free Radic. Biol. Med. 33, 1693–1702. bination of memantine and tea polyphenol for neuroprotection against brain
Alkon, D.L., Sun, M.K., Nelson, T.J., 2007. PKC signaling deficits: a mechanistic hypoth- excitotoxicity. J. Neurosci. Res. 86, 2696–2704.
esis for the origins of Alzheimer’s disease. Trends Pharmacol. Sci. 28, 51–60. Chen, G.J., Xu, J., Lahousse, S.A., Caggiano, N.L., de la Monte, S.M., 2003. Transient
Aluise, C.D., Sultana, R., Tangpong, J., Vore, M., St Clair, D., Moscow, J.A., Butterfield, hypoxia causes Alzheimer-type molecular and biochemical abnormalities in
D.A., 2010. Chemo brain (chemo fog) as a potential side effect of doxorubicin cortical neurons: potential strategies for neuroprotection. J. Alzheimers Dis. 5,
administration: role of cytokine-induced, oxidative/nitrosative stress in cogni- 209–228.
tive dysfunction. Adv. Exp. Med. Biol. 678, 147–156. Chen, J., Zhou, Y., Mueller-Steiner, S., Chen, L.F., Kwon, H., Yi, S., Mucke, L., Gan,
Amazzal, L., Lapotre, A., Quignon, F., Bagrel, D., 2007. Mangiferin protects against 1- L., 2005. SIRT1 protects against microglia-dependent amyloid-beta toxicity
methyl-4-phenylpyridinium toxicity mediated by oxidative stress in N2A cells. through inhibiting NF-kappaB signaling. J. Biol. Chem. 280, 40364–40374.
Neurosci. Lett. 418, 159–164. Choi, C.W., Choi, Y.H., Cha, M.R., Kim, Y.S., Yon, G.H., Hong, K.S., Park, W.K., Kim, Y.H.,
Anekonda, T.S., 2006. Resveratrol – a boon for treating Alzheimer’s disease? Brain Ryu, S.Y., 2011. In vitro BACE-1 inhibitory activity of resveratrol oligomers from
Res. Rev. 52, 316–326. the seed extract of Paeonia lactiflora. Planta Med. 77, 374–376.
Anekonda, T.S., Reddy, P.H., 2006. Neuronal protection by sirtuins in Alzheimer’s Choi, D.W., Monyer, H., Giffard, R.G., Goldberg, M.P., Christine, C.W., 1990. Acute
disease. J. Neurochem. 96, 305–313. brain injury, NMDA receptors, and hydrogen ions: observations in cortical cell
Arts, I.C., Hollman, P.C., 2005. Polyphenols and disease risk in epidemiologic studies. cultures. Adv. Exp. Med. Biol. 268, 501–504.
Am. J. Clin. Nutr. 81, 317S–325S. Choi, Y.T., Jung, C.H., Lee, S.R., Bae, J.H., Baek, W.K., Suh, M.H., Park, J., Park, C.W.,
Ataie, A., Sabetkasaei, M., Haghparast, A., Moghaddam, A.H., Kazeminejad, B., Suh, S.I., 2001. The green tea polyphenol (−)-epigallocatechin gallate attenuates
2010. Neuroprotective effects of the polyphenolic antioxidant agent, curcumin, beta-amyloid-induced neurotoxicity in cultured hippocampal neurons. Life Sci.
against homocysteine-induced cognitive impairment and oxidative stress in the 70, 603–614.
rat. Pharmacol. Biochem. Behav. 96, 378–385. Cirillo, G., Hampel, S., Klingeler, R., Puoci, F., Iemma, F., Curcio, M., Parisi, O.I., Spizzirri,
Avramovich-Tirosh, Y., Reznichenko, L., Mit, T., Zheng, H., Fridkin, M., Weinreb, U.G., Picci, N., Leonhardt, A., Ritschel, M., Buchner, B., 2011. Antioxidant multi-
O., Mandel, S., Youdim, M.B., 2007. Neurorescue activity, APP regulation and walled carbon nanotubes by free radical grafting of gallic acid: new materials
amyloid-beta peptide reduction by novel multi-functional brain permeable for biomedical applications. J. Pharm. Pharmacol. 63, 179–188.
iron- chelating-antioxidants, M-30 and green tea polyphenol. EGCG. Curr. Coma, M., Sereno, L., Da Rocha-Souto, B., Scotton, T.C., Espana, J., Sanchez, M.B.,
Alzheimer Res. 4, 403–411. Rodriguez, M., Agullo, J., Guardia-Laguarta, C., Garcia-Alloza, M., Borrelli, L.A.,
Barzilai, A., Rahamimoff, H., 1983. Inhibition of Ca2+ -transport ATPase from synap- Clarimon, J., Lleo, A., Bacskai, B.J., Saura, C.A., Hyman, B.T., Gomez-Isla, T., 2010.
tosomal vesicles by flavonoids. Biochim. Biophys. Acta 730, 245–254. Triflusal reduces dense-core plaque load, associated axonal alterations and
Benaki, D., Stathopoulou, K., Leondiadis, L., Ferderigos, N., Pelecanou, M., Mikros, inflammatory changes, and rescues cognition in a transgenic mouse model of
E., 2009. Detection of interactions of the beta-amyloid peptide with small Alzheimer’s disease. Neurobiol. Dis. 38, 482–491.
molecules employing transferred NOEs. J. Pept. Sci. 15, 435–441. Commenges, D., Scotet, V., Renaud, S., Jacqmin-Gadda, H., Barberger-Gateau, P., Dar-
Bieschke, J., Russ, J., Friedrich, R.P., Ehrnhoefer, D.E., Wobst, H., Neugebauer, K., tigues, J.F., 2000. Intake of flavonoids and risk of dementia. Eur. J. Epidemiol. 16,
Wanker, E.E., 2010. EGCG remodels mature alpha-synuclein and amyloid-beta 357–363.
fibrils and reduces cellular toxicity. Proc. Natl. Acad. Sci. U.S.A. 107, 7710–7715. Conseil, G., Baubichon-Cortay, H., Dayan, G., Jault, J.M., Barron, D., Di Pietro, A., 1998.
Bjelakovic, G., Gluud, L.L., Nikolova, D., Bjelakovic, M., Nagorni, A., Gluud, C., 2011. Flavonoids: a class of modulators with bifunctional interactions at vicinal ATP-
Antioxidant supplements for liver diseases. Cochrane Database Syst. Rev. 3, and steroid-binding sites on mouse P-glycoprotein. Proc. Natl. Acad. Sci. U.S.A.
CD007749. 95, 9831–9836.
Boege, F., Straub, T., Kehr, A., Boesenberg, C., Christiansen, K., Andersen, A., Jakob, Cordier, W., Steenkamp, V., 2011. Herbal remedies affecting coagulation: a review.
F., Kohrle, J., 1996. Selected novel flavones inhibit the DNA binding or the DNA Pharm. Biol., doi:10.3109/13880209.2011.611145, in press.
religation step of eukaryotic topoisomerase I. J. Biol. Chem. 271, 2262–2270. Dade, M.M., Schinella, G.R., Fioravanti, D.E., Alfiotournier, H., 2011. Antioxidant and
Bohnen, N.I., Kaufer, D.I., Hendrickson, R., Ivanco, L.S., Lopresti, B., Davis, J.G., Con- cytotoxic properties of an aqueous extract from the Argentinean plant Hedeoma
stantine, G., Mathis, C.A., Moore, R.Y., DeKosky, S.T., 2005. Cognitive correlates multiflorum. Pharm. Biol. 49, 633–639.
of alterations in acetylcholinesterase in Alzheimer’s disease. Neurosci. Lett. 380, Dai, Q., Borenstein, A.R., Wu, Y., Jackson, J.C., Larson, E.B., 2006. Fruit and vegetable
127–132. juices and Alzheimer’s disease: the Kame Project. Am. J. Med. 119, 751–759.
Bonfoco, E., Krainc, D., Ankarcrona, M., Nicotera, P., Lipton, S.A., 1995. Apoptosis and Dasgupta, B., Milbrandt, J., 2007. Resveratrol stimulates AMP kinase activity in neu-
necrosis: two distinct events induced, respectively, by mild and intense insults rons. Proc. Natl. Acad. Sci. U.S.A. 104, 7217–7222.
with N-methyl-d-aspartate or nitric oxide/superoxide in cortical cell cultures. Datta, S.R., Brunet, A., Greenberg, M.E., 1999. Cellular survival: a play in three Akts.
Proc. Natl. Acad. Sci. U.S.A. 92, 7162–7166. Genes Dev. 13, 2905–2927.
Bonni, A., Brunet, A., West, A.E., Datta, S.R., Takasu, M.A., Greenberg, M.E., 1999. Davis, K.L., Mohs, R.C., Marin, D., Purohit, D.P., Perl, D.P., Lantz, M., Austin, G.,
Cell survival promoted by the Ras-MAPK signaling pathway by transcription- Haroutunian, V., 1999. Cholinergic markers in elderly patients with early signs
dependent and -independent mechanisms. Science 286, 1358–1362. of Alzheimer disease. JAMA 281, 1401–1406.
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 341

Dawson, V.L., Dawson, T.M., London, E.D., Bredt, D.S., Snyder, S.H., 1991. Nitric oxide Hafner-Bratkovic, I., Gaspersic, J., Smid, L.M., Bresjanac, M., Jerala, R., 2008. Curcumin
mediates glutamate neurotoxicity in primary cortical cultures. Proc. Natl. Acad. binds to the alpha-helical intermediate and to the amyloid form of prion protein
Sci. U.S.A. 88, 6368–6371. – a new mechanism for the inhibition of PrP(Sc) accumulation. J. Neurochem.
Dekermendjian, K., Kahnberg, P., Witt, M.R., Sterner, O., Nielsen, M., Liljefors, T., 1999. 104, 1553–1564.
Structure–activity relationships and molecular modeling analysis of flavonoids Hagiwara, A., Hirose, M., Takahashi, S., Ogawa, K., Shirai, T., Ito, N., 1991. Forestomach
binding to the benzodiazepine site of the rat brain GABA(A) receptor complex. and kidney carcinogenicity of caffeic acid in F344 rats and C57BL/6N × C3H/HeN
J. Med. Chem. 42, 4343–4350. F1 mice. Cancer Res. 51, 5655–5660.
Delclos, K.B., Bucci, T.J., Lomax, L.G., Latendresse, J.R., Warbritton, A., Weis, C.C., New- Halliwell, B., 1992. Reactive oxygen species and the central nervous system. J. Neu-
bold, R.R., 2001. Effects of dietary genistein exposure during development on rochem. 59, 1609–1623.
male and female CD (Sprague–Dawley) rats. Reprod. Toxicol. 15, 647–663. Halliwell, B., 2001. Role of free radicals in the neurodegenerative diseases: thera-
Della-Morte, D., Dave, K.R., DeFazio, R.A., Bao, Y.C., Raval, A.P., Perez-Pinzon, M.A., peutic implications for antioxidant treatment. Drugs Aging 18, 685–716.
2009. Resveratrol pretreatment protects rat brain from cerebral ischemic dam- Halliwell, B., Zhao, K., Whiteman, M., 2000. The gastrointestinal tract: a major site
age via a sirtuin 1-uncoupling protein 2 pathway. Neuroscience 159, 993–1002. of antioxidant action? Free Radic. Res. 33, 819–830.
Deprez, S., Mila, I., Huneau, J.F., Tome, D., Scalbert, A., 2001. Transport of proan- Han, Y.S., Zheng, W.H., Bastianetto, S., Chabot, J.G., Quirion, R., 2004. Neuropro-
thocyanidin dimer, trimer, and polymer across monolayers of human intestinal tective effects of resveratrol against beta-amyloid-induced neurotoxicity in rat
epithelial Caco-2 cells. Antioxid. Redox Signal. 3, 957–967. hippocampal neurons: involvement of protein kinase C. Br. J. Pharmacol. 141,
Desmond, D.W., Moroney, J.T., Sano, M., Stern, Y., 2002. Incidence of dementia after 997–1005.
ischemic stroke: results of a longitudinal study. Stroke 33, 2254–2260. Hanson, E.S., Foot, L.M., Leibold, E.A., 1999. Hypoxia post-translationally activates
Di Pietro, A., Godinot, C., Bouillant, M.L., Gautheron, D.C., 1975. Pig heart mitochon- iron-regulatory protein 2. J. Biol. Chem. 274, 5047–5052.
drial ATPase: properties of purified and membrane-bound enzyme. Effects of Hanson, E.S., Leibold, E.A., 1999. Regulation of the iron regulatory proteins by reac-
flavonoids. Biochimie 57, 959–967. tive nitrogen and oxygen species. Gene Expr. 7, 367–376.
Doerge, D.R., Chang, H.C., 2002. Inactivation of thyroid peroxidase by soy isoflavones, Hanyu, H., Asano, T., Sakurai, H., Tanaka, Y., Takasaki, M., Abe, K., 2002. MR analysis of
in vitro and in vivo. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 777, the substantia innominata in normal aging, Alzheimer disease, and other types
269–279. of dementia. AJNR Am. J. Neuroradiol. 23, 27–32.
Doerge, D.R., Sheehan, D.M., 2002. Goitrogenic and estrogenic activity of soy Harikumar, K.B., Aggarwal, B.B., 2008. Resveratrol: a multitargeted agent for age-
isoflavones. Environ. Health Perspect. 110 (Suppl 3), 349–353. associated chronic diseases. Cell Cycle 7, 1020–1035.
Domingo, D.S., Camouse, M.M., Hsia, A.H., Matsui, M., Maes, D., Ward, N.L., Cooper, Harish, G., Venkateshappa, C., Mythri, R.B., Dubey, S.K., Mishra, K., Singh, N., Vali,
K.D., Baron, E.D., 2010. Anti-angiogenic effects of epigallocatechin-3-gallate in S., Bharath, M.M., 2010. Bioconjugates of curcumin display improved protec-
human skin. Int. J. Clin. Exp. Pathol. 3, 705–709. tion against glutathione depletion mediated oxidative stress in a dopaminergic
Donovan, J.L., Manach, C., Rios, L., Morand, C., Scalbert, A., Remesy, C., 2002. Pro- neuronal cell line: implications for Parkinson’s disease. Bioorg. Med. Chem. 18,
cyanidins are not bioavailable in rats fed a single meal containing a grapeseed 2631–2638.
extract or the procyanidin dimer B3. Br. J. Nutr. 87, 299–306. Heneka, M.T., O‘Banion, M.K., 2007. Inflammatory processes in Alzheimer’s disease.
Duennwald, M.L., Shorter, J., 2010. Countering amyloid polymorphism and drug J. Neuroimmunol. 184, 69–91.
resistance with minimal drug cocktails. Prion 4, 244–251. Higuchi, A., Yonemitsu, K., Koreeda, A., Tsunenari, S., 2003. Inhibitory activity of
Dunnick, J.K., Hailey, J.R., 1992. Toxicity and carcinogenicity studies of quercetin, a epigallocatechin gallate (EGCG) in paraquat-induced microsomal lipid peroxi-
natural component of foods. Fundam. Appl. Toxicol. 19, 423–431. dation – a mechanism of protective effects of EGCg against paraquat toxicity.
Ehrnhoefer, D.E., Bieschke, J., Boeddrich, A., Herbst, M., Masino, L., Lurz, R., Toxicology 183, 143–149.
Engemann, S., Pastore, A., Wanker, E.E., 2008. EGCG redirects amyloidogenic Hirose, M., Hoshiya, T., Mizoguchi, Y., Nakamura, A., Akagi, K., Shirai, T., 2001.
polypeptides into unstructured, off-pathway oligomers. Nat. Struct. Mol. Biol. Green tea catechins enhance tumor development in the colon without effects
15, 558–566. in the lung or thyroid after pretreatment with 1,2-dimethylhydrazine or 2,2 -
Ehrnhoefer, D.E., Duennwald, M., Markovic, P., Wacker, J.L., Engemann, S., Roark, M., dihydroxy-di-n-propylnitrosamine in male F344 rats. Cancer Lett. 168, 23–29.
Legleiter, J., Marsh, J.L., Thompson, L.M., Lindquist, S., Muchowski, P.J., Wanker, Hofer, T., Wenger, H., Gassmann, M., 2002. Oxygen sensing, HIF-1alpha stabilization
E.E., 2006. Green tea (−)-epigallocatechin-gallate modulates early events in and potential therapeutic strategies. Pflugers Arch. 443, 503–507.
huntingtin misfolding and reduces toxicity in Huntington’s disease models. Hollman, P.C., de Vries, J.H., van Leeuwen, S.D., Mengelers, M.J., Katan, M.B., 1995.
Hum. Mol. Genet. 15, 2743–2751. Absorption of dietary quercetin glycosides and quercetin in healthy ileostomy
Esch, F.S., Keim, P.S., Beattie, E.C., Blacher, R.W., Culwell, A.R., Oltersdorf, T., McClure, volunteers. Am. J. Clin. Nutr. 62, 1276–1282.
D., Ward, P.J., 1990. Cleavage of amyloid beta peptide during constitutive pro- Holt, R.R., Lazarus, S.A., Sullards, M.C., Zhu, Q.Y., Schramm, D.D., Hammerstone, J.F.,
cessing of its precursor. Science 248, 1122–1124. Fraga, C.G., Schmitz, H.H., Keen, C.L., 2002. Procyanidin dimer B2 [epicatechin-
Etcheberrigaray, R., Tan, M., Dewachter, I., Kuiperi, C., Van der Auwera, I., Wera, S., (4beta-8)-epicatechin] in human plasma after the consumption of a flavanol-
Qiao, L., Bank, B., Nelson, T.J., Kozikowski, A.P., Van Leuven, F., Alkon, D.L., 2004. rich cocoa. Am. J. Clin. Nutr. 76, 798–804.
Therapeutic effects of PKC activators in Alzheimer’s disease transgenic mice. Huang, Y.H., Zhang, Q.H., 2010. Genistein reduced the neural apoptosis in the brain
Proc. Natl. Acad. Sci. U.S.A. 101, 11141–11146. of ovariectomised rats by modulating mitochondrial oxidative stress. Br. J. Nutr.
Fan, M.M., Raymond, L.A., 2007. N-methyl-d-aspartate (NMDA) receptor function 104, 1297–1303.
and excitotoxicity in Huntington’s disease. Prog. Neurobiol. 81, 272–293. Hudson, S.A., Ecroyd, H., Dehle, F.C., Musgrave, I.F., Carver, J.A., 2009. (−)-
Ferreira, A.C., Lisboa, P.C., Oliveira, K.J., Lima, L.P., Barros, I.A., Carvalho, D.P., 2002. Epigallocatechin-3-gallate (EGCG) maintains kappa-casein in its pre-fibrillar
Inhibition of thyroid type 1 deiodinase activity by flavonoids. Food Chem. Toxi- state without redirecting its aggregation pathway. J. Mol. Biol. 392, 689–700.
col. 40, 913–917. Hughes Jr., C.L., 1988. Phytochemical mimicry of reproductive hormones and mod-
Fisher, A., 2008. Cholinergic treatments with emphasis on m1 muscarinic agonists as ulation of herbivore fertility by phytoestrogens. Environ. Health Perspect. 78,
potential disease-modifying agents for Alzheimer’s disease. Neurotherapeutics 171–174.
5, 433–442. Huo, C., Wan, S.B., Lam, W.H., Li, L., Wang, Z., Landis-Piwowar, K.R., Chen, D., Dou,
Fukui, M., Choi, H.J., Zhu, B.T., 2010. Mechanism for the protective effect of resvera- Q.P., Chan, T.H., 2008. The challenge of developing green tea polyphenols as
trol against oxidative stress-induced neuronal death. Free Radic. Biol. Med. 49, therapeutic agents. Inflammopharmacology 16, 248–252.
800–813. Inoue, H., Akiyama, S., Maeda-Yamamoto, M., Nesumi, A., Tanaka, T., Murakami, A.,
Gao, Z.B., Chen, X.Q., Hu, G.Y., 2006. Inhibition of excitatory synaptic transmission 2011. High-dose green tea polyphenols induce nephrotoxicity in dextran sulfate
by trans-resveratrol in rat hippocampus. Brain Res. 1111, 41–47. sodium-induced colitis mice by down-regulation of antioxidant enzymes and
Gardana, C., Canzi, E., Simonetti, P., 2009. The role of diet in the metabolism of heat-shock protein expressions. Cell Stress Chaperones 16, 653–662.
daidzein by human faecal microbiota sampled from Italian volunteers. J. Nutr. Iqbal, K., Liu, F., Gong, C.X., Alonso Adel, C., Grundke-Iqbal, I., 2009. Mechanisms of
Biochem. 20, 940–947. tau-induced neurodegeneration. Acta Neuropathol. 118, 53–69.
Ghanim, H., Sia, C.L., Korzeniewski, K., Lohano, T., Abuaysheh, S., Marum- Iqbal, K., Wang, X., Blanchard, J., Liu, F., Gong, C.X., Grundke-Iqbal, I., 2010.
ganti, A., Chaudhuri, A., Dandona, P., 2011. A resveratrol and polyphe- Alzheimer’s disease neurofibrillary degeneration: pivotal and multifactorial.
nol preparation suppresses oxidative and inflammatory stress response Biochem. Soc. Trans. 38, 962–966.
to a high-fat, high-carbohydrate meal. J. Clin. Endocrinol. Metab. 96, Ivan, M., Kondo, K., Yang, H., Kim, W., Valiando, J., Ohh, M., Salic, A., Asara, J.M., Lane,
1409–1414. W.S., Kaelin Jr., W.G., 2001. HIFalpha targeted for VHL-mediated destruction by
Gotz, M.E., Freyberger, A., Riederer, P., 1990. Oxidative stress: a role in the patho- proline hydroxylation: implications for O2 sensing. Science 292, 464–468.
genesis of Parkinson’s disease. J. Neural Transm. Suppl. 29, 241–249. Jaakkola, P., Mole, D.R., Tian, Y.M., Wilson, M.I., Gielbert, J., Gaskell, S.J., Kriegsheim,
Gotz, M.E., Kunig, G., Riederer, P., Youdim, M.B., 1994. Oxidative stress: free radical A., Hebestreit, H.F., Mukherji, M., Schofield, C.J., Maxwell, P.H., Pugh, C.W., Rat-
production in neural degeneration. Pharmacol. Ther. 63, 37–122. cliffe, P.J., 2001. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation
Guo, A.J., Xie, H.Q., Choi, R.C., Zheng, K.Y., Bi, C.W., Xu, S.L., Dong, T.T., Tsim, K.W., complex by O2 -regulated prolyl hydroxylation. Science 292, 468–472.
2010. Galangin, a flavonol derived from Rhizoma Alpiniae Officinarum, inhibits Jang, J.H., Surh, Y.J., 2003. Protective effect of resveratrol on beta-amyloid-induced
acetylcholinesterase activity in vitro. Chem. Biol. Interact. 187, 246–248. oxidative PC12 cell death. Free Radic. Biol. Med. 34, 1100–1110.
Guo, Q., Zhao, B., Li, M., Shen, S., Xin, W., 1996. Studies on protective mechanisms of Jendroska, K., Hoffmann, O.M., Patt, S., 1997. Amyloid beta peptide and precur-
four components of green tea polyphenols against lipid peroxidation in synap- sor protein (APP) in mild and severe brain ischemia. Ann. N.Y. Acad. Sci. 826,
tosomes. Biochim. Biophys. Acta 1304, 210–222. 401–405.
Guo, Q., Zhao, B., Shen, S., Hou, J., Hu, J., Xin, W., 1999. ESR study on the Jeon, S.Y., Kwon, S.H., Seong, Y.H., Bae, K., Hur, J.M., Lee, Y.Y., Suh, D.Y., Song, K.S.,
structure–antioxidant activity relationship of tea catechins and their epimers. 2007. Beta-secretase (BACE1)-inhibiting stilbenoids from Smilax Rhizoma. Phy-
Biochim. Biophys. Acta 1427, 13–23. tomedicine 14, 403–408.
342 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

Jones, E., Hughes, R.E., 1982. Quercetin, flavonoids and the life-span of mice. Exp. Lambert, J.D., Sang, S., Hong, J., Kwon, S.J., Lee, M.J., Ho, C.T., Yang, C.S., 2006. Per-
Gerontol. 17, 213–217. acetylation as a means of enhancing in vitro bioactivity and bioavailability of
Joseph, J.A., Shukitt-Hale, B., Brewer, G.J., Weikel, K.A., Kalt, W., Fisher, D.R., 2010. Dif- epigallocatechin-3-gallate. Drug Metab. Dispos. 34, 2111–2116.
ferential protection among fractionated blueberry polyphenolic families against Larner, A.J., 2010. Cholinesterase inhibitors: beyond Alzheimer’s disease. Expert Rev.
DA-, Abeta(42)- and LPS-induced decrements in Ca(2+) buffering in primary Neurother. 10, 1699–1705.
hippocampal cells. J. Agric. Food Chem. 58, 8196–8204. Lee, H., Bae, J.H., Lee, S.R., 2004. Protective effect of green tea polyphenol EGCG
Joseph, T.B., Wang, S.W., Liu, X., Kulkarni, K.H., Wang, J., Xu, H., Hu, M., 2007. Disposi- against neuronal damage and brain edema after unilateral cerebral ischemia in
tion of flavonoids via enteric recycling: enzyme stability affects characterization gerbils. J. Neurosci. Res. 77, 892–900.
of prunetin glucuronidation across species, organs, and UGT isoforms. Mol. Lee, H.H., Lin, C.T., Yang, L.L., 2007. Neuroprotection and free radical scavenging
Pharm. 4, 883–894. effects of Osmanthus fragrans. J. Biomed. Sci. 14, 819–827.
Kalfon, L., Youdim, M.B., Mandel, S.A., 2007. Green tea polyphenol (−)- Lee, J.W., Lee, Y.K., Ban, J.O., Ha, T.Y., Yun, Y.P., Han, S.B., Oh, K.W., Hong, J.T., 2009.
epigallocatechin-3-gallate promotes the rapid protein kinase C- and Green tea (−)-epigallocatechin-3-gallate inhibits beta-amyloid-induced cogni-
proteasome-mediated degradation of Bad: implications for neuroprotection. J. tive dysfunction through modification of secretase activity via inhibition of ERK
Neurochem. 100, 992–1002. and NF-kappaB pathways in mice. J. Nutr. 139, 1987–1993.
Kang, K.S., Wen, Y., Yamabe, N., Fukui, M., Bishop, S.C., Zhu, B.T., 2010. Dual ben- Lemus-Molina, Y., Sanchez-Gomez, M.V., Delgado-Hernandez, R., Matute, C., 2009.
eficial effects of (−)-epigallocatechin-3-gallate on levodopa methylation and Mangifera indica L. extract attenuates glutamate-induced neurotoxicity on rat
hippocampal neurodegeneration: in vitro and in vivo studies. PLoS One 5, cortical neurons. Neurotoxicology 30, 1053–1058.
e11951. Levey, A.I., Kitt, C.A., Simonds, W.F., Price, D.L., Brann, M.R., 1991. Identification and
Kantengwa, S., Polla, B.S., 1991. Flavonoids, but not protein kinase C inhibitors, pre- localization of muscarinic acetylcholine receptor proteins in brain with subtype-
vent stress protein synthesis during erythrophagocytosis. Biochem. Biophys. specific antibodies. J. Neurosci. 11, 3218–3226.
Res. Commun. 180, 308–314. Levites, Y., Amit, T., Mandel, S., Youdim, M.B., 2003. Neuroprotection and neurores-
Karuppagounder, S.S., Pinto, J.T., Xu, H., Chen, H.L., Beal, M.F., Gibson, G.E., 2009. cue against Abeta toxicity and PKC-dependent release of nonamyloidogenic
Dietary supplementation with resveratrol reduces plaque pathology in a trans- soluble precursor protein by green tea polyphenol (−)-epigallocatechin-3-
genic model of Alzheimer’s disease. Neurochem. Int. 54, 111–118. gallate. FASEB J. 17, 952–954.
Kawahara, M., Kuroda, Y., 2000. Molecular mechanism of neurodegeneration Levites, Y., Amit, T., Youdim, M.B., Mandel, S., 2002a. Involvement of protein
induced by Alzheimer’s beta-amyloid protein: channel formation and disruption kinase C activation and cell survival/cell cycle genes in green tea polyphe-
of calcium homeostasis. Brain Res. Bull. 53, 389–397. nol (−)-epigallocatechin 3-gallate neuroprotective action. J. Biol. Chem. 277,
Kawahara, M., Kuroda, Y., Arispe, N., Rojas, E., 2000. Alzheimer’s beta-amyloid, 30574–30580.
human islet amylin, and prion protein fragment evoke intracellular free cal- Levites, Y., Weinreb, O., Maor, G., Youdim, M.B., Mandel, S., 2001. Green
cium elevations by a common mechanism in a hypothalamic GnRH neuronal tea polyphenol (−)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-
cell line. J. Biol. Chem. 275, 14077–14083. 1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J. Neu-
Kay, C.D., 2010. The future of flavonoid research. Br. J. Nutr. 104 (Suppl 3), S91–S95. rochem. 78, 1073–1082.
Kelloff, G.J., Crowell, J.A., Hawk, E.T., Steele, V.E., Lubet, R.A., Boone, C.W., Covey, J.M., Levites, Y., Youdim, M.B., Maor, G., Mandel, S., 2002b. Attenuation of 6-
Doody, L.A., Omenn, G.S., Greenwald, P., Hong, W.K., Parkinson, D.R., Bagheri, hydroxydopamine (6-OHDA)-induced nuclear factor-kappaB (NF-kappaB)
D., Baxter, G.T., Blunden, M., Doeltz, M.K., Eisenhauer, K.M., Johnson, K., Knapp, activation and cell death by tea extracts in neuronal cultures. Biochem. Phar-
G.G., Longfellow, D.G., Malone, W.F., Nayfield, S.G., Seifried, H.E., Swall, L.M., Sig- macol. 63, 21–29.
man, C.C., 1996. Strategy and planning for chemopreventive drug development: Levy, M., Porat, Y., Bacharach, E., Shalev, D.E., Gazit, E., 2008. Phenolsul-
clinical development plans II. J. Cell. Biochem. Suppl. 26, 54–71. fonphthalein, but not phenolphthalein, inhibits amyloid fibril formation:
Kelsey, N.A., Wilkins, H.M., Linseman, D.A., 2010. Nutraceutical antioxidants as novel implications for the modulation of amyloid self-assembly. Biochemistry 47,
neuroprotective agents. Molecules 15, 7792–7814. 5896–5904.
Kennedy, D.O., Wightman, E.L., Reay, J.L., Lietz, G., Okello, E.J., Wilde, A., Haskell, C.F., Li, J., Wu, H.M., Zhou, R.L., Liu, G.J., Dong, B.R., 2008. Huperzine A for Alzheimer’s
2010. Effects of resveratrol on cerebral blood flow variables and cognitive perfor- disease. Cochrane Database Syst. Rev., CD005592.
mance in humans: a double-blind, placebo-controlled, crossover investigation. Li, M.H., Jang, J.H., Sun, B., Surh, Y.J., 2004. Protective effects of oligomers of grape
Am. J. Clin. Nutr. 91, 1590–1597. seed polyphenols against beta-amyloid-induced oxidative cell death. Ann. N.Y.
Kim, E.Y., Ham, S.K., Shigenaga, M.K., Han, O., 2008. Bioactive dietary polypheno- Acad. Sci. 1030, 317–329.
lic compounds reduce nonheme iron transport across human intestinal cell Liang, Y.Q., Tang, X.C., 2004. Comparative effects of huperzine A, donepezil and
monolayers. J. Nutr. 138, 1647–1651. rivastigmine on cortical acetylcholine level and acetylcholinesterase activity in
Kim, H.K., Kim, M., Kim, S., Chung, J.H., 2004. Effects of green tea polyphenol on rats. Neurosci. Lett. 361, 56–59.
cognitive and acetylcholinesterase activities. Biosci. Biotechnol. Biochem. 68, Lilja, J.J., Kivisto, K.T., Backman, J.T., Neuvonen, P.J., 2000. Effect of grapefruit juice
1977–1979. dose on grapefruit juice-triazolam interaction: repeated consumption prolongs
Kim, J.Y., Lee, W.S., Kim, Y.S., Curtis-Long, M.J., Lee, B.W., Ryu, Y.B., Park, K.H., 2011. triazolam half-life. Eur. J. Clin. Pharmacol. 56, 411–415.
Isolation of cholinesterase-inhibiting flavonoids from Morus lhou. J. Agric. Food Lin, H.J., Lai, C.C., Lee Chao, P.D., Fan, S.S., Tsai, Y., Huang, S.Y., Wan, L., Tsai, F.J.,
Chem. 2007. Aloe-emodin metabolites protected N-methyl-d-aspartate-treated reti-
Kim, S.J., Jeong, H.J., Lee, K.M., Myung, N.Y., An, N.H., Yang, W.M., Park, S.K., Lee, nal ganglion cells by Cu–Zn superoxide dismutase. J. Ocul. Pharmacol. Ther. 23,
H.J., Hong, S.H., Kim, H.M., Um, J.Y., 2007. Epigallocatechin-3-gallate suppresses 152–171.
NF-kappaB activation and phosphorylation of p38 MAPK and JNK in human Lin, M.S., Hung, K.S., Chiu, W.T., Sun, Y.Y., Tsai, S.H., Lin, J.W., Lee, Y.H., 2011. Curcumin
astrocytoma U373MG cells. J. Nutr. Biochem. 18, 587–596. enhances neuronal survival in N-methyl-d-aspartic acid toxicity by inducing
Kitisripanya, N., Saparpakorn, P., Wolschann, P., Hannongbua, S., 2011. Binding RANTES expression in astrocytes via PI-3K and MAPK signaling pathways. Prog.
of huperzine A and galanthamine to acetylcholinesterase, based on ONIOM Neuropsychopharmacol. Biol. Psychiatry 35, 931–938.
method. Nanomedicine 7, 60–68. Lin, M.T., Beal, M.F., 2006. Mitochondrial dysfunction and oxidative stress in neu-
Klein, J.C., Eggers, C., Kalbe, E., Weisenbach, S., Hohmann, C., Vollmar, S., Baudrexel, S., rodegenerative diseases. Nature 443, 787–795.
Diederich, N.J., Heiss, W.D., Hilker, R., 2010. Neurotransmitter changes in demen- Lin, Y.S., Tsai, Y.J., Tsay, J.S., Lin, J.K., 2003. Factors affecting the levels of tea polyphe-
tia with Lewy bodies and Parkinson disease dementia in vivo. Neurology 74, nols and caffeine in tea leaves. J. Agric. Food Chem. 51, 1864–1873.
885–892. Lipton, S.A., 2007. Pathologically-activated therapeutics for neuroprotection: mech-
Koh, S.H., Lee, S.M., Kim, H.Y., Lee, K.Y., Lee, Y.J., Kim, H.T., Kim, J., Kim, M.H., Hwang, anism of NMDA receptor block by memantine and S-nitrosylation. Curr. Drug
M.S., Song, C., Yang, K.W., Lee, K.W., Kim, S.H., Kim, O.H., 2006. The effect of Targets 8, 621–632.
epigallocatechin gallate on suppressing disease progression of ALS model mice. Lipton, S.A., Choi, Y.B., Pan, Z.H., Lei, S.Z., Chen, H.S., Sucher, N.J., Loscalzo, J., Singel,
Neurosci. Lett. 395, 103–107. D.J., Stamler, J.S., 1993. A redox-based mechanism for the neuroprotective and
Krikorian, R., Nash, T.A., Shidler, M.D., Shukitt-Hale, B., Joseph, J.A., 2010. Concord neurodestructive effects of nitric oxide and related nitroso-compounds. Nature
grape juice supplementation improves memory function in older adults with 364, 626–632.
mild cognitive impairment. Br. J. Nutr. 103, 730–734. Lipton, S.A., Rosenberg, P.A., 1994. Excitatory amino acids as a final common pathway
Ksiezak-Reding, H., Ho, L., Santa-Maria, I., Diaz-Ruiz, C., Wang, J., Pasinetti, for neurologic disorders. N. Engl. J. Med. 330, 613–622.
G.M., 2010. Ultrastructural alterations of Alzheimer’s disease paired Liu, Y.Q., Xin, T.R., Liang, J.J., Wang, W.M., Zhang, Y.Y., 2010. Memory performance,
helical filaments by grape seed-derived polyphenols. Neurobiol. Aging, brain excitatory amino acid and acetylcholinesterase activity of chronically alu-
doi:10.1016/j.neurobiolaging.2010.11.006, in press. minum exposed mice in response to soy isoflavones treatment. Phytother. Res.
Kumar, P., Padi, S.S., Naidu, P.S., Kumar, A., 2007. Possible neuroprotective mecha- 24, 1451–1456.
nisms of curcumin in attenuating 3-nitropropionic acid-induced neurotoxicity. Lu, J., Zhang, K., Chen, S., Wen, W., 2009. Grape seed extract inhibits VEGF
Methods Find. Exp. Clin. Pharmacol. 29, 19–25. expression via reducing HIF-1alpha protein expression. Carcinogenesis 30,
Kwak, H.M., Jeon, S.Y., Sohng, B.H., Kim, J.G., Lee, J.M., Lee, K.B., Jeong, H.H., Hur, J.M., 636–644.
Kang, Y.H., Song, K.S., 2005. beta-Secretase (BACE1) inhibitors from pomegranate Lu, J.H., Guo, J., Yang, W.H., 2006. Effects of green tea polyphenol on the behaviour of
(Punica granatum) husk. Arch. Pharm. Res. 28, 1328–1332. Alzheimer’ s disease like mice induced by d-galactose and Abeta 25–35. Zhong
Lafon-Cazal, M., Pietri, S., Culcasi, M., Bockaert, J., 1993. NMDA-dependent superox- Yao Cai 29, 352–354.
ide production and neurotoxicity. Nature 364, 535–537. Lu, K.T., Ko, M.C., Chen, B.Y., Huang, J.C., Hsieh, C.W., Lee, M.C., Chiou, R.Y., Wung,
Lahiri, D.K., Rogers, J.T., Greig, N.H., Sambamurti, K., 2004. Rationale for the devel- B.S., Peng, C.H., Yang, Y.L., 2008. Neuroprotective effects of resveratrol on MPTP-
opment of cholinesterase inhibitors as anti-Alzheimer agents. Curr. Pharm. Des. induced neuron loss mediated by free radical scavenging. J. Agric. Food Chem.
10, 3111–3119. 56, 6910–6913.
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 343

Ma, Q., Kim, E.Y., Han, O., 2010. Bioactive dietary polyphenols decrease heme iron Ogura, H., Kosasa, T., Kuriya, Y., Yamanishi, Y., 2000. Comparison of inhibitory activi-
absorption by decreasing basolateral iron release in human intestinal Caco-2 ties of donepezil and other cholinesterase inhibitors on acetylcholinesterase and
cells. J. Nutr. 140, 1117–1121. butyrylcholinesterase in vitro. Methods Find. Exp. Clin. Pharmacol. 22, 609–613.
Ma, X., Tan, C., Zhu, D., Gang, D.R., Xiao, P., 2007. Huperzine A from Huperzia species Okawara, M., Katsuki, H., Kurimoto, E., Shibata, H., Kume, T., Akaike, A., 2007. Resver-
– an ethnopharmacolgical review. J. Ethnopharmacol. 113, 15–34. atrol protects dopaminergic neurons in midbrain slice culture from multiple
Maher, P., Dargusch, R., Bodai, L., Gerard, P.E., Purcell, J.M., Marsh, J.L., 2011. ERK insults. Biochem. Pharmacol. 73, 550–560.
activation by the polyphenols fisetin and resveratrol provides neuroprotection Ono, K., Hasegawa, K., Naiki, H., Yamada, M., 2004. Anti-amyloidogenic activity of
in multiple models of Huntington’s disease. Hum. Mol. Genet. 20, 261–270. tannic acid and its activity to destabilize Alzheimer’s beta-amyloid fibrils in vitro.
Man, S.C., Durairajan, S.S., Kum, W.F., Lu, J.H., Huang, J.D., Cheng, C.F., Chung, V., Xu, Biochim. Biophys. Acta 1690, 193–202.
M., Li, M., 2008. Systematic review on the efficacy and safety of herbal medicines Ono, K., Yoshiike, Y., Takashima, A., Hasegawa, K., Naiki, H., Yamada, M., 2003.
for Alzheimer’s disease. J. Alzheimers Dis. 14, 209–223. Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in
Manach, C., Williamson, G., Morand, C., Scalbert, A., Remesy, C., 2005. Bioavailability vitro: implications for the prevention and therapeutics of Alzheimer’s disease.
and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. J. Neurochem. 87, 172–181.
Am. J. Clin. Nutr. 81, 230S–242S. Ortega-Arellano, H.F., Jimenez-Del-Rio, M., Velez-Pardo, C., 2011. Life span and
Manczak, M., Mao, P., Calkins, M.J., Cornea, A., Reddy, A.P., Murphy, M.P., Szeto, locomotor activity modification by glucose and polyphenols in Drosophila
H.H., Park, B., Reddy, P.H., 2010. Mitochondria-targeted antioxidants protect melanogaster chronically exposed to oxidative stress-stimuli: implications in
against amyloid-beta toxicity in Alzheimer’s disease neurons. J. Alzheimers Dis. Parkinson’s disease. Neurochem. Res. 36, 1073–1086.
20 (Suppl. 2), S609–S631. Ossowska, K., Smialowska, M., Kuter, K., Wieronska, J., Zieba, B., Wardas, J., Nowak, P.,
Mandel, S., Amit, T., Reznichenko, L., Weinreb, O., Youdim, M.B., 2006. Green tea cate- Dabrowska, J., Bortel, A., Biedka, I., Schulze, G., Rommelspacher, H., 2006. Degen-
chins as brain-permeable, natural iron chelators-antioxidants for the treatment eration of dopaminergic mesocortical neurons and activation of compensatory
of neurodegenerative disorders. Mol. Nutr. Food Res. 50, 229–234. processes induced by a long-term paraquat administration in rats: implications
Mandel, S., Maor, G., Youdim, M.B., 2004a. Iron and alpha-synuclein in the substantia for Parkinson’s disease. Neuroscience 141, 2155–2165.
nigra of MPTP-treated mice: effect of neuroprotective drugs R-apomorphine Pallas, M., Casadesus, G., Smith, M.A., Coto-Montes, A., Pelegri, C., Vilaplana, J.,
and green tea polyphenol (−)-epigallocatechin-3-gallate. J. Mol. Neurosci. 24, Camins, A., 2009. Resveratrol and neurodegenerative diseases: activation of
401–416. SIRT1 as the potential pathway towards neuroprotection. Curr. Neurovasc. Res.
Mandel, S., Weinreb, O., Amit, T., Youdim, M.B., 2004b. Cell signaling pathways in 6, 70–81.
the neuroprotective actions of the green tea polyphenol (−)-epigallocatechin- Pannala, A.S., Rice-Evans, C.A., Halliwell, B., Singh, S., 1997. Inhibition of
3-gallate: implications for neurodegenerative diseases. J. Neurochem. 88, peroxynitrite-mediated tyrosine nitration by catechin polyphenols. Biochem.
1555–1569. Biophys. Res. Commun. 232, 164–168.
Mandel, S.A., Avramovich-Tirosh, Y., Reznichenko, L., Zheng, H., Weinreb, O., Amit, Papandreou, M.A., Dimakopoulou, A., Linardaki, Z.I., Cordopatis, P., Klimis-Zacas, D.,
T., Youdim, M.B., 2005. Multifunctional activities of green tea catechins in neu- Margarity, M., Lamari, F.N., 2009. Effect of a polyphenol-rich wild blueberry
roprotection. Modulation of cell survival genes, iron-dependent oxidative stress extract on cognitive performance of mice, brain antioxidant markers and acetyl-
and PKC signaling pathway. Neurosignals 14, 46–60. cholinesterase activity. Behav. Brain Res. 198, 352–358.
Marambaud, P., Zhao, H., Davies, P., 2005. Resveratrol promotes clearance of Park, J.W., Jang, Y.H., Kim, J.M., Lee, H., Park, W.K., Lim, M.B., Chu, Y.K., Lo, E.H.,
Alzheimer’s disease amyloid-beta peptides. J. Biol. Chem. 280, 37377–37382. Lee, S.R., 2009. Green tea polyphenol (−)-epigallocatechin gallate reduces neu-
Martin, D., Salinas, M., Lopez-Valdaliso, R., Serrano, E., Recuero, M., Cuadrado, A., ronal cell damage and up-regulation of MMP-9 activity in hippocampal CA1
2001. Effect of the Alzheimer amyloid fragment Abeta(25–35) on Akt/PKB kinase and CA2 areas following transient global cerebral ischemia. J. Neurosci. Res. 87,
and survival of PC12 cells. J. Neurochem. 78, 1000–1008. 567–575.
Mattson, M.P., 1991. Evidence for the involvement of protein kinase C in neu- Peng, Y., Jiang, L., Lee, D.Y., Schachter, S.C., Ma, Z., Lemere, C.A., 2006. Effects of
rodegenerative changes in cultured human cortical neurons. Exp. Neurol. 112, huperzine A on amyloid precursor protein processing and beta-amyloid gen-
95–103. eration in human embryonic kidney 293 APP Swedish mutant cells. J. Neurosci.
Mattson, M.P., 2004. Pathways towards and away from Alzheimer’s disease. Nature Res. 84, 903–911.
430, 631–639. Peng, Y., Lee, D.Y., Jiang, L., Ma, Z., Schachter, S.C., Lemere, C.A., 2007. Huperzine
Medina, J.H., Viola, H., Wolfman, C., Marder, M., Wasowski, C., Calvo, D., Paladini, A.C., A regulates amyloid precursor protein processing via protein kinase C and
1997. Overview – flavonoids: a new family of benzodiazepine receptor ligands. mitogen-activated protein kinase pathways in neuroblastoma SK-N-SH cells
Neurochem. Res. 22, 419–425. over-expressing wild type human amyloid precursor protein 695. Neuroscience
Mendez, M.A., Anthony, M.S., Arab, L., 2002. Soy-based formulae and infant growth 150, 386–395.
and development: a review. J. Nutr. 132, 2127–2130. Perry, E.K., Tomlinson, B.E., Blessed, G., Bergmann, K., Gibson, P.H., Perry, R.H., 1978.
Mennen, L.I., Walker, R., Bennetau-Pelissero, C., Scalbert, A., 2005. Risks and safety Correlation of cholinergic abnormalities with senile plaques and mental test
of polyphenol consumption. Am. J. Clin. Nutr. 81, 326S–329S. scores in senile dementia. Br. Med. J. 2, 1457–1459.
Mielke, K., Herdegen, T., 2000. JNK and p38 stresskinases – degenerative effectors of Possemiers, S., Bolca, S., Verstraete, W., Heyerick, A., 2011. The intestinal micro-
signal-transduction-cascades in the nervous system. Prog. Neurobiol. 61, 45–60. biome: a separate organ inside the body with the metabolic potential to
Min, B.S., Cuong, T.D., Lee, J.S., Shin, B.S., Woo, M.H., Hung, T.M., 2010. Cholinesterase influence the bioactivity of botanicals. Fitoterapia 82, 53–66.
inhibitors from Cleistocalyx operculatus buds. Arch. Pharm. Res. 33, 1665–1670. Ramos, S., 2007. Effects of dietary flavonoids on apoptotic pathways related to cancer
Miyake, Y., Shimoi, K., Kumazawa, S., Yamamoto, K., Kinae, N., Osawa, T., 2000. Iden- chemoprevention. J. Nutr. Biochem. 18, 427–442.
tification and antioxidant activity of flavonoid metabolites in plasma and urine Rastmanesh, R., 2011. High polyphenol, low probiotic diet for weight loss because
of eriocitrin-treated rats. J. Agric. Food Chem. 48, 3217–3224. of intestinal microbiota interaction. Chem. Biol. Interact. 189, 1–8.
Morand, C., Manach, C., Crespy, V., Remesy, C., 2000. Quercetin 3-O-beta-glucoside Ratcliffe, P.J., O‘Rourke, J.F., Maxwell, P.H., Pugh, C.W., 1998. Oxygen sensing,
is better absorbed than other quercetin forms and is not present in rat plasma. hypoxia-inducible factor-1 and the regulation of mammalian gene expression.
Free Radic. Res. 33, 667–676. J. Exp. Biol. 201, 1153–1162.
Morel, I., Lescoat, G., Cogrel, P., Sergent, O., Pasdeloup, N., Brissot, P., Cillard, P., Ray, B., Bisht, S., Maitra, A., Lahiri, D.K., 2011. Neuroprotective and neurorescue
Cillard, J., 1993. Antioxidant and iron-chelating activities of the flavonoids cate- effects of a novel polymeric nanoparticle formulation of curcumin (NanoCurc)
chin, quercetin and diosmetin on iron-loaded rat hepatocyte cultures. Biochem. in the neuronal cell culture and animal model: implications for Alzheimer’s
Pharmacol. 45, 13–19. disease. J. Alzheimers Dis. 23, 61–77.
Mythri, R.B., Harish, G., Dubey, S.K., Misra, K., Bharath, M.M., 2011. Glutamoyl Reddy, P.H., 2006. Amyloid precursor protein-mediated free radicals and oxida-
diester of the dietary polyphenol curcumin offers improved protection against tive damage: implications for the development and progression of Alzheimer’s
peroxynitrite-mediated nitrosative stress and damage of brain mitochon- disease. J. Neurochem. 96, 1–13.
dria in vitro: implications for Parkinson’s disease. Mol. Cell. Biochem. 347, Revuelta, M.P., Cantabrana, B., Hidalgo, A., 1997. Depolarization-dependent effect
135–143. of flavonoids in rat uterine smooth muscle contraction elicited by CaCl2 . Gen.
Nanjo, F., Goto, K., Seto, R., Suzuki, M., Sakai, M., Hara, Y., 1996. Scavenging effects Pharmacol. 29, 847–857.
of tea catechins and their derivatives on 1,1-diphenyl-2-picrylhydrazyl radical. Rezg, R., Mornagui, B., El-Fazaa, S., Gharbi, N., 2008. Caffeic acid attenuates malathion
Free Radic. Biol. Med. 21, 895–902. induced metabolic disruption in rat liver, involvement of acetylcholinesterase
Nanjo, F., Mori, M., Goto, K., Hara, Y., 1999. Radical scavenging activity of activity. Toxicology 250, 27–31.
tea catechins and their related compounds. Biosci. Biotechnol. Biochem. 63, Reznichenko, L., Amit, T., Zheng, H., Avramovich-Tirosh, Y., Youdim, M.B., Weinreb,
1621–1623. O., Mandel, S., 2006. Reduction of iron-regulated amyloid precursor protein and
Ng, T.P., Feng, L., Niti, M., Kua, E.H., Yap, K.B., 2008. Tea consumption and cognitive beta-amyloid peptide by (−)-epigallocatechin-3-gallate in cell cultures: impli-
impairment and decline in older Chinese adults. Am. J. Clin. Nutr. 88, 224–231. cations for iron chelation in Alzheimer’s disease. J. Neurochem. 97, 527–536.
Nunan, J., Small, D.H., 2000. Regulation of APP cleavage by alpha-, beta- and gamma- Rice-Evans, C.A., Miller, N.J., Paganga, G., 1996. Structure-antioxidant activity rela-
secretases. FEBS Lett. 483, 6–10. tionships of flavonoids and phenolic acids. Free Radic. Biol. Med. 20, 933–956.
Obregon, D.F., Rezai-Zadeh, K., Bai, Y., Sun, N., Hou, H., Ehrhart, J., Zeng, J., Mori, T., Riederer, P., Sofic, E., Rausch, W.D., Schmidt, B., Reynolds, G.P., Jellinger, K., Youdim,
Arendash, G.W., Shytle, D., Town, T., Tan, J., 2006. ADAM10 activation is required M.B., 1989. Transition metals, ferritin, glutathione, and ascorbic acid in parkin-
for green tea (−)-epigallocatechin-3-gallate-induced alpha-secretase cleavage sonian brains. J. Neurochem. 52, 515–520.
of amyloid precursor protein. J. Biol. Chem. 281, 16419–16427. Ritz, M.F., Ratajczak, P., Curin, Y., Cam, E., Mendelowitsch, A., Pinet, F., Andriantsi-
Ogunshola, O.O., Antoniou, X., 2009. Contribution of hypoxia to Alzheimer’s dis- tohaina, R., 2008. Chronic treatment with red wine polyphenol compounds
ease: is HIF-1alpha a mediator of neurodegeneration? Cell. Mol. Life Sci. 66, mediates neuroprotection in a rat model of ischemic cerebral stroke. J. Nutr.
3555–3563. 138, 519–525.
344 A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345

Riviere, C., Delaunay, J.C., Immel, F., Cullin, C., Monti, J.P., 2009. The polyphenol piceid Sousa, T., Paterson, R., Moore, V., Carlsson, A., Abrahamsson, B., Basit, A.W., 2008.
destabilizes preformed amyloid fibrils and oligomers in vitro: hypothesis on The gastrointestinal microbiota as a site for the biotransformation of drugs. Int.
possible molecular mechanisms. Neurochem. Res. 34, 1120–1128. J. Pharm. 363, 1–25.
Riviere, C., Richard, T., Quentin, L., Krisa, S., Merillon, J.M., Monti, J.P., 2007. Inhibitory Spencer, J.P., Abd El Mohsen, M.M., Minihane, A.M., Mathers, J.C., 2008. Biomarkers
activity of stilbenes on Alzheimer’s beta-amyloid fibrils in vitro. Bioorg. Med. of the intake of dietary polyphenols: strengths, limitations and application in
Chem. 15, 1160–1167. nutrition research. Br. J. Nutr. 99, 12–22.
Riviere, C., Richard, T., Vitrac, X., Merillon, J.M., Valls, J., Monti, J.P., 2008. New Spencer, J.P., Rice-Evans, C., Williams, R.J., 2003. Modulation of pro-survival
polyphenols active on beta-amyloid aggregation. Bioorg. Med. Chem. Lett. 18, Akt/protein kinase B and ERK1/2 signaling cascades by quercetin and its in vivo
828–831. metabolites underlie their action on neuronal viability. J. Biol. Chem. 278,
Roberson, E.D., Mucke, L., 2006. 100 years and counting: prospects for defeating 34783–34793.
Alzheimer’s disease. Science 314, 781–784. Spencer, J.P., Schroeter, H., Crossthwaithe, A.J., Kuhnle, G., Williams, R.J., Rice-Evans,
Rodgers, E.E., Theibert, A.B., 2002. Functions of PI 3-kinase in development of the C., 2001a. Contrasting influences of glucuronidation and O-methylation of epi-
nervous system. Int. J. Dev. Neurosci. 20, 187–197. catechin on hydrogen peroxide-induced cell death in neurons and fibroblasts.
Romeo, L., Intrieri, M., D‘Agata, V., Mangano, N.G., Oriani, G., Ontario, M.L., Free Radic. Biol. Med. 31, 1139–1146.
Scapagnini, G., 2009. The major green tea polyphenol, (−)-epigallocatechin- Spencer, J.P., Schroeter, H., Rechner, A.R., Rice-Evans, C., 2001b. Bioavailability of
3-gallate, induces heme oxygenase in rat neurons and acts as an effective flavan-3-ols and procyanidins: gastrointestinal tract influences and their rele-
neuroprotective agent against oxidative stress. J. Am. Coll. Nutr. 28 (Suppl), vance to bioactive forms in vivo. Antioxid. Redox Signal. 3, 1023–1039.
492S–499S. Srividhya, R., Jyothilakshmi, V., Arulmathi, K., Senthilkumaran, V., Kalaiselvi, P., 2008.
Rosenblat, M., Belinky, P., Vaya, J., Levy, R., Hayek, T., Coleman, R., Merchav, S., Avi- Attenuation of senescence-induced oxidative exacerbations in aged rat brain by
ram, M., 1999. Macrophage enrichment with the isoflavan glabridin inhibits (−)-epigallocatechin-3-gallate. Int. J. Dev. Neurosci. 26, 217–223.
NADPH oxidase-induced cell-mediated oxidation of low density lipoprotein. A Suganuma, M., Okabe, S., Oniyama, M., Tada, Y., Ito, H., Fujiki, H., 1998. Wide distri-
possible role for protein kinase C. J. Biol. Chem. 274, 13790–13799. bution of [3H] (−)-epigallocatechin gallate, a cancer preventive tea polyphenol,
Rosengarten, B., Dannhardt, V., Burr, O., Pohler, M., Rosengarten, S., Oechsner, M., in mouse tissue. Carcinogenesis 19, 1771–1776.
Reuter, I., 2010. Neurovascular coupling in Parkinson’s disease patients: effects Sun, Q.Q., Xu, S.S., Pan, J.L., Guo, H.M., Cao, W.Q., 1999. Huperzine-A capsules enhance
of dementia and acetylcholinesterase inhibitor treatment. J. Alzheimers Dis. 22, memory and learning performance in 34 pairs of matched adolescent students.
415–421. Zhongguo Yao Li Xue Bao 20, 601–603.
Rosi, S., Ramirez-Amaya, V., Hauss-Wegrzyniak, B., Wenk, G.L., 2004. Chronic brain Taniguchi, S., Suzuki, N., Masuda, M., Hisanaga, S., Iwatsubo, T., Goedert, M.,
inflammation leads to a decline in hippocampal NMDA-R1 receptors. J. Neuroin- Hasegawa, M., 2005. Inhibition of heparin-induced tau filament formation by
flamm. 1, 12. phenothiazines, polyphenols, and porphyrins. J. Biol. Chem. 280, 7614–7623.
Sabarinathan, D., Mahalakshmi, P., Vanisree, A.J., 2011. Naringenin, a flavanone Tanzi, R.E., Bertram, L., 2005. Twenty years of the Alzheimer’s disease amyloid
inhibits the proliferation of cerebrally implanted C6 glioma cells in rats. Chem. hypothesis: a genetic perspective. Cell 120, 545–555.
Biol. Interact. 189, 26–36. Teipel, S.J., Flatz, W.H., Heinsen, H., Bokde, A.L., Schoenberg, S.O., Stockel, S., Dietrich,
Sakata, Y., Zhuang, H., Kwansa, H., Koehler, R.C., Dore, S., 2010. Resveratrol protects O., Reiser, M.F., Moller, H.J., Hampel, H., 2005. Measurement of basal forebrain
against experimental stroke: putative neuroprotective role of heme oxygenase atrophy in Alzheimer’s disease using MRI. Brain 128, 2626–2644.
1. Exp. Neurol. 224, 325–329. Temme, E.H., Van Hoydonck, P.G., 2002. Tea consumption and iron status. Eur. J. Clin.
Salah, N., Miller, N.J., Paganga, G., Tijburg, L., Bolwell, G.P., Rice-Evans, C., 1995. Nutr. 56, 379–386.
Polyphenolic flavanols as scavengers of aqueous phase radicals and as chain- Terao, J., Yamaguchi, S., Shirai, M., Miyoshi, M., Moon, J.H., Oshima, S., Inakuma, T.,
breaking antioxidants. Arch. Biochem. Biophys. 322, 339–346. Tsushida, T., Kato, Y., 2001. Protection by quercetin and quercetin 3-O-beta-
Savaskan, E., Olivieri, G., Meier, F., Seifritz, E., Wirz-Justice, A., Muller-Spahn, F., d-glucuronide of peroxynitrite-induced antioxidant consumption in human
2003. Red wine ingredient resveratrol protects from beta-amyloid neurotoxi- plasma low-density lipoprotein. Free Radic. Res. 35, 925–931.
city. Gerontology 49, 380–383. Terzuoli, E., Donnini, S., Giachetti, A., Iniguez, M.A., Fresno, M., Melillo, G., Ziche, M.,
Scalbert, A., Morand, C., Manach, C., Remesy, C., 2002. Absorption and metabolism 2010. Inhibition of hypoxia inducible factor-1alpha by dihydroxyphenylethanol,
of polyphenols in the gut and impact on health. Biomed. Pharmacother. 56, a product from olive oil, blocks microsomal prostaglandin-E synthase-1/vascular
276–282. endothelial growth factor expression and reduces tumor angiogenesis. Clin.
Scalbert, A., Williamson, G., 2000. Dietary intake and bioavailability of polyphenols. Cancer Res. 16, 4207–4216.
J. Nutr. 130, 2073S–2085S. Tesch, G.H., Lim, A.K., 2011. Recent insights into diabetic renal injury from the db/db
Schilter, B., Andersson, C., Anton, R., Constable, A., Kleiner, J., O‘Brien, J., Renwick, mouse model of type 2 diabetic nephropathy. Am. J. Physiol. Renal Physiol. 300,
A.G., Korver, O., Smit, F., Walker, R., 2003. Guidance for the safety assessment F301–F310.
of botanicals and botanical preparations for use in food and food supplements. Thomas, R., Kim, M.H., 2005. Epigallocatechin gallate inhibits HIF-1alpha degrada-
Food Chem. Toxicol. 41, 1625–1649. tion in prostate cancer cells. Biochem. Biophys. Res. Commun. 334, 543–548.
Selkoe, D.J., 1998. The cell biology of beta-amyloid precursor protein and presenilin Thompson, M., Williams, C.R., Elliot, G.E., 1976. Stability of flavonoid complexes of
in Alzheimer’s disease. Trends Cell Biol. 8, 447–453. copper(II) and flavonoid antioxidant activity. Anal. Chim. Acta 85, 375–381.
Selkoe, D.J., 2001. Alzheimer’s disease: genes, proteins, and therapy. Physiol. Rev. Tota, S., Awasthi, H., Kamat, P.K., Nath, C., Hanif, K., 2010. Protective effect of
81, 741–766. quercetin against intracerebral streptozotocin induced reduction in cerebral
Semenza, G.L., 1998. Hypoxia-inducible factor 1: master regulator of O2 homeostasis. blood flow and impairment of memory in mice. Behav. Brain Res. 209, 73–79.
Curr. Opin. Genet. Dev. 8, 588–594. Tu, S.H., Ku, C.Y., Ho, C.T., Chen, C.S., Huang, C.S., Lee, C.H., Chen, L.C., Pan, M.H., Chang,
Sharpe, R.M., Franks, S., 2002. Environment, lifestyle and infertility – an inter- H.W., Chang, C.H., Chang, Y.J., Wei, P.L., Wu, C.H., Ho, Y.S., 2011. Tea polyphenol
generational issue. Nat. Cell Biol. 4 (Suppl.), s33–s40. (−)-epigallocatechin-3-gallate inhibits nicotine- and estrogen-induced alpha9-
Shi, J., Yang, S.H., Stubley, L., Day, A.L., Simpkins, J.W., 2000. Hypoperfusion induces nicotinic acetylcholine receptor upregulation in human breast cancer cells. Mol.
overexpression of beta-amyloid precursor protein mRNA in a focal ischemic Nutr. Food Res. 55, 455–466.
rodent model. Brain Res. 853, 1–4. Ursini, F., Maiorino, M., Morazzoni, P., Roveri, A., Pifferi, G., 1994. A novel antioxidant
Shim, S.S., Hammonds, M.D., Kee, B.S., 2008. Potentiation of the NMDA receptor in flavonoid (IdB 1031) affecting molecular mechanisms of cellular activation. Free
the treatment of schizophrenia: focused on the glycine site. Eur. Arch. Psychiatry Radic. Biol. Med. 16, 547–553.
Clin. Neurosci. 258, 16–27. van Acker, S.A., van den Berg, D.J., Tromp, M.N., Griffioen, D.H., van Bennekom, W.P.,
Shimmyo, Y., Kihara, T., Akaike, A., Niidome, T., Sugimoto, H., 2008a. Flavonols van der Vijgh, W.J., Bast, A., 1996. Structural aspects of antioxidant activity of
and flavones as BACE-1 inhibitors: structure–activity relationship in cell-free, flavonoids. Free Radic. Biol. Med. 20, 331–342.
cell-based and in silico studies reveal novel pharmacophore features. Biochim. van het Hof, K.H., Wiseman, S.A., Yang, C.S., Tijburg, L.B., 1999. Plasma and lipoprotein
Biophys. Acta 1780, 819–825. levels of tea catechins following repeated tea consumption. Proc. Soc. Exp. Biol.
Shimmyo, Y., Kihara, T., Akaike, A., Niidome, T., Sugimoto, H., 2008b. Multifunction Med. 220, 203–209.
of myricetin on A beta: neuroprotection via a conformational change of A beta Vauzour, D., Vafeiadou, K., Rice-Evans, C., Williams, R.J., Spencer, J.P., 2007. Activation
and reduction of A beta via the interference of secretases. J. Neurosci. Res. 86, of pro-survival Akt and ERK1/2 signalling pathways underlie the anti-apoptotic
368–377. effects of flavanones in cortical neurons. J. Neurochem. 103, 1355–1367.
Shirai, M., Moon, J.H., Tsushida, T., Terao, J., 2001. Inhibitory effect of a quercetin Veronese, M.L., Gillen, L.P., Burke, J.P., Dorval, E.P., Hauck, W.W., Pequignot, E., Wald-
metabolite, quercetin 3-O-beta-d-glucuronide, on lipid peroxidation in liposo- man, S.A., Greenberg, H.E., 2003. Exposure-dependent inhibition of intestinal
mal membranes. J. Agric. Food Chem. 49, 5602–5608. and hepatic CYP3A4 in vivo by grapefruit juice. J. Clin. Pharmacol. 43, 831–839.
Singh, M., Arseneault, M., Sanderson, T., Murthy, V., Ramassamy, C., 2008. Challenges Vingtdeux, V., Giliberto, L., Zhao, H., Chandakkar, P., Wu, Q., Simon, J.E., Janle, E.M.,
for research on polyphenols from foods in Alzheimer’s disease: bioavailability, Lobo, J., Ferruzzi, M.G., Davies, P., Marambaud, P., 2010. AMP-activated pro-
metabolism, and cellular and molecular mechanisms. J. Agric. Food Chem. 56, tein kinase signaling activation by resveratrol modulates amyloid-beta peptide
4855–4873. metabolism. J. Biol. Chem. 285, 9100–9113.
Smith, A., Giunta, B., Bickford, P.C., Fountain, M., Tan, J., Shytle, R.D., 2010. Nanoli- Vislocky, L.M., Fernandez, M.L., 2010. Biomedical effects of grape products. Nutr.
pidic particles improve the bioavailability and alpha-secretase inducing ability Rev. 68, 656–670.
of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer’s disease. Wallin, A.K., Blennow, K., Andreasen, N., Minthon, L., 2006. CSF biomarkers for
Int. J. Pharm. 389, 207–212. Alzheimer’s Disease: levels of beta-amyloid, tau, phosphorylated tau relate to
Soucek, T., Cumming, R., Dargusch, R., Maher, P., Schubert, D., 2003. The regulation of clinical symptoms and survival. Dement. Geriatr. Cogn. Disord. 21, 131–138.
glucose metabolism by HIF-1 mediates a neuroprotective response to amyloid Wang, B.S., Wang, H., Wei, Z.H., Song, Y.Y., Zhang, L., Chen, H.Z., 2009a. Effi-
beta peptide. Neuron 39, 43–56. cacy and safety of natural acetylcholinesterase inhibitor huperzine A in the
A. Ebrahimi, H. Schluesener / Ageing Research Reviews 11 (2012) 329–345 345

treatment of Alzheimer’s disease: an updated meta-analysis. J. Neural. Transm. Wisniewski, A.B., Klein, S.L., Lakshmanan, Y., Gearhart, J.P., 2003. Exposure to genis-
116, 457–465. tein during gestation and lactation demasculinizes the reproductive system in
Wang, C.Y., Zheng, W., Wang, T., Xie, J.W., Wang, S.L., Zhao, B.L., Teng, W.P., Wang, rats. J. Urol. 169, 1582–1586.
Z.Y., 2011. Huperzine A activates Wnt/beta-catenin signaling and enhances the Xu, S.S., Cai, Z.Y., Qu, Z.W., Yang, R.M., Cai, Y.L., Wang, G.Q., Su, X.Q., Zhong, X.S., Cheng,
nonamyloidogenic pathway in an Alzheimer transgenic mouse model. Neu- R.Y., Xu, W.A., Li, J.X., Feng, B., 1999. Huperzine-A in capsules and tablets for
ropsychopharmacology 36, 1073–1089. treating patients with Alzheimer disease. Zhongguo Yao Li Xue Bao 20, 486–490.
Wang, G.L., Semenza, G.L., 1993. General involvement of hypoxia-inducible fac- Xu, S.S., Gao, Z.X., Weng, Z., Du, Z.M., Xu, W.A., Yang, J.S., Zhang, M.L., Tong, Z.H., Fang,
tor 1 in transcriptional response to hypoxia. Proc. Natl. Acad. Sci. U.S.A. 90, Y.S., Chai, X.S., et al., 1995. Efficacy of tablet huperzine-A on memory, cognition,
4304–4308. and behavior in Alzheimer’s disease. Zhongguo Yao Li Xue Bao 16, 391–395.
Wang, H., Tang, X.C., 1998a. Anticholinesterase effects of huperzine A, E2020, and Xu, Z., Chen, S., Li, X., Luo, G., Li, L., Le, W., 2006. Neuroprotective effects of (−)-
tacrine in rats. Zhongguo Yao Li Xue Bao 19, 27–30. epigallocatechin-3-gallate in a transgenic mouse model of amyotrophic lateral
Wang, J., Pantopoulos, K., 2005. The pathway for IRP2 degradation involving 2- sclerosis. Neurochem. Res. 31, 1263–1269.
oxoglutarate-dependent oxygenase(s) does not require the E3 ubiquitin ligase Yamamoto, N., Moon, J.H., Tsushida, T., Nagao, A., Terao, J., 1999. Inhibitory effect of
activity of pVHL. Biochim. Biophys. Acta 1743, 79–85. quercetin metabolites and their related derivatives on copper ion-induced lipid
Wang, J., Pfleger, C.M., Friedman, L., Vittorino, R., Zhao, W., Qian, X., Conley, L., Ho, peroxidation in human low-density lipoprotein. Arch. Biochem. Biophys. 372,
L., Pasinetti, G.M., 2010a. Potential application of grape derived polyphenols in 347–354.
Huntington’s disease. Transl. Neurosci. 1, 95–100. Yang, J., Li, Q., Zhou, X.D., Kolosov, V.P., Perelman, J.M., 2011. Naringenin attenuates
Wang, J., Santa-Maria, I., Ho, L., Ksiezak-Reding, H., Ono, K., Teplow, D.B., Pasinetti, mucous hypersecretion by modulating reactive oxygen species production and
G.M., 2010b. Grape derived polyphenols attenuate tau neuropathology in a inhibiting NF-kappaB activity via EGFR-PI3K-Akt/ERK MAPKinase signaling in
mouse model of Alzheimer’s disease. J. Alzheimers Dis. 22, 653–661. human airway epithelial cells. Mol. Cell. Biochem. 351, 29–40.
Wang, J., Zhang, H.Y., Tang, X.C., 2010c. Huperzine a improves chronic inflammation Yazawa, K., Kihara, T., Shen, H., Shimmyo, Y., Niidome, T., Sugimoto, H., 2006. Distinct
and cognitive decline in rats with cerebral hypoperfusion. J. Neurosci. Res. 88, mechanisms underlie distinct polyphenol-induced neuroprotection. FEBS Lett.
807–815. 580, 6623–6628.
Wang, L.S., Zhou, J., Shao, X.M., Tang, X.C., 2003. Huperzine A attenuates cognitive Yuan, J., Yankner, B.A., 2000. Apoptosis in the nervous system. Nature 407, 802–809.
deficits and brain injury after hypoxia-ischemic brain damage in neonatal rats. Zangara, A., 2003. The psychopharmacology of huperzine A: an alkaloid with cog-
Zhonghua Er Ke Za Zhi 41, 42–45. nitive enhancing and neuroprotective properties of interest in the treatment of
Wang, Q., Sun, A.Y., Simonyi, A., Jensen, M.D., Shelat, P.B., Rottinghaus, G.E., Alzheimer’s disease. Pharmacol. Biochem. Behav. 75, 675–686.
MacDonald, R.S., Miller, D.K., Lubahn, D.E., Weisman, G.A., Sun, G.Y., 2005. Zeng, X.J., Yu, S.P., Zhang, L., Wei, L., 2010. Neuroprotective effect of the endogenous
Neuroprotective mechanisms of curcumin against cerebral ischemia-induced neural peptide apelin in cultured mouse cortical neurons. Exp. Cell Res. 316,
neuronal apoptosis and behavioral deficits. J. Neurosci. Res. 82, 138–148. 1773–1783.
Wang, R., Tang, X.C., 2005. Neuroprotective effects of huperzine A. A natural Zha, J., Harada, H., Yang, E., Jockel, J., Korsmeyer, S.J., 1996. Serine phosphorylation
cholinesterase inhibitor for the treatment of Alzheimer’s disease. Neurosignals of death agonist BAD in response to survival factor results in binding to 14-3-3
14, 71–82. not BCL-X(L). Cell 87, 619–628.
Wang, R., Yan, H., Tang, X.C., 2006a. Progress in studies of huperzine A, a natural Zhang, F., Shi, J.S., Zhou, H., Wilson, B., Hong, J.S., Gao, H.M., 2010. Resveratrol protects
cholinesterase inhibitor from Chinese herbal medicine. Acta Pharmacol. Sin. 27, dopamine neurons against lipopolysaccharide-induced neurotoxicity through
1–26. its anti-inflammatory actions. Mol. Pharmacol. 78, 466–477.
Wang, R., Zhang, H.Y., Tang, X.C., 2001. Huperzine A attenuates cognitive dysfunction Zhang, H., Schools, G.P., Lei, T., Wang, W., Kimelberg, H.K., Zhou, M., 2008. Resveratrol
and neuronal degeneration caused by beta-amyloid protein-(1–40) in rat. Eur. attenuates early pyramidal neuron excitability impairment and death in acute
J. Pharmacol. 421, 149–156. rat hippocampal slices caused by oxygen-glucose deprivation. Exp. Neurol. 212,
Wang, R., Zhang, Y.W., Zhang, X., Liu, R., Hong, S., Xia, K., Xia, J., Zhang, Z., Xu, H., 44–52.
2006b. Transcriptional regulation of APH-1A and increased gamma-secretase Zhang, L., Cao, H., Wen, J., Xu, M., 2009. Green tea polyphenol (−)-epigallocatechin-
cleavage of APP and Notch by HIF-1 and hypoxia. FASEB J. 20, 1275–1277. 3-gallate enhances the inhibitory effect of huperzine A on acetylcholinesterase
Wang, T., Tang, X.C., 1998b. Reversal of scopolamine-induced deficits in radial maze by increasing the affinity with serum albumin. Nutr. Neurosci. 12, 142–148.
performance by (−)-huperzine A: comparison with E2020 and tacrine. Eur. J. Zhang, S., Zhang, Z., Sandhu, G., Ma, X., Yang, X., Geiger, J.D., Kong, J., 2007a. Evidence
Pharmacol. 349, 137–142. of oxidative stress-induced BNIP3 expression in amyloid beta neurotoxicity.
Wang, Y., Zeng, Q.G., Zhang, Z.B., Yan, R.M., Wang, L.Y., Zhu, D., 2010d. Isolation Brain Res. 1138, 221–230.
and characterization of endophytic huperzine A-producing fungi from Huperzia Zhang, X., Zhou, K., Wang, R., Cui, J., Lipton, S.A., Liao, F.F., Xu, H., Zhang,
serrata. J. Ind. Microbiol. Biotechnol. Y.W., 2007b. Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated hypoxia
Wang, Y.E., Yue, D.X., Tang, X.C., 1986. Anti-cholinesterase activity of huperzine A. increases BACE1 expression and beta-amyloid generation. J. Biol. Chem. 282,
Zhongguo Yao Li Xue Bao 7, 110–113. 10873–10880.
Wang, Y.J., Thomas, P., Zhong, J.H., Bi, F.F., Kosaraju, S., Pollard, A., Fenech, M., Zhou, Zhang, Z., Wang, X., Chen, Q., Shu, L., Wang, J., Shan, G., 2002. [Clinical efficacy and
X.F., 2009b. Consumption of grape seed extract prevents amyloid-beta depo- safety of huperzine Alpha in treatment of mild to moderate Alzheimer disease,
sition and attenuates inflammation in brain of an Alzheimer’s disease mouse. a placebo-controlled, double-blind, randomized trial]. Zhonghua Yi Xue Za Zhi
Neurotox. Res. 15, 3–14. 82, 941–944.
Warden, B.A., Smith, L.S., Beecher, G.R., Balentine, D.A., Clevidence, B.A., 2001. Cat- Zhang, Z.J., Tong, Y., Wang, X.Y., Yao, S.M., Jin, G.X., Wang, X.P., 2007c. Huperzine A
echins are bioavailable in men and women drinking black tea throughout the as add-on therapy in patients with treatment-resistant schizophrenia: an open-
day. J. Nutr. 131, 1731–1737. labeled trial. Schizophr. Res. 92, 273–275.
Waxman, E.A., Lynch, D.R., 2005. N-methyl-d-aspartate receptor subtypes: multiple Zhao, B., Guo, Q., Xin, W., 2001. Free radical scavenging by green tea polyphenols.
roles in excitotoxicity and neurological disease. Neuroscientist 11, 37–49. Methods Enzymol. 335, 217–231.
Wei, J., Walton, E.A., Milici, A., Buccafusco, J.J., 1994. m1–m5 muscarinic receptor Zhao, Q., Tang, X.C., 2002. Effects of huperzine A on acetylcholinesterase isoforms
distribution in rat CNS by RT-PCR and HPLC. J. Neurochem. 63, 815–821. in vitro: comparison with tacrine, donepezil, rivastigmine and physostigmine.
Weinreb, O., Amit, T., Mandel, S., Youdim, M.B., 2009. Neuroprotective molecu- Eur. J. Pharmacol. 455, 101–107.
lar mechanisms of (−) epigallocatechin-3-gallate: a reflective outcome of its Zheng, L.T., Ock, J., Kwon, B.M., Suk, K., 2008. Suppressive effects of flavonoid
antioxidant, iron chelating and neuritogenic properties. Genes Nutr. 4, 283–296. fisetin on lipopolysaccharide-induced microglial activation and neurotoxicity.
Weinreb, O., Amit, T., Youdim, M.B., 2007. A novel approach of proteomics and tran- Int. Immunopharmacol. 8, 484–494.
scriptomics to study the mechanism of action of the antioxidant-iron chelator Zhou, Y.D., Kim, Y.P., Li, X.C., Baerson, S.R., Agarwal, A.K., Hodges, T.W., Ferreira,
green tea polyphenol (−)-epigallocatechin-3-gallate. Free Radic. Biol. Med. 43, D., Nagle, D.G., 2004. Hypoxia-inducible factor-1 activation by (−)-epicatechin
546–556. gallate: potential adverse effects of cancer chemoprevention with high-dose
Weinreb, O., Mandel, S., Amit, T., Youdim, M.B., 2004. Neurological mechanisms of green tea extracts. J. Nat. Prod. 67, 2063–2069.
green tea polyphenols in Alzheimer’s and Parkinson’s diseases. J. Nutr. Biochem. Zhou, Y.Q., Yang, Z.L., Xu, L., Li, P., Hu, Y.Z., 2009. Akebia saponin D, a saponin com-
15, 506–516. ponent from Dipsacus asper Wall, protects PC 12 cells against amyloid-beta
Wenger, R.H., Gassmann, M., 1997. Oxygen(es) and the hypoxia-inducible factor-1. induced cytotoxicity. Cell Biol. Int. 33, 1102–1110.
Biol. Chem. 378, 609–616. Zhu, D., Wang, J., Zeng, Q., Zhang, Z., Yan, R., 2010. A novel endophytic Huperzine
Whitehouse, P.J., 1998. The cholinergic deficit in Alzheimer’s disease. J. Clin. Psychi- A-producing fungus, Shiraia sp. Slf14, isolated from Huperzia serrata. J. Appl.
atry 59 (Suppl. 13), 19–22. Microbiol. 109, 1469–1478.
Williams, R.J., Spencer, J.P., Rice-Evans, C., 2004. Flavonoids: antioxidants or sig- Zijp, I.M., Korver, O., Tijburg, L.B., 2000. Effect of tea and other dietary factors on iron
nalling molecules? Free Radic. Biol. Med. 36, 838–849. absorption. Crit. Rev. Food Sci. Nutr. 40, 371–398.

You might also like