<<

J Basic Clin Physiol Pharmacol 2017; 28(5): 413–423

Review

Thanh Tung Bui* and Thanh Hai Nguyen Natural product for the treatment of Alzheimer’s disease https://doi.org/10.1515/jbcpp-2016-0147 Received September 27, 2016; accepted May 28, 2017; previously Pathogenesis of Alzheimer’s disease published online July 14, 2017 Until now, the pathogenesis of AD is not yet completely Abstract: Alzheimer’s disease (AD) is related to increas- understood. The genetic susceptibility and environmental ing age. It is mainly characterized by progressive neu- factors are responsible for late onset sporadic AD, which rodegenerative disease, which damages memory and is the most common form of the disease. Many studies cognitive function. Natural products offer many options have attempted to understand the disease mechanism and to reduce the progress and symptoms of many kinds of develop a disease-modifying drug. Two main factors are diseases, including AD. Meanwhile, natural compound responsible for the development of AD: β-amyloid protein structures, including lignans, flavonoids, tannins, poly- and abnormal tau protein, or both of them. AD is charac- phenols, triterpenes, sterols, and , have anti- teristized by the overproduction of β-amyloid proteins (Aβ) inflammatory, antioxidant, anti-amyloidogenic, and and hyperphosphorylated Tau protein, which can lead to anticholinesterase activities. In this review, we summa- the loss of synaptic connections and neurons in the hip- rize the pathogenesis and targets for treatment of AD. We pocampus and cerebral cortex, a decline in cognitive func- also present several medicinal plants and isolated com- tion, and [2]. The accumulation of β-amyloid pounds that are used for preventing and reducing symp- protein and neurofibrillary tangles have been shown in toms of AD. AD patients. The overproduction and accumulation of Aβ lead to oxidation, lipid peroxidation, inflammation, dis- Keywords: ; Alzheimer’s turbance of cell functions, and apoptosis and formation of disease; β-amyloid; natural product. neurofibrillary tangles [3]. The tau protein is hyperphos- phorylated, which leads to the formation of tangles depos- ited in the hippocampus and, ultimately, to cell death [4]. Many biochemical changes within cells, such as oxidative Introduction stress, inflammation, metabolic disturbances, disruption of Ca2+ homeostasis, and the accumulation of unfolded/ Alzheimer’s disease (AD) is a disease related to aging. It mis-folded proteins, have been demonstrated to induce is neurodegenerative disease that leads to memory loss neuronal cell death in AD patients [5]. The two mecha- and impaired cognitive function. The reports of the World nisms involved in AD are oxidative damage and inflam- Health Organization in 2012 showed that over 35.6 million matory process, and both of these have been studied well people in the world have dementia, and about 60%–70% in the literature. of this population has AD. The same report also predicted that the number of people with dementia, and those with AD, shall continue to increase [1]. Acetylcholinesterase inhibitor for the treatment of Alzheimer’s disease

*Corresponding author: Thanh Tung Bui, School of Medicine and hypothesis is an important hypothesis that is Pharmacy, Vietnam National University, Hanoi, Office 506, Building used to explain the pathogenesis of AD. This hypothesis Y1, 144 XuanThuy, Cau Giay, Ha Noi, Vietnam, Tel.: +84-4-85876172, Fax: +84-0437450188, E-mail: [email protected] explains that AD is caused by a decrease in neurotrans- Thanh Hai Nguyen: School of Medicine and Pharmacy, Viet Nam mitter (Ach). Many drugs used in the treat- National University, Hanoi, Vietnam ment of AD are based on cholinergic hypothesis [6]. ACh, 414 Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease serotonin, and norepinephrine are neurotransmitter defi- stress in AD patients. Hence, antioxidant treatment is a cits in the brain that occur because of cell death. Many promising strategy for inhibiting disease progression. studies have shown that a low level of neurotransmitters A recent study has shown the link between antioxidant in a cholinergic system is responsible for cognitive decline intake and reduced symptoms of dementia [16]. Treatment and memory loss in AD patients [6–9]. Acetylcholinester- with antioxidants, such as those found n vitamin E, vitamin ase (AChE) is a main enzyme that degrades the ACh neu- C, selegiline, estrogen, and Ginkgo biloba, may inhibit the rotransmitter. Various therapeutic strategies elevate ACh development of AD [17]. Furthermore, some medicinal neurotransmitter levels, then increase cholinergic trans- plants with antioxidant activities also exhibit potential for mission by inhibiting ACh hydrolysis with AChE inhibitors, AD treatment. Jung et al. have reported that three major stimulating nicotinic and muscarinic receptors or using alkaloids in Coptidis rhizoma, including groenlandicine, cholinomimetic substances [9]. Drugs such as , berberine, and palmatine, can potentially exhibit anti-AD , , or inhibit effects by inhibiting AChE activity and Aβ accumulation, AChE, augment the level of Ach, and improves cholinergic and also present antioxidant capacities to inhibit ROS and transmission. These drugs have been used to alleviate the RNS levels [13]. Silibinin, an isolated flavonoid from Silybum symptoms of AD, which are caused by the degeneration of marianum, also has an antioxidant property. Silibinin has cholinergic neurons and injured transmission. However, been shown to prevent memory impairment and oxidative the inhibition of AChE has not been very effective in the damage induced by Aβ in mice model, making it a poten- treatment of Alzheimer [9]. AChE inhibitors have many tial agent for AD treatment [18]. Therefore, further studies side effects, such as nausea, vomiting, diarrhea, abdomi- related to medicinal plants that have antioxidant properties nal pain, dyspepsia, and skin rash [8]. Tacrine has also for the treatment of AD should be conducted. been found to induce hepatotoxicity in clinical trials [10]. was taken out from the drug market due to many disadvantages. Therefore, finding new AChE inhibi- tors, which may be found in medicinal plant resources, Amyloid hypothesis and Alzheimer with less adverse effects is important. drugs targeting β-amyloid

The amyloid hypothesis was developed in 1991. It states that the extracellular β-amyloid deposits are the main Antioxidants for the treatment cause of AD [19]. β-amyloid, a short fragment of the of Alzheimer’s disease amyloid precursor protein (APP), is a product of a process- ing error of the APP in the brain. The accumulated clus- Many studies have shown that brain tissues in AD patients ters of β-amyloid proteins are called amyloid plaques. The are subjected to oxidative stress. Oxidative stress leads to amyloid plaques in the brain provoke the destruction of protein oxidation, lipid oxidation, DNA oxidation, and gly- neuronal cells and leads to AD. APP is a long protein con- coxidation [11]. The imbalance in the production of reactive sisting of up to 771 amino acids. APP is cut by two differ- oxygen species (ROS) and antioxidative defense system, ent leading to the generation of β-amyloid [20]. which is responsible for ROS removal, is characterized by The first β-secretase cuts the APP and then the second oxidative stress. It leads to the age-related neurodegenera- gamma-secretase enzyme cuts one more time to generate tion and cognitive decline [12]. ROS, such as superoxide β-amyloid, which may have a length of 38, 40, or 42 amino − anion radical (O2˙ ), peroxide (H2O2), hydroxyl acids. The length of 42 amino acids causes β-amyloid to be 1 radical (˙OH), singlet oxygen ( O2), alkoxyl radicals (RO˙), chemically stickier than the other lengths, thus leading to peroxyl radicals (ROO˙), and reactive nitrogen species clumps and the formation of plaques. (RNS), such as peroxynitrites (ONOO −), enrich many β-Amyloid plaques also provoke the formation of human degenerative diseases [13]. Oxidative stress occurs tangles of tau proteins. These tangles also damage the when ROS and RNS levels exceed the removal capacity of neuronal cells, which lead to dementia [21]. The formation the antioxidant system and cause cell dysfunc- of β-amyloid plaques also leads to oxidative stress in the tion, leading to several pathological conditions or aging neurons, which in turn, aggravates the development of AD [14]. High levels of protein oxidation, lipid oxidation, DNA [22]. Aβ has several isoforms, but the form of 42 amino acids oxidation, and glycoxidation are the main manifestations has been mainly found in amyloid plaques. Other isoforms during the course of AD [15]. The imbalance of ROS levels have also been found, such as Aβ1−40 and Aβ25−35. Many and antioxidative defense system typically causes oxidative studies have used Aβ1−42 and Aβ25−35 to produce oxidative Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease 415 stress to study the neuroprotection of natural products [32]. Many studies have been conducted to investigate the using different cell lines [23]. When neuronal cell is exposed effects of total medicinal plant extracts on AD as well as to Aβ, this leads to increased ROS production, mitochon- to isolate and identify the active compounds involved [33]. dria dysfunction, and apoptosis as well as the down reg- Many compounds, including lignans, flavonoids, tannins, ulation of antioxidant genes, leading to neuronal cell polyphenols, triterpenes, sterols, and alkaloids, have dysfunction and worsened AD symptoms [24]. APOE4, an shown various beneficial pharmacological activities, such isoform of apolipoprotein, is a major genetic risk factor for as antiinflammatory, anti-amyloidogenic, anticholinester- AD. This APOE4 may induce β-amyloid accumulation in the ase, and antioxidant [32]. Some compounds, such as aged brain [19]. As amyloid plaques are increased in AD patients. garlic extract, , melatonin, , Ginkgo Hence, researchers have proposed that drugs preventing biloba extract, green tea, and vitamins C and E, have been β-amyloid accumulation may be a potential treatment for used in patients with AD and yielded positive results [34– AD. PBT2, a drug candidate, is a second-­generation of eight- 36]. Below, we summarize information on some medicinal hydroxyquinoline analog for AD. PBT2 has been shown in plants and isolated compounds that are used for the treat- animal models to effectively detoxify Aβ and increase the ment of AD. clearance of β-amyloid [25]. In a clinical trial, PBT2 at a dose of 250 mg also decreased levels of Aβ1−42 in the brain [26]. Therefore, drugs that can inhibit β-amyloid accumulation may be a strategy for AD treatment. Medicinal plants used for the The β-amyloid plaques in the brain damages neu- ronal cells by inducing inflammatory reaction. AD is also treatment of Alzheimer’s disease characterized by neuroinflammation features. Cytokines, such as interleukin-1, interleukin-6 (IL-6), and tumor Curcuma longa necrosis factor alpha, have been shown to play a role in the neuroinflammatory process. These cytokines’ expres- Turmeric of Curcuma longa is a rhizomatous perennial sions are increased by β-amyloid peptide. In return, these plant of the ginger family, Zingiberaceae. The active cytokines estimate the accumulation of β-amyloid peptide compounds are water-insoluble curcuminoids, includ- [27]. Some researchers suggested that these cytokines may ing curcumin, demethoxycurcumin, and bis-demethox- play an important role in the progression of AD [28]. The ycurcumin [37]. Curcumin is the main curcuminoid and prolonged use of non-steroidal anti-inflammatory drugs it is responsible for the yellow color of the turmeric root (NSAIDs) has reduced the development of AD and delayed [38]. Curcumin has anti-inflammatory, antioxidant, anti- the onset of the disease [29]. The mechanism of NSAIDs in tumor, antibacterial activities, among others [39, 40]. A decreasing symptoms of Alzheimer may be related to the previous review has shown that curcumin may be a prom- inhibition of the (COX) and the activation ising compound for the treatment of AD [41]. According of peroxisome proliferator-activated γ (PPARγ) to another study, when aged mice with β-amyloid plaque [29]. NSAIDs have been shown to suppress the expression accumulation were fed with curcumin, this reduced the of COX, which in turn, stimulates the synthesis of prosta- amount of plaque deposition [42]. Curcumin can decrease glandins and decreases the production of cytokines [30]. oxidative damage status in the brain [43]. In an Alzhei- An in vivo model of transgenic mice with AD and in vitro mer transgenic mouse model, curcumin has been shown model of cell cultures of peripheral, glial, and neuronal to reverse the β-amyloid pathology [44, 45]. Some symp- NSAIDs, such as , indomethacin, and , toms of AD were also reduced by curcumin’s antioxidant decrease the level of Aβ [29]. However, the beneficial effect and anti-inflammatory properties [45]. In vitro curcumin of NSAIDs in AD treatment are still being discussed due to may inhibit lipid peroxidation and neutralize ROS levels, the limited evidence presented [31]. making it even more potent than vitamin E [46, 47]. Curcumin (with a dose of 5–10 μM) protected PC12 cells against Aβ-induced neurotoxicity by inhibiting oxidative damage and tau hyperphosphorylation [42]. In a clini- Herbal medicine for the treatment cal trial, curcumin has shown several beneficial effects of Alzheimer’s disease on healthy middle aged people, including lowering the plasma β-amyloid protein concentrations [48]. More Medicinal plants have been shown to decrease the pro- experiments are needed to evaluate the exact mechanism gress and symptoms of various diseases, including AD of C. longa. 416 Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease

Bacopa monnieri memory in rats were significantly enhanced by feeding them with the ethanolic extract of C. pluricaulis and its Bacopa monnieri is commonly used in traditional medicine ethyl acetate and water fractions [60]. The oral admin- as a nerve tonic, diuretic, and a cardiotonic agent as well as istration of C. pluricaulis­ also alleviated the neurotoxic for the treatment of asthma and rheumatism. It also applied effect of scopolamine by reducing the induction of for epilepsy, insomnia [49]. The main compounds of B. mon- protein and mRNA levels of tau and AβPP [61]. Unfortu- nieri are saponins and triterpenoids. These compounds, nately, no clinical data exist on the effects of C. pluricaulis such as bacopasides III, bacopasides IV, bacopasides V, on AD patients. Further investigations are thus needed to bacosides A, bacosides B, bacosaponins A, bacosaponins B, explore the potential favorable effects of C. pluricaulis on and bacosaponins C, have been isolated from B. monnieri. AD treatment. The presence of some saponin glycosides, such as jujubo- genin bisdesmosides bacopasaponins D, bacopasaponins E, and bacopasaponins F, in B. monnieri has also been reported. Centella asiatica Furthermore, other compounds, including alkaloids, sterols, betulic acid, polyphenols, and sulfhydryl, which have anti- Traditional medicines have used Centella asiatica for rejuve- oxidant activities, have also been found in B. monnieri [50]. nating the neuronal cells. It has also been used for increas- Traditional medicine has used B. monnieri for improving ing intelligence, longevity, and memory [52]. Asiatic acid memory and cognitive function [51]. Many studies related and asiaticoside have been isolated from C. asiatica, and to neuropharmacological effects and nootropic action of these compounds have shown the ability to reduce H2O2- B. monnieri extracts have been conducted extensively [52]. induced cell cytotoxicity, decrease free radical levels, and In the hippocampus, B. ­monnieri augments protein kinase inhibit β-amyloid cell damages in vitro. C. asiatica extracts activity, which provides a nootropic action [53]. In an reduced the β-amyloid pathology and decreased the oxida- animal Alzheimer model, rats fed with B. monnieri extract tive stress response in the brains of a mice model of AD showed decreased cholinergic degeneration and exhibited [62]. It has already been reported that C. asiatica ethanol cognition-enhancing effect [54]. Another study reported extracts can protect neuronal cells against Aβ1−40-induced that B. monnieri inhibited the AChE activity and increased neurotoxicity. C. asiatica can also decrease ROS produc- ACh levels [55]. Moreover, B. monnieri extracts also protected tion and modulate the antioxidative defense system in neuronal cells from damages caused by β-amyloids. More- cells by enhancing the activities of superoxide dismutase, over, neuronal cells treated with B. monnieri extract exhib- catalase, peroxidase, glutathione reductase, ited lower levels of ROS, suggesting that B. monnieri reduced and levels of glutathione and [63]. intracellular oxidative stress [56]. A clinical trial of AD These activities suggest the important role of C. asiatica for patients showed that the polyherbal formulation containing the prevention and treatment of AD [64]. B. monnieri extract effectively improved the cognitive func- tions and decreased the level of inflammation and oxidative stress in patients [57]. Nevertheless, detailed investigations Ginkgo biloba are needed to further assess the potential neuroprotective action of B. monnieri against AD. Ginkgo biloba leaves have been used in traditional medi- cine as an agent for improving memory and age-related deterioration. The phytochemicals of Ginkgo biloba Convolvulus pluricaulis leaf extracts contain flavonoids, organic acids, and ter- enoids, which provide neuroprotective activity. The pro- The phytochemicals of Convolvulus pluricaulis reveal tective mechanism of Ginkgo biloba leaf extracts against that it may contain active compounds, such as triterpe- β-amyloid has been shown to induce cytotoxicity and noids, flavonol glycosides, anthocyanins, and steroids. may be related to the capacity to scavenge free radicals, They provide the nootropic and memory-enhancing reduce mitochondrial dysfunction, activate JNK and ERK activity for C. pluricaulis [58]. Another study reported pathways, and prevent neuronal apoptosis. Bilobalide that C. pluricaulis­ may calm the nerves by regulating is a principal terpenoid compound isolated from Ginkgo stress hormone, adrenaline, and cortisol levels in the biloba leaves; it has potent protective effects on neurons body [58]. Traditional medicines have used C. pluricau- and Schwann cells [65]. Bilobalide has been shown to lis for nervous system-related diseases, including stress, reduce the expression of p53, Bax, and caspase-3 proteins anxiety, mental fatigue, and insomnia [59]. Learning and as well as inhibite ROS-induced apoptosis in PC12 cells Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease 417

[66]. Bilobalide also stimulated neurogenesis and synap- Others togenesis by increasing the levels of transcription factor phosphorylated CREB and neurotrophin BDNF in neu- Some natural product compounds have also been isolated ronal cells [67]. Furthermore, the clinical trial of market- from medicinal plants for the treatment of AD. ing a dietary supplement containing Ginkgo biloba extract was beneficial in treating cognitive decline in the aging Quercetin process and in the early stages of AD [68].

Zingiber officinaleis

Zingiber officinaleis has been widely used in food supple- ments as extracts or as ingredients of ginger tea. Principal­ compounds, such as gingerols, shagols, bisabolene, ­zingiberene, and monoterpenes, have been isolated from Z. officinaleis [69]. In vitro assays have shown the AChE Quercetin is a flavonoid compound found in a wide variety inhibitory activity of Z. officinaleis. Inhibiting the AChE of medicinal plants, such as apples, onions, berries, green enzyme increases ACh levels in synapses, augments the tea, and red wine. Quercetin has a strong antioxidant activity of cholinergic pathways, and enhances cognitive activity by scavenging ROS [76]. In addition, it has other functions in AD patients. Furthermore, Z. officinaleis has beneficial properties, including anticancer, antiviral, anti- the ability to inhibit lipid peroxidation and provide the inflammatory, and antiamyloidogenic properties [77, 78]. protective effect against AD. An in vivo assay has reported Quercetin at a dose of 10 μM has shown antiamyloidogenic reduced levels of lipid peroxidation in rats fed with activity by inhibiting the accumulation of β-amyloids [79]. Z. officinaleis extract. The mechanism may be explained Quercetin also reduced the Aβ-induced apoptosis in neu- by the ability of Z. officinaleis extract to reduce the over- ronal cells. However, at a higher dose (40 μM), quercetin stimulation of N-methyl-D-aspartate (NMDA) receptors may induce cytotoxicity [80]. A recent study has shown and prevent the formation of free radicals [70]. that nanoencapsulated quercetin in zein nanoparticles significantly improved cognition and memory impair- ments on senescence-accelerated P8 mice. The mecha- Allium sativum nism may be related to the decreased expression of the hippocampal astrocyte marker GFAP [81]. Allium sativum has been used in food and medicinal ­supplements. Borek et al. have investigated the neuropro- tective effect of the aged garlic extract in a Tg2576 mice Epigallocatechin-3-gallate model, in which mice fed with aged garlic extracts exhib- ited enhanced memory in the hippocampal region [71]. The mechanism has scavenged the free radicals; increased enzyme antioxidants, such as superoxide dismutase, cata- lase, glutathione peroxidase, and glutathione levels; inhib- ited lipid peroxidation level; and reduced inflammatory [72]. Moreover, aged garlic extracts inhib- ited 3-hydroxy-3-methylglutaryl-CoA reductase and reduced cholesterol synthesis. Aged garlic extracts can also protect neurons from β-amyloid neurotoxicity and apoptosis, pre- venting cognitive decline, ischemia or reperfusion-related neuronal death, and enhancing learning and memory retention [73]. A recent study has reported that aged garlic Epigallocatechin-3-gallate is a flavonoid-type catechin extracts were useful in enhancing the short-term recog- found in Camellia sinensis. Epigallocatechin-3-gallate nition memory and reducing the neuroinflammation in has potent antioxidant activity, and many studies have Aβ-induced rats [74] as well as in attenuating neuroinflam- investigated the effects of epigallocatechin-3-gallate on matory responses in microglial cells [75]. various diseases, including cancer and cardiovascular and 418 Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease neurodegenerative diseases [82]. Epigallocatechin-3-­gallate Resveratrol has been shown to increase glutathione peroxidase activity, inhibit the AChE activity and inhibit NO metabolite forma- tion and ROS generation in streptozotocin-induced­ demen- tia in mice [83]. Epigallocatechin-3-gallate also increased memory formation and inhibited γ-secretase enzyme activity in mutant PS2 Alzheimer mice [84]. Furthermore, epigallocatechin-3-gallate prevented LPS-induced memory loss and apoptosis by decreasing amyloid precursor protein expression, inhibiting beta-site APP cleaving enzyme 1 Resveratrol is a polyphenolic compound belonging to activity, and reducing β-amyloid accumulation. Moreover, the group stilbenes. Resveratrol can be found in red it has also been shown to reduce the expression of inflam- wine, nuts, and the skin of grapes and other fruits [94]. matory factors, such as tumor necrosis factor-α (TNF-α), Many studies have shown that it has anti-cancer, anti- interleukin 1β, IL-6, inducible synthase (iNOS), inflammatory, antioxidant, and protective cardiovascular -2 (COX-2), soluble intracellular adhesion, properties, can lower blood glucose levels, and neuropro- and molecule-1 macrophage colony-stimulating factor, and tective effects [95]. Resveratrol has a potent antioxidant to prevent astrocyte activation in neuronal cells [85, 86]. activity by scavenging ROS, increasing gluthatione levels, Epigallocatechin-3-gallate has also been shown to reduce and improving the endogenous antioxidants [96]. Resver- Aβ accumulation and elevate neprilysin enzyme expres- atrol can also reduce the level of β-amyloids by inducing sion, which is the rate-limiting degradation enzyme of Aβ in the non-amyloidogenic cleavage of APP and enhancing senescence-accelerated P8 mice [87]. the clearance of β-amyloid [95]. Resveratrol (15, 45, and 135 mg/kg) also inhibited AChE activity in neuronal cells [97]. A randomized, double-blind, placebo-controlled Berberine trial of resveratrol for AD provided evidence that resvera- trol is safe, well-tolerated, and has the ability to reduce CSF Aβ40 and plasma Aβ40 levels [98]. Recently, He et al. [99] provided evidence proving that resveratrol protects against hyperphosphorylation and/or mediates dephos- phorylation of the tau protein. Moroever, they found that resveratrol ameliorated the deleterious effects in a rat model for AD by alleviating cholinergic pathways, thus reducing oxidative stress and improving spatial memory [100]. Evidence that resveratrol may modulate neuro- inflammation and induces adaptive immunity by the acti- vation of SIRT1 has also been presented [101]. Berberine is a quaternary ammonium salt and a type of isoquinoline alkaloids, isolated from Coptis chinensis [88]. Berberine has many biological activities, such as antioxi- dant activity, inhibition of AChE and , monoamine oxidase, and cholesterol-­lowering activity [88]. Tg mice fed with berberine at a dose of 100 mg/kg via oral administration showed significantly increasing learning and spatial memory [89]. BV2 microglia cells treated with berberine significantly reduced the β-amyloid-induced expressions of IL-6, COX-2, and iNOS [90]. In addition, ber- berine also strongly reduced the expression of NF-κB by inhibiting the PI3K/protein kinase B and MAPK pathways Huperzine A is a sesquiterpene group found in [91]. A recent study has shown that berberine can signifi- Huperzia serrate. Traditional medicine has used Huper- cantly ameliorate memory impairment, reduce Aβ and APP zine A for the treatment of fever and swelling. H. serrate levels, reduce Aβ plaque deposition in the hippocampus extract can also be used as a food supplement for improv- [92], and prevent the increase of AChE activity [93]. ing memory [102]. Huperzine A has a strong effect on the Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease 419 inhibition of AChE. Its mechanism is similar to those of [112]. may also prevent locomotor activity, rivastigmine, donepezil, and galantamine, which have short-term spatial memory, and biochemical alterations of been used for AD treatment. The new tacrine-Huperzine brain tissue found in a rat model of AD by reducing lipid A combination (called Huprines) is a potent AChE inhibi- peroxidation and inflammatory process [113]. Clinical trial tors that can significantly reduce Aβ-induced memory studies are needed to further demonstrate the efficiency of injury [103]. Clinical trials have shown that huperzine A rosmarinic acid against AD. has very low minimal adverse reactions, such as gastro- enteric symptoms, dizziness, headaches, nausea, and Luteolin reduced heart rate. This is an advantage of huperzine A compared with other drug AChE inhibitors for AD treat- ment [102]. Huperzine A can potentially inhibit several apoptotic factors, including caspase-3, Bax, and p53. Huperzine A also regulated the expression and secretion of the [104]. Huperzine A enhanced learning capacity and memory in Tg mice in a Morris water maze test. The mechanism can be explained by the inhi- bition of PKC/MAPK, γ-secretases, and BACE as well as in the increase of phospho GSK-3 [105]. Huperzine A also Luteolin is a flavonoid compound found in many medici- reduced the β-amyloid plaques and oligomeric A amount nal plants, such as Magnoliophyta, Pteridophyta, Bryo- in the cortex and hippocampus [106]. Moreover, huperzine phyta, and Pinophyta [114]. Luteolin has been shown to A can inhibit the NMDA receptor and potassium chanel have many biological activities, such as anti-inflamma- in the brain [107]. A phase II trial of huperzine A in mild tory, antioxidant, anticancer, antimicrobial, and neu- to moderate AD showed that huperzine A 200 μg did not roprotective effects [115]. Luteolin can also reduce the demonstrate cognitive benefits; however, the huperzine A -induced hyperphosphorylation of the tau protein, 400 μg can improve the Alzheimer’s Disease Assessment the mechanism of which may be explained by its antioxi- Scale-cognitive [108]. Still, further studies are needed to dant activity and ability to regulate the tau phosphatase/ fully elucidate the efficiency of huperzine A against AD. kinase system [116]. Furthermore, luteolin can decrease the expression of amyloid precursor protein and reduced Rosmarinic acid the formation of β-amyloids [117]. Luteolin can also also inhibit apoptosis by decreasing intracellular ROS genera- tion; improving antioxidant endogenous system; such as augmenting SOD, CAT and GPx activities; and activating the NRF2 pathway [118]. Luteolin has also been shown to have the ability to relieve the cognitive dysfunction, improve significantly antioxidant system, decrease the lipid peroxide production, and inhibit inflammatory reaction in the brain tissue of rats induced by chronic Rosmarinic acid is a polyphenol-type carboxylic acid cerebral hypoperfusion [119]. In another study, luteolin existed in many speies [109]. Rosmarinic acid also improved the cognition function and memory on a has many pharmacological activities, such as antioxidant, streptozotocin-induced AD rat model [120]. Despite these antibacterial, anti-inflammatory, anticancer, antiviral, studies, further clinical trial data are needed to confirm and neuroprotective effects [110]. Rosmarinic acid may the protective effects of luteolin against AD. significantly prevent β-amyloid-induced memory loss, the mechanism of which may be attributed to its capacity to inhibit NF-κB and TNF-α expressions [111]. Rosmarinic acid has also been shown to protect neuronal PC12 cells Conclusions to avoid beta-amyloid-induced cytotoxicity. Further- more, it may decrease the hyperphosphorylation of the Many valuable medicinal plants can be applied to reduce tau protein. Inhibiting apoptotic pathways by rosmarinic dementia and treat AD. The main chemical compounds, acid may be explained due to its capacity to inhibit ROS such as flavonoids and alkaloids, have been shown formation, caspase-3 activation, and DNA fragmentation to have strong effects against AD. Therefore, studying 420 Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease medicinal plants in depth and finding new active com- 16. Grundman M, Delaney P. Antioxidant strategies for Alzheimer’s pounds against AD is necessary. disease. Proc Nutr Soc 2002;61:191–202. 17. Staehelin HB. Micronutrients and Alzheimer’s disease. Proc Nutr Soc 2005;64:565v70. Author contributions: All the authors have accepted 18. Lu P, Mamiya T, Lu L, Mouri A, Zou L, Nagai T, et al. Silibinin responsibility for the entire content of this submitted prevents amyloid β peptide-induced memory impairment and manuscript and approved its submission. oxidative stress in mice. Br J Pharmacol 2009;157:1270–7. Research funding: None declared. 19. Hardy J, Allsop D. Amyloid deposition as the central event in Employment or leadership: None declared. the aetiology of Alzheimer’s disease. Trends Pharmacol Sci 1991;12:383–8. Honorarium: None declared. 20. Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Competing interests: The funding organization(s) played Physiol Rev 2001;81:741–66. no role in the study design; in the collection, analysis, and 21. Nikolaev A, McLaughlin T, O’Leary DD, Tessier-Lavigne M. APP interpretation of data; in the writing of the report; or in the binds DR6 to trigger axon pruning and neuron death via distinct decision to submit the report for publication. caspases. Nature 2009;457:981–9. 22. West MJ, Coleman PD, Flood DG, Troncoso JC. Differences in the pattern of hippocampal neuronal loss in normal ageing and Alzheimer’s disease. Lancet 1994;344:769–72. 23. Martín S, González-Burgos E, Carretero ME, Gómez-Serranillos References MP. Neuroprotective properties of Spanish red wine and its isolated polyphenols on astrocytes. Food Chem 2011;128:40–8. 1. Blennow K, de Leon MJ, Zetterberg H. Alzheimer’s disease. 24. Moreira PI, Honda K, Liu Q, Aliev G, Oliveira CR, Santos MS, et al. Lancet 2006;368:387–403. Alzheimer’s disease and oxidative stress: the old problem remains 2. Zhang L, Yu H, Zhao X, Lin X, Tan C, Cao G, et al. Neuroprotective unsolved. Curr Med Chem-Cent Nerv Syst Agents 2005;5:51–62. effects of salidroside against beta-amyloid-induced oxidative 25. Bush AI. Drug development based on the metals hypothesis of stress in SH-SY5Y human neuroblastoma cells. Neurochem Int Alzheimer’s disease. J Alzheimer’s Dis 2008;15:223–40. 2010;57:547–55. 26. Faux NG, Ritchie CW, Gunn A, Rembach A, Tsatsanis A, Bedo J, 3. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s et al. PBT2 rapidly improves cognition in Alzheimer’s Disease: disease: progress and problems on the road to therapeutics. additional phase II analyses. J Alzheimer’s Dis 2010;20:509–16. Science 2002;297:353–6. 27. Cacquevel M, Lebeurrier N, Cheenne S, Vivien D. Cytokines in 4. Chun W, Johnson G. The role of tau phosphorylation and cleav- neuroinflammation and Alzheimer’s disease. Curr Drug Targets age in neuronal cell death. Front Biosci 2006;12:733–56. 2004;5:529–34. 5. Marchbanks R. Biochemistry of Alzheimer’s dementia. J Neuro- 28. Moore AH, O’Banion MK. Neuroinflammation and anti-­ chem 1982;39:9–15. inflammatory therapy for Alzheimer’s disease. Adv Drug Deliv 6. Francis PT, Palmer AM, Snape M, Wilcock GK. The cholinergic Rev 2002;54:1627–56. hypothesis of Alzheimer’s disease: a review of progress. J Neurol 29. Gasparini L, Ongini E, Wenk G. Non-steroidal anti-inflammatory Neurosurg Psychiatry 1999;66:137–47. drugs (NSAIDs) in Alzheimer’s disease: old and new mecha- 7. Pappas BA, Bayley PJ, Bui BK, Hansen LA, Thal LJ. acetyl- nisms of action. J Neurochem 2004;91:521–36. transferase activity and cognitive domain scores of Alzheimer’s 30. Breitner JC. Inflammatory processes and antiinflammatory drugs patients. Neurobiol Aging 2000;21:11–7. in Alzheimer’s disease: a current appraisal. Neurobiol Aging 8. McGleenon B, Dynan K, Passmore A. ­Acetylcholinesterase 1996;17:789–94. inhibitors in Alzheimer’s disease. Br J Clin Pharmacol 31. Miguel-Álvarez M, Santos-Lozano A, Sanchis-Gomar F, Fiuza- 1999;48:471–80. Luces C, Pareja-Galeano H, Garatachea N, et al. Non-steroidal 9. Upadhyaya P, Seth V, Ahmad M. Therapy of Alzheimer’s disease: anti-inflammatory drugs as a treatment for Alzheimer’s disease: an update. Afr J Pharm Pharmacol 2010;4:408–21. a systematic review and meta-analysis of treatment effect. Drugs 10. Watkins PB, Zimmerman HJ, Knapp MJ, Gracon SI, Lewis KW. Aging 2015;32:139–47. Hepatotoxic effects of tacrine administration in patients with 32. Howes MJR, Perry NS, Houghton PJ. Plants with traditional uses Alzheimer’s disease. J Am Med Assoc 1994;271:992–8. and activities, relevant to the management of Alzheimer’s dis- 11. Nunomura A, Castellani RJ, Zhu X, Moreira PI, Perry G, Smith MA. ease and other cognitive disorders. Phytother Res 2003;17:1–18. Involvement of oxidative stress in Alzheimer disease. J Neuro- 33. Ansari N, Khodagholi F. Natural products as promising drug pathol Exp Neurol 2006;65:631–41. candidates for the treatment of Alzheimer’s disease: molecular 12. Harman D. The aging process. Proc Natl Acad Sci 1981;78:7124–8. mechanism aspect. Curr Neuropharmacol 2013;11:414–29. 13. Jung HA, Min B-S, Yokozawa T, Lee J-H, Kim YS, Choi JS. Anti-­ 34. Olajide OJ, Yawson EO, Gbadamosi IT, Arogundade TT, Lambe Alzheimer and antioxidant activities of Coptidis Rhizoma E, Obasi K, et al. Ascorbic acid ameliorates behavioural deficits ­alkaloids. Biol Pharm Bull 2009;32:1433–8. and neuropathological alterations in rat model of Alzheimer’s 14. Yu BP. Cellular defenses against damage from reactive oxygen disease. Environ Toxicol Pharmacol 2017;50:200–11. species. Physiol Rev 1994;74:139–62. 35. D’Onofrio G, Sancarlo D, Ruan Q, Yu Z, Panza F, Daniele A, et al. 15. Lovell MA, Markesbery WR. Oxidative DNA damage in mild cog- Phytochemicals in the treatment of Alzheimer’s disease: a sys- nitive impairment and late-stage Alzheimer’s disease. Nucleic tematic review. Curr Drug Targets 2016;17. DOI: 10.2174/138945 Acids Res 2007;35:7497–504. 0117666161102121553. Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease 421

36. Ataie A, Shadifar M, Ataee R. Polyphenolic antioxidants and monnieri in Alzheimer’s disease model. J Ethnopharmacol neuronal regeneration. Basic Clin Neurosci 2016;7:81–90. 2010;127:26–31. 37. Aggarwal BB, Sundaram C, Malani N, Ichikawa H. Curcumin: the 55. Bhattacharya S, Bhattacharya A, Kumar A, Ghosal S. Antioxidant Indian solid gold. The molecular targets and therapeutic uses activity of Bacopa monniera in rat frontal cortex, striatum and of curcumin in health and disease. USA: Springer Science & hippocampus. Phytother Res 2000;14:174–9. ­Business Media, 2007:1–75. 56. Limpeanchob N, Jaipan S, Rattanakaruna S, Phrompittayarat 38. Begum AN, Jones MR, Lim GP, Morihara T, Kim P, Heath DD, et al. W, Ingkaninan K. Neuroprotective effect of Bacopa monnieri Curcumin structure-function, bioavailability, and efficacy in on beta-amyloid-induced cell death in primary cortical culture. models of neuroinflammation and Alzheimer’s disease. J Pharm J Ethnopharmacol 2008;120:112–7. Exp Ther 2008;326:196–208. 57. Sadhu A, Upadhyay P, Agrawal A, Ilango K, Karmakar D, 39. Carvalho AC, Gomes AC, Pereira-Wilson C, Lima CF. Chapter 35 – Singh GP, et al. Management of cognitive determinants in mechanisms of action of curcumin on aging: nutritional and senile dementia of Alzheimer’s type: therapeutic ­potential pharmacological applications A2 – Malavolta, Marco. In: Moc- of a novel polyherbal drug product. Clin Drug Investig chegiani E, editor. Molecular basis of nutrition and aging. San 2014;34:857–69. Diego: Academic Press, 2016:491–511. 58. Sethiya NK, Nahata A, Mishra SH, Dixit VK. An update on 40. Jiang S, Han J, Li T, Xin Z, Ma Z, Di W, et al. Curcumin as a poten- Shankhpushpi, a cognition-boosting Ayurvedic medicine. Zhong tial protective compound against cardiac diseases. Pharmacol Xi Yi Jie He Xue Bao 2009;7:1001–22. Res 2017;119:373–83. 59. Malik J, Karan M, Vasisht K. Nootropic, anxiolytic and CNS- 41. Hamaguchi T, Ono K, Yamada M. Review: curcumin and Alzhei- depressant studies on different plant sources of shankhpushpi. mer’s disease. CNS Neurosci Ther 2010;16:285–97. Pharm Biol 2011;49:1234–42. 42. Veldman ER, Jia Z, Halldin C, Svedberg MM. Amyloid binding 60. Nahata A, Patil U, Dixit V. Effect of Convulvulus pluricaulis properties of curcumin analogues in Alzheimer’s disease post- Choisy. on learning behaviour and memory enhancement activ- mortem brain tissue. Neurosci Lett 2016;630:183–8. ity in rodents. Nat Prod Res 2008;22:1472–82. 43. Motaghinejad M, Motevalian M, Fatima S, Hashemi H, 61. Bihaqi SW, Singh AP, Tiwari M. Supplementation of Convolvulus Gholami M. Curcumin confers neuroprotection against - pluricaulis attenuates scopolamine-induced increased tau and induced hippocampal neurodegeneration via CREB-BDNF Amyloid precursor protein (AβPP) expression in rat brain. Indian ­pathway in rats. Biomed Pharmacother 2017;87:721–40. J Pharmacol 2012;44:593. 44. Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry 62. Veerendra Kumar MH, Gupta YK. Effect of Centella asiatica on spice curcumin reduces oxidative damage and amyloid pathology cognition and oxidative stress in an intracerebroventricular in an Alzheimer transgenic mouse. J Neurosci 2001;21:8370–7. streptozotocin model of Alzheimer’s disease in rats. Clin Exp 45. Yang F, Lim GP, Begum AN, Ubeda OJ, Simmons MR, Ambegaokar Pharmacol Physiol 2003;30:336–42. SS, et al. Curcumin inhibits formation of amyloid beta oligomers 63. Chen C-L, Tsai W-H, Chen C-J, Pan T-M. Centella asiatica extract and fibrils, binds plaques, and reduces amyloid in vivo. J Biol protects against amyloid β 1–40-induced neurotoxicity in Chem 2005;280:5892–901. neuronal cells by activating the antioxidative defence system. 46. Butterfield D, Castegna A, Pocernich C, Drake J, Scapagnini J Tradit Complement Med 2016;6:362–9. G, Calabrese V. Nutritional approaches to ­combat oxida- 64. Dhanasekaran M, Holcomb LA, Hitt AR, Tharakan B, Porter JW, tive stress in Alzheimer’s disease. J Nutritional Biochem Young KA, et al. Centella asiatica extract selectively decreases 2002;13:444. amyloid beta levels in hippocampus of Alzheimer’s disease 47. Sahu PK. Design, structure activity relationship, cytotoxicity animal model. Phytother Res 2009;23:14–9. and evaluation of antioxidant activity of curcumin derivatives/ 65. Defeudis FV. Bilobalide and neuroprotection. Pharmacol Res analogues. Eur J Med Chem 2016;121:510–6. 2002;46:565–8. 48. DiSilvestro RA, Joseph E, Zhao S, Bomser J. Diverse effects of a 66. Zhou L-J, Zhu X-Z. Reactive oxygen species-induced apoptosis in low dose supplement of lipidated curcumin in healthy middle PC12 cells and protective effect of bilobalide. J Pharm Exp Ther aged people. Nutr J 2012;11:79. 2000;293:982–8. 49. Gohil K, Patel J. A review on Bacopa monniera: current research 67. Tchantchou F, Lacor PN, Cao Z, Lao L, Hou Y, Cui C, et al. Stimula- and future prospects. Int J Green Pharm 2010;4:1. tion of neurogenesis and synaptogenesis by bilobalide and 50. Russo A, Borrelli F. Bacopa monniera, a reputed nootropic plant: quercetin via common final pathway in hippocampal neurons. an overview. Phytomedicine 2005;12:305–17. J Alzheimers Dis 2009;18:787–98. 51. Dhanasekaran M, Tharakan B, Holcomb LA, Hitt AR, Young 68. Yakoot M, Salem A, Helmy S. Effect of Memo®, a natural formula KA, Manyam BV. Neuroprotective mechanisms of ayurvedic combination, on Mini-Mental State Examination scores in antidementia botanical Bacopa monniera. Phytother Res patients with mild cognitive impairment. Clin Interv Aging 2007;21:965–9. 2013;8:975–81. 52. MS Bharath M. Exploring the role of “Brahmi” (Bocopa monnieri 69. Ali BH, Blunden G, Tanira MO, Nemmar A. Some phytochemical, and Centella asiatica) in brain function and therapy. Recent Pat pharmacological and toxicological properties of ginger (Zingiber Endocr Metab Immune Drug Discov 2011;5:33–49. officinale Roscoe): a review of recent research. Food Chem Toxi- 53. Singh H, Dhawan B. Effect of Bacopa monniera Linn. (Brāhmi) col 2008;46:409–20. extract on avoidance responses in rat. J Ethnopharmacol 70. Oboh G, Ademiluyi AO, Akinyemi AJ. Inhibition of acetylcho- 1982;5:205–14. linesterase activities and some pro-oxidant induced lipid peroxi- 54. Uabundit N, Wattanathorn J, Mucimapura S, Ingkaninan K. dation in rat brain by two varieties of ginger (Zingiber officinale). Cognitive enhancement and neuroprotective effects of Bacopa Exp Toxicol Pathol 2012;64:315–9. 422 Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease

71. Essa MM, Vijayan RK, Castellano-Gonzalez G, Memon MA, Braidy 88. Kulkarni S, Dhir A. Berberine: a plant alkaloid with therapeutic N, Guillemin GJ. Neuroprotective effect of natural products potential for central nervous system disorders. Phytother Res against Alzheimer’s disease. Neurochem Res 2012;37:1829–42. 2010;24:317–24. 72. Borek C. Antioxidant health effects of aged garlic extract. J Nutr 89. Durairajan SS, Liu L-F, Lu J-H, Chen L-L, Yuan Q, Chung SK, et al. 2001;131:1010S–5S. Berberine ameliorates β-amyloid pathology, gliosis, and cogni- 73. Borek C. Garlic reduces dementia and heart-disease risk. J Nutr tive impairment in an Alzheimer’s disease transgenic mouse 2006;136:810S–2S. model. Neurobiol Aging 2012;33:2903–19. 74. Nillert N, Pannangrong W, Welbat JU, Chaijaroonkhanarak W, 90. Zhu F, Qian C. Berberine chloride can ameliorate the spatial Sripanidkulchai K, Sripanidkulchai B. Neuroprotective effects of memory impairment and increase the expression of interleu- aged garlic extract on cognitive dysfunction and neuroinflamma- kin-1beta and inducible nitric oxide synthase in the rat model tion induced by β-amyloid in rats. Nutrients 2017;9:24. of Alzheimer’s disease. BMC Neurosci 2006;7:78. 75. Qu Z, Mossine VV, Cui J, Sun GY, Gu Z. Protective effects of AGE 91. Jia L, Liu J, Song Z, Pan X, Chen L, Cui X, et al. Berberine sup- and its components on neuroinflammation and neurodegenera- presses amyloid-beta-induced inflammatory response in micro- tion. Neuromol Med 2016;18:474–82. glia by inhibiting nuclear factor-kappaB and mitogen-activated 76. Ossola B, Kääriäinen TM, Männistö PT. The multiple faces protein kinase signalling pathways. J Pharm Pharmacol of quercetin in neuroprotection. Expert Opin Drug Saf 2012;64:1510–21. 2009;8:397–409. 92. Huang M, Jiang X, Liang Y, Liu Q, Chen S, Guo Y. Berberine 77. Russo M, Spagnuolo C, Tedesco I, Bilotto S, Russo GL. The improves cognitive impairment by promoting autophagic flavonoid quercetin in disease prevention and therapy: facts and clearance and inhibiting production of β-amyloid in APP/ fancies. Biochem Pharmacol 2012;83:6–15. tau/PS1 mouse model of Alzheimer’s disease. Exp Gerontol 78. Bischoff SC. Quercetin: potentials in the prevention and therapy 2017;91:25–33. of disease. Curr Opin Clin Nutr Metab Care 2008;11:733–40. 93. de Oliveira JS, Abdalla FH, Dornelles GL, Adefegha SA, Palma 79. Jiménez-Aliaga K, Bermejo-Bescós P, Benedí J, Martín-Aragón TV, Signor C, et al. Berberine protects against memory S. Quercetin and rutin exhibit antiamyloidogenic and fibril- impairment and anxiogenic-like behavior in rats submitted to disaggregating effects in vitro and potent antioxidant activity in sporadic Alzheimer’s-like dementia: Involvement of acetylcho- APPswe cells. Life Sci 2011;89:939–45. linesterase and cell death. NeuroToxicol 2016;57:241–50. 80. Ansari MA, Abdul HM, Joshi G, Opii WO, Butterfield DA. 94. Bhat KP, Kosmeder JW, Pezzuto JM. Biological effects of res- Protective effect of quercetin in primary neurons against Aβ veratrol. Antioxid Redox Signal 2001;3:1041–64. (1–42): relevance to Alzheimer’s disease. J Nutr Biochem 95. Li F, Gong Q, Dong H, Shi J. Resveratrol, a neuroprotec- 2009;20:269–75. tive ­supplement for Alzheimer’s disease. Curr Pharm Des 81. Puerta E, Suárez-Santiago JE, Santos-Magalhães NS, Ramirez 2012;18:27–33. MJ, Irache JM. Effect of the oral administration of nanoencapsu- 96. Kumar A, Naidu P, Seghal N, Padi S. Neuroprotective effects of lated quercetin on a mouse model of Alzheimer’s disease. Int J resveratrol against intracerebroventricular colchicine-induced Pharm 2017;517:50–7. cognitive impairment and oxidative stress in rats. Pharmacol- 82. Ahmad N, Feyes DK, Agarwal R, Mukhtar H, Nieminen A-L. Green ogy 2006;79:17–26. tea constituent epigallocatechin-3-gallate and induction of 97. Luo L, Huang YM. [Effect of resveratrol on the cognitive ability apoptosis and cell cycle arrest in human carcinoma cells. J Natl of Alzheimeros mice]. Zhong Nan Da Xue Xue Bao. Yi Xue Ban Cancer Ins 1997;89:1881–6. J Centr South University. Med Sci 2006;31:566–9. 83. Biasibetti R, Tramontina AC, Costa AP, Dutra MF, Quincozes-­ 98. Turner RS, Thomas RG, Craft S, Van Dyck CH, Mintzer J, Santos A, Nardin P, et al. Green tea (−) epigallocatechin-3-gal- Reynolds BA, et al. A randomized, double-blind, placebo- late reverses oxidative stress and reduces acetylcholinesterase controlled trial of resveratrol for Alzheimer disease. Neurology activity in a streptozotocin-induced model of dementia. Behav 2015;85:1383–91. Brain Res 2013;236:186–93. 99. He X, Li Z, Rizak JD, Wu S, Wang Z, He R, et al. Resveratrol 84. Lee JW, Lee YK, Ban JO, Ha TY, Yun YP, Han SB, et al. Green attenuates formaldehyde induced hyperphosphorylation tea (-)-epigallocatechin-3-gallate inhibits β-amyloid-induced of tau protein and cytotoxicity in N2a cells. Front Neurosci cognitive dysfunction through modification of secretase activ- 2017;10:598. ity via inhibition of ERK and NF-κB pathways in mice. J Nutr 100. Karthick C, Periyasamy S, Jayachandran KS, Anusuyadevi M. 2009;139:1987–93. Intrahippocampal administration of induced cho- 85. Li R, Huang YG, Fang D, Le WD. ( −)-Epigallocatechin gallate linergic dysfunction via NR2A/NR2B expression: implications inhibits lipopolysaccharide-induced microglial activation and of resveratrol against Alzheimer disease pathophysiology. protects against inflammation-mediated dopaminergic neuronal Front Mol Neurosci 2016;9:28. injury. J Neurosci Res 2004;78:723–31. 101. Moussa C, Hebron M, Huang X, Ahn J, Rissman RA, Aisen PS, 86. Lee Y-J, Choi D-Y, Yun Y-P, Han SB, Oh K-W, Hong JT. Epigallocate- et al. Resveratrol regulates neuro-inflammation and induces chin-3-gallate prevents systemic inflammation-induced memory adaptive immunity in Alzheimer’s disease. J Neuroinflamm deficiency and amyloidogenesis via its anti-neuroinflammatory 2017;14:1. properties. J Nutr Biochem 2013;24:298–310. 102. Ha GT, Wong RK, Zhang Y. Huperzine a as potential treatment of 87. Chang X, Rong C, Chen Y, Yang C, Hu Q, Mo Y, et al. Alzheimer’s disease: an assessment on chemistry, pharmacol- ­(−)-Epigallocatechin-3-gallate attenuates cognitive deterioration ogy, and clinical studies. Chem Biodivers 2011;8:1189–204. in Alzheimer’s disease model mice by upregulating neprilysin 103. Ratia M, Gimenez-Llort L, Camps P, Munoz-Torrero D, Perez B, expression. Exp Cell Res 2015;334:136–45. Clos M, et al. and huperzine A improve cognition and Bui and Nguyen: Natural product for the treatment of Alzheimer’s disease 423

regulate some neurochemical processes related with Alzhei- PC12 cells from amyloid-β peptide-induced neurotoxicity. mer’s disease in triple transgenic mice (3xTg-AD). Neurode- J Pharm Exp Ther 2006;317:1143–9. gener Dis 2012;11:129–40. 113. Gok DK, Ozturk N, Er H, Aslan M, Demir N, Derin N, et al. Effects 104. Zhang HY, Tang XC. Neuroprotective effects of huperzine A: of rosmarinic acid on cognitive and biochemical alterations in new therapeutic targets for neurodegenerative disease. Trends ovariectomized rats treated with D-galactose. Folia Histochem Pharmacol Sci 2006;27:619–25. Cyto 2015;53:283–93. 105. Wang Y, Tang XC, Zhang HY. Huperzine A alleviates synaptic 114. Lopez-Lazaro M. Distribution and biological activities of the deficits and modulates amyloidogenic and nonamyloidogenic flavonoid luteolin. Mini Rev Med Chem 2009;9:31–59. pathways in APPswe/PS1dE9 transgenic mice. J Neurosci Res 115. Seelinger G, Merfort I, Schempp CM. Anti-oxidant, anti- 2012;90:508–17. inflammatory and anti-allergic activities of luteolin. Planta Med 106. Wang C-Y, Zheng W, Wang T, Xie J-W, Wang S-L, Zhao B-L, 2008;74:1667–77. et al. Huperzine A activates Wnt/β-catenin signaling and 116. Zhou F, Chen S, Xiong J, Li Y, Qu L. Luteolin reduces zinc- enhances the nonamyloidogenic pathway in an Alzhei- induced tau phosphorylation at Ser262/356 in an ROS- mer transgenic mouse model. Neuropsychopharmacology dependent manner in SH-SY5Y cells. Biol Trace Elem Res 2011;36:1073–89. 2012;149:273–9. 107. Gordon RK, Nigam SV, Weitz JA, Dave JR, Doctor BP, Ved HS. 117. Liu R, Meng F, Zhang L, Liu A, Qin H, Lan X, et al. Luteolin The NMDA receptor ion channel: a site for binding of isolated from the medicinal plant Elsholtzia rugulosa (Labia- ­Huperzine A. J Appl Toxicol 2001;21(S1):S47–51. tae) prevents copper-mediated toxicity in β-amyloid precur- 108. Rafii M, Walsh S, Little JT, Behan K, Reynolds B, Ward C, et al. sor protein Swedish mutation overexpressing SH-SY5Y cells. A phase II trial of huperzine A in mild to moderate Alzheimer Molecules 2011;16:2084–96. disease. Neurology 2011;76:1389–94. 118. Hwang Y-J, Lee E-J, Kim H-R, Hwang K-A. Molecular mecha- 109. Shekarchi M, Hajimehdipoor H, Saeidnia S, Gohari AR, Hamed- nisms of luteolin-7-O-glucoside-induced growth inhibition ani MP. Comparative study of rosmarinic acid content in some on human liver cancer cells: G2/M cell cycle arrest and plants of Labiatae family. Pharmacogn Mag 2012;8:37–41. caspase-­independent apoptotic signaling pathways. BMB Rep 110. Petersen M, Simmonds MS. Rosmarinic acid. Phytochemistry 2013;46:611–6. 2003;62:121–5. 119. Fu X, Zhang J, Guo L, Xu Y, Sun L, Wang S, et al. Protective role 111. Alkam T, Nitta A, Mizoguchi H, Itoh A, Nabeshima T. A natural of luteolin against cognitive dysfunction induced by chronic scavenger of peroxynitrites, rosmarinic acid, protects against cerebral hypoperfusion in rats. Pharmacol Biochem Behav impairment of memory induced by Aβ 25–35. Behav Brain Res 2014;126:122–30. 2007;180:139–45. 120. Wang H, Wang H, Cheng H, Che Z. Ameliorating effect of luteo- 112. Iuvone T, De Filippis D, Esposito G, D’Amico A, Izzo AA. The lin on memory impairment in an Alzheimer’s disease model. spice sage and its active ingredient rosmarinic acid protect Mol Med Rep 2016;13:4215–20.