Next Article in Journal
Biomarker Signatures of Two Phenotypical Prefrailty Types in the Irish Longitudinal Study on Ageing
Previous Article in Journal
Feasibility of a Small Group Otago Exercise Program for Older Adults Living with Dementia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Multifaceted Role of Neuroprotective Plants in Alzheimer’s Disease Treatment

Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA
*
Author to whom correspondence should be addressed.
Submission received: 4 February 2022 / Revised: 23 February 2022 / Accepted: 25 February 2022 / Published: 26 February 2022
(This article belongs to the Section Geriatric Neurology)

Abstract

:
Alzheimer’s disease (AD) is an age-related, progressive neurodegenerative disorder characterized by impaired cognition, memory loss, and altered personality. Many of the available pharmaceutical treatments do not alter the onset of disease progression. Recently, alternatives to developed drug candidates have been explored including medicinal plants and herbal treatments for the treatment of AD. This article examines the role of herbal plant extracts and the neuroprotective effects as alternative modes of intervention for AD progression. These extracts contain key metabolites that culminate alterations in AD progression. The traditional plant extracts explored in this article induce a variety of beneficial properties, including antioxidants, anti-inflammatory, and enhanced cognition, while also inducing activity on AD drug targets such as Aβ degradation. While these neuroprotective aspects for AD are relatively recent, there is great potential in the drug discovery aspect of these plant extracts for future use in AD treatment.

1. Introduction

Alzheimer’s disease (AD) is a heterogeneous neurodegenerative disorder currently affecting more than 6 million individuals in the United States and approximately 50 million individuals worldwide [1]. Furthermore, as a neurodegenerative disorder, AD irreversibly and gradually hinders cognition, memory, and the ability to carry out activities of daily living [2]. Although the etiology of AD has not been completely elucidated, there are various potentially causative factors that have been identified. In general, genetic factors have been associated with 5–10% of familial AD, with the remaining 90–95% being sporadic [3]. Furthermore, apolipoprotein E4 (ApoE4) homozygosity or heterozygosity has also been determined to significantly elevate the risk of AD development [4]. Nonetheless, age is considered the most critical risk factor for AD, with approximately 3% of individuals in the 65–74 age range and 17% of individuals in the 75–84 age range suffering from this disease [5,6,7]. Scientific efforts to discover a cure for AD have been expensive and disappointing, with current drugs mainly addressing the symptoms of the disorder and having limited effectiveness once the disease progresses in severity [8,9]. As such, despite the high prevalence of AD, only five drugs, consisting of galantamine, rivastigmine, donepezil, memantine, and a combination of donepezil and memantine, have been previously FDA-approved for its treatment [10]. Nonetheless, as of now, a new drug, aducanumab, has obtained accelerated approval [11].
The pathology of AD manifests with degeneration of the neurons, along with loss of synapses in the cortex, the hippocampus, and the subcortical structures [12]. In turn, this leads to the gross atrophy of the affected regions, yielding memory loss, inability to learn novel information, executive dysfunction, and an incapacity to fulfill daily living activities [13]. Furthermore, the pathological hallmarks of AD consist of neurofibrillary tangles and neuritic plaques, which are associated with cytoskeletal changes attributed to the hyperphosphorylation of microtubule-associated tau and the accumulation of Amyloid-beta in the brain [14]. The accumulation of Aβ has been suggested to prompt neurodegeneration, thereby leading to the clinical dementia often witnessed in AD patients [15].
In traditional medicine, herbal remedies have often been utilized for health promotion and treatment [16]. However, there has been minimal scientific attention in the usage of these traditional medications. The bioactive components present in traditional herbs help alleviate a variety of AD symptoms including poor cognition and memory loss. These components provide neuroprotection and improved memory through their mechanisms of action [17]. This review provides an overview of herb-derived therapeutics for AD that relieve the symptoms.

2. AD Pathological Mechanisms

AD is a progressive age-related neurodegenerative disorder that is responsible for most cases of dementia [5]. Besides the environmental and genetic factors, which are presupposed to contribute to AD etiology, there are numerous hypotheses which have been elucidated to attempt to explicate AD. The most prevalent hypotheses are the Aβ cascade hypothesis, the inflammation hypothesis, the cholinergic hypothesis, the tau hypothesis, and the oxidative hypothesis [18,19,20]. The Aβ cascade hypothesis implicates Aβ peptides as the AD causative agent due to the extracellular deposition of the Aβ peptides as senile plaques, with the neurofibrillary tangles resulting in neuronal loss, dementia, and vascular damage [21]. Neurofribrillary tangles are also considered an AD hallmark and principally consist of tau, which is a microtubule-associated scaffold protein [22]. The aggregation of tau impairs the axons, leading to neurodegeneration [23]. Currently, the inflammation hypothesis has gained prominence as one of the major AD pathological factors, encompassing the immune response sustained in the brain [24]. Continuous activation of the brain’s immune cells, such as microglia, leads to the release and production of various proinflammatory cytokines, which aids tau and Aβ pathologies and leads to the loss of neurons [24]. Damage to the cholinergic neurons has also been accepted as a critical pathological chance associated with AD-related cognitive impairment [25]. As such, the cholinergic hypothesis proposes that cholinergic neuronal dysfunction in the brain could substantially contribute to the AD-related cognitive decline [25]. In general, this hypothesis is supported by the usage of cholinesterase inhibitors in the treatment of AD [26]. Furthermore, oxidative stress has also been determined to have a crucial role in AD pathogenesis [27]. In fact, a vast amount of evidence indicates that AD is perpetually accompanied by elevated oxidative cellular stress in the brain, yielded by the elevated production of free radicals, reduced polyunsaturated fatty acid, elevated protein and DNA oxidation, elevated lipid peroxidation, and the aggregation and accumulation of Aβ, which cause oxidative stress [28].

3. Herbal Neuroprotective Strategies

Currently, there is a vast amount of evidence suggesting that the primary pathological causative factor of AD is Aβ accumulation [29]. As such, reducing the amount of Aβ has been a major target in the development of AD therapeutics [30,31,32]. Nonetheless, effective therapeutic regimens for AD may necessitate the utilization of numerous neuroprotective agents [33,34]. As of now, various molecular targets have been recognized as mediating pathophysiological processes [35]. The identification of these targets could potentially allow the development of high-yield neuroprotective techniques for AD treatment [35]. Potential neuroprotective mechanisms fixate on inhibiting deleterious intraneuronal pathways triggered by toxic stimuli and Aβ via the interaction with neuronal targets [36]. Popular neuroprotective strategies for AD management consist of discovering small molecules with the ability to block these Aβ interactions with intracellular and extracellular targets to avoid caspase activation and the expression of proapoptotic proteins, to reduce the stress kinase signaling cascades, to inhibit tau protein phosphorylation, to counterpoise cholinergic functional loss, and to boost neuronal plasticity while blocking excitotoxicity [37,38,39,40,41]. In general, natural products are advantageous because some have the ability to exhibit their neuroprotective effects through a plethora of differing approaches. As such, herbal medicine for AD could be multidimensional when the herb being utilized has various effective bioactive compounds [42]. Additionally, the synergistic action of herbal extracts and mixtures also has the potential to eliminate severe adverse effects correlated with the utilization of a single xenobiotic product, allowing for a broader spectrum of activity and reducing the chances of pathogenic resistance [43].

4. Herbal Neuroprotective Effects

Numerous natural bioactive compounds have been demonstrated to have neuroprotective roles via antioxidation, antineuroinflammatory, anti- Aβ and tau aggregation, as well as targeting cholinergic neurotransmission [44,45,46]. As of now, it is reasonable to presume that AD onset and progression could be delayed or slowed down via the utilization of neuroprotective natural products that elicit their effects by affecting various pathological targets [47]. Moreover, herbal administration and combinations for medical usage has been widely utilized in traditional Chinese medicine, Native American medicine, and Ayurveda [48,49]. For AD, a wide variety of plant extracts has been utilized and tested in clinical studies (Table 1).
Withania somnifera, common name ashwagandha, is an evergreen shrub in the Solanaceae family [78,79]. W. somnifera is one of the most conspicuous herbs prescribed for AD [80,81]. In general, it is prescribed as a nerve tonic and energy booster [81]. W. somnifera, as an adaptogen, has been demonstrated to possess free radical scavenging and antioxidant and immune-boosting activities [78]. W. somnifera contains a plethora of bioactive compounds of medical interest, such as withanolides A-Y, withanone, withasomniferols A-C, dehydrowithanolide-R, withasomidienone, and other ergostane-type steroidal lactones [82,83]. The plant also includes alkaloids, beta-sitosterol, and phytosterols sitoindosides VII-X [83]. Some of these constituents have been demonstrated to scavenge free radicals produced during AD pathological progression [83,84]. Molecular modeling research studies have elucidated that withanamides A and C have the ability to bind to the active motif of Abeta-25-35 and avert the formation of fibrils [85]. Subsequently, these compounds have been shown to protect neuronal rat cells and PC-12 cells from Beta-amyloid-induced neuronal death [51,86]. Consequently, therapeutic treatments that consist of W. somnifera’s methanol extractions have been shown to trigger the outgrowth of neurites in a time- and dose-dependent approach in human neuroblastoma cells [87]. Another research study consisting of cultured cortical rat neurons displayed a diminishment of pre- and postsynaptic stimuli, as well as dendritic and axonal atrophy, when treated with Aβ peptides [88]. Consequent therapeutic treatment with withanolide A displayed significant regenerative properties of dendrites and axons, and even displayed restorative properties of the pre- and postsynapses in the cultured neurons [88].
Withanolide A has been shown to inhibit Abeta25-35-induced axonal and dendritic degeneration in the hippocampus and cerebral cortex, while also seemingly restoring synapses and Aβ -peptide-induced memory impairment in mice [89]. Additionally, the ameliorative outcomes were retained after treatment discontinuation [89]. Subsequently, aqueous extracts of W. somnifera elevated the activity of choline acetyl transferase and acetylcholine in rats, potentially explicating the memory- and cognition-enhancing effects [87,90]. Consequently, root extract treatments led to low-density lipoprotein receptor-related protein upregulation, thereby boosting Aβ clearance and ameliorating AD pathology in APP/PS1 mice [91]. Similarly, administering semipurified extracts of W. somnifera orally inhibited Aβ peptide accumulation and reversed behavioral impairment in APP/PS1 mice models of AD [91]. These ameliorative effects were mediated via the enhancement of liver low-density lipoprotein receptor-related protein. Consequently, Drosophila melanogaster models of AD have also elucidated that W. somnifera treatment could mitigate Abeta toxicity while seemingly boosting longevity [92]. Nonetheless, although a vast amount of literature has reported the ameliorative therapeutic effects of W. somnifera, clinical data related to its use for cognitive impairment are limited [93].
In a double-blind, randomized, placebo-controlled study encompassing 50 subjects with mild cognitive impairment (MCI), the subjects were treated with 300 mg of W. somnifera root extracts twice daily or with a placebo for eight continuous weeks [94]. At the end of the eight weeks, the W. somnifera-treated group showed significant enhancements in information-processing speed, attention span, and executive function [94]. As such, these two studies provide evidence for W. somnifera’s enhancing roles in memory and executive function in subjects with MCI [91,94].
Bacopa monnieri, common names waterhyssop or brahmi, is a perennial plant in the Plantaginaceae family [95]. B. monnieri is a nootropic herb with low toxicity and has been traditionally utilized as a memory booster and neural tonic in a plethora of ailments [95]. There have been studies that have elucidated evidence for the role of B. monnieri in epilepsy, dementia, and Parkinson’s disease attenuation [96,97]. Furthermore, it has also been utilized for stress, asthma, and insomnia [98,99]. The bioactive phytochemicals present in B. monnieri encompass sterols, polyphenols, sulfhydryl compounds, saponins, bacosides A and B, betulic acid, bacopasides II, IV, and V, and bacosaponins A, B, C, D, and E [100]. The neuroprotective activity of B. monnieri could potentially be due to the bioactivity of the phytochemicals, thereby explaining its usage in traditional medicine. In fact, research studies conducted in vitro and in vivo demonstrate that these phytochemicals contain free radical scavenging and antioxidant activities via the blockage of lipid peroxidation in various brain areas [57,101]. B. monnieri elicits its role via the reduction in divalent metals, reduction in lipid peroxide formation, inhibition of lipoxygenase, and through its scavenging activity of reactive oxygen species [102].
Various studies have seemingly elucidated the role of B. monnieri in terms of intellect and memory [103,104]. In order to illuminate the neuroprotective activities of B. monnieri in rat models of AD, researchers conducted studies examining the administration of 20, 40, and 80 mg/kg alcoholic extracts of B. monnieri on rats for 2 weeks prior to and 1 week after intracerebroventricular ethylcholine aziridinium ion administration [56]. The Morris water maze was utilized to test spatial memory, and histological assays were utilized to assess the density of cholinergic neurons [56]. In this case, the B. monnieri extract appeared to enhance the escape latency time in the Morris water maze and blocked the diminishment of cholinergic neuron density [56]. Another conducted study demonstrated the backtracking of colchine-induced cognitive impairments by B. monnieri extracts [105]. Similarly, B. monnieri extracts attenuated oxidative damage caused by colchicine via the reduction in the protein carbonyl content while also restoring antioxidant enzyme activity [105].
The majority of the research studies examining the cognitive enhancement elicited by B. monnieri in humans have been centered on normal geriatric individuals. In a randomized, double-blind, placebo-controlled clinical trial encompassing 35 subjects of 55 years and older, the subjects were administered a 125 mg dosage of B. monnieri extract or a placebo twice a day for an interval of 12 weeks, with a placebo period that consisted of an additional four weeks [58]. The researchers conducted numerous memory tests focused on logical memory, visual reproduction, paired-association learning, general information, orientation, digit forward, and digit backward subtests [68]. The subjects were then given a score on each subtest, with the total memory consisting of the result of all of the subtests [58]. Treatment with B. monnieri extract significantly improved paired-association learning, mental control, and logical memory in the subjects in comparison to the placebo groups at 8 and 12 weeks following trial initiation [58]. These results propose that B. monnieri extracts could be beneficial in age-associated memory deficit treatment.
In another study conducted by Dimpel et al., ten subjects were administered 500 mg of Sideritis scardica extract, 320 mg of B. monnieri extract, or a mixture via the utilization of a crossover design [106]. S. scardica extracts, which are abundant in a plethora of flavonoids, have been demonstrated to inhibit aggregation and toxicity of amyloid-beta in Caenorhabditis elegans models of AD [107]. Consequently, the d2-concentration test was utilized to characterize thinking, performance, tension with selective orientation of perception, and wakefulness of the subjects [106]. The results of the treatment group elucidate that S. scardica extract yielded improved d2-concentration tests scores when combined with low-dose B. monnieri extract. Likewise, B. monnieri alone exhibited enhanced effects after continuous treatment, proposing repetitive dosing of B. monnieri as a potential therapeutic alternative for subjects with MCI.
Additionally, another multicenter, prospective, and noncomparative clinical trial encompassing 104 MCI subjects and a duration of 60 days was carried out [108]. The subjects were administered B. monnieri in combination with vitamin E, phosphatidylserine, and astaxanthin [108]. Mnemonic and cognitive performances were tested with the clock-drawing test and the Alzheimer’s Disease Assessment Scale–Cognitive Subscale (ADAS-cog), which have the ability to assess the risk of MCI-to-AD advancement [108,109]. The researchers noticed significant enhancement in the clock-drawing test and the Alzheimer’s Disease Assessment Scale–Cognitive Subscale after 60 days when compared to the baseline of the subjects [108]. Although memory is greatly affected by a plethora of factors, such as hormones, cyclic AMP, neurotransmitters, focus, attention, synapse formation, nutrients, and protein transcription, a couple of these processes could potentially be modulated by B. monnieri in isolation or in combination with other compounds as partially shown in these studies [56,57,101,110,111].
Ginkgo biloba, common name gingko or maidenhair tree, is a tree species in the Ginkgoaceae family native to China [112]. The leaf extract of G. biloba has been prominently utilized for its ability to enhance memory and deter age-related impairments [113]. G. biloba contains terpenoids and flavonoids, which are considered the main pharmacologically active constituents [114]. The majority of the clinical trials conducted utilizing G. biloba extract included a combination of ginkgolic acids, terpene lactones, and flavonoid glycosides [115,116,117]. G. biloba extract has demonstrated ameliorative effects in AD, tinnitus, and cancer treatments [117,118,119]. The proposed mechanisms of G. biloba extract include Beta-amyloid peptide aggregation inhibitory effects, antioxidant activities, antiplatelet-activating factor activity, and reduced expression of the peripheral benzodiazepine receptor, which is helpful for stress relief [120,121,122].
G. biloba extract has been shown to reverse nitric oxide and Beta-amyloid toxicity in vitro while simultaneously decreasing apoptosis in vivo and in vitro [120,123,124]. Therapeutic treatment with G. biloba extract has also shown the ability to improve spatial learning and memory of aluminum-intoxicated rats, potentially due to a reduced APP expression and caspase-3 in the brains of treated-rats [125]. Modest cognitive function enhancement was noted in AD subjects in numerous randomized, placebo-controlled, double-blind clinical trials [126,127]. Furthermore, a meta-analysis was conducted by Hashiguchi et al. examining the effects of G. biloba treatment in nine studies (seven of which utilized the Syndrome Kurztest and ADAS-Cog) at doses of 120 mg with a 26-week duration for efficacy parameters [128]. Their meta-analysis showed that the standard mean differences in seven of the studies favored G. biloba treatment over the administered placebo, although two of the studies did not show statistically significant differences between G. biloba and placebo treatment in ADAS-Cog [128]. In the AD and vascular dementia subgroups, the standard mean differences in the Syndrome Kurztest favored G. biloba over placebo treatment, with 240 mg daily dosages having significantly enhanced standard mean differences in the Syndrome Kurztest [128].
Centella asiatica, common names Asiatic pennyworth, Indian pennyworth, Gotu kola, and kodavan, is a perennial plant in the Apiaceae family [129]. C. asiatica is native to Asian wetlands and has been utilized as a medicinal herb for cogniceutical and nutraceutical purposes [129]. C. asiatica is presupposed to heal skin, boost kidney and liver health, and strengthen the brain [130]. C. asiatica is also considered to be a rejuvenating plant for brain and nerve cells, since it is thought to augment intelligence and enhance memory [131]. In vitro research studies utilizing madasiatic acid, madecassoside, asiatic acid, and asiaticosides C. asiatica derivatives have elucidated that these compounds have the ability to block hydrogen peroxide and beta-amyloid-induced cellular death, as well as to reduce the concentration of free radicals, thereby proposing a possible role for C. asiatica in AD treatment and prevention [64,132,133].
Ethanolic extracts of C. asiatica have also elicited neurite outgrowths in SH-SY5Y human cells in the presence of the nerve growth factor and expedited regeneration of the axons in rats [63]. C. asiatica leaf extracts also demonstrated enhanced memory and learning in rats via the modulation of various neurotransmitters in rat brains, inclusive of noradrenaline, 5-hydroxytryptamine, and dopamine [134]. This advocated for the possible therapeutic role of C. asiatica in the therapeutic treatment of AD-associated cognitive decline.
Furthermore, by utilizing double-transgenic PS/APP mice, which spontaneously generate Aβ plaques, studies have demonstrated that treatment with 2.5 mg/kg of C. asiatica extract significantly reduced hippocampal Abeta1-40 and Abeta-1-42 levels [65]. Consequently, long-term treatment with higher dosages of aqueous C. asiatica extract eventuated in a significant decline in Congo red-positive amyloid fibrillar plaques [65]. Additionally, there was significant scavenging activity of reactive oxygen species with the lowest dosage utilized, which was 25 μg, and 83% of the reactive oxygen species were removed with a 250 μg dosage [65].
Various asiatic acid derivatives, as some of the most common phytochemicals found in C. asiatica, demonstrated significant cognitive-enhancing activities when utilizing a scopolamine-induced model of memory impairment [135]. Scopolamine was utilized because it yields transient memory deficiencies analogous to the early stages of AD [135]. Utilizing the Morris water maze test and passive avoidance, the study demonstrated that pretreatment with three differing asiatic acid derivatives significantly enhanced memory in comparison to the scopolamine-treated mice that were not given any derivatives [135]. The improvement in cognition witnessed in the treated mice was attributed to elevated choline acetyltransferase activity, yielding enhanced acetylcholine synthesis [135].
Subsequently, in a double-blind, randomized, placebo-controlled research study, C. asiatica extract was administered twice a day for two months to 28 healthy subjects at 250, 500, and 750 mg dosages [136]. Mood and cognitive performance were noted at baseline, after the first administration, after one month, and after two months post-treatment [136]. The experimental results elucidate that the highest dosage indeed enhanced working memory, and improvements in self-rated mood were also noted post-treatment, implying that C. asiatica could be helpful in mitigating age-associated cognitive impairment while also boosting mood in healthy geriatric subjects [136].
Crocus sativus, also known as autumn crocus or saffron crocus, is a flowing plant in the Iridaceae family [137]. It is prominent for producing the crimson-colored spice saffron from the filaments that grow within the flower [138]. Saffron is widely cultivated in various countries, such as Greece, Iran, and India [139]. Furthermore, the spice has a wide variety of applications in the cosmetic, textile, and medicinal industries [139,140]. Safranal, a carboxaldehyde, is a major constituent of saffron [68]. In vivo and in vitro research studies have demonstrated that the phytochemicals found in saffron contain antiamyloidogenic, antioxidant, and anti-inflammatory properties [67,68].
In order to examine saffron’s efficacy in therapeutic treatment of mild to moderate AD, a double-blind clinical study enrolled 46 subjects that were randomly assigned to obtain 30 mg/day of saffron or a placebo [141]. After 16 weeks, saffron significantly improved subject outcomes in cognitive performance compared to the placebo when observing their Alzheimer’s Disease Assessment Scale–Cognitive Subscale scores [141]. Additionally, there were no significant differences in the treatment or placebo-controlled groups in terms of adverse effects reported [141]. Subsequent to this study, researchers distinguished donepezil, the cholinesterase inhibitor, with a saffron extract, in subjects suffering mild to moderate AD [69]. In a double-blind, randomized, controlled clinical trial with a duration of 22 weeks, 54 Persian-speaking subjects were randomly dispensed 10 mg/day of donepezil or 30 mg/day of saffron [69]. After the conclusion of the study, it was determined that saffron had similar effects in enhancing cognitive function and less side effects in comparison to donepezil [69]. The researchers proposed that saffron’s ability to treat mild to moderate AD could potentially be due to its inhibitory activity towards deposition and aggregation of beta-amyloid plaques [69].
A pilot study examining efficacy and safety compared memantine with saffron extract in terms of cognitive defect reductions [142]. A total of 68 subjects with moderate to severe AD were recruited and enrolled in a double-blind, randomized, parallel-group research study [142]. The subjects were administered 20 mg/day of memantine or 30 mg/day of saffron extract capsules for 12 months [142]. The saffron extract was comparable with memantine in decreasing cognitive decline and had a seemingly low adverse effect rate in subjects with moderate to severe AD [142]. Due to these studies, saffron could be considered an herbal spice with the ability to enhance the completing activities of daily living as well as boosting cognitive functions in subjects with MCI and AD [69,141,142]. Subsequently, the studies demonstrated no severe or prominent adverse effects, making saffron a suitable natural therapeutic for these patients.
Curcuma longa, or turmeric, is a flowering plant native to Southeast Asia and the Indian subcontinent in the Zingiberaceae family [143]. The curcuminoid polyphenolic compounds in this rhizome plant lead to the display of the bright orange-yellow color [144]. In general, turmeric is antibacterial, antiseptic, and anti-inflammatory and has a long history of medicinal usage for allergies, boosting immunity, stimulating digestion, preventing infection, boosting liver detoxification, and balancing cholesterol levels [144,145,146]. The bioactive components of turmeric consist of water-soluble curcuminoids and turmerone oil [147]. The curcuminoids encompass cyclocurcumin, bisdemethoxycurcumin, demethoxycurcumin, and curcumin [148]. Moreover, curcumin is the main curcuminoid associated with reduced AD risk due to its anti-inflammatory activity [149]. In vitro studies have elucidated that curcumin has the ability to neutralize reactive oxygen species and block lipid peroxidation, with curcumin being determined to have a greater potency than vitamin E [150,151].
Administering curcumin orally to aged mice with advanced deposits of plaque yielded a significant diminishment in the plaque loads [152]. Additionally, curcumin was also associated with reduced inflammation, amyloid pathology, and oxidative damage in mice models of AD [152,153]. Likewise, administering curcumin via peripheral injections crossed the blood–brain barrier and decreased plaque levels and blocked subsequent plaque development [152]. Furthermore, various studies using animal models of AD have demonstrated enhanced cognitive function in curcumin-treated groups, potentially due to curcumin’s ability to decrease the levels of Aβ plaques, as well as its antioxidant and anti-inflammatory properties [154,155]. To illustrate this, a double-transgenic APP/PS1 AD model allowed researchers to inspect the effects of two different curcumin dosages, including a 160 ppm low dose and 1000 ppm high dose for a period of 6 months [156]. Both doses reported significant cognitive enhancement, but the higher curcumin dosage generated better cognitive improvement [156]. Likewise, curcumin decreased the deposits of Aβ, potentially via autophagy promotion [156]. Nonetheless, due to curcumin’s inefficient penetration of the BBB, nominal bioavailability, and quick gastrointestinal metabolism, various curcumin analogs have been tested [157,158,159]. Even though the derivatives provided differing improved outcomes based on the disease model, they were all effective at decreasing plaque-associated pathology and enhancing cognitive function [160,161,162].
Curcumin also has the ability to reverse cognitive deficits in numerous animal models of AD [163,164,165]. Regardless of the administration route, higher dosages have been determined to be more efficacious than lower dosages, and cognitive enhancement was more prominent when curcumin was administered with piperine, which has a plethora of pharmacological and positive health effects against chronic disorders [146]. Subsequently, there is evidence suggesting that metals similar to iron, zinc, or copper may have a role in AD-related pathogenesis [166]. The metals are found concentrated in AD-affected brains and trigger oxidative neurotoxicity and amyloid aggregation [167]. Curcumin has been shown to produce strong metal complexes, thereby blocking mentally induced Aβ inflammation, toxicity, and aggregation [163,168].
Nonetheless, clinical studies conducted utilizing curcumin have been limited in number. Although research has demonstrated that curcumin in combination with vitamin C, B vitamins, piperine, and other dietary supplements has neuroprotective and synergistic effects, further research is necessary to conclusively state curcumin’s ability at ameliorating cognitive function in humans [146,162,169]. As such, if future studies elevate curcumin’s systemic bioavailability and enhance BBB penetration, curcumin could be a promising therapeutic for AD.
Fuzhisan, a Chinese herbal complex comprising Anemone altaica in the Araceae family, Glycyrrhiza uralensis in the Leguminosae family, Scutellaria baicalensis in the Labiatae family, and Ginseng root in the Araliaceae family, has been clinically utilized for senile dementia [75]. Fuzhisan’s neuroprotective effects have been proposed to be associated with anti- Aβ accumulation and anti-apoptotic effects, as well as elevating acetylcholine levels and enhancing neurotropic effects [170]. Consequently, studies have shown that Fuzhisan inhibits Abeta25-35-induced neurotoxicity, with the induction of the ADAM10 and SIRT1-Fox0 pathway serving a potential role in neuroprotection, since PC12 cells treated with Fuzhisan demonstrated significantly elevated levels of ADAM10 [170]. Patel et al. elucidated the protective effects of SIRT1 on AD, showing that caloric restrictions diminished plaque formation and Aβ levels in the brains of AD transgenic mice models [171]. Consequently, deficiencies in SIRT1 have also been correlated with enhanced levels of phosphorylated tau in the neurons and the number of neurofibrillary tangles in the brains of AD subjects [171,172]. In an in vivo study in aged rats, Fuzhisan administered at 0.3, 0.6, and 1.2 g/kg/d ameliorated cognitive function. Western blot and spectrophotometry results demonstrate that Fuzhisan elevated the production and activity of choline O-acetyltransferase and acetylcholine contents in the hippocampus [75]. Furthermore, a research study conducted by Bi et al. assessed the glucose metabolism and cognitive performance by ADAS-Cog in 22 patients, with 12 receiving 10 mg/day of Fuzhisan and 10 receiving a placebo [76]. These studies, especially the randomized, placebo controlled human trials suggest that various herbal products could potentially ameliorate cognitive function (Table 2).

5. Medicinal Plants for AD with Limited Studies

Currently, there are a plethora of other medicinal plants that show preventative and therapeutic activity for AD [174]. Nonetheless, in vitro and in vivo studies are limited, with most data deriving from observational studies. These plants include Commiphora wightii, Tinospora cordifolia, Hypericum perforatum, Rhodiola rosea, Moringa oleifera, Convolvolus pluricaulis, Hericium erinaceus, Camellia sinensis, and others [174,175,176,177,178,179]. Similarly, there are also neuroprotective natural products that could be obtained from food. To illustrate this, Allium sativum in the Alliaceae family has shown to be a potent anti-neuroinflammatory, antioxidant, and regulator of neurotransmitter signaling when referring to aged garlic extract, which could ameliorate AD pathogenesis [180,181]. Likewise, juice and extracts from Punica granatum have also shown neuroprotective effects in animal models, potentially by counteracting oxidative damage, minimizing inflammation of the brain and soluble Aβ 42 and hippocampal amyloid deposition [182,183].
Consequently, natural products, such as cannabidiol (CBD) and tetrahydrocannabinol (THC), have also shown to be effective in vitro and in vivo [184,185,186,187]. In PC12 neuronal cells, CBD has shown protection against oxidative stress, Aβ-induced neurotoxicity, inhibition of tau hyperphosphorylation, prevention of proinflammatory gene transcription, and inhibition of Aβ-induced tau hyperphosphorylation [188,189]. In vivo, CBD has shown attenuation of Aβ-induced neuroinflammatory responses by minimizing proinflammatory gene and mediator expression, as well as minimized reactive gliosis [187,190]. In vivo studies using CBD and THC have shown enhanced memory in the active avoidance and two-object recognition tasks [186]. Additionally, it has been shown to reduce soluble Abeta42 levels and alter the plaque composition [186].

6. Future Directions in Herbal Medicine

Currently, approximately 6.2 million Americans aged 65+ are suffering from Alzheimer’s dementia [1]. Additionally, that number is expected to grow to 13.8 million Americans by 2060 if effective therapeutics are not developed and utilized to halt, prevent, or slow AD-related pathogenesis [1]. Consequently, AD has an astoundingly high economic burden, with an approximated USD 305 billion having been spent on AD treatment in 2020 [191]. As such, finding effective therapeutic treatments are necessary to enhance patient outcomes. Even though various novel approaches have been found for symptomatic treatment and numerous disease-modifying therapies are under development, most clinical trials related to AD have not been successful [192]. Due to this, a significant deviation from monotherapeutic treatments has taken place to favor multitherapeutic, individualized, and comprehensive approaches since AD is a highly heterogeneous disorder [193,194].
Tau and Aβ have been shown to boost the loss of blood–brain–barrier (BBB) integrity [195]. Thus, a critical challenge in AD drug delivery relies on circumventing the BBB, which averts the entry of a plethora of possible therapeutic agents [195]. In general, the most common administration route is oral, but it has not been clearly elucidated whether herbal components can access the central nervous system from systemic circulation. Furthermore, rapid metabolism, limited solubility in aqueous environment, and incomplete distribution in the CNS are further limits that must be overcome. Thus, intranasal administration is an efficient and noninvasive administration route that could bypass the BBB and directly target the central nervous system [196]. Utilizing this method of delivery, herbs in medicated oil or dry powder forms could be administered directly to the subject. Medicated oils could also contain a mixture of lipid-soluble and lipophilic molecules to warrant the synergistic interactions between the varied herbal components. The benefits of intranasal delivery comprise brain injury avoidance, surmounting the requirement of implanting delivery devises, and reducing the systemic-administration-associated side effects [196,197]. Utilizing the intranasal administration technique, researchers have successfully treated memory loss in transgenic mice models of AD [198]. However, this method also has some limitations, such as a particularly small volume of administrated drugs, the limited surface area in the olfactory epithelium, and a relatively short retention time for drug absorption [199,200]. Thus, further research is required to support and enhance the usage of intranasal administration for herbal delivery.
In general, a large amount of evidence has shown that various herbs and natural bioactive products could be effective in AD treatment while having minimal severe adverse effects [201]. Even though it is not fully understood, the AD pathological process is proposed to be multifactorial [202]. As such, neuroprotective techniques encompassing a plethora of mechanisms of action are crucial for AD treatment and prevention. Natural product extracts and mixtures, with various bioactive compounds and neuroprotective mechanisms, are advantageous in drug discovery for AD. However, more research is necessary to address the concerns associated with herbal medicine and natural medicine for AD. Consequently, although the use of some herbs, such as those of C. longa and B. monnieri, have demonstrated slight clinical improvements, many natural products such as polyphenols remain of interest in the treatment of neurodegenerative disorders [203]. Additionally, some limitations include chemical instability, since curcumin and resveratrol are chemically unstable [204]. Likewise, low bioavailability, such as that observed with curcumin, is also a major issue [157]. Therefore, it has been challenging to translate effective preclinical results. Nonetheless, numerous studies have attempted to improve bioavailability by employing nanocarriers and nanotechnology, which could potentially enhance clinical efficacy and therapeutic response [205]. One notable example has been that of nanolipid epigallocatechin-3-gallate particles, which have demonstrated the ability to increase α-secretase, enhancing their ability in vitro and increasing epigallocatechin-3-gallate’s oral bioavailability by greater than two-fold [206]. As such, more comprehensive quality control and practical guidelines, in addition to novel strategies and approaches to promote central nervous system access of the herbal neuroprotective agents, could potentially allow natural product therapy to play an essential role in AD preventative and therapeutic measures.

Author Contributions

Conceptualization, T.Z.; writing—original draft preparation, T.Z.; writing—review and editing, J.K.; visualization, T.Z.; supervision, C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alzheimer’s Association. 2021 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2021, 17, 327–406. [Google Scholar] [CrossRef] [PubMed]
  2. Corey-Bloom, J. The ABC of Alzheimer’s disease: Cognitive changes and their management in Alzheimer’s disease and related dementias. Int. Psychogeriatr. 2002, 14 (Suppl. 1), 51–75. [Google Scholar] [CrossRef] [PubMed]
  3. Bekris, L.M.; Yu, C.E.; Bird, T.D.; Tsuang, D.W. Genetics of Alzheimer disease. J. Geriatr. Psychiatry Neurol. 2010, 23, 213–227. [Google Scholar] [CrossRef] [Green Version]
  4. Huang, Y. Roles of apolipoprotein E4 (ApoE4) in the pathogenesis of Alzheimer’s disease: Lessons from ApoE mouse models. Biochem. Soc. Trans. 2011, 39, 924–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Guerreiro, R.; Bras, J. The age factor in Alzheimer’s disease. Genome Med. 2015, 7, 106. [Google Scholar] [CrossRef] [Green Version]
  6. Hebert, L.E.; Weuve, J.; Scherr, P.A.; Evans, D.A. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology 2013, 80, 1778–1783. [Google Scholar] [CrossRef] [Green Version]
  7. Farrer, L.A.; Cupples, L.A.; Haines, J.L.; Hyman, B.; Kukull, W.A.; Mayeux, R.; Myers, R.H.; Pericak-Vance, M.A.; Risch, N.; van Duijn, C.M. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA 1997, 278, 1349–1356. [Google Scholar] [CrossRef]
  8. Reitz, C. Toward precision medicine in Alzheimer’s disease. Ann. Transl. Med. 2016, 4, 107. [Google Scholar] [CrossRef] [Green Version]
  9. Oxford, A.E.; Stewart, E.S.; Rohn, T.T. Clinical Trials in Alzheimer’s Disease: A Hurdle in the Path of Remedy. Int. J. Alzheimer’s Dis. 2020, 2020, 5380346. [Google Scholar] [CrossRef] [Green Version]
  10. Tan, C.C.; Yu, J.T.; Wang, H.F.; Tan, M.S.; Meng, X.F.; Wang, C.; Jiang, T.; Zhu, X.C.; Tan, L. Efficacy and safety of donepezil, galantamine, rivastigmine, and memantine for the treatment of Alzheimer’s disease: A systematic review and meta-analysis. J. Alzheimer’s Dis. 2014, 41, 615–631. [Google Scholar] [CrossRef]
  11. Dunn, B.; Stein, P.; Temple, R.; Cavazzoni, P. An Appropriate Use of Accelerated Approval—Aducanumab for Alzheimer’s Disease. N. Engl. J. Med. 2021, 385, 856–857. [Google Scholar] [CrossRef] [PubMed]
  12. Vickers, J.C.; Dickson, T.C.; Adlard, P.A.; Saunders, H.L.; King, C.E.; McCormack, G. The cause of neuronal degeneration in Alzheimer’s disease. Prog. Neurobiol. 2000, 60, 139–165. [Google Scholar] [CrossRef]
  13. Whitwell, J.L. Progression of atrophy in Alzheimer’s disease and related disorders. Neurotox. Res. 2010, 18, 339–346. [Google Scholar] [CrossRef] [PubMed]
  14. De-Paula, V.J.; Radanovic, M.; Diniz, B.S.; Forlenza, O.V. Alzheimer’s disease. Subcell. Biochem. 2012, 65, 329–352. [Google Scholar] [CrossRef] [PubMed]
  15. Näslund, J.; Haroutunian, V.; Mohs, R.; Davis, K.L.; Davies, P.; Greengard, P.; Buxbaum, J.D. Correlation between elevated levels of amyloid beta-peptide in the brain and cognitive decline. JAMA 2000, 283, 1571–1577. [Google Scholar] [CrossRef] [PubMed]
  16. Bent, S. Herbal medicine in the United States: Review of efficacy, safety, and regulation: Grand rounds at University of California, San Francisco Medical Center. J. Gen. Intern. Med. 2008, 23, 854–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Eckert, G.P. Traditional used Plants against Cognitive Decline and Alzheimer Disease. Front. Pharmacol. 2010, 1, 138. [Google Scholar] [CrossRef] [Green Version]
  18. Ricciarelli, R.; Fedele, E. The Amyloid Cascade Hypothesis in Alzheimer’s Disease: It’s Time to Change Our Mind. Curr. Neuropharmacol. 2017, 15, 926–935. [Google Scholar] [CrossRef] [Green Version]
  19. Maccioni, R.B.; Farías, G.; Morales, I.; Navarrete, L. The revitalized tau hypothesis on Alzheimer’s disease. Arch. Med. Res. 2010, 41, 226–231. [Google Scholar] [CrossRef]
  20. Liu, P.P.; Xie, Y.; Meng, X.Y.; Kang, J.S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct. Target. Ther. 2019, 4, 29. [Google Scholar] [CrossRef]
  21. Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef]
  22. Busche, M.A.; Wegmann, S.; Dujardin, S.; Commins, C.; Schiantarelli, J.; Klickstein, N.; Kamath, T.V.; Carlson, G.A.; Nelken, I.; Hyman, B.T. Tau impairs neural circuits, dominating amyloid-β effects, in Alzheimer models in vivo. Nat. Neurosci. 2019, 22, 57–64. [Google Scholar] [CrossRef]
  23. Frost, B.; Jacks, R.L.; Diamond, M.I. Propagation of tau misfolding from the outside to the inside of a cell. J. Biol. Chem. 2009, 284, 12845–12852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef] [PubMed]
  25. Francis, P.T.; Palmer, A.M.; Snape, M.; Wilcock, G.K. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry 1999, 66, 137–147. [Google Scholar] [CrossRef] [PubMed]
  26. Birks, J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst. Rev. 2006, 1, CD005593. [Google Scholar] [CrossRef]
  27. Huang, W.J.; Zhang, X.; Chen, W.W. Role of oxidative stress in Alzheimer’s disease. Biomed. Rep. 2016, 4, 519–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Markesbery, W.R. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic. Biol. Med. 1997, 23, 134–147. [Google Scholar] [CrossRef]
  29. Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353–356. [Google Scholar] [CrossRef] [Green Version]
  30. Izzo, N.J.; Staniszewski, A.; To, L.; Fa, M.; Teich, A.F.; Saeed, F.; Wostein, H.; Walko, T.; Vaswani, A.; Wardius, M.; et al. Alzheimer’s therapeutics targeting amyloid beta 1-42 oligomers I: Abeta 42 oligomer binding to specific neuronal receptors is displaced by drug candidates that improve cognitive deficits. PLoS ONE 2014, 9, e111898. [Google Scholar] [CrossRef]
  31. Conway, K.A.; Baxter, E.W.; Felsenstein, K.M.; Reitz, A.B. Emerging beta-amyloid therapies for the treatment of Alzheimer’s disease. Curr. Pharm. Des. 2003, 9, 427–447. [Google Scholar] [CrossRef]
  32. Tickler, A.K.; Wade, J.D.; Separovic, F. The role of Abeta peptides in Alzheimer’s disease. Protein Pept. Lett. 2005, 12, 513–519. [Google Scholar] [CrossRef] [PubMed]
  33. Uddin, M.S.; Al Mamun, A.; Kabir, M.T.; Ashraf, G.M.; Bin-Jumah, M.N.; Abdel-Daim, M.M. Multi-Target Drug Candidates for Multifactorial Alzheimer’s Disease: AChE and NMDAR as Molecular Targets. Mol. Neurobiol. 2021, 58, 281–303. [Google Scholar] [CrossRef] [PubMed]
  34. González, J.F.; Alcántara, A.R.; Doadrio, A.L.; Sánchez-Montero, J.M. Developments with multi-target drugs for Alzheimer’s disease: An overview of the current discovery approaches. Expert Opin. Drug Discov. 2019, 14, 879–891. [Google Scholar] [CrossRef] [PubMed]
  35. Longo, F.M.; Massa, S.M. Neuroprotective strategies in Alzheimer’s disease. NeuroRx 2004, 1, 117–127. [Google Scholar] [CrossRef] [PubMed]
  36. Niikura, T.; Tajima, H.; Kita, Y. Neuronal cell death in Alzheimer’s disease and a neuroprotective factor, humanin. Curr. Neuropharmacol. 2006, 4, 139–147. [Google Scholar] [CrossRef]
  37. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [Green Version]
  38. Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115. [Google Scholar] [CrossRef] [Green Version]
  39. Ding, Y.; Zhao, J.; Zhang, X.; Wang, S.; Viola, K.L.; Chow, F.E.; Zhang, Y.; Lippa, C.; Klein, W.L.; Gong, Y. Amyloid Beta Oligomers Target to Extracellular and Intracellular Neuronal Synaptic Proteins in Alzheimer’s Disease. Front. Neurol. 2019, 10, 1140. [Google Scholar] [CrossRef] [Green Version]
  40. Quiroz-Baez, R.; Ferrera, P.; Rosendo-Gutiérrez, R.; Morán, J.; Bermúdez-Rattoni, F.; Arias, C. Caspase-12 activation is involved in amyloid-β protein-induced synaptic toxicity. J. Alzheimer’s Dis. 2011, 26, 467–476. [Google Scholar] [CrossRef]
  41. Iqbal, K.; Liu, F.; Gong, C.X.; Grundke-Iqbal, I. Tau in Alzheimer disease and related tauopathies. Curr. Alzheimer Res. 2010, 7, 656–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Pradeep, S.; Jain, A.S.; Dharmashekara, C.; Prasad, S.K.; Kollur, S.P.; Syed, A.; Shivamallu, C. Alzheimer’s Disease and Herbal Combination Therapy: A Comprehensive Review. J. Alzheimer’s Dis. Rep. 2020, 4, 417–429. [Google Scholar] [CrossRef] [PubMed]
  43. Olila, D.; Opuda-Asibo, J. Antibacterial and antifungal activities of extracts of Zanthoxylum chalybeum and Warburgia ugandensis, Ugandan medicinal plants. Afr. Health Sci. 2001, 1, 66–72. [Google Scholar] [PubMed]
  44. Droy-Lefaix, M.T. Effect of the antioxidant action of Ginkgo biloba extract (EGb 761) on aging and oxidative stress. Age 1997, 20, 141–149. [Google Scholar] [CrossRef] [Green Version]
  45. Shal, B.; Ding, W.; Ali, H.; Kim, Y.S.; Khan, S. Anti-neuroinflammatory Potential of Natural Products in Attenuation of Alzheimer’s Disease. Front. Pharmacol. 2018, 9, 548. [Google Scholar] [CrossRef]
  46. Noori, T.; Dehpour, A.R.; Sureda, A.; Sobarzo-Sanchez, E.; Shirooie, S. Role of natural products for the treatment of Alzheimer’s disease. Eur. J. Pharmacol. 2021, 898, 173974. [Google Scholar] [CrossRef]
  47. Koynova, R.; Tenchov, B. Natural Product Formulations for the Prevention and Treatment of Alzheimer’s disease: A Patent Review. Recent Pat. Drug Deliv. Formul. 2018, 12, 23–39. [Google Scholar] [CrossRef]
  48. Patwardhan, B.; Warude, D.; Pushpangadan, P.; Bhatt, N. Ayurveda and traditional Chinese medicine: A comparative overview. Evid.-Based Complement. Altern. Med. 2005, 2, 465–473. [Google Scholar] [CrossRef] [Green Version]
  49. Koithan, M.; Farrell, C. Indigenous Native American Healing Traditions. J. Nurse Pract. 2010, 6, 477–478. [Google Scholar] [CrossRef] [Green Version]
  50. Singh, M.; Ramassamy, C. Screening of neuroprotective activity of Indian medicinal plant. J. Nutr Sci. 2017, 6, e54. [Google Scholar] [CrossRef] [Green Version]
  51. Jayaprakasam, B.; Padmanabhan, K.; Nair, M.G. Withanamides in Withania somnifera fruit protect PC-12 cells from beta-amyloid responsible for Alzheimer’s disease. Phytother. Res. 2010, 24, 859–863. [Google Scholar] [CrossRef] [PubMed]
  52. Pratte, M.A.; Nanavati, K.B.; Young, V.; Morley, C.P. An alternative treatment for anxiety: A systematic review of human trial results reported for the Ayurvedic herb ashwagandha (Withania somnifera). J. Altern. Complement. Med. 2014, 20, 901–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Pingali, U.; Pilli, R.; Fatima, N. Effect of standardized aqueous extract of Withania somnifera on tests of cognitive and psychomotor performance in healthy human participants. Pharmacogn. Res. 2014, 6, 12–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chengappa, K.N.; Bowie, C.R.; Schlicht, P.J.; Fleet, D.; Brar, J.S.; Jindal, R. Randomized placebo-controlled adjunctive study of an extract of withania somnifera for cognitive dysfunction in bipolar disorder. J. Clin. Psychiatry 2013, 74, 1076–1083. [Google Scholar] [CrossRef] [Green Version]
  55. Russo, A.; Borrelli, F. Bacopa monniera, a reputed nootropic plant: An overview. Phytomedicine 2005, 12, 305–317. [Google Scholar] [CrossRef]
  56. Uabundit, N.; Wattanathorn, J.; Mucimapura, S.; Ingkaninan, K. Cognitive enhancement and neuroprotective effects of Bacopa monnieri in Alzheimer’s disease model. J. Ethnopharmacol. 2010, 127, 26–31. [Google Scholar] [CrossRef]
  57. Limpeanchob, N.; Jaipan, S.; Rattanakaruna, S.; Phrompittayarat, W.; Ingkaninan, K. Neuroprotective effect of Bacopa monnieri on beta-amyloid-induced cell death in primary cortical culture. J. Ethnopharmacol. 2008, 120, 112–117. [Google Scholar] [CrossRef]
  58. Raghav, S.; Singh, H.; Dalal, P.K.; Srivastava, J.S.; Asthana, O.P. Randomized controlled trial of standardized Bacopa monniera extract in age-associated memory impairment. Indian J. Psychiatry 2006, 48, 238–242. [Google Scholar] [CrossRef] [Green Version]
  59. Osman, N.M.; Amer, A.S.; Abdelwahab, S. Effects of Ginko biloba leaf extract on the neurogenesis of the hippocampal dentate gyrus in the elderly mice. Anat. Sci. Int. 2016, 91, 280–289. [Google Scholar] [CrossRef]
  60. Zhao, J.; Li, K.; Wang, Y.; Li, D.; Wang, Q.; Xie, S.; Wang, J.; Zuo, Z. Enhanced anti-amnestic effect of donepezil by Ginkgo biloba extract (EGb 761) via further improvement in pro-cholinergic and antioxidative activities. J. Ethnopharmacol. 2021, 269, 113711. [Google Scholar] [CrossRef]
  61. Liu, H.; Ye, M.; Guo, H. An Updated Review of Randomized Clinical Trials Testing the Improvement of Cognitive Function of. Front. Pharmacol. 2019, 10, 1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Mahadevan, S.; Park, Y. Multifaceted therapeutic benefits of Ginkgo biloba L.: Chemistry, efficacy, safety, and uses. J. Food Sci. 2008, 73, R14–R19. [Google Scholar] [CrossRef]
  63. Soumyanath, A.; Zhong, Y.P.; Henson, E.; Wadsworth, T.; Bishop, J.; Gold, B.G.; Quinn, J.F. Centella asiatica Extract Improves Behavioral Deficits in a Mouse Model of Alzheimer’s Disease: Investigation of a Possible Mechanism of Action. Int. J. Alzheimer’s Dis. 2012, 2012, 381974. [Google Scholar] [CrossRef] [Green Version]
  64. Veerendra Kumar, M.H.; Gupta, Y.K. Effect of Centella asiatica on cognition and oxidative stress in an intracerebroventricular streptozotocin model of Alzheimer’s disease in rats. Clin. Exp. Pharmacol. Physiol. 2003, 30, 336–342. [Google Scholar] [CrossRef] [PubMed]
  65. Dhanasekaran, M.; Holcomb, L.A.; Hitt, A.R.; Tharakan, B.; Porter, J.W.; Young, K.A.; Manyam, B.V. Centella asiatica extract selectively decreases amyloid beta levels in hippocampus of Alzheimer’s disease animal model. Phytother. Res. 2009, 23, 14–19. [Google Scholar] [CrossRef]
  66. Shinomol, G.K.; Bharath, M.M. Exploring the Role of “Brahmi” (Bacopa monnieri and Centella asiatica) in Brain Function and Therapy. Recent Pat. Endocr. Metab. Immune Drug Discov. 2011, 5, 33–49. [Google Scholar] [CrossRef] [Green Version]
  67. Adalier, N.; Parker, H. Vitamin E, Turmeric and Saffron in Treatment of Alzheimer’s Disease. Antioxidants 2016, 5, 40. [Google Scholar] [CrossRef] [Green Version]
  68. Khazdair, M.R.; Boskabady, M.H.; Hosseini, M.; Rezaee, R.; Tsatsakis, A.M. The effects of Crocus sativus (saffron) and its constituents on nervous system: A review. Avicenna J. Phytomed. 2015, 5, 376–391. [Google Scholar]
  69. Akhondzadeh, S.; Shafiee Sabet, M.; Harirchian, M.H.; Togha, M.; Cheraghmakani, H.; Razeghi, S.; Hejazi, S.S.; Yousefi, M.H.; Alimardani, R.; Jamshidi, A.; et al. A 22-week, multicenter, randomized, double-blind controlled trial of Crocus sativus in the treatment of mild-to-moderate Alzheimer’s disease. Psychopharmacology 2010, 207, 637–643. [Google Scholar] [CrossRef]
  70. Batarseh, Y.S.; Bharate, S.S.; Kumar, V.; Kumar, A.; Vishwakarma, R.A.; Bharate, S.B.; Kaddoumi, A. Crocus sativus Extract Tightens the Blood-Brain Barrier, Reduces Amyloid β Load and Related Toxicity in 5XFAD Mice. ACS Chem. Neurosci. 2017, 8, 1756–1766. [Google Scholar] [CrossRef]
  71. Chen, M.; Du, Z.Y.; Zheng, X.; Li, D.L.; Zhou, R.P.; Zhang, K. Use of curcumin in diagnosis, prevention, and treatment of Alzheimer’s disease. Neural Regen Res. 2018, 13, 742–752. [Google Scholar] [CrossRef]
  72. Zhang, X.; Tian, Y.; Li, Z.; Tian, X.; Sun, H.; Liu, H.; Moore, A.; Ran, C. Design and synthesis of curcumin analogues for in vivo fluorescence imaging and inhibiting copper-induced cross-linking of amyloid beta species in Alzheimer’s disease. J. Am. Chem. Soc. 2013, 135, 16397–16409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zhang, X.; Yin, W.K.; Shi, X.D.; Li, Y. Curcumin activates Wnt/β-catenin signaling pathway through inhibiting the activity of GSK-3β in APPswe transfected SY5Y cells. Eur. J. Pharm. Sci. 2011, 42, 540–546. [Google Scholar] [CrossRef] [PubMed]
  74. Teter, B.; Morihara, T.; Lim, G.P.; Chu, T.; Jones, M.R.; Zuo, X.; Paul, R.M.; Frautschy, S.A.; Cole, G.M. Curcumin restores innate immune Alzheimer’s disease risk gene expression to ameliorate Alzheimer pathogenesis. Neurobiol. Dis. 2019, 127, 432–448. [Google Scholar] [CrossRef] [PubMed]
  75. Li, X.L.; Wang, D.S.; Zhao, B.Q.; Li, Q.; Qu, H.Y.; Zhang, T.; Zhou, J.P.; Sun, M.J. Effects of Chinese herbal medicine fuzhisan on aged rats. Exp. Gerontol. 2008, 43, 853–858. [Google Scholar] [CrossRef]
  76. Bi, M.; Tong, S.; Zhang, Z.; Ma, Q.; Zhang, S.; Luo, Z.; Zhang, Y.; Li, X.; Wang, D. Changes in cerebral glucose metabolism in patients with mild-to-moderate Alzheimer’s disease: A pilot study with the Chinese herbal medicine fuzhisan. Neurosci. Lett. 2011, 501, 35–40. [Google Scholar] [CrossRef]
  77. Gao, R.; Wang, Y.; Pan, Q.; Huang, G.; Li, N.; Mou, J.; Wang, D. Fuzhisan, a chinese herbal medicine, suppresses beta-secretase gene transcription via upregulation of SIRT1 expression in N2a-APP695 cells. Int. J. Clin. Exp. Med. 2015, 8, 7231–7240. [Google Scholar]
  78. Singh, N.; Bhalla, M.; de Jager, P.; Gilca, M. An overview on ashwagandha: A Rasayana (rejuvenator) of Ayurveda. Afr. J. Tradit. Complement. Altern. Med. 2011, 8, 208–213. [Google Scholar] [CrossRef]
  79. Zahiruddin, S.; Basist, P.; Parveen, A.; Parveen, R.; Khan, W.; Ahmad, S. Ashwagandha in brain disorders: A review of recent developments. J. Ethnopharmacol. 2020, 257, 112876. [Google Scholar] [CrossRef]
  80. Dar, N.J. Neurodegenerative diseases and Withania somnifera (L.): An update. J. Ethnopharmacol. 2020, 256, 112769. [Google Scholar] [CrossRef]
  81. Rao, R.V.; Descamps, O.; John, V.; Bredesen, D.E. Ayurvedic medicinal plants for Alzheimer’s disease: A review. Alzheimer’s Res. Ther. 2012, 4, 22. [Google Scholar] [CrossRef]
  82. Matsuda, H.; Murakami, T.; Kishi, A.; Yoshikawa, M. Structures of withanosides I, II, III, IV, V, VI, and VII, new withanolide glycosides, from the roots of Indian Withania somnifera DUNAL. and inhibitory activity for tachyphylaxis to clonidine in isolated guinea-pig ileum. Bioorg. Med. Chem. 2001, 9, 1499–1507. [Google Scholar] [CrossRef]
  83. Mishra, L.C.; Singh, B.B.; Dagenais, S. Scientific basis for the therapeutic use of Withania somnifera (ashwagandha): A review. Altern. Med. Rev. 2000, 5, 334–346. [Google Scholar] [PubMed]
  84. Sumathi, S.; Padma, P.R.; Gathampari, S.; Vidhya, S. Free radical scavenging activity of different parts of withania somnifera. Anc. Sci. Life 2007, 26, 30–34. [Google Scholar] [PubMed]
  85. Kumar, S.; Harris, R.J.; Seal, C.J.; Okello, E.J. An aqueous extract of Withania somnifera root inhibits amyloid β fibril formation in vitro. Phytother. Res. 2012, 26, 113–117. [Google Scholar] [CrossRef]
  86. Parihar, M.S.; Hemnani, T. Phenolic antioxidants attenuate hippocampal neuronal cell damage against kainic acid induced excitotoxicity. J. Biosci. 2003, 28, 121–128. [Google Scholar] [CrossRef]
  87. Kuboyama, T.; Tohda, C.; Zhao, J.; Nakamura, N.; Hattori, M.; Komatsu, K. Axon- or dendrite-predominant outgrowth induced by constituents from Ashwagandha. Neuroreport 2002, 13, 1715–1720. [Google Scholar] [CrossRef] [PubMed]
  88. Tohda, C.; Kuboyama, T.; Komatsu, K. Dendrite extension by methanol extract of Ashwagandha (roots of Withania somnifera) in SK-N-SH cells. Neuroreport 2000, 11, 1981–1985. [Google Scholar] [CrossRef]
  89. Kuboyama, T.; Tohda, C.; Komatsu, K. Neuritic regeneration and synaptic reconstruction induced by withanolide A. Br. J. Pharmacol. 2005, 144, 961–971. [Google Scholar] [CrossRef] [Green Version]
  90. Tohda, C.; Kuboyama, T.; Komatsu, K. Search for natural products related to regeneration of the neuronal network. Neurosignals 2005, 14, 34–45. [Google Scholar] [CrossRef]
  91. Sehgal, N.; Gupta, A.; Valli, R.K.; Joshi, S.D.; Mills, J.T.; Hamel, E.; Khanna, P.; Jain, S.C.; Thakur, S.S.; Ravindranath, V. Withania somnifera reverses Alzheimer’s disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc. Natl. Acad. Sci. USA 2012, 109, 3510–3515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Halim, M.A.; Rosli, I.M.; Jaafar, S.S.M.; Ooi, H.-M.; Long, P.-W.; Shamsuddin, S.; Najimudin, N.; Azzam, G. Withania somnifera Showed Neuroprotective Effect and Increase Longevity in Drosophila Alzheimer’s Disease Model; Cold Spring Harbor Laboratory: Cold Spring Harbor, LI, USA, 2020. [Google Scholar]
  93. Ng, Q.X.; Loke, W.; Foo, N.X.; Tan, W.J.; Chan, H.W.; Lim, D.Y.; Yeo, W.S. A systematic review of the clinical use of Withania somnifera (Ashwagandha) to ameliorate cognitive dysfunction. Phytother. Res. 2020, 34, 583–590. [Google Scholar] [CrossRef]
  94. Choudhary, D.; Bhattacharyya, S.; Bose, S. Efficacy and Safety of Ashwagandha (Withania somnifera (L.) Dunal) Root Extract in Improving Memory and Cognitive Functions. J. Diet. Suppl. 2017, 14, 599–612. [Google Scholar] [CrossRef] [PubMed]
  95. Aguiar, S.; Borowski, T. Neuropharmacological review of the nootropic herb Bacopa monnieri. Rejuvenation Res. 2013, 16, 313–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Mathew, J.; Balakrishnan, S.; Antony, S.; Abraham, P.M.; Paulose, C.S. Decreased GABA receptor in the cerebral cortex of epileptic rats: Effect of Bacopa monnieri and Bacoside-A. J. Biomed. Sci. 2012, 19, 25. [Google Scholar] [CrossRef] [PubMed]
  97. Jadiya, P.; Khan, A.; Sammi, S.R.; Kaur, S.; Mir, S.S.; Nazir, A. Anti-Parkinsonian effects of Bacopa monnieri: Insights from transgenic and pharmacological Caenorhabditis elegans models of Parkinson’s disease. Biochem. Biophys. Res. Commun. 2011, 413, 605–610. [Google Scholar] [CrossRef] [PubMed]
  98. Sokołowska, L.; Bylka, W. Bacopa Monnieri-activity and applications in medicine. Wiad. Lek. 2015, 68, 358–362. [Google Scholar]
  99. Channa, S.; Dar, A.; Anjum, S.; Yaqoob, M.; Atta Ur, R. Anti-inflammatory activity of Bacopa monniera in rodents. J. Ethnopharmacol. 2006, 104, 286–289. [Google Scholar] [CrossRef]
  100. Jeyasri, R.; Muthuramalingam, P.; Suba, V.; Ramesh, M.; Chen, J.T. Bacopa monnieri and Their Bioactive Compounds Inferred Multi-Target Treatment Strategy for Neurological Diseases: A Cheminformatics and System Pharmacology Approach. Biomolecules 2020, 10, 536. [Google Scholar] [CrossRef] [Green Version]
  101. Chaudhari, K.S.; Tiwari, N.R.; Tiwari, R.R.; Sharma, R.S. Neurocognitive Effect of Nootropic Drug Brahmi (Bacopa monnieri) in Alzheimer’s Disease. Ann. Neurosci. 2017, 24, 111–122. [Google Scholar] [CrossRef] [Green Version]
  102. Dhanasekaran, M.; Tharakan, B.; Holcomb, L.A.; Hitt, A.R.; Young, K.A.; Manyam, B.V. Neuroprotective mechanisms of ayurvedic antidementia botanical Bacopa monniera. Phytother. Res. 2007, 21, 965–969. [Google Scholar] [CrossRef] [PubMed]
  103. Roodenrys, S.; Booth, D.; Bulzomi, S.; Phipps, A.; Micallef, C.; Smoker, J. Chronic effects of Brahmi (Bacopa monnieri) on human memory. Neuropsychopharmacology 2002, 27, 279–281. [Google Scholar] [CrossRef] [Green Version]
  104. Kumar, N.; Abichandani, L.G.; Thawani, V.; Gharpure, K.J.; Naidu, M.U.; Venkat Ramana, G. Efficacy of Standardized Extract of Bacopa monnieri (Bacognize®) on Cognitive Functions of Medical Students: A Six-Week, Randomized Placebo-Controlled Trial. Evid.-Based Complement. Altern. Med. 2016, 2016, 4103423. [Google Scholar] [CrossRef] [Green Version]
  105. Saini, N.; Singh, D.; Sandhir, R. Neuroprotective effects of Bacopa monnieri in experimental model of dementia. Neurochem. Res. 2012, 37, 1928–1937. [Google Scholar] [CrossRef] [PubMed]
  106. Dimpel, W.; Schombert, L.; Biller, A. Psychophysiological Effects of Sideritis and Bacopa Extract and Three Combinations Thereof—A Quantitative EEG Study in Subjects Suffering from Mild Cognitive Impairment (MCI). Adv. Alzheimer’s Dis. 2016, 5, 64876. [Google Scholar] [CrossRef] [Green Version]
  107. Heiner, F.; Feistel, B.; Wink, M. Sideritis scardica extracts inhibit aggregation and toxicity of amyloid-β in Caenorhabditis elegans used as a model for Alzheimer’s disease. PeerJ 2018, 6, e4683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Zanotta, D.; Puricelli, S.; Bonoldi, G. Cognitive effects of a dietary supplement made from extract of Bacopa monnieri, astaxanthin, phosphatidylserine, and vitamin E in subjects with mild cognitive impairment: A noncomparative, exploratory clinical study. Neuropsychiatr. Dis. Treat. 2014, 10, 225–230. [Google Scholar] [CrossRef] [Green Version]
  109. Sunderland, T.; Hill, J.L.; Mellow, A.M.; Lawlor, B.A.; Gundersheimer, J.; Newhouse, P.A.; Grafman, J.H. Clock drawing in Alzheimer’s disease. A novel measure of dementia severity. J. Am. Geriatr. Soc. 1989, 37, 725–729. [Google Scholar] [CrossRef]
  110. Francis, P.T. The interplay of neurotransmitters in Alzheimer’s disease. CNS Spectr. 2005, 10, 6–9. [Google Scholar] [CrossRef] [PubMed]
  111. Prabhakar, S.; Vishnu, V.Y.; Modi, M.; Mohanty, M.; Sharma, A.; Medhi, B.; Mittal, B.R.; Khandelwal, N.; Goyal, M.K.; Lal, V.; et al. Efficacy of Bacopa Monnieri (Brahmi) and Donepezil in Alzheimer’s Disease and Mild Cognitive Impairment: A Randomized Double-Blind Parallel Phase 2b Study. Ann. Indian Acad. Neurol. 2020, 23, 767–773. [Google Scholar] [CrossRef]
  112. McKenna, D.J.; Jones, K.; Hughes, K. Efficacy, safety, and use of ginkgo biloba in clinical and preclinical applications. Altern. Ther. Health Med. 2001, 7, 70–86, 88–90. [Google Scholar] [PubMed]
  113. Silberstein, R.B.; Pipingas, A.; Song, J.; Camfield, D.A.; Nathan, P.J.; Stough, C. Examining brain-cognition effects of ginkgo biloba extract: Brain activation in the left temporal and left prefrontal cortex in an object working memory task. Evid.-Based Complement. Altern. Med. 2011, 2011, 164139. [Google Scholar] [CrossRef]
  114. Singh, S.K.; Srivastav, S.; Castellani, R.J.; Plascencia-Villa, G.; Perry, G. Neuroprotective and Antioxidant Effect of Ginkgo biloba Extract Against AD and Other Neurological Disorders. Neurotherapeutics 2019, 16, 666–674. [Google Scholar] [CrossRef] [PubMed]
  115. Diamond, B.J.; Shiflett, S.C.; Feiwel, N.; Matheis, R.J.; Noskin, O.; Richards, J.A.; Schoenberger, N.E. Ginkgo biloba extract: Mechanisms and clinical indications. Arch. Phys. Med. Rehabil. 2000, 81, 668–678. [Google Scholar] [CrossRef]
  116. Li, Z.Y.; Chung, Y.H.; Shin, E.J.; Dang, D.K.; Jeong, J.H.; Ko, S.K.; Nah, S.Y.; Baik, T.G.; Jhoo, J.H.; Ong, W.Y.; et al. YY-1224, a terpene trilactone-strengthened Ginkgo biloba, attenuates neurodegenerative changes induced by β-amyloid (1-42) or double transgenic overexpression of APP and PS1 via inhibition of cyclooxygenase-2. J. Neuroinflamm. 2017, 14, 94. [Google Scholar] [CrossRef]
  117. Yang, G.; Wang, Y.; Sun, J.; Zhang, K.; Liu, J. Ginkgo Biloba for Mild Cognitive Impairment and Alzheimer’s Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr. Top. Med. Chem. 2016, 16, 520–528. [Google Scholar] [CrossRef]
  118. Mahmoudian-Sani, M.R.; Hashemzadeh-Chaleshtori, M.; Asadi-Samani, M.; Yang, Q. Ginkgo biloba in the treatment of tinnitus: An updated literature review. Int. Tinnitus J. 2017, 21, 58–62. [Google Scholar] [CrossRef] [Green Version]
  119. Park, Y.J.; Kim, M.J.; Kim, H.R.; Yi, M.S.; Chung, K.H.; Oh, S.M. Chemopreventive effects of Ginkgo biloba extract in estrogen-negative human breast cancer cells. Arch. Pharm. Res. 2013, 36, 102–108. [Google Scholar] [CrossRef]
  120. Bastianetto, S.; Ramassamy, C.; Doré, S.; Christen, Y.; Poirier, J.; Quirion, R. The Ginkgo biloba extract (EGb 761) protects hippocampal neurons against cell death induced by beta-amyloid. Eur. J. Neurosci. 2000, 12, 1882–1890. [Google Scholar] [CrossRef]
  121. Ryu, K.H.; Han, H.Y.; Lee, S.Y.; Jeon, S.D.; Im, G.J.; Lee, B.Y.; Kim, K.; Lim, K.M.; Chung, J.H. Ginkgo biloba extract enhances antiplatelet and antithrombotic effects of cilostazol without prolongation of bleeding time. Thromb. Res. 2009, 124, 328–334. [Google Scholar] [CrossRef]
  122. Amri, H.; Ogwuegbu, S.O.; Boujrad, N.; Drieu, K.; Papadopoulos, V. In vivo regulation of peripheral-type benzodiazepine receptor and glucocorticoid synthesis by Ginkgo biloba extract EGb 761 and isolated ginkgolides. Endocrinology 1996, 137, 5707–5718. [Google Scholar] [CrossRef] [PubMed]
  123. Smith, J.V.; Burdick, A.J.; Golik, P.; Khan, I.; Wallace, D.; Luo, Y. Anti-apoptotic properties of Ginkgo biloba extract EGb 761 in differentiated PC12 cells. Cell. Mol. Biol. 2002, 48, 699–707. [Google Scholar] [PubMed]
  124. Bastianetto, S.; Zheng, W.H.; Quirion, R. The Ginkgo biloba extract (EGb 761) protects and rescues hippocampal cells against nitric oxide-induced toxicity: Involvement of its flavonoid constituents and protein kinase C. J. Neurochem. 2000, 74, 2268–2277. [Google Scholar] [CrossRef] [PubMed]
  125. Gong, Q.H.; Wu, Q.; Huang, X.N.; Sun, A.S.; Shi, J.S. Protective effects of Ginkgo biloba leaf extract on aluminum-induced brain dysfunction in rats. Life Sci. 2005, 77, 140–148. [Google Scholar] [CrossRef]
  126. Le Bars, P.L.; Katz, M.M.; Berman, N.; Itil, T.M.; Freedman, A.M.; Schatzberg, A.F. A placebo-controlled, double-blind, randomized trial of an extract of Ginkgo biloba for dementia. North American EGb Study Group. JAMA 1997, 278, 1327–1332. [Google Scholar] [CrossRef]
  127. Janssen, I.M.; Sturtz, S.; Skipka, G.; Zentner, A.; Velasco Garrido, M.; Busse, R. Ginkgo biloba in Alzheimer’s disease: A systematic review. Wien. Med. Wochenschr. 2010, 160, 539–546. [Google Scholar] [CrossRef] [PubMed]
  128. Hashiguchi, M.; Ohta, Y.; Shimizu, M.; Maruyama, J.; Mochizuki, M. Meta-analysis of the efficacy and safety of Ginkgo biloba extract for the treatment of dementia. J. Pharm. Health Care Sci. 2015, 1, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Orhan, I.E. Centella asiatica (L.) Urban: From Traditional Medicine to Modern Medicine with Neuroprotective Potential. Evid.-Based Complement. Altern. Med. 2012, 2012, 946259. [Google Scholar] [CrossRef] [Green Version]
  130. Gohil, K.J.; Patel, J.A.; Gajjar, A.K. Pharmacological Review on Centella asiatica: A Potential Herbal Cure-all. Indian J. Pharm. Sci. 2010, 72, 546–556. [Google Scholar] [CrossRef] [Green Version]
  131. Lokanathan, Y.; Omar, N.; Ahmad Puzi, N.N.; Saim, A.; Hj Idrus, R. Recent Updates in Neuroprotective and Neuroregenerative Potential of Centella asiatica. Malays. J. Med. Sci. 2016, 23, 4–14. [Google Scholar]
  132. Xu, Y.; Cao, Z.; Khan, I.; Luo, Y. Gotu Kola (Centella Asiatica) extract enhances phosphorylation of cyclic AMP response element binding protein in neuroblastoma cells expressing amyloid beta peptide. J. Alzheimer’s Dis. 2008, 13, 341–349. [Google Scholar] [CrossRef]
  133. da Rocha, M.D.; Viegas, F.P.; Campos, H.C.; Nicastro, P.C.; Fossaluzza, P.C.; Fraga, C.A.; Barreiro, E.J.; Viegas, C., Jr. The role of natural products in the discovery of new drug candidates for the treatment of neurodegenerative disorders II: Alzheimer’s disease. CNS Neurol. Disord. Drug Targets 2011, 10, 251–270. [Google Scholar] [CrossRef] [PubMed]
  134. Mohandas Rao, K.G.; Muddanna Rao, S.; Gurumadhva Rao, S. Centella asiatica (L.) leaf extract treatment during the growth spurt period enhances hippocampal CA3 neuronal dendritic arborization in rats. Evid.-Based Complement. Altern. Med. 2006, 3, 349–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kim, S.R.; Koo, K.A.; Lee, M.K.; Park, H.G.; Jew, S.S.; Cha, K.H.; Kim, Y.C. Asiatic acid derivatives enhance cognitive performance partly by improving acetylcholine synthesis. J. Pharm. Pharmacol. 2004, 56, 1275–1282. [Google Scholar] [CrossRef]
  136. Wattanathorn, J.; Mator, L.; Muchimapura, S.; Tongun, T.; Pasuriwong, O.; Piyawatkul, N.; Yimtae, K.; Sripanidkulchai, B.; Singkhoraard, J. Positive modulation of cognition and mood in the healthy elderly volunteer following the administration of Centella asiatica. J. Ethnopharmacol. 2008, 116, 325–332. [Google Scholar] [CrossRef]
  137. Abu-Izneid, T.; Rauf, A.; Khalil, A.A.; Olatunde, A.; Khalid, A.; Alhumaydhi, F.A.; Aljohani, A.S.M.; Sahab Uddin, M.; Heydari, M.; Khayrullin, M.; et al. Nutritional and health beneficial properties of saffron (Crocus sativus L): A comprehensive review. Crit. Rev. Food Sci. Nutr. 2020, 1–24. [Google Scholar] [CrossRef]
  138. Serrano-Díaz, J.; Sánchez, A.M.; Maggi, L.; Martínez-Tomé, M.; García-Diz, L.; Murcia, M.A.; Alonso, G.L. Increasing the applications of Crocus sativus flowers as natural antioxidants. J. Food Sci. 2012, 77, C1162–C1168. [Google Scholar] [CrossRef] [PubMed]
  139. Srivastava, R.; Ahmed, H.; Dixit, R.K.; Saraf, S.A. Crocus sativus L.: A comprehensive review. Pharmacogn. Rev. 2010, 4, 200–208. [Google Scholar] [CrossRef] [Green Version]
  140. Bathaie, S.Z.; Mousavi, S.Z. New applications and mechanisms of action of saffron and its important ingredients. Crit. Rev. Food Sci. Nutr. 2010, 50, 761–786. [Google Scholar] [CrossRef]
  141. Akhondzadeh, S.; Sabet, M.S.; Harirchian, M.H.; Togha, M.; Cheraghmakani, H.; Razeghi, S.; Hejazi, S.; Yousefi, M.H.; Alimardani, R.; Jamshidi, A.; et al. Saffron in the treatment of patients with mild to moderate Alzheimer’s disease: A 16-week, randomized and placebo-controlled trial. J. Clin. Pharm. Ther. 2010, 35, 581–588. [Google Scholar] [CrossRef]
  142. Farokhnia, M.; Shafiee Sabet, M.; Iranpour, N.; Gougol, A.; Yekehtaz, H.; Alimardani, R.; Farsad, F.; Kamalipour, M.; Akhondzadeh, S. Comparing the efficacy and safety of Crocus sativus L. with memantine in patients with moderate to severe Alzheimer’s disease: A double-blind randomized clinical trial. Hum. Psychopharmacol. 2014, 29, 351–359. [Google Scholar] [CrossRef] [PubMed]
  143. Kim, D.S.; Park, S.Y.; Kim, J.K. Curcuminoids from Curcuma longa L. (Zingiberaceae) that protect PC12 rat pheochromocytoma and normal human umbilical vein endothelial cells from betaA(1-42) insult. Neurosci. Lett. 2001, 303, 57–61. [Google Scholar] [CrossRef]
  144. Jurenka, J.S. Anti-inflammatory properties of curcumin, a major constituent of Curcuma longa: A review of preclinical and clinical research. Altern. Med. Rev. 2009, 14, 141–153. [Google Scholar]
  145. Ammon, H.P.; Wahl, M.A. Pharmacology of Curcuma longa. Planta Med. 1991, 57, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  147. Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: The golden pigment from golden spice. Cancer Res. Treat. 2014, 46, 2–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Sharifi-Rad, J.; Rayess, Y.E.; Rizk, A.A.; Sadaka, C.; Zgheib, R.; Zam, W.; Sestito, S.; Rapposelli, S.; Neffe-Skocińska, K.; Zielińska, D.; et al. Turmeric and Its Major Compound Curcumin on Health: Bioactive Effects and Safety Profiles for Food, Pharmaceutical, Biotechnological and Medicinal Applications. Front. Pharmacol. 2020, 11, 01021. [Google Scholar] [CrossRef] [PubMed]
  149. Mishra, S.; Palanivelu, K. The effect of curcumin (turmeric) on Alzheimer’s disease: An overview. Ann. Indian Acad. Neurol. 2008, 11, 13–19. [Google Scholar] [CrossRef]
  150. Epstein, J.; Sanderson, I.R.; Macdonald, T.T. Curcumin as a therapeutic agent: The evidence from in vitro, animal and human studies. Br. J. Nutr. 2010, 103, 1545–1557. [Google Scholar] [CrossRef] [Green Version]
  151. Maheshwari, R.K.; Singh, A.K.; Gaddipati, J.; Srimal, R.C. Multiple biological activities of curcumin: A short review. Life Sci. 2006, 78, 2081–2087. [Google Scholar] [CrossRef]
  152. Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; et al. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Lim, G.P.; Chu, T.; Yang, F.; Beech, W.; Frautschy, S.A.; Cole, G.M. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J. Neurosci. 2001, 21, 8370–8377. [Google Scholar] [CrossRef] [PubMed]
  154. Zhu, L.N.; Mei, X.; Zhang, Z.G.; Xie, Y.P.; Lang, F. Curcumin intervention for cognitive function in different types of people: A systematic review and meta-analysis. Phytother. Res. 2019, 33, 524–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Kou, J.; Wang, M.; Shi, J.; Zhang, H.; Pu, X.; Song, S.; Yang, C.; Yan, Y.; Döring, Y.; Xie, X.; et al. Curcumin Reduces Cognitive Deficits by Inhibiting Neuroinflammation through the Endoplasmic Reticulum Stress Pathway in Apolipoprotein E4 Transgenic Mice. ACS Omega 2021, 6, 6654–6662. [Google Scholar] [CrossRef]
  156. Wang, C.; Zhang, X.; Teng, Z.; Zhang, T.; Li, Y. Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin-induced autophagy in APP/PS1 double transgenic mice. Eur. J. Pharmacol. 2014, 740, 312–320. [Google Scholar] [CrossRef]
  157. Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B. Bioavailability of curcumin: Problems and promises. Mol. Pharm. 2007, 4, 807–818. [Google Scholar] [CrossRef]
  158. Vyas, A.; Dandawate, P.; Padhye, S.; Ahmad, A.; Sarkar, F. Perspectives on new synthetic curcumin analogs and their potential anticancer properties. Curr. Pharm. Des. 2013, 19, 2047–2069. [Google Scholar]
  159. Chainoglou, E.; Hadjipavlou-Litina, D. Curcumin analogues and derivatives with anti-proliferative and anti-inflammatory activity: Structural characteristics and molecular targets. Expert Opin. Drug Discov. 2019, 14, 821–842. [Google Scholar] [CrossRef]
  160. Katsori, A.M.; Chatzopoulou, M.; Dimas, K.; Kontogiorgis, C.; Patsilinakos, A.; Trangas, T.; Hadjipavlou-Litina, D. Curcumin analogues as possible anti-proliferative & anti-inflammatory agents. Eur. J. Med. Chem. 2011, 46, 2722–2735. [Google Scholar] [CrossRef]
  161. Wan, Y.; Liang, Y.; Liang, F.; Shen, N.; Shinozuka, K.; Yu, J.T.; Ran, C.; Quan, Q.; Tanzi, R.E.; Zhang, C. A Curcumin Analog Reduces Levels of the Alzheimer’s Disease-Associated Amyloid-β Protein by Modulating AβPP Processing and Autophagy. J. Alzheimer’s Dis. 2019, 72, 761–771. [Google Scholar] [CrossRef]
  162. Voulgaropoulou, S.D.; van Amelsvoort, T.; Prickaerts, J.; Vingerhoets, C. The effect of curcumin on cognition in Alzheimer’s disease and healthy aging: A systematic review of pre-clinical and clinical studies. Brain Res. 2019, 1725, 146476. [Google Scholar] [CrossRef] [PubMed]
  163. Baum, L.; Ng, A. Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer’s disease animal models. J. Alzheimer’s Dis. 2004, 6, 367–377. [Google Scholar] [CrossRef] [PubMed]
  164. Wang, P.; Su, C.; Li, R.; Wang, H.; Ren, Y.; Sun, H.; Yang, J.; Sun, J.; Shi, J.; Tian, J.; et al. Mechanisms and effects of curcumin on spatial learning and memory improvement in APPswe/PS1dE9 mice. J. Neurosci. Res. 2014, 92, 218–231. [Google Scholar] [CrossRef] [PubMed]
  165. Bassani, T.B.; Turnes, J.M.; Moura, E.L.R.; Bonato, J.M.; Cóppola-Segovia, V.; Zanata, S.M.; Oliveira, R.; Vital, M. Effects of curcumin on short-term spatial and recognition memory, adult neurogenesis and neuroinflammation in a streptozotocin-induced rat model of dementia of Alzheimer’s type. Behav. Brain Res. 2017, 335, 41–54. [Google Scholar] [CrossRef] [PubMed]
  166. Zatta, P.; Drago, D.; Bolognin, S.; Sensi, S.L. Alzheimer’s disease, metal ions and metal homeostatic therapy. Trends Pharmacol. Sci. 2009, 30, 346–355. [Google Scholar] [CrossRef] [PubMed]
  167. Cristóvão, J.S.; Santos, R.; Gomes, C.M. Metals and Neuronal Metal Binding Proteins Implicated in Alzheimer’s Disease. Oxid. Med. Cell Longev. 2016, 2016, 9812178. [Google Scholar] [CrossRef] [Green Version]
  168. Yan, F.S.; Sun, J.L.; Xie, W.H.; Shen, L.; Ji, H.F. Neuroprotective Effects and Mechanisms of Curcumin-Cu(II) and -Zn(II) Complexes Systems and Their Pharmacological Implications. Nutrients 2017, 10, 28. [Google Scholar] [CrossRef] [Green Version]
  169. Parachikova, A.; Green, K.N.; Hendrix, C.; LaFerla, F.M. Formulation of a medical food cocktail for Alzheimer’s disease: Beneficial effects on cognition and neuropathology in a mouse model of the disease. PLoS ONE 2010, 5, e14015. [Google Scholar] [CrossRef]
  170. Guo, P.; Wang, D.; Wang, X.; Feng, H.; Tang, Y.; Sun, R.; Zheng, Y.; Dong, L.; Zhao, J.; Zhang, X.; et al. Effect and mechanism of fuzhisan and donepezil on the sirtuin 1 pathway and amyloid precursor protein metabolism in PC12 cells. Mol. Med. Rep. 2016, 13, 3539–3546. [Google Scholar] [CrossRef]
  171. Patel, N.V.; Gordon, M.N.; Connor, K.E.; Good, R.A.; Engelman, R.W.; Mason, J.; Morgan, D.G.; Morgan, T.E.; Finch, C.E. Caloric restriction attenuates Abeta-deposition in Alzheimer transgenic models. Neurobiol. Aging 2005, 26, 995–1000. [Google Scholar] [CrossRef]
  172. Sydow, A.; Van der Jeugd, A.; Zheng, F.; Ahmed, T.; Balschun, D.; Petrova, O.; Drexler, D.; Zhou, L.; Rune, G.; Mandelkow, E.; et al. Tau-induced defects in synaptic plasticity, learning, and memory are reversible in transgenic mice after switching off the toxic Tau mutant. J. Neurosci. 2011, 31, 2511–2525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Baum, L.; Lam, C.W.; Cheung, S.K.; Kwok, T.; Lui, V.; Tsoh, J.; Lam, L.; Leung, V.; Hui, E.; Ng, C.; et al. Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J. Clin. Psychopharmacol. 2008, 28, 110–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Perry, E.K.; Pickering, A.T.; Wang, W.W.; Houghton, P.J.; Perry, N.S. Medicinal plants and Alzheimer’s disease: From ethnobotany to phytotherapy. J. Pharm. Pharmacol. 1999, 51, 527–534. [Google Scholar] [CrossRef]
  175. Adams, M.; Gmünder, F.; Hamburger, M. Plants traditionally used in age related brain disorders—A survey of ethnobotanical literature. J. Ethnopharmacol. 2007, 113, 363–381. [Google Scholar] [CrossRef]
  176. Howes, M.J.; Houghton, P.J. Ethnobotanical treatment strategies against Alzheimer’s disease. Curr. Alzheimer Res. 2012, 9, 67–85. [Google Scholar] [CrossRef]
  177. Nabavi, S.F.; Braidy, N.; Orhan, I.E.; Badiee, A.; Daglia, M.; Nabavi, S.M. Rhodiola rosea L. and Alzheimer’s Disease: From Farm to Pharmacy. Phytother. Res. 2016, 30, 532–539. [Google Scholar] [CrossRef] [PubMed]
  178. Sutalangka, C.; Wattanathorn, J.; Muchimapura, S.; Thukham-mee, W. Moringa oleifera mitigates memory impairment and neurodegeneration in animal model of age-related dementia. Oxid. Med. Cell. Longev. 2013, 2013, 695936. [Google Scholar] [CrossRef] [Green Version]
  179. Farooqui, A.A.; Farooqui, T.; Madan, A.; Ong, J.H.; Ong, W.Y. Ayurvedic Medicine for the Treatment of Dementia: Mechanistic Aspects. Evid.-Based Complement. Altern. Med. 2018, 2018, 2481076. [Google Scholar] [CrossRef] [Green Version]
  180. Chauhan, N.B. Anti-amyloidogenic effect of Allium sativum in Alzheimer’s transgenic model Tg2576. J. Herb. Pharmacother. 2003, 3, 95–107. [Google Scholar] [CrossRef]
  181. Ray, B.; Chauhan, N.B.; Lahiri, D.K. The “aged garlic extract” (AGE) and one of its active ingredients S-allyl-L-cysteine (SAC) as potential preventive and therapeutic agents for Alzheimer’s disease (AD). Curr. Med. Chem. 2011, 18, 3306–3313. [Google Scholar] [CrossRef]
  182. Choi, S.J.; Lee, J.H.; Heo, H.J.; Cho, H.Y.; Kim, H.K.; Kim, C.J.; Kim, M.O.; Suh, S.H.; Shin, D.H. Punica granatum protects against oxidative stress in PC12 cells and oxidative stress-induced Alzheimer’s symptoms in mice. J. Med. Food 2011, 14, 695–701. [Google Scholar] [CrossRef] [PubMed]
  183. Ahmed, A.H.; Subaiea, G.M.; Eid, A.; Li, L.; Seeram, N.P.; Zawia, N.H. Pomegranate extract modulates processing of amyloid-β precursor protein in an aged Alzheimer’s disease animal model. Curr. Alzheimer Res. 2014, 11, 834–843. [Google Scholar] [CrossRef] [PubMed]
  184. Cao, C.; Li, Y.; Liu, H.; Bai, G.; Mayl, J.; Lin, X.; Sutherland, K.; Nabar, N.; Cai, J. The potential therapeutic effects of THC on Alzheimer’s disease. J. Alzheimer’s Dis. 2014, 42, 973–984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Scuderi, C.; Steardo, L.; Esposito, G. Cannabidiol promotes amyloid precursor protein ubiquitination and reduction of beta amyloid expression in SHSY5YAPP+ cells through PPARγ involvement. Phytother. Res. 2014, 28, 1007–1013. [Google Scholar] [CrossRef]
  186. Aso, E.; Sánchez-Pla, A.; Vegas-Lozano, E.; Maldonado, R.; Ferrer, I. Cannabis-based medicine reduces multiple pathological processes in AβPP/PS1 mice. J. Alzheimer’s Dis. 2015, 43, 977–991. [Google Scholar] [CrossRef] [Green Version]
  187. Esposito, G.; Scuderi, C.; Valenza, M.; Togna, G.I.; Latina, V.; De Filippis, D.; Cipriano, M.; Carratù, M.R.; Iuvone, T.; Steardo, L. Cannabidiol reduces Aβ-induced neuroinflammation and promotes hippocampal neurogenesis through PPARγ involvement. PLoS ONE 2011, 6, e28668. [Google Scholar] [CrossRef]
  188. Vallée, A.; Lecarpentier, Y.; Guillevin, R.; Vallée, J.N. Effects of cannabidiol interactions with Wnt/β-catenin pathway and PPARγ on oxidative stress and neuroinflammation in Alzheimer’s disease. Acta Biochim. Biophys. Sin. 2017, 49, 853–866. [Google Scholar] [CrossRef] [Green Version]
  189. Iuvone, T.; Esposito, G.; Esposito, R.; Santamaria, R.; Di Rosa, M.; Izzo, A.A. Neuroprotective effect of cannabidiol, a non-psychoactive component from Cannabis sativa, on beta-amyloid-induced toxicity in PC12 cells. J. Neurochem. 2004, 89, 134–141. [Google Scholar] [CrossRef]
  190. Esposito, G.; Scuderi, C.; Savani, C.; Steardo, L., Jr.; De Filippis, D.; Cottone, P.; Iuvone, T.; Cuomo, V.; Steardo, L. Cannabidiol in vivo blunts beta-amyloid induced neuroinflammation by suppressing IL-1beta and iNOS expression. Br. J. Pharmacol. 2007, 151, 1272–1279. [Google Scholar] [CrossRef] [Green Version]
  191. Wong, W. Economic burden of Alzheimer disease and managed care considerations. Am. J. Manag. Care 2020, 26, S177–S183. [Google Scholar] [CrossRef]
  192. Rafii, M.S.; Aisen, P.S. Alzheimer’s Disease Clinical Trials: Moving Toward Successful Prevention. CNS Drugs 2019, 33, 99–106. [Google Scholar] [CrossRef] [PubMed]
  193. Folch, J.; Petrov, D.; Ettcheto, M.; Abad, S.; Sánchez-López, E.; García, M.L.; Olloquequi, J.; Beas-Zarate, C.; Auladell, C.; Camins, A. Current Research Therapeutic Strategies for Alzheimer’s Disease Treatment. Neural Plast. 2016, 2016, 8501693. [Google Scholar] [CrossRef] [Green Version]
  194. Ngandu, T.; Lehtisalo, J.; Solomon, A.; Levälahti, E.; Ahtiluoto, S.; Antikainen, R.; Bäckman, L.; Hänninen, T.; Jula, A.; Laatikainen, T.; et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): A randomised controlled trial. Lancet 2015, 385, 2255–2263. [Google Scholar] [CrossRef]
  195. Zenaro, E.; Piacentino, G.; Constantin, G. The blood-brain barrier in Alzheimer’s disease. Neurobiol. Dis. 2017, 107, 41–56. [Google Scholar] [CrossRef] [Green Version]
  196. Türker, S.; Onur, E.; Ozer, Y. Nasal route and drug delivery systems. Pharm. World Sci. 2004, 26, 137–142. [Google Scholar] [CrossRef] [PubMed]
  197. Keller, L.A.; Merkel, O.; Popp, A. Intranasal drug delivery: Opportunities and toxicologic challenges during drug development. Drug Deliv. Transl. Res. 2021, 1, 1–23. [Google Scholar] [CrossRef] [PubMed]
  198. Miyake, M.M.; Bleier, B.S. The blood-brain barrier and nasal drug delivery to the central nervous system. Am. J. Rhinol. Allergy 2015, 29, 124–127. [Google Scholar] [CrossRef] [PubMed]
  199. Erdő, F.; Bors, L.A.; Farkas, D.; Bajza, Á.; Gizurarson, S. Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res. Bull. 2018, 143, 155–170. [Google Scholar] [CrossRef]
  200. Wu, H.; Hu, K.; Jiang, X. From nose to brain: Understanding transport capacity and transport rate of drugs. Expert Opin. Drug Deliv. 2008, 5, 1159–1168. [Google Scholar] [CrossRef]
  201. Akhondzadeh, S.; Abbasi, S.H. Herbal medicine in the treatment of Alzheimer’s disease. Am. J. Alzheimer’s Dis. Other Demen. 2006, 21, 113–118. [Google Scholar] [CrossRef]
  202. Iqbal, K.; Grundke-Iqbal, I. Alzheimer’s disease, a multifactorial disorder seeking multitherapies. Alzheimer’s Dement. 2010, 6, 420–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Chen, X.; Drew, J.; Berney, W.; Lei, W. Neuroprotective Natural Products for Alzheimer’s Disease. Cells 2021, 10, 1309. [Google Scholar] [CrossRef]
  204. Renaud, J.; Martinoli, M.G. Considerations for the Use of Polyphenols as Therapies in Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 1883. [Google Scholar] [CrossRef] [Green Version]
  205. Kumar, A.; Ahuja, A.; Ali, J.; Baboota, S. Curcumin-loaded lipid nanocarrier for improving bioavailability, stability and cytotoxicity against malignant glioma cells. Drug Deliv. 2016, 23, 214–229. [Google Scholar] [CrossRef] [PubMed]
  206. Smith, A.; Giunta, B.; Bickford, P.C.; Fountain, M.; Tan, J.; Shytle, R.D. Nanolipidic particles improve the bioavailability and alpha-secretase inducing ability of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer’s disease. Int. J. Pharm. 2010, 389, 207–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. The neuroprotective effects of herbs administrated for AD treatment. Neuroprotective herbs act via various mechanisms to elicit their ameliorative effects on AD patients. The neurotherapeutic effects of the herbs aid in the restoration and enhancement of memory and cognitive function.
Table 1. The neuroprotective effects of herbs administrated for AD treatment. Neuroprotective herbs act via various mechanisms to elicit their ameliorative effects on AD patients. The neurotherapeutic effects of the herbs aid in the restoration and enhancement of memory and cognitive function.
HerbNeuroprotective EffectType of Research StudyReferences
Withania somniferaAnti-inflammatory, antioxidant, inhibits abeta production and neurite outgrowths, boosts neural regeneration, reverses dysfunction of the mitochondria, enhances processing speed, social cognition, auditory–verbal working memoryIn vitro, in vivo, clinical research studies[50,51,52,53,54]
Bacopa monnieriAnti-inflammatory, enhances attention, memory, executive function, inhibits abeta production, enhances cardiac function, antioxidantIn vitro, in vivo, clinical research studies[55,56,57,58]
Gingko bilobaEnhances mitochondrial function, antioxidant, boosts neurogenesis, stimulates cerebral blood flowIn vitro, pre-clinical and clinical research studies[59,60,61,62]
Centella asiaticaDecreases oxidative stress, apoptosis, abeta levels, boosts dendritic growth, enhances memory and improves moodIn vitro, in vivo, clinical research studies[63,64,65,66]
Crocus sativusAntiamyloidogenic, antidepressant, neuroprotective effects, immune system modulation, antioxidantIn vitro, in vivo, clinical research studies[67,68,69,70]
Curcuma longaAnti-inflammatory, antioxidant, antimicrobial, inhibits abeta production, anti-apoptosisIn vitro, in vivo, pre-clinical and clinical research studies[71,72,73,74]
Fuzhisan (Anemone altaica, Glycyrrhiza uralensis, Scutellaria baicalensis, Panax ginseng)Anti-abeta accumulation, antiapoptosis, neurotropic effects, and enhances acetylcholine levelsin vitro, in vivo, clinical research studies[75,76,77]
Table 2. Neurotherapeutic effects of Withania somnifera, Bacopa monnieri, Gingko biloba, Centella asiatica, Crocus sativus, Curcuma longa, and Fuzhisan in randomized, placebo controlled human trials. In these trials, no severe adverse effects were noted for any of the administered extracts or natural compounds.
Table 2. Neurotherapeutic effects of Withania somnifera, Bacopa monnieri, Gingko biloba, Centella asiatica, Crocus sativus, Curcuma longa, and Fuzhisan in randomized, placebo controlled human trials. In these trials, no severe adverse effects were noted for any of the administered extracts or natural compounds.
HerbConducted ByStudy DesignSample
Size
Dosage
Regimen
Metrics TestedFindings
Withania somniferaPingali et al. [53]Randomized, placebo
controlled, double-blind study
20250 mg twice daily for 14 daysReaction time, choice discrimination, digit symbol substitution, digit vigilance, card sorting tests, finger-tapping testSignificant improvement in reaction time, choice discrimination, digit vigilance, card sorting tests, and digit symbol substation
No effect in
finger-tapping test
Bacopa
monnieri
Raghav et al. [58]Randomized, placebo
controlled, double-blind study
40125 mg twice daily for 12 weeks, followed by 4 weeks of placebo (16-week total duration)Mental control, logical memory, digit forward, digit backward, visual reproduction and paired-associate learningSignificant improvement in mental control, logical memory, and paired-associate learning
Gingko
biloba
Le Bars et al. [126]Randomized, placebo
controlled, double-blind study
309120 mg once daily for 52 weeksADAS-CogModest
improvement in cognitive performance measured by ADAS-Cog and noted by caregivers
Centella
asiatica
Wattanathorn et al. [136]Randomized, placebo
controlled, double-blind study
28250, 500, and 750 mg once daily for 8 weeksBond–Lader mood scale, alert factor, content factor, calm factor, word recognition, image recognitionIncreased accuracy in word recognition, image recognition, alertness, and calmness, with high dosage showing greater effects. No significant difference in content factor between treatment and placebo groups.
Crocus
sativus
Akhondzadeh et al. [69]Randomized, placebo
controlled, double-blind study
2215 mg twice daily for 22 weeksADAS-CogEffectively similar to 10 mg daily of donepezil in the ADAS-Cog scale
Curcuma longaBaum et al. [173]Randomized, placebo
controlled, double-blind study
341000 to 4000 mg once daily for 26 weeksADAS-CogNo cognitive decline in enrolled subjects during study duration
Fuzhisan (Anemone altaica, Glycyrrhiza uralensis, Scutellaria baicalensis, Panax ginseng)Bi et al. [76]Randomized, placebo
controlled, double-blind study
2210 mg once daily for 12 weeksADAS-Cog, neuropsychiatric indexSignificantly improved ADAS-Cog and neuropsychiatric index scores compared to placebo
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zieneldien, T.; Kim, J.; Cao, C. The Multifaceted Role of Neuroprotective Plants in Alzheimer’s Disease Treatment. Geriatrics 2022, 7, 24. https://0-doi-org.brum.beds.ac.uk/10.3390/geriatrics7020024

AMA Style

Zieneldien T, Kim J, Cao C. The Multifaceted Role of Neuroprotective Plants in Alzheimer’s Disease Treatment. Geriatrics. 2022; 7(2):24. https://0-doi-org.brum.beds.ac.uk/10.3390/geriatrics7020024

Chicago/Turabian Style

Zieneldien, Tarek, Janice Kim, and Chuanhai Cao. 2022. "The Multifaceted Role of Neuroprotective Plants in Alzheimer’s Disease Treatment" Geriatrics 7, no. 2: 24. https://0-doi-org.brum.beds.ac.uk/10.3390/geriatrics7020024

Article Metrics

Back to TopTop