Next Article in Journal
Differential Expression of miRNAs and Their Predicted Target Genes Indicates That Gene Expression in Atlantic Salmon Gill Is Post-Transcriptionally Regulated by miRNAs in the Parr-Smolt Transformation and Adaptation to Sea Water
Previous Article in Journal
Ketogenic and Low FODMAP Diet in Therapeutic Management of a Young Autistic Patient with Epilepsy and Dysmetabolism Poorly Responsive to Therapies: Clinical Response and Effects of Intestinal Microbiota
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Forest Biomass as a Promising Source of Bioactive Essential Oil and Phenolic Compounds for Alzheimer’s Disease Therapy

by
Patrícia Moreira
1,2,
Patrícia Matos
2,3,4,
Artur Figueirinha
2,3,4,
Lígia Salgueiro
2,4,
Maria Teresa Batista
4,
Pedro Costa Branco
5,
Maria Teresa Cruz
1,2 and
Cláudia Fragão Pereira
1,6,*
1
CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
2
Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
3
LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
4
CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
5
RAIZ—Forest and Paper Research Institute, 3800-783 Aveiro, Portugal
6
Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(15), 8812; https://doi.org/10.3390/ijms23158812
Submission received: 19 July 2022 / Revised: 4 August 2022 / Accepted: 6 August 2022 / Published: 8 August 2022
(This article belongs to the Section Molecular Biology)

Abstract

:
Alzheimer’s disease (AD) is the most common neurodegenerative disorder affecting elderly people worldwide. Currently, there are no effective treatments for AD able to prevent disease progression, highlighting the urgency of finding new therapeutic strategies to stop or delay this pathology. Several plants exhibit potential as source of safe and multi-target new therapeutic molecules for AD treatment. Meanwhile, Eucalyptus globulus extracts revealed important pharmacological activities, namely antioxidant and anti-inflammatory properties, which can contribute to the reported neuroprotective effects. This review summarizes the chemical composition of essential oil (EO) and phenolic extracts obtained from Eucalyptus globulus leaves, disclosing major compounds and their effects on AD-relevant pathological features, including deposition of amyloid-β (Aβ) in senile plaques and hyperphosphorylated tau in neurofibrillary tangles (NFTs), abnormalities in GABAergic, cholinergic and glutamatergic neurotransmission, inflammation, and oxidative stress. In general, 1,8-cineole is the major compound identified in EO, and ellagic acid, quercetin, and rutin were described as main compounds in phenolic extracts from Eucalyptus globulus leaves. EO and phenolic extracts, and especially their major compounds, were found to prevent several pathological cellular processes and to improve cognitive function in AD animal models. Therefore, Eucalyptus globulus leaves are a relevant source of biological active and safe molecules that could be used as raw material for nutraceuticals and plant-based medicinal products useful for AD prevention and treatment.

1. Introduction

Alzheimer’s disease (AD) is a multifactorial age-related neurodegenerative disorder that is characterized by loss of memory and impairment of other cognitive functions as well as behavioral alterations [1,2]. AD is the most common form of dementia in the elderly affecting 50 million people worldwide and is expected to impact on 152 million people in 2050 [3], highlighting the urgency of developing disease-modifying strategies able to prevent or delay its progression. The characteristic neuropathological hallmarks, which have been implicated in AD pathophysiology, are the intracellular accumulation of neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein and the extracellular deposition of amyloid-β (Aβ) peptide in senile plaques [4]. In addition to formation and deposition of Aβ and hyperphosphorylated tau, several other molecular alterations have been described in AD, including neuronal dystrophy, synaptic loss, oxidative stress, inflammation, and depletion of acetylcholine (ACh) levels, among many others [5]. In the last decades, several efforts have been made towards the identification of novel therapeutic targets in AD because the treatments approved by Food and Drug Administration (FDA) and European Medicines Agency (EMA) only alleviate symptoms without altering the progression of the disease and also have multiple side effects. Interestingly, two of the five approved drugs are from natural origin (galantamine and physostigmine-derivative rivastigmine) [6]; therefore, the relevance of plants as sources of new safe and multi-target therapeutic agents for AD has stimulated the research worldwide [7,8,9]. In accordance, over the past 20 years, about 50% of the over 1000 different compounds that have been studied as candidates for AD treatment are molecules obtained from natural sources, and some have already been tested in clinical trials [10,11]. Eucalyptus globulus (E. globulus) is a tree native to Australia and extensively cultivated in many countries of Europe, including Portugal [12]. The exploration of eucalyptus by the pulp, paper, and wood industry generates large quantities of residual biomass (bark, leaves, and branches) whose valorization can represent a significant contribution to the circular economy. The potential of some by-products of E. globulus as a source of bioactive compounds has been demonstrated. In fact, E. globulus leaves are traditionally used for treatment of respiratory threats. Furthermore, several compounds obtained from E. globulus leaves, such as essential oils (EOs) and phenolic compounds, have been shown to be important antimicrobial agents as well as to exhibit antioxidant and anti-inflammatory properties, among other relevant biological activities [12,13], reinforcing the interest in deepening further studies focused on this by-product. In fact, due to the already demonstrated properties, these compounds have been used against bacteria and fungi infections, for pain relief, and to deal with immune system-related diseases besides respiratory problems [14]. Additionally, there are some evidence that these compounds could be applied to the skin to deal with conditions, which is supported by our recent work demonstrating their beneficial effect against several skin alterations such as aging and pigmentation [15]. The therapeutic potential for AD of some of these compounds was also reported in a few studies due to their inhibitory effect on acetylcholinesterase (AChE) and neuroprotective effects [13,16]. This review summarizes the chemical composition and the potential of EOs and phenolic compounds extracted from E. globulus leaves, particularly the major components, as promising therapeutic agents for AD.

2. Chemical Composition of Essential Oil and Phenolic Compounds from E. globulus Leaves

The International Standard Organization on Essential Oils (ISO 9235: 2013) and the European Pharmacopoeia define an EO as the product obtained from plant raw material by hydrodistillation, steam distillation or dry distillation, or by a suitable mechanical process (for Citrus fruits). EOs are usually complex mixtures of volatile compounds present in different concentrations [17]. Monoterpenes and sesquiterpenes are usually the main groups found in EOs, and in some cases, phenylpropanoids are also important components [18]. The yield and chemical composition of EOs depend on several extrinsic (ecological and environmental aspects) and intrinsic (sexual, seasonal, ontogenetic, and genetic variations) factors [19]. Generally, the chemical characterization of EOs is performed by gas chromatography-mass spectrometry (GC-MS) techniques, and the quality of EOs is evaluated by comparison with analytical monographs published by the European Pharmacopoeia. This compendium determines the chromatographic profile of the EO obtained from E. globulus leaves by gas chromatography, establishing the range of the main constituents that the EO should contain at least 70% 1,8-cineole, 4–12% limonene, 1–9% α-pinene, less than 1.5% β-pinene and α-phellandrene, and less than 0.1% camphor. Concerning the EO yield, it is reported to range from 1.5 to 3.5%. Several chemical profiles have been described in the literature, with significant variations of the main compounds, as well as the EO yield, which can change according to several factors, namely the geographical region of the plants, the maturity state, and the condition of the leaves (fresh or dry), as summarized in the Table 1. According to these studies, the EOs of E. globulus leaves from several world regions are enriched in 1,8-cineole, and high content in this compound was reported in EOs obtained from this specie planted in Argentina (98.9%) [20], Tunisia (95.6%) [21], Italy (95.5%) [22], Brazil (90.0%) [23], Australia (90.0%) [24], India (85.0%) [25], Ethiopia (81.6%) [26], Morocco (80.0%) [27], Algeria (78.5%) [28], and Portugal (74.6%) [29]. Only two EOs have a completely different chemical profile with absence or very low levels of 1,8-cineole [30,31].
According to the Encyclopedia of Food Sciences and Nutrition, phenolic compounds present hydroxylated aromatic rings, in which the hydroxyl group is directly attached to the phenyl, substituted phenyl, or other aryl group [117]. Phenolic compounds are a large group of secondary metabolites produced by plants in response to environmental stresses, such as pathogen infection, high light, low temperatures, nutrient deficiency, and predators. Plants constitutively contain these compounds, which are a varied group of phytochemicals [118]. Phenolic compounds can be divided in several classes, namely phenolic acids (hydroxycinnamic acids, hydroxybenzoic acids), flavonoids, and tannins [119]. Usually, the analysis of the phenolic compounds is performed by high-performance liquid chromatography (HPLC), which offers high sensitivity and great efficiency, but gas chromatography and capillary electrophoresis can also be used. Different detection systems can be combined with these techniques, and mass spectrometry is the preferred system [120]. Regarding the phenolic compounds from E. globulus leaves, the chemical composition is very heterogeneous and can vary according to geographical region and the extractive solvent, as reported in several studies (Table 2). The predominant phenolic compounds from E. globulus leaves are phenolic acids, namely the ellagic acid and flavonoids, particularly of flavonol subclass, specifically quercetin and its glycoside rutin. Ellagitannins are predominant among the less abundant phenolic compounds in E. globulus leaves.
The more abundant compounds found in EOs and phenolic extracts obtained from E. globulus leaves (Figure 1) play an important role in their biological activities. The research of E. globulus has mainly focused on the composition and biological activities of EOs obtained from leaves, and only few studies disclosed the chemical composition of the leaves’ phenolic extracts and their biological properties. Therefore, since E. globulus extracts and EOs obtained from leaves are rich in bioactive compounds, their potential for formulation of food and plant-based medicinal products should be explored. However, the analysis of the biological properties of extracts and/or isolated compounds is a key step in assessing their potential of valorization.

3. Role of Essential Oil and Phenolic Compounds from E. globulus Leaves in Alzheimer’s Disease

Table 3 lists neuroprotective effects of EO, phenolic extracts and its major constituents obtained from E. globulus leaves against various neurodegeneration model systems.

3.1. Aβ Formation and Tau Hyperphosphorylation

According to the “Amyloid Cascade Hypothesis”, accumulation and oligomerization of Aβ peptide in the brain plays a major role in AD pathophysiology [131]. Aβ is a short fragment formed by the amyloidogenic proteolytic cleavage of the amyloid precursor protein (APP) [132] (Figure 2), which exhibits toxic effects on neuronal and glia cells in both oligomeric and fibrillar forms. Therefore, several approaches have been designed to decrease Aβ peptide formation from APP, and the most studied targets are β-secretase (BACE) and the γ-secretase complex. APP cleavage is performed by these two enzymes at variable sites to form numerous fragments of Aβ [133,134]. There are two isoforms of BACE [132]: BACE-1 [135] and BACE-2 [136]. The inhibition of BACE-1 is the most attractive therapeutic approach in AD because Aβ production from APP cleavage in the brain mainly results from the action of this β-secretase isoform. The membrane fragment formed upon BACE1 action is then cleaved by γ-secretase, generating Aβ fragments, namely Aβ1-40 and Aβ1-42 [137]. The inhibition of γ-secretase is also a valuable strategy but is less attractive than β-secretase due to fact that it is a multiprotein complex. Unfortunately, serious side effects were revealed in the clinical trials performed with secretase inhibitors [138,139] since the inhibition of these two enzymes can interfere with the processing of other substrates [140,141]. For example, γ-secretase inhibition has adverse side effects on Notch signaling that may cause severe gastrointestinal toxicity and the β-secretase inhibition can affect negatively the central or peripheral myelinization. Besides secretases inhibitors and modulators aimed to reduce Aβ formation, there are other therapeutic strategies under development to halt AD progression, such as prevention of Aβ oligomerization and aggregation into plaques, Aβ vaccination to promote Aβ clearance, and inhibition of its accumulation [142]. In fact, recently, FDA approved the commercialization of a new drug for AD treatment, the aducanumab, which is an antibody capable of removing Aβ plaques from the brain and the first drug capable of interfering with the neurodegenerative process of the disease.
Hyperphosphorylated tau-enriched NFTs are another neuropathological hallmark of AD. Under physiologic conditions, tau is the principal microtubule (MT)-associated protein that cooperates with tubulin to regulate MTs stability, which is crucial to axonal transport and thus to neuronal functioning [143]. In AD, hyperphosphorylated tau loses the capacity to bind MTs and forms NFTs that contribute to the neurodegenerative process [144,145] (Figure 3). Overproduction of inflammatory mediators has been shown to activate kinases such as cyclin-dependent kinase-5 (CDK-5) and glycogen synthase kinase-3β (GSK-3β), which consequently lead to tau phosphorylation [146,147]. In AD, GSK-3β plays a crucial role in tau hyperphosphorylation [148], but it was also demonstrated to contribute to Aβ aggregation and deposition into senile plaques [149]. With this in mind, GSK-3β inhibitors could represent a promising treatment strategy for AD.
In the last years, some studies revealed that phenolic compounds can interfere with both amyloid and tau pathologies, supporting their beneficial role in AD. However, there is no information in the literature about the effect of EO from E. globulus leaves and its major compound 1,8-cineole on AD. Ellagic acid was found as a potential BACE-1 inhibitor as well as a protective strategy against Aβ deposition and tau hyperphosphorylation. In a screening for anti-dementia agents from natural products, Kwak and collaborators (2005) reported that ellagic acid was a moderate BACE-1 specific inhibitor [150] and in vitro studies showed that ellagic acid promoted a significant loss of oligomers levels and was able to prevent Aβ-induced toxicity [151,152]. Accordingly, ellagic acid treatment in a sporadic AD rat model induced by streptozotocin (STZ) administration markedly decreased brain Aβ levels, suggesting its potential to delay amyloidogenesis [153]. Finally, it was reported that ellagic acid decreased APP and BACE-1 expression levels as well as Aβ deposition in the hippocampus of APP/PS1 transgenic mice, a model of familial AD [154]. This study also described the inhibition of tau hyperphosphorylation by ellagic acid mediated by the activation of the protein kinase B (Akt)/GSK-3β signaling pathway.
Regarding quercetin and rutin, several studies identified quercetin and rutin as BACE-1 inhibitors and provided strong evidences that both compounds are able to reduce Aβ deposition and quercetin to decrease tau hyperphosphorylation and aggregation, proving its neuroprotective effects. In different in vitro AD models, both compounds showed to prevent Aβ fibrils formation and cytotoxicity [155,156,157,158], and rutin was identified as a BACE-1 inhibitor that specifically prevents APP cleavage, decreasing production of the sAPPβ fragment [155,159,160]. In primary cortical neurons, quercetin was also described to act as a potent BACE-1 inhibitor and to decrease Aβ levels [161]. However, an in vitro study of Paris and co-authors provided evidences that quercetin inhibits Aβ and sAPPβ production by regulating BACE-1 expression and not by acting directly as an inhibitor of its activity [162]. Furthermore, a combined in vitro cell-based/in silico screening reported that quercetin shows potent Aβ anti-aggregation activity [163]. Moreover, in HT22 hippocampal neurons as well as in differentiated SH-SY5Y, quercetin reduced okadaic acid (OA)-induced tau hyperphosphorylation, inhibited the activity of CD-K5, attenuated the rise of intracellular calcium, and inhibited neuronal apoptosis via suppression of phosphoinositide 3-kinase (PI3K)/Akt/GSK-3β, mitogen-activated protein kinases (MAPKs), Bcl-2-associated X (BAX), and caspase-3 activities as well as nuclear factor-κB (NF-κB) activation [164,165,166]. Furthermore, quercetin was found to prevent tau phosphorylation through AMP-activated protein kinase (AMPK) activation and GSK-3β inhibition in OA-treated SH-SY5Y cells and in the hippocampus of mice fed with a high-fat diet [167]. Quercetin also inhibited Aβ fibrillization but not its toxic oligomerization in a C. elegans model of Aβ deposition [168] by activation of macroautophagy and proteasomal degradation pathways [169]. In accordance, senile plaques were reduced by quercetin in the cerebral cortex and hippocampus of APP/PS1 mice [170]. Other in vivo studies revealed that quercetin decreased extracellular β-amyloidosis, tauopathy, astrogliosis, and microgliosis in the hippocampus and amygdala of 3xTg-AD mice, decreasing the number of paired helical filaments (PHF), Aβ levels, and BACE1-mediated cleavage of APP [171,172]. In quercetin-treated 3xTg-AD mice, reactive microglia and Aβ aggregates were reduced [173], and the oral administration of quercetin increased brain apolipoprotein E (ApoE) and decreased Aβ levels in the cerebral cortex of 5xFAD mice model [174]. Moreover, increased Aβ clearance and decreased astrogliosis were observed in APP/PS1 mice receiving a quercetin-enriched diet during the early-middle stage of AD-like pathology progression [175]. In Aβ-injected mice, an animal model of sporadic AD, it was also demonstrated that protein levels of APP and BACE as well as of p-tau were reduced by quercetin [176]. Finally, it was recently reported that quercetin administration decreased the amount of Aβ in the hippocampal CA1 regions of Aβ-injected rats [177]. Additionally, the oral administration of rutin decreased oligomeric Aβ levels in brain of APP/PS1 transgenic mice [178].

3.2. Oxidative Stress

The presence of oxidative stress markers in the AD brain has been pointed out as another relevant AD hallmark. Oxidative stress is caused by an imbalance between the production of reactive oxygen species (ROS) and/or reactive nitrogen species (RNS) and the removal capacity of the antioxidant system, promoting damage to lipids, proteins, ribonucleic acids (RNA), and deoxyribonucleic acids (DNA) [179,180]. Despite the mechanisms by which the redox balance is altered in AD and the sources of ROS/RNS remain unknown, numerous studies suggest that Aβ is a potent trigger of oxidative stress that is, at least in part, mediated by the disruption of mitochondrial function and subsequent generation of oxidant species [181] (Figure 4). Therefore, development of novel antioxidant strategies is required to prevent AD progression.
Many studies reported the antioxidant properties of EOs from E. globulus, which contribute to its neuroprotective effects [17,116]. For example, an in vitro study performed by Mizuno (2015) found that hydrogen peroxide (H2O2)-induced neuronal death was attenuated by the EO of E. globulus [182]. Moreover, Yadav (2019) showed that E. globulus oil alleviated depressive and cognitive symptoms of ketamine-induced psychosis in rats mediated by its antioxidant effect in the cerebral cortex and hippocampus, where the levels of reduced glutathione (GSH) were restored [77]. In both studies, the reported protective effect of EO from E. globulus might be due to the presence of 1,8-cineole, which was shown to be the major component. In fact, Ryu (2014) showed that 1,8-cineole may attenuate oxidative stress in cortical neuronal/glial cells through its antioxidant capacity as ROS scavenger and activator of superoxide dismutase (SOD) [183]. Additionally, an in vitro study using a neuronal cell model, performed by Khan and colleagues in 2014, demonstrated that Aβ-induced neuronal toxicity was prevented by 1,8-cineole pretreatment. The loss of mitochondrial membrane potential as well as ROS accumulation were attenuated by 1,8-cineole, supporting its anti-oxidative properties [184]. On the other hand, as observed above in the previous section, α-pinene is also present in the EO of E. globulus leaves and in vivo studies revealed its antioxidant effect. Lee (2017) demonstrated that α-pinene increased protein levels of antioxidant enzymes, namely the heme oxygenase-1 (HO-1) and manganese superoxide dismutase (MnSOD) in the hippocampus of the scopolamine-induced AD mice model via activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) [185], which is a transcription factor that stimulates an antioxidant defense response. Nrf2 levels decrease with age, and reduced Nrf2 levels were reported in AD animal models and postmortem human brain tissue from patients [186]. Interestingly, recent studies revealed that Nrf2 activators may delay the progression and ameliorate the symptoms of the disease, suggesting that Nrf2 inducers might be relevant therapeutic molecules for AD [187].
González-Burgos (2018) investigated the antioxidant activity of different extracts (acetone, ethanol, and methanol) from E. globulus leaves and concluded that the extracts rich in phenolic compounds were effective to prevent H2O2-induced oxidative stress and preserve cell viability, increasing the activity of antioxidant enzymes and GSH levels as well as decreasing lipid peroxidation and ROS production in SH-SY5Y cells [13]. As mentioned before, the ellagic acid is one of the most predominant compounds found in phenolic extracts from E. globulus leaves. In fact, several studies reported the antioxidant properties of ellagic acid with significant impact on the progression of AD pathology, particularly through the activation of several antioxidant enzymes, reducing lipid peroxidation and free radical scavenging activity. Kabiraj and collaborators (2014) showed that ellagic acid is able to scavenge peroxynitrite, protecting PC12 cells against rotenone-induced cell death and also to reduce ROS and RNS production in these neuronal-like cells. Moreover, these authors demonstrated that ellagic acid suppressed apoptosis caused by rotenone by reducing poly (ADP-ribose) polymerase-1 (PARP) cleavage, which is a hallmark of apoptotic cell death [188]. Shen and co-authors (2017) also found that ellagic acid protected PC12 cells from Aβ-induced damage by inhibiting ROS production and reducing calcium ion influx [152]. Furthermore, ellagic acid pretreatment in intrahippocampal Aβ-microinjected rats, a model that mimics early-onset AD, mitigated oxidative stress by increasing the antioxidants catalase (CAT) and GSH and reducing the levels of malondialdehyde (MDA), a lipid peroxidation product [189]. Other study of Jha (2018) used STZ to induce a sporadic AD-like phenotype in rats and observed a decrease in oxidative stress profile after treatment with ellagic acid. Ellagic acid-treated animals revealed higher brain levels of mitochondrial ATPase and a marked dose-dependent free radical scavenging activity. In addition, this study reported attenuation of MDA levels together with an increase in GSH levels and activation of CAT in animals treated with STZ in the presence of ellagic acid [153]. Consistently, two other studies reported that ellagic acid administration reduced the production of thiobarbituric acid reactive substances (TBARS) and prevented the depletion of the antioxidant GSH and inhibition of SOD and CAT activities in STZ-treated rats [190,191]. Furthermore, an in vivo study performed by Uzar (2012) demonstrated that ellagic acid protected neurons against oxidative damage in STZ-induced diabetic rats, decreasing lipid peroxidation and total oxidant status and oxidative stress index. Additionally, ellagic acid attenuated the effects of STZ on activated CAT and paraoxanase-1 (PON-1) enzymes [192].
Quercetin and its glycoside rutin are two abundant compounds found in phenolic extracts of eucalyptus leaves, and several in vitro and in vivo studies have investigated their neuroprotective potential in AD. Both compounds were reported to attenuate oxidative stress in different AD models, mainly by decreasing ROS production and lipid peroxidation and increasing GSH content and the activity of several antioxidant enzymes. In APPswe cells, which are a cellular model of AD consisting of cells transfected with Swedish mutated human APP, Jimenez-Aliaga and collaborators (2011) demonstrated that quercetin and rutin decrease ROS generation and lipid peroxidation and increase intracellular GSH content, improving the redox status of APPswe cells treated with H2O2 [155]. In addition, rutin and quercetin were found to have free radical scavenging activity and to ameliorate Aβ-induced neuronal death in mouse primary cortical neuronal cultures [193]. Moreover, rutin attenuated mitochondrial damage and reduced the levels of ROS and oxidized glutathione (GSSG) as well as the formation of MDA and stimulated the activity of the antioxidant enzymes CAT, SOD, GSH, and glutathione peroxidase (GPx) in microglia cells exposed to Aβ [156]. Rutin was also demonstrated to inhibit amylin-induced neurotoxicity in SH-SY5Y cells, reducing the formation of ROS, GSSG, and MDA; attenuating mitochondrial damage and increasing the GSH/GSSG ratio; and enhancing the antioxidant activity of SOD, CAT, and GPx [194]. Additionally, quercetin was shown to preserve cell viability in PC12 cells treated with H2O2 [195]. An in vitro study with primary hippocampal cultures described that low doses of quercetin significantly attenuated Aβ-induced cytotoxicity, lipid peroxidation, protein oxidation, and apoptosis; however, higher dosages were reported to potentiate neuronal dysfunction [196]. Later studies demonstrated that quercetin protected rat primary hippocampal neurons against H2O2- or Aβ-induced neurotoxicity, attenuating ROS accumulation and depolarization of the mitochondrial membrane [197]. The role of quercetin in OA-induced oxidative stress in HT22 hippocampal cells was investigated, and it was found that pre-treatment with quercetin activates SOD, avoids GSH depletion, and decreases ROS production and MDA levels. The alterations in membrane potential caused by OA were reversed by quercetin, further supporting its neuroprotective action [164]. Quercetin was also reported to raise intracellular GSH content and prevent oxidative/nitrosative damage to DNA, lipids, and proteins in SH-SY5Y cells exposed to a neurotoxin [198]. On the other hand, rutin pretreatment was shown to decrease TBARS and PARP activity and increase GSH content and the activity of GPx, glutathione reductase, and CAT enzymes in the hippocampus of rats treated with STZ [199]. The effect of rutin was investigated in APPswe/PS1dE9 transgenic mice, and it was demonstrated that it decreased GSSG and MDA levels and increased SOD activity and GSH/GSSG ratio [178]. Moreover, lipid peroxidation was decreased in the brain, liver, and kidneys by treatment with rutin in an AD mouse model induced by Aβ injection [200]. Oxidative damage was attenuated by rutin treatment in rats with chronic cerebral hypoperfusion, namely GPx activity were increased, and the levels of MDA and protein carbonyls were decreased in rutin-treated animals [201]. Furthermore, it was recently demonstrated that pretreatment with rutin reduced CAT, GSH, and SOD protein levels in rats injected with doxorubicin [202]. Furthermore, an in vivo study performed by Tota and collaborators showed that quercetin restored cerebral blood flow and adenosine triphosphate (ATP) content after STZ administration in mice and reduced oxidative and nitrosative stress as demonstrated by a reduction in MDA and by an increase in GSH content [203]. It was also reported that quercetin treatment reduced MDA levels in the brain of STZ-induced diabetic rats [204]. In addition, quercetin decreased MDA generation in brain homogenates of mice treated with trimethyltin and showed strong antioxidant capacity determined through free radical scavenging activity assays [205]. Furthermore, lipid peroxidation was shown to be significantly inhibited by quercetin in the brain of Aβ-injected mice [206]. Indeed, increased SOD, CAT, and GSH and decreased MDA levels were observed in the brain of Aβ-injected rats treated with quercetin, concomitantly with activation of the antioxidant Nrf2/HO-1 pathway [177]. Quercetin ameliorated mitochondrial dysfunction, as evidenced by restoration of mitochondrial membrane potential and ROS and ATP levels in mitochondria isolated from the hippocampus of APP/PS1 transgenic mice. Furthermore, the activity of AMPK, which is a master regulator of cellular energy and metabolism, was significantly increased by quercetin [170]. Recent studies demonstrated that quercetin prevented the mitochondrial apoptotic pathway and neuronal degeneration by a mechanism involving regulation of BAX/Bcl2 and reduction of caspase-3 activity, cytochrome c release, and PARP cleavage in the brain of mice treated with lipopolysaccharide (LPS) [207]. Finally, reduction of MDA levels in animals injected with Aβ by rutin and quercetin were associated with upregulation of cAMP-response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) [208,209], which is an important regulator of neuronal growth and synaptic plasticity. CREB is one of the essential regulators of BDNF since its phosphorylated form binds to a specific sequence in the BDNF promoter and controls its transcription [210].
Several evidences support a crosstalk between oxidative stress and endoplasmic reticulum (ER) stress. In AD, the accumulation of misfolded proteins in susceptible brain regions suggests that the impairment of ER proteostasis machinery is involved in AD pathophysiology [211]. Therefore, ER stress can be considered as a therapeutic target for AD treatment. Under conditions of misfolded proteins overload within the ER lumen, ER stress sensors initiate the unfolded protein response (UPR) to reestablish homeostasis. This pathway comprises the activation of three ER trans-membrane proteins, namely inositol-requiring enzyme 1α (IRE1α), PKR-like ER kinase (PERK), and activating transcription factor 6 (ATF6) [210,212]. IRE1α activation promotes splicing of X-Box-binding protein 1 (XBP1)-mRNA [212] and the spliced XBP1 accumulated inside the nucleus upregulates crucial genes to reestablish global proteostasis under ER stress [213]. Furthermore, IRE1α can also activate relevant signaling mediators, namely c-Jun N-terminal kinase (JNK), which regulates autophagy and apoptosis [214]. ATF6 is an ER-membrane-bound transcription factor that triggers the transcription of ER molecular chaperones [210]. PERK also acts as an ER stress sensor, and under stress conditions, the α-subunit of eukaryotic initiation factor 2 (eIF2α) is oligomerized and phosphorylated by PERK [212]. This inhibits global protein translation, decreasing the overload of misfolded proteins [215,216]. Moreover, eIF2α phosphorylation increases translation of the activating transcription factor 4 (ATF4), which encodes genes of autophagy and proteins responsible for cell redox and metabolic regulation [216]. In addition, under chronic ER stress, ATF4 upregulates the transcription factor C/EBP homologous protein (CHOP), GADD34, and numerous members of Bcl2 family such as BAX and BAK, two central apoptotic regulators [217]. GADD34 can revert the eIF2α phosphorylation in a feed-forward cycle to close PERK signaling [218]. There are some evidences that quercetin ameliorates ER stress in AD models. In 2015, Hayakawa and colleagues reported that quercetin can rescue proteostasis, decreasing eIF2α phosphorylation, ATF4 expression, and Aβ secretion through GADD34 upregulation in cells upon autophagy impairment or ER stress conditions, which was confirmed in vivo using an AD mouse model [219]. In addition, quercetin repressed ER stress by reducing phosphorylation of eIF2α, PERK, and IRE1α; suppressed oxidative stress by reducing intracellular ROS production; and restored mitochondrial membrane potential in OA-treated SH-SY5Y cells. The same study also reported reduced IRE1α and PERK phosphorylation in mice exposed to high-fat diets [167]. A recent study performed by Woo and co-authors in Aβ-injected mice revealed that quercetin attenuates oxidative stress, namely ROS and TBARS generation. Under these conditions, a decrease was observed in the levels of ER stress markers such as phosphorylated eIF2α and PERK, XBP1, and CHOP as well as of pro-apoptotic Bax, phosphorylated JNK, and cleaved caspases-3 and -9 together with upregulation of the anti-apoptotic protein Bcl2 [176].

3.3. Inflammation

Recent evidences suggest that inflammation has a fundamental role in AD pathogenesis; therefore, controlling the interactions between the nervous and the immune system might be crucial to prevent or delay the disease [220]. Brain inflammation seems to play a neuroprotective role in acute-phase responses but becomes deleterious during a chronic response to toxic insults [221]. Activated microglia release a diversity of proinflammatory and toxic products, including ROS, nitric oxide (NO), and cytokines such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α), which play a significant role in the neuroinflammatory process. Aβ peptide increases the levels of cytokines, including TNF-α and IL-1β, and in turn, elevated levels of IL-1β potentiate Aβ accumulation [220,222]. Additionally, elevated levels of IL-1β can increase the production of other cytokines, such as IL-6, which activates the CDK-5 kinase that can lead to tau hyperphosphorylation [223]. Neuroinflammation has emerged as a third relevant hallmark in AD that can act as a link between amyloid and tau pathologies [224] (Figure 5). In fact, immune-related cells and proteins have been reported to be located within close proximity to senile plaques [225,226], and some evidences indicate that the prolonged use of nonsteroidal anti-inflammatory drugs (NSAIDs) reduce the risk to develop AD and delays the progression of the disease [227], possibly due to the inhibition of cyclooxygenases (COX) and activation of peroxisome proliferator-activated receptor γ (PPARγ) [227]. COX expression is repressed by NSAIDs, which declines the synthesis of prostaglandins and decreases the secretion of cytokines [228]. There are also evidences that NSAIDs decrease the level of Aβ in neuronal cell cultures and transgenic mice modelling AD [227]. Nevertheless, additional studies are required to confirm the beneficial effect of NSAIDs in AD.
E. globulus EO and its major component were recently reported to have anti-inflammatory activity relevant in the AD context. It has been previously reported that the expression of proinflammatory cytokines TNF-α, IL-1β, and IL-6 was lowered by 1,8-cineole in cells exposed to Aβ, and 1,8-cineole also succeeds in reducing NO accumulation and downregulating inducible NO synthase (iNOS), COX-2, and NF-κB [184]. More recently, the EO from E. globulus was demonstrated to reduce the serum levels of TNF-α in rats with psychosis, in the absence of any other significant alteration in inflammatory markers [77].
There are some evidences that extracts from eucalyptus leaves and ellagic acid reduce inflammation through depletion of TNF-α levels in AD models. Akhtar and collaborators extracted eucalyptus leaves with ethanol and detected anti-inflammatory activity, as shown by inhibition of TNF-α and NO production in macrophages exposed to LPS and interferon-γ (INF-γ) [229]. An in vitro study performed in cultured primary murine cortical microglia demonstrated that ellagic acid decreases Aβ-induced TNF-α secretion [230]. Another in vivo study showed that the reduction of hippocampal nuclear/cytoplasmatic Nrf2 ratio in Aβ-microinjected rats was reversed by ellagic acid treatment, which also reverted the alterations in NF-κB and TLR4 expression [189]. Moreover, ellagic acid was shown to prevent the accumulation of TNF-α detected in the STZ-induced AD rat model [190,191].
The anti-inflammatory effects of quercetin and rutin in AD models has been reported in several studies, which describe a decrease in NO production and in the expression of proinflammatory cytokines. Regarding in vitro studies, Wang and co-authors observed that rutin reduced NO formation and iNOS activity and also modulated the production of proinflammatory cytokines by decreasing TNF-α and IL-1β generation in microglia cells treated with Aβ [156]. Similarly, rutin was showed to reduce the production of NO, iNOS activity, and release of the pro-inflammatory cytokines TNF-α and IL-1β in amylin-treated SH-SY5Y cells, attenuating neurotoxicity [194]. Additionally, a study performed in LPS-stimulated microglia cells reported that rutin decreases expression levels of TNF- α, IL-1β, IL-6, and iNOS as well as the secretion of IL-6, TNF-α, and NO and increases the production of interleukin-10 (IL-10), the M2 regulatory cytokine, as well as arginase. Moreover, rutin also restored LPS-induced upregulation of COX-2, interleukin-18 (IL-18), and transforming growth factor-β (TGF-β) [231]. Similarly, in vitro studies have also linked quercetin’s neuroprotective effect with its anti-inflammatory activity. For example, quercetin was shown to prevent the release of TNF-α and IL-6 from activated microglia and astrocytes and attenuated the activation of proinflammatory signaling pathways such as MAPK and NF-κB [198]. Thioredoxin-interacting protein (TXNIP) is a crucial node in ER stress and NLR family pyrin domain containing 3 (NLRP3) inflammasome, which activates caspase-1, leading to IL-1β secretion to cause inflammation in cells or tissues [232]. NLRP3 inflammasome is a protein complex that comprises NLRP3, the adaptor protein apoptosis-associated speck-like protein containing a C-terminal caspase-activation-and-recruitment domain (CARD) (ASC), and the precursor pro-caspase-1. Consistent with this, quercetin suppressed TXNIP expression and NLRP3 inflammasome activation indicated by downregulation of NLRP3, ASC, and procaspase-1 in OA-treated SH-SY5Y cells. Quercetin effectively reduced IL-1β and IL-6 production in neuronal cells and restored NLRP3 activity and reduced IL-1β and TNF-α production in mice exposed to a high-fat diet [167]. Quercetin also attenuated neuroinflammation in a mouse model of AD decreasing IL-1β and monocyte chemoattractant protein-1 (MCP-1) levels [233]. A study using quercetin-treated 3xTg-AD mice showed a reduction in reactive microglia and astrocytes, glial fibrillary acidic protein (GFAP), iNOS, and COX-2 immunoreactivity as well as IL-1β levels in hippocampal lysates [173]. Quercetin also reduced LPS-induced gliosis and the levels of various inflammatory markers, such as TNF-α, COX-2, and iNOS, in the cortex and hippocampus of adult mice [207]. Finally, quercetin decreased NO formation in STZ and Aβ- injected mice [203,206]. In vivo studies with rutin also disclosed its anti-inflammatory activity in AD context. Indeed, rutin ameliorated STZ-induced inflammation in rats by decreasing NO levels and the expression of GFAP, interleukin-8 (IL-8), COX-2, iNOS, and NF-κB [199]. Rutin also inhibited glial activation; reduced the levels of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6; and prevented neuronal damage in rats with chronic cerebral hypoperfusion [201]. Other study showed that chronic treatment with rutin decreases TNF-α levels in the hippocampus and frontal cortex of rats injected with doxorubicin [202]. The oral administration of rutin was also found to downregulate microgliosis and astrocytosis and to reduce IL-1β and IL-6 levels in the brain of the APP/PS1 transgenic AD mice model [178]. Additionally, the NO formation was reduced by rutin in Aβ-injected mice [200].

3.4. Cholinesterase Activity

The “Cholinergic Hypothesis” is central to explain AD pathophysiology (Figure 6). This hypothesis considers that cholinergic neurons are affected in AD, leading to a decrease in the synthesis of the neurotransmitter ACh and subsequent release to the synaptic cleft, resulting in cognitive decline and memory loss [234,235]. Therefore, inhibitors of AChE and butyrylcholinesterase (BChE) enzymes that degrade ACh can represent a therapeutic strategy to increase the levels of ACh in the synaptic cleft and its binding to post-synaptic receptors, thus potentiating cholinergic neurotransmission. In fact, three of the four drugs approved for the relief of AD symptoms are AChE inhibitors, namely donepezil, rivastigmine, and galantamine [5]. The active sites of AChE/BChE enzymes bind these cholinesterase inhibitors in a reversible manner and avoid ACh degradation, facilitating cholinergic neurotransmission. Thus, AD symptoms are ameliorated due to the rise of ACh concentration in the synaptic cleft [236]. However, the efficacy of cholinesterase inhibitors in AD treatment is limited, and side effects have been reported, such as nausea, abdominal pain, diarrhea, dyspepsia, vomiting, and skin rash [237]. Hence, the discovery of new cholinesterase inhibitors from medicinal plant sources concomitantly presenting less adverse effects can be a valuable strategy.
The effective in vitro inhibition of AChE activity by E. globulus EO has been described [16]. Moreover, studies in cellular models detected anti-cholinesterase activity of 1,8-cineole and α-pinene [238,239]. In addition, the AChE inhibitory activity of eucalyptus EO in the hippocampus region of rat’s brain with psychotic symptoms was recently reported [77]. Additionally, mRNA levels of enzymes involved in ACh metabolism were evaluated in the cortex of scopolamine-induced amnesic animals, and it was observed that the administration of α-pinene reverted the decrease in the mRNA levels of choline acetyltransferase (ChAT), which is responsible for the formation of ACh [185]. However, mRNA levels of AChE were not altered by scopolamine treatment in the presence or absence of α-pinene. These studies revealed the neuroprotective potential of E. globulus EO and its major compounds due to their capacity to inhibit AChE activity.
Ellagic acid was recently described to reduce AChE activity in the brain of animals injected with Aβ [189] or with STZ [153,190], supporting the ability of ellagic acid to reduce cerebral ACh degradation and its neuroprotective role.
Quercetin and rutin also demonstrated to inhibit AChE activity, revealing neuroprotective effects, particularly in AD. In fact, a study of Ademosun and colleagues showed that both compounds significantly decrease AChE and BChE activities in rat brain homogenates, but quercetin showed a higher inhibitory ability than rutin [240]. One docking study concluded that rutin exhibited an elevated docking score against AChE in comparison with quercetin, suggesting that rutin is a promising drug candidate for AD [241]. Rutin treatment was also found to alleviate ACh depletion and ChAT inhibition as well as the activation of AChE caused by cerebral hypoperfusion in rats [201]. On the other hand, in vitro studies demonstrated that quercetin has a strong inhibitory effect against AChE and BChE enzymes [160,242,243], and a relevant role of quercetin as an AChE inhibitor has been described, supporting its therapeutic potential for AD [244,245,246]. Accordingly, several other in vitro studies found similar or higher AchE inhibitory activity of quercetin over conventional AchE inhibitors [247]. It was observed that quercetin has significant AChE inhibitory activity almost similar to that of huperzine A [248] or donepezil [249], which are well-known AChE inhibitors. In addition, these results were confirmed in vivo, and quercetin has been reported to attenuate the AChE activity in the brain of STZ-treated mice [203,204]. Another study revealed that quercetin suppressed AChE activation in a dose-dependent manner in brain tissues of mice exposed to neurotoxic trimethyltin [205]. Finally, Liu and co-authors showed that quercetin was able to restore cortical ACh levels and inhibit AChE activity in Aβ-injected mice [209].

3.5. GABAergic and Glutamatergic Dysfunction

γ-Aminobutyric acid (GABA) is a major inhibitory neurotransmitter in the human brain, which plays a relevant role in cognitive functions [250] (Figure 7A). Significant reductions in cerebral GABA levels have been described in AD patients as well as in AD animal models [251]. GABAA is one isoform of GABA receptors, and some studies have demonstrated decreased GABAA/benzodiazepine (BZD) receptor density [252,253] and expression levels [254] in the brain of AD patients. Interestingly, the role of selective GABAA agonists to counteract Aβ-induced toxicity was showed [255] suggesting that the GABAergic system is involved in the pathophysiology of AD and therefore may be a potential therapeutic target for this neurodegenerative disorder. Recently, it was found that eucalyptus oil increases brain GABA levels [77], and α-pinene acts as a partial modulator of GABAA-BZD receptors and binds directly to the BZD binding site of the GABAA receptor [256].
Glutamate is an excitatory neurotransmitter typically present in the hippocampus and cerebral cortex that plays an important role in learning and memory [257] (Figure 7B). There are two types of post-synaptic glutamate receptors, ionotropic and metabotropic G protein-coupled receptors, which modulate calcium and sodium influx into neuronal cells [258]. However, excessive activation of glutamate receptors, in particular the N-methyl-D-aspartate (NMDA) subtype of ionotropic receptors, provokes excitotoxic neuronal death [259]. In AD, an excessive activation of the NMDA receptor has been described and contributes to the neurodegenerative process in consequence of the excessive influx of calcium [259]. Numerous evidences suggest that blocking excitotoxicity might be beneficial in AD. Indeed, memantine, which was approved by FDA and EMA for the treatment of AD symptoms, is an uncompetitive NMDA receptor antagonist that blocks excitoxicity with minimal side effects due to the preservation of normal glutamatergic transmission [260]. A study using computational models proposed 1,8-cineole as a good candidate for NMDA antagonism comparing its molecular features with the conventional ligand memantine [261].
These promising findings suggest that the effect of E. globulus EO on GABAergic and glutamatergic transmission should also be explored as therapeutic strategies for AD. Nevertheless, there is no information in the literature about phenolic compounds and AD-associated perturbation of GABAergic and glutamatergic neurotransmission.

3.6. Impaired Learning and Memory

Learning is the process of acquiring new information, while memory is the process of storing this information to use it for future purposes (Figure 8). Cognition is defined as the combination of learning and memory and is strictly dependent on the concerted action of several neurotransmitters.
In AD, the stage and severity of the disease are determined by the compromise in cognition [17]. The deterioration of cholinergic neurons have been reported to be implicated in cognitive deficits in AD patients [262]. Accordingly, anticholinergic agents such as scopolamine have been reported to induce memory deficits [263], and on the other hand, an improvement of the cholinergic system can revert alterations in cognition [264]. Based on this, and as previously stated, AChE and BChE inhibitors demonstrated to revert cognitive symptoms and have been approved for AD treatment.
EO from E. globulus was recently demonstrated to be able to restore learning and memory function in rats treated with ketamine that induces psychosis [77]. In addition, the administration of α-pinene attenuated learning and memory impairments induced in rats treated with scopolamine [185].
Several in vivo studies also support the beneficial effects of ellagic acid in cognition of AD animal models. For example, it was demonstrated that ellagic acid efficiently prevents scopolamine- and diazepam-induced cognitive impairments without affecting animals’ locomotion [265]. Treatment with ellagic acid also showed to ameliorate memory and spatial learning alterations in the APP/PS1 transgenic AD mice model [154]. Moreover, the study of Kiasalari and collaborators described that ellagic acid ameliorates learning and memory performance in Aβ-injected rats [189]. Regarding the STZ-induced AD rat model, it was observed that ellagic acid prevents the STZ-induced cognitive deficits in animals without affecting locomotor activity and motor coordination [153,190,191].
Strong evidences support that quercetin and rutin prevent cognitive impairments in several AD animal models. Pretreatment with quercetin and rutin prevented scopolamine-induced memory impairment in zebrafish without locomotor alterations [266]. Rutin also ameliorated deficits in learning and memory in STZ rats [199] as well as the problems in spatial learning and memory, in working memory, and also in contextual memory in rats with chronic cerebral hypoperfusion [201]. In AD transgenic mice, it was demonstrated that rutin decreased spatial memory deficits [178] and alleviated cognition and memory impairments in Aβ-injected mice [200]. Furthermore, rutin restored short- and long-term episodic memory in scopolamine- and doxorubicin-treated rats without interfering with the locomotor activity of the animals [202,267]. Quercetin administration in aged and LPS-treated mice also enhanced the memory capacity in the absence of alterations in locomotion [207,268]. Furthermore, quercetin avoided STZ-induced memory impairment in mice [203] and enhanced spatial memory in rats [269]. Quercetin also prevented the impairment of memory and the anxiogenic-like behavior induced in STZ-diabetic rats [204]. Additionally, quercetin treatment attenuated trimethyltin-induced memory impairment in mice [205]. Moreover, in a study with mice exposed to a high-fat diet, quercetin administration enhanced cognition [167]. Another study also showed that a quercetin-enriched diet during the early-middle pathology stages ameliorated cognitive dysfunction in APP/PS1 mice [175]. In addition, beneficial effects of quercetin in learning, memory deficits, and cognitive function were demonstrated in APP/PS1, APP23, and 3xTg-AD transgenic mice models of AD [170,171,172,219]. Furthermore, quercetin administration in Aβ-induced amnesic mice enhanced learning and memory performance [206,209,270]. Finally, two studies with rats injected with Aβ also demonstrated the capacity of quercetin to enhance learning and memory [177,271]. Importantly, in early-stage AD patients, memory recall assessed using the Revised Hasegawa Dementia Scale was demonstrated to be enhanced by the intake of quercetin [272].
Table 3. Neuroprotective effects of EO, phenolic extracts, and the major constituents obtained from E. globulus leaves against various neurodegeneration model systems.
Table 3. Neuroprotective effects of EO, phenolic extracts, and the major constituents obtained from E. globulus leaves against various neurodegeneration model systems.
CompoundModelDose and DurationEffectsReference
EOCell freeIC50 = 0.1298 mg/mLInhibited AChE activity[16]
GT1-7 cells treated with H2O225 ppm, 24 hAttenuated neuronal death[182]
Wistar albino rats treated with ketamine500 and 1000 mg/kg/day, p.o., 21 daysFacilitated GABA release, increased GSH levels, inhibited dopamine neurotransmission, decreased TNF-α levels, and diminished AChE activity
Restored learning and memory function
[77]
Computational-Candidate for NMDA antagonism[261]
CineolCell freeIC50 = 840 μMInhibited AChE activity[239]
Differentiated PC12 cells treated with Aβ25-352.5, 5 and 10 μM, 24 hRestored cell viability
Reduced mitochondria membrane potential and ROS and NO levels
Lowered expression of TNF-α, IL-1β, IL-6, iNOS, COX-2, and NF-κB
[184]
Primary rat cortical neurons/glial10 μM, 4 hIncreased SOD activity and reduced ROS production[183]
α-pineneComputational-Partially modulated GABAA-BZD receptors
Directly bound to the BZD binding site of GABAA receptor
[256]
Brain slices10 µM
C57BL/6N mice treated with pentobarbital100 mg/kg, p.o.
C57BL/6 mice treated with scopolamine10 mg/kg, i.p.Improved cognitive dysfunction
Increased expression of ChAT
Increased protein levels of antioxidant enzymes
Activated Nrf2
[185]
Phenolic extractsSH-SY5Y cells treated with H2O25, 10, 25 and 50 µg/mL, 24 hIncreased cell viability, GSH levels, and antioxidant enzymes activity
Decreased ROS production and lipid peroxidation levels
[13]
RAW264.7 cells treated with LPS and INF-γ51 and 83 µg/mL, 24 hInhibited NO and TNF-α production[229]
Ellagic acidCell freeIC50 = 39 μMInhibited BACE-1 activity[150]
SH-SY5Y cells treated with Aβ1-425 and 10 μM, 48 hPromoted oligomers loss
Prevented neuronal death
[151]
PC12 cells treated with Aβ25-350.5, 2.5, and 5 µM, 12 hAttenuated Aβ-induced toxicity
Inhibited ROS production and reduced calcium ion influx
PC12 cells treated with rotenone10 µM, 24 hAttenuated cell death
Reduced ROS and RNS production
Suppressed apoptosis
[188]
Primary murine cortical microglia treated with Aβ1-4210 µM, 24 hDecreased TNF-α secretion[230]
APP/PS1 transgenic mice50 mg/kg/day, i.g., 60 daysAmeliorated learning and memory deficits
Reduced neuronal apoptosis and amyloid deposition
Inhibited tau hyperphosphorylation and decreased GSK-3β activity
[154]
Wistar rats treated with Aβ25-3550 and 100 mg/kg/day, i.p., 7 daysImproved learning and memory deficits
Mitigated oxidative stress by increasing CAT and GSH and reducing MDA levels
Reduced AChE activity
Modulated NF-κB/Nrf2/TLR4 signaling pathway
[189]
Wistar rats treated with STZ50 mg/kg/day, p.o., 30 daysDecreased brain Aβ levels
Revealed marked dose-dependent free radical scavenging effect and higher BMA levels
Reduced AChE activity
Prevented cognitive disfunction
[153]
Wistar rats treated with STZ35 mg/kg/day, p.o., 4 weeksReduced TBARS production and prevented the depletion of GSH and the inhibition of SOD and CAT activities
Increased TNF-α levels
Reduced AChE activity
Restored memory deficits
[190]
Wistar rats treated with STZ17.5 and 35 mg/kg/day, p.o., 28 daysReduced TBARS production and prevented the depletion of GSH
Increased TNF-α levels
Restored memory deficits
[191]
Diabetic rats treated with STZ50 mg/kg/day, p.o., 21 daysDecreased lipid peroxidation and oxidative stress index
Increased antioxidant enzymes
Attenuated NO production
[192]
Wistar rats treated with scopolamine and diazepam30 and 100 mg/kg/day, i.p., 10 daysPrevented cognitive deficits[265]
QuercetinComputational-Candidate as AChE inhibitor [247,249]
Cell free50 µMInhibited and destabilized Aβ fibril formation [158]
Cell free1 mg/mL (76.2% and 46.8% inhibition)Inhibited AChE and BChE activities[242]
Cell freeIC50 = 181 µM and
IC50 = 203 µM
Inhibited AChE and BChE activities[240]
Cell freeIC50 = 354 µM and
IC50 = 421 µM
Inhibited AChE and BChE activities[243]
Cell freeIC50 = 19.8 µMInhibited AChE activity[244]
Cell freeIC50 = 3.6 µMInhibited AChE activity[245]
Cell freeIC50 = 14.4 µMInhibited AChE activity[246]
Cell freeIC50 = 51 µMInhibited AchE activity[248]
Cell free
Bacterial cells
IC50 = 124.6 μMDecreased Aβ aggregation [163]
Cell free-Inhibited Aβ fibril formation[157]
HT22 cells treated with Aβ25-35-Attenuated neuronal death
HT22 cells treated with OA5 and 10 µM, 12 hAttenuated neuronal death
Decreased levels of SOD, mitochondria membrane potential, GPx, MDA, and ROS
Inhibited hyperphosphorylation of tau protein
Inhibited apoptosis via the reduction of Bax and up-regulation of cleaved caspase 3 via the inhibition of PI3K/Akt/GSK-3β, MAPKs, and activation of NF-κB
[164]
HT22 cells treated with OA5 and 10 µM, 12 hAttenuated tau protein hyperphosphorylation
Inhibited the activity of CD-K5
Attenuated intracellular calcium rise
[165]
Differentiated SH-SY5Y cells treated with OA100 nM, 6 hDecreased tau phosphorylation levels[166]
SH-SY5Y cells treated with OA10 µM, 6 hSuppressed ER stress with decreased phosphorylation of IRE1α and PERK
Decreased ROS production and restored mitochondria membrane potential
Inhibited TXNIP and NLRP3 inflammasome activation and downregulated ASC and pro-caspase-1
[167]
C57BL/6J mice exposed to high-fat diets50 mg/kg/day, p.o., 10 weeksReduced IL-1β and IL-6 production
Attenuated tau phosphorylation
Reduced IL-1β and TNF-α production
Enhanced AMPK activity
Inhibited IRE1α and PERK phosphorylation, NLRP3 expression, and tau phosphorylation
Improved cognitive disorder
Cell free1, 5 and 10 µMInhibited the formation of Aβ fibrils and disaggregated Aβ fibrils[155]
APPswe-transfected SH-SY5Y cells25, 50, and 100 nM, 24 hDecreased ROS production and lipid peroxidation
Increased GSH content and the redox status
Cell freeIC50 = 55 μM and IC50 = 19 µM
IC50 = 0.55 µM
Inhibited AChE and BChE activities
Inhibited BACE activity
[160]
SH-SY5Y cells treated with L-DOPA10, 50, 250, and 1000 µM, 24 hAttenuated neuronal death
7W CHO cells
overexpressing APP
10, 25, and 50 µM, 24 hInhibited Aβ and sAPPβ production
Regulated BACE expression
[162]
SH-SY5Y cells treated with TNF-α20 µM, 30 min
SH-SY5Y, U373, and THP-1 cells treated with LPS and INF-γ or INF-γ33 µM, 8 hReduced oxidative/nitrative damage to DNA, lipids, and proteins
Increased intracellular GSH content
Reduced the release of TNF-α and IL-6
Attenuated the activation of MAPK and NF-kB
[198]
PC12 cells treated with H2O210, 30, 60 and 100 µM, 2 hPreserved cell viability[195]
Cell freeIC50 = 5.4 μMInhibited BACE activity[161]
Primary rat E18 cortical neurons20 μM, 24 hDecreased Aβ levels
Primary rat hippocampal neurons treated with Aβ1-425 and 10 μM, 24 hAttenuated neuronal death, protein oxidation, lipid peroxidation, and apoptosis[196]
Primary rat hippocampal neurons treated with Aβ1-42 and H2O210 μM, 24 hAttenuated neuronal death, ROS accumulation, and depolarization of mitochondrial membrane[197]
Primary mouse cortical neurons treated with
Aβ25-35
30 μM, 24 and 48 hDemonstrated free radical scavenging activity
Ameliorated neuronal death
[193]
Cell free250 µMInhibited Aβ fibrilization[168]
C. elegans treated with Aβ1-4273 µM, ~12 daysIncreased % of survival
C. elegans treated with Aβ1-42100 µM, 48 hIncreased proteasomal activity
Enhanced the flow of proteins through the macroautophagy
pathway
[169]
Zebrafish treated with scopolamine50 mg/kg/single dose, i.p.Attenuated memory deficits[266]
APP/PS1 transgenic mice20 and 40 mg/kg/day, 16 weeksImproved cognitive deficits
Reduced scattered senile plaques
Ameliorated mitochondrial dysfunction by restoration of mitochondrial
membrane potential, ROS, and ATP levels
Increased AMPK activity
[170]
APP/PS1 transgenic mice2 mg/g diet, 12 monthsIncreased Aβ clearance and reduced astrogliosis
Ameliorated cognitive dysfunction
[175]
APP/PS1 transgenic mice1% in mouse chow, 10 monthsAttenuated neuroinflammation by reducing IL-1β and MCP-1 levels[233]
APP23 transgenic mice0.5% in mouse chow, 52 weeksReduced eIF2α phosphorylation and ATF4 expression through GADD34 induction
Improved memory deficits
[219]
3xTg-AD mice25 mg/kg/48 h, i.p., 3 monthsDecreased extracellular β-amyloidosis, tauopathy, astrogliosis, and microgliosis
Reduced PHF and Aβ levels
Decreased BACE-1-mediated cleavage of APP
Improved performance on learning and spatial memory
[171]
3xTg-AD mice100 mg/kg/48 h, p.o., 12 months
Reduced β-amyloidosis and tauopathy
Improved cognitive deficits
[172]
3xTg-AD mice25 mg/kg/48 h,
i.p., 3 months
Decreased reactive microglia and Aβ
Reduced GFAP, iNOS, COX-2, and IL-1β
[173]
5xFAD mice500 mg/kg/day, oral gavage, 10 daysIncreased brain ApoE and reduced Aβ levels[174]
ICR mice injected with Aβ1-42 50 and 100 mg/kg/day, p.o., 1 monthImproved learning and memory loss[270]
ICR mice injected with Aβ25-3550 mg/kg/day, p.o., 2 weeksDecreased protein levels of APP, BACE, and p-tau
Reduced oxidative stress such as ROS and TBARS levels
Decreased the protein levels of ER stress markers GRP78, p-PERK, p-eIF2α, XBP1, and CHOP and the proapoptotic molecules Bax, p-JNK, and cleaved caspases-3 and -9
[176]
Mice injected with Aβ25-35 30 mg/kg/day, p.o., 14 daysDecreased NO formation and lipid peroxidation
Improved cognitive function
[206]
Kunming mice injected with Aβ25-355, 10, 20 and 40 mg/kg/day, oral gavage, 8 days Regulated ERK/CREB/BDNF pathway
Restored ACh levels and inhibited AChE activity
Improved the learning and memory capabilities
[209]
ICR mice treated with trimethyltin5, 10, and 20 mg/kg/day, 3 weeksDecreased MDA generation and showed antioxidant capacity
Inhibited AChE activity
Improved cognitive deficits
[205]
C57BL/6N mice treated with LPS30 mg/kg/day, i.p., 2 weeksPrevented the mitochondrial apoptotic pathway and neuronal degeneration by regulating Bax/Bcl2, decreasing activated cytochrome c and caspase-3 activity, and cleaving PARP-1
Reduced activated gliosis and levels of various inflammatory markers such as TNF-α, COX-2, and iNOS
Improved memory performance
[207]
Swiss mice treated with LPS25, 50, and 100 mg/kg/day, i.p., 7 daysReversed memory deficits[268]
Sprague–Dawley rats injected with Aβ1-42 100 mg/kg/day, p.o., 19 daysReduced Aβ levels
Increased SOD, CAT, and GSH and decreased MDA levels
Increased Nrf2/HO-1 pathway
Improved cognitive deficits
[177]
Wistar rats injected with Aβ1-4240 mg/kg/day, p.o., 1 monthAlleviated learning and memory deficits[271]
Wistar rats treated with STZ5, 25, and 50 mg/kg/day, oral gavage, 40 daysReduced MDA levels
Prevented the increase in AChE activity
Prevented memory deficits
[204]
Wistar rats treated with STZ40 and 80 mg/kg/day, i.p., 12 daysEnhanced spatial memory[269]
Human early-stage
AD patients
80 mg/patient/day, p.o., 4 weeksEnhanced memory recall [272]
RutinComputational-Candidate as AChE inhibitor[241]
Cell free10 µMInhibited BACE activity[159]
Cell freeIC50 = 0.219 mM and IC50 = 0.288 mMInhibited AChE and BChE activities[240]
Cell free
APPswe-transfected SH-SY5Y cells
1, 5, and 10 µM
100 µM
25, 50, and 100 nM, 24 h
Inhibited the formation of Aβ fibrils and disaggregated Aβ fibrils
Inhibited BACE activity
Decreased ROS production and lipid peroxidation
Increased GSH content and the redox status
[155]
Cell free50 and 200 µMInhibited Aβ fibrillization and attenuated Aβ-induced cytotoxicity[156]
SH-SY5Y and BV-2 cells treated with Aβ1-420.8 and 8 µM, 24 hDecreased ROS, NO, GSSG, and MDA formation
Reduced iNOS activity and attenuated mitochondrial damage
Increased GSH/GSSG ratio
Enhanced SOD, CAT, and GPx activities
Decreased TNF-α and IL-1β generation
Cell free
SH-SY5Y cells treated with L-DOPA
IC50 = 3.8 nM
10, 50, 250, and 1000 µM, 24 h
Inhibited BACE activity
Attenuated neuronal death
[160]
SH-SY5Y cells treated with amylin0.8, 4, and 8 µM, 24 and 48 hAttenuated neuronal death
Decreased the production of ROS, NO, GSSG, MDA, and pro-inflammatory cytokines TNF-α and IL-1β
Attenuated mitochondrial damage and increased the GSH/GSSG ratio
Enhanced the antioxidant enzyme activity of SOD, CAT, and GPx
Reduced iNOS activity
[194]
Primary mouse cortical neurons treated with
Aβ25-35
30 μM, 24 and 48 hDemonstrated free radical scavenging activity
Ameliorated neuronal death
[193]
Primary rat microglia treated with LPS50 mM, 24 hDecreased expression levels of TNF- α, IL-1β, IL-6, and iNOS
Reduced the production of IL-6, TNF-α, and NO
Increased production of the M2 regulatory cytokine IL-10 and arginase
Restored upregulation of COX-2, IL-18, and TGF-β
[231]
Zebrafish treated with scopolamine50 mg/kg/single dose, i.p.Attenuated memory deficits[266]
APP/PS1 transgenic mice100 mg/kg/day, p.o., 6 weeksDecreased oligomeric Aβ level
Increased SOD activity and GSH/GSSG ratio
Reduced GSSG and MDA levels
Downregulated microgliosis and astrocytosis
Decreased IL-1β and IL-6 levels
Attenuated memory deficits
[178]
ICR mice injected with Aβ25-35100 mg/kg/day, p.o., 14 daysDecreased NO formation and lipid peroxidation
Attenuated cognitive deficits
[200]
Swiss albino mice treated with STZ2.5, 5, and 10 mg/kg/day, p.o., 21 daysRestored cerebral blood flow and ATP content
Reduced MDA and NO levels and increased GSH content
Attenuated elevated AChE activity
Prevented memory impairment
[203]
Wistar rats injected with Aβ1-42100 mg/kg/day, i.p., 3 weeksIncreased ERK, CREB, and BDNF expression and decreased MDA level
Improved memory deficits
[208]
Wistar rats treated with STZ25 mg/kg/day, p.o., 3 weeksDecreased TBARS, PARP activity, and NO level
Increased GSH content and activities of GPx, glutathione reductase, and CAT
Reduced the expression of COX-2, GFAP, IL-8, iNOS, and NF-kB
Improved cognitive deficits
[199]
Sprague–Dawley rats with chronic
cerebral hypoperfusion
50 mg/kg/day, i.p., 12 weeksAttenuated oxidative damage, namely increased GPx activity and decreased MDA levels and protein carbonyls
Inhibited glial activation; reduced the levels of TNF-α, IL-1β, and IL-6; and prevented neuronal damage
Alleviated ACh depletion, ChAT inhibition, and AChE activation
Improved cognitive deficits
[201]
Wistar rats injected with doxorubicin50 mg/kg/day, p.o., 50 daysReduced CAT, GSH, SOD, and TNF-α levels
Prevented memory deficits
[202]
Wistar rats treated with scopolamine50 and 100 mg/kg/day, p.o., 15 daysImproved short- and long-term episodic memory deficits[267]

4. Conclusions

Several efforts have been made to develop alternatives to the current therapies for AD treatment, which only alleviate symptoms without altering the progression of the disease, increasing efficiency, and decreasing side effects. A promising strategy to identify novel disease-modifying therapies is to test compounds extracted from natural resources. The present review discusses findings obtained in in vitro and in vivo studies performed with EO, phenolic extracts, and the major constituents obtained from E. globulus leaves in what concerns therapeutic effect and mechanism of action. Overall, 1,8-cineole was found to be the major compound present in EO, and ellagic acid, quercetin, and rutin are the main components of phenolic extracts from E. globulus, which were demonstrated to efficiently prevent or attenuate several AD-related hallmarks, namely amyloid and tau pathologies, oxidative stress and neuroinflammation, neurotransmission deficits, and also memory and learning impairments. The information reviewed herein suggests that extracts from E. globulus leaves could be used as raw material to develop efficient and safe nutraceuticals and/or plant-based medicinal products useful for AD prevention and novel therapies able to modify the progression of the disease. However, further studies are required to further confirm the beneficial effects described for extracts from E. globulus leaves in AD.

Author Contributions

Conceptualization, M.T.C. and C.F.P.; formal analysis, P.M. (Patrícia Moreira) and P.M. (Patrícia Matos); investigation, P. Moreira; writing—original draft preparation, P.M. (Patrícia Moreira); writing—review and editing, P.M. (Patrícia Moreira), A.F., L.S., M.T.B., M.T.C. and C.F.P.; supervision, P.C.B., M.T.C. and C.F.P.; funding acquisition, P.C.B., M.T.C. and C.F.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was carried out under the Project inpactus—innovative products and technologies from eucalyptus, Project NO. 21874 (POCI-01-0247-FEDER-021874), funded by Portugal 2020 through the European Regional Development Fund (ERDF) in the frame of COMPETE 2020 NO. 246/AXIS II/2017 and Portuguese national funds via the Foundation for Science and Technology (FCT) through the projects UIDB/04539/2020 and UIDP/04539/2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. De Strooper, B.; Karran, E. The Cellular Phase of Alzheimer’s Disease. Cell 2016, 164, 603–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mirakhur, A.; Craig, D.; Hart, D.J.; McLlroy, S.P.; Passmore, A.P. Behavioural and psychological syndromes in Alzheimer’s disease. Int J. Geriatr. Psychiatry 2004, 19, 1035–1039. [Google Scholar] [CrossRef] [PubMed]
  3. Patterson, C. World Alzheimer Report 2018: The State of the Art of Dementia Research: New Frontiers; Alzheimer’s Disease International (ADI): London, UK, 2018. [Google Scholar]
  4. Braak, H.; de Vos, R.A.; Jansen, E.N.; Bratzke, H.; Braak, E. Neuropathological hallmarks of Alzheimer’s and Parkinson’s diseases. Prog. Brain. Res. 1998, 117, 267–285. [Google Scholar] [PubMed]
  5. Sharma, P.; Srivastava, P.; Seth, A.; Tripathi, P.N.; Banerjee, A.G.; Shrivastava, S.K. Comprehensive review of mechanisms of pathogenesis involved in Alzheimer’s disease and potential therapeutic strategies. Prog. Neurobiol. 2019, 174, 53–89. [Google Scholar] [CrossRef] [PubMed]
  6. Russo, P.; Frustaci, A.; Del Bufalo, A.; Fini, M.; Cesario, A. Multitarget drugs of plants origin acting on Alzheimer’s disease. Curr. Med. Chem. 2013, 20, 1686–1693. [Google Scholar] [CrossRef]
  7. Kalasz, H.; Ojha, S.; Tekes, K.; Szoke, E.; Mohanraj, R.; Fahim, M.; Adeghate, E.; Adem, A. Pharmacognostical Sources of Popular Medicine to Treat Alzheimer’s Disease. Open Med. Chem. J. 2018, 12, 23–35. [Google Scholar] [CrossRef] [Green Version]
  8. Park, S.Y. Potential therapeutic agents against Alzheimer’s disease from natural sources. Arch. Pharm. Res. 2010, 33, 1589–1609. [Google Scholar] [CrossRef]
  9. Ansari, N.; Khodagholi, F. Natural products as promising drug candidates for the treatment of Alzheimer’s disease: Molecular mechanism aspect. Curr. Neuropharmacol. 2013, 11, 414–429. [Google Scholar] [CrossRef] [Green Version]
  10. Cacabelos, R.; Torrellas, C.; Carrera, I.; Cacabelos, P.; Corzo, L.; Fernandez-Novoa, L.; Tellado, I.; Carril, J.C.; Aliev, G. Novel Therapeutic Strategies for Dementia. CNS Neurol. Disord. Drug Targets 2016, 15, 141–241. [Google Scholar] [CrossRef]
  11. Koynova, R.; Tenchov, B. Natural Product Formulations for the Prevention and Treatment of Alzheimer’s disease: A Patent Review. Recent. Pat. Drug Deliv. 2018, 12, 23–39. [Google Scholar] [CrossRef]
  12. Barbosa, L.C.; Filomeno, C.A.; Teixeira, R.R. Chemical Variability and Biological Activities of Eucalyptus spp. Essential Oils. Molecules 2016, 21, 1671. [Google Scholar] [CrossRef] [Green Version]
  13. Gonzalez-Burgos, E.; Liaudanskas, M.; Viskelis, J.; Zvikas, V.; Janulis, V.; Gomez-Serranillos, M.P. Antioxidant activity, neuroprotective properties and bioactive constituents analysis of varying polarity extracts from Eucalyptus globulus leaves. J. Food Drug Anal. 2018, 26, 1293–1302. [Google Scholar] [CrossRef] [PubMed]
  14. Dhakad, A.K.; Pandey, V.V.; Beg, S.; Rawat, J.M.; Singh, A. Biological, medicinal and toxicological significance of Eucalyptus leaf essential oil: A review. J. Sci. Food Agric. 2018, 98, 833–848. [Google Scholar] [CrossRef] [PubMed]
  15. Moreira, P.; Sousa, F.J.; Matos, P.; Brites, G.S.; Goncalves, M.J.; Cavaleiro, C.; Figueirinha, A.; Salgueiro, L.; Batista, M.T.; Branco, P.C.; et al. Chemical Composition and Effect against Skin Alterations of Bioactive Extracts Obtained by the Hydrodistillation of Eucalyptus globulus Leaves. Pharmaceutics 2022, 14, 561. [Google Scholar] [CrossRef]
  16. Aazza, S.; Lyoussi, B.; Miguel, M.G. Antioxidant and antiacetylcholinesterase activities of some commercial essential oils and their major compounds. Molecules 2011, 16, 7672–7690. [Google Scholar] [CrossRef] [Green Version]
  17. Ayaz, M.; Sadiq, A.; Junaid, M.; Ullah, F.; Subhan, F.; Ahmed, J. Neuroprotective and Anti-Aging Potentials of Essential Oils from Aromatic and Medicinal Plants. Front. Aging Neurosci. 2017, 9, 168. [Google Scholar] [CrossRef] [Green Version]
  18. Bakkali, F.; Averbeck, S.; Averbeck, D.; Idaomar, M. Biological effects of essential oils—A review. Food Chem. Toxicol. 2008, 46, 446–475. [Google Scholar] [CrossRef]
  19. Faleiro, M.L.; Miguel, M.G. Use of Essential Oils and Their Components against Multidrug-Resistant Bacteria. In Fighting Multidrug Resistance with Herbal Extracts, Essential Oils and Their Components; Rai, M.K., Kon, K.V., Eds.; Academic Press: Cambridge, MA, USA, 2013; pp. 65–94. [Google Scholar]
  20. Zunino, M.P.; Areco, V.A.; Zygadlo, J.A. Insecticidal activity of three essential oils against two new important soybean pests: Sternechus pinguis (Fabricius) and Rhyssomatus subtilis Fiedler (Coleoptera: Curculionidae). Boletín Latinoam. Caribe Plantas Med. Aromáticas 2012, 11, 269–277. [Google Scholar]
  21. Noumi, E.; Snoussi, M.; Hajlaoui, H.; Trabelsi, N.; Ksouri, R.; Valentin, E.; Bakhrouf, A. Chemical composition, antioxidant and antifungal potential of Melaleuca alternifolia (tea tree) and Eucalyptus globulus essential oils against oral Candida species. J. Med. Plants Res. 2011, 5, 4147–4156. [Google Scholar]
  22. Tardugno, R.; Pellati, F.; Iseppi, R.; Bondi, M.; Bruzzesi, G.; Benvenuti, S. Phytochemical composition and in vitro screening of the antimicrobial activity of essential oils on oral pathogenic bacteria. Nat. Prod. Res. 2018, 32, 544–551. [Google Scholar] [CrossRef]
  23. Vilela, G.R.; de Almeida, G.S.; D’Arce, M.A.B.R.; Moraes, M.H.D.; Brito, J.O.; da Silva, M.F.d.G.; Silva, S.C.; de Stefano Piedade, S.M.; Calori-Domingues, M.A.; da Gloria, E.M. Activity of essential oil and its major compound, 1, 8-cineole, from Eucalyptus globulus Labill., against the storage fungi Aspergillus flavus Link and Aspergillus parasiticus Speare. J. Stored Prod. Res. 2009, 45, 108–111. [Google Scholar] [CrossRef]
  24. Yang, Y.C.; Choi, H.Y.; Choi, W.S.; Clark, J.M.; Ahn, Y.J. Ovicidal and adulticidal activity of Eucalyptus globulus leaf oil terpenoids against Pediculus humanus capitis (Anoplura: Pediculidae). J. Agric. Food Chem. 2004, 52, 2507–2511. [Google Scholar] [CrossRef] [PubMed]
  25. Chauhan, N.; Malik, A.; Sharma, S. Repellency potential of essential oils against housefly, Musca domestica L. Environ. Sci. Pollut. Res. Int. 2018, 25, 4707–4714. [Google Scholar] [CrossRef] [PubMed]
  26. Manguro, L.O.; Opiyo, S.A.; Asefa, A.; Dagne, E.; Muchori, P.W. Chemical Constituents of Essential Oils from Three Eucalyptus Species Acclimatized in Ethiopia and Kenya. J. Essent. Oil Bear. Plants 2010, 13, 561–567. [Google Scholar] [CrossRef]
  27. Ait-Ouazzou, A.; Loran, S.; Bakkali, M.; Laglaoui, A.; Rota, C.; Herrera, A.; Pagan, R.; Conchello, P. Chemical composition and antimicrobial activity of essential oils of Thymus algeriensis, Eucalyptus globulus and Rosmarinus officinalis from Morocco. J. Sci. Food Agric. 2011, 91, 2643–2651. [Google Scholar] [CrossRef]
  28. Atmani-Merabet, G.; Belkhiri, A.; Dems, M.A.; Lalaouna, A.; Khalfaoui, Z.; Mosbah, B. Chemical composition, toxicity, and acaricidal activity of Eucalyptus globulus essential oil from Algeria. Curr. Issues Pharm. Med. Sci. 2018, 31, 89–93. [Google Scholar] [CrossRef] [Green Version]
  29. Vieira, M.; Bessa, L.J.; Martins, M.R.; Arantes, S.; Teixeira, A.P.S.; Mendes, A.; Martins da Costa, P.; Belo, A.D.F. Chemical Composition, Antibacterial, Antibiofilm and Synergistic Properties of Essential Oils from Eucalyptus globulus Labill. and Seven Mediterranean Aromatic Plants. Chem. Biodivers. 2017, 14, e1700006–e1700018. [Google Scholar] [CrossRef] [Green Version]
  30. Djenane, D.; Yangüela, J.; Amrouche, T.; Boubrit, S.; Boussad, N.; Roncalés, P. Chemical composition and antimicrobial effects of essential oils of Eucalyptus globulus, Myrtus communis and Satureja hortensis against Escherichia coli O157: H7 and Staphylococcus aureus in minced beef. Food Sci. Technol. Int. 2011, 17, 505–515. [Google Scholar] [CrossRef]
  31. Singh, A.; Ahmad, A.; Bushra, R. Supercritical carbon dioxide extraction of essential oils from leaves of Eucalyptus globulus L., their analysis and application. Anal. Methods 2016, 8, 1339–1350. [Google Scholar] [CrossRef]
  32. Benayache, S.; Benayache, F.; Benyahia, S.; Chalchat, J.-C.; Garry, R.-P. Leaf oils of some Eucalyptus species growing in Algeria. J. Essent. Oil Res. 2001, 13, 210–213. [Google Scholar] [CrossRef]
  33. Harkat-Madouri, L.; Asma, B.; Madani, K.; Said, Z.B.-O.S.; Rigou, P.; Grenier, D.; Allalou, H.; Remini, H.; Adjaoud, A.; Boulekbache-Makhlouf, L. Chemical composition, antibacterial and antioxidant activities of essential oil of Eucalyptus globulus from Algeria. Ind. Crops Prod. 2015, 78, 148–153. [Google Scholar] [CrossRef]
  34. Nadjib, B.M.; Amine, F.M.; Abdelkrim, K.; Fairouz, S.; Maamar, M. Liquid and vapour phase antibacterial activity of Eucalyptus globulus essential oil= susceptibility of selected respiratory tract pathogens. Am. J. Infect. Dis. 2014, 10, 105. [Google Scholar] [CrossRef] [Green Version]
  35. Daroui-Mokaddem, H.; Kabouche, A.; Bouacha, M.; Soumati, B.; El-Azzouny, A.; Bruneau, C.; Kabouche, Z. GC/MS analysis and antimicrobial activity of the essential oil of fresh leaves of Eucalytus globulus, and leaves and stems of Smyrnium olusatrum from Constantine (Algeria). Nat. Prod. Commun. 2010, 5, 1934578X1000501031. [Google Scholar] [CrossRef] [Green Version]
  36. Toudert-Taleb, K.; Hedjal-Chebheb, M.; Hami, H.; Debras, J.; Kellouche, A. Composition of essential oils extracted from six aromatic plants of Kabylian origin (Algeria) and evaluation of their bioactivity on Callosobruchus maculatus (Fabricius, 1775) (Coleoptera: Bruchidae). Afr. Entomol. 2014, 22, 417–427. [Google Scholar] [CrossRef]
  37. Viturro, C.I.; Molina, A.C.; Heit, C.I. Volatile components of Eucalyptus globulus Labill ssp. bicostata from Jujuy, Argentina. J. Essent. Oil Res. 2003, 15, 206–208. [Google Scholar] [CrossRef]
  38. Alzogaray, R.A.; Lucia, A.; Zerba, E.N.; Masuh, H.M. Insecticidal activity of essential oils from eleven Eucalyptus spp. and two hybrids: Lethal and sublethal effects of their major components on Blattella germanica. J. Econ. Entomol. 2011, 104, 595–600. [Google Scholar] [CrossRef]
  39. Lucia, A.; Licastro, S.; Zerba, E.; Masuh, H. Yield, chemical composition, and bioactivity of essential oils from 12 species of Eucalyptus on Aedes aegypti larvae. Entomol. Exp. Appl. 2008, 129, 107–114. [Google Scholar] [CrossRef]
  40. Toloza, A.C.; Lucía, A.; Zerba, E.; Masuh, H.; Picollo, M.I. Eucalyptus essential oil toxicity against permethrin-resistant Pediculus humanus capitis (Phthiraptera: Pediculidae). Parasitol. Res. 2010, 106, 409–414. [Google Scholar] [CrossRef]
  41. Peschiutta, M.L.; Pizzolitto, R.P.; Ordano, M.A.; Zaio, Y.; Zygadlo, J.A. Laboratory evaluation of insecticidal activity of plant essential oils against the vine mealybug, Planococcus ficus. J. Insect Sci. 2017, 56, 79–83. [Google Scholar]
  42. Yeom, H.-J.; Kang, J.; Kim, S.-W.; Park, I.-K. Fumigant and contact toxicity of Myrtaceae plant essential oils and blends of their constituents against adults of German cockroach (Blattella germanica) and their acetylcholinesterase inhibitory activity. Pestic. Biochem. Physiol. 2013, 107, 200–206. [Google Scholar] [CrossRef]
  43. Lee, S.-E.; Choi, W.-S.; Lee, H.-S.; Park, B.-S. Cross-resistance of a chlorpyrifos-methyl resistant strain of Oryzaephilus surinamensis (Coleoptera: Cucujidae) to fumigant toxicity of essential oil extracted from Eucalyptus globulus and its major monoterpene, 1, 8-cineole. J. Stored Prod. Res. 2000, 36, 383–389. [Google Scholar] [CrossRef]
  44. Aldoghaim, F.S.; Flematti, G.R.; Hammer, K.A. Antimicrobial activity of several cineole-rich Western Australian Eucalyptus essential oils. Microorganisms 2018, 6, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Park, H.-M.; Kim, J.; Chang, K.-S.; Kim, B.-S.; Yang, Y.-J.; Kim, G.-H.; Shin, S.-C.; Park, I.-K. Larvicidal activity of Myrtaceae essential oils and their components against Aedes aegypti, acute toxicity on Daphnia magna, and aqueous residue. J. Med. Entomol. 2011, 48, 405–410. [Google Scholar] [CrossRef] [PubMed]
  46. Baranska, M.; Schulz, H.; Reitzenstein, S.; Uhlemann, U.; Strehle, M.; Krüger, H.; Quilitzsch, R.; Foley, W.; Popp, J. Vibrational spectroscopic studies to acquire a quality control method of Eucalyptus essential oils. Biopolym. Orig. Res. Biomol. 2005, 78, 237–248. [Google Scholar] [CrossRef]
  47. Mayaud, L.; Carricajo, A.; Zhiri, A.; Aubert, G. Comparison of bacteriostatic and bactericidal activity of 13 essential oils against strains with varying sensitivity to antibiotics. Lett. Appl. Microbiol. 2008, 47, 167–173. [Google Scholar] [CrossRef]
  48. Araújo, A.; Oliveira, J.V.d.; França, S.M.; Navarro, D.M.; Barbosa, D.R.; Dutra, K.d.A. Toxicity and repellency of essential oils in the management of Sitophilus zeamais. Rev. Bras. Eng. Agrícola Ambient. 2019, 23, 372–377. [Google Scholar] [CrossRef] [Green Version]
  49. Maciel, M.; Morais, S.; Bevilaqua, C.; Silva, R.; Barros, R.; Sousa, R.; Sousa, L.; Brito, E.; Souza-Neto, M. Chemical composition of Eucalyptus spp. essential oils and their insecticidal effects on Lutzomyia longipalpis. Vet. Parasitol. 2010, 167, 1–7. [Google Scholar] [CrossRef] [Green Version]
  50. Ambrosio, C.M.; de Alencar, S.M.; Moreno, A.M.; Da Gloria, E.M. Evaluation of the selective antibacterial activity of Eucalyptus globulus and Pimenta pseudocaryophyllus essential oils individually and in combination on Enterococcus faecalis and Lactobacillus rhamnosus. Can. J. Microbiol. 2018, 64, 844–855. [Google Scholar] [CrossRef]
  51. Castro, J.C.; Endo, E.H.; de Souza, M.R.; Zanqueta, E.B.; Polonio, J.C.; Pamphile, J.A.; Ueda-Nakamura, T.; Nakamura, C.V.; Dias Filho, B.P.; de Abreu Filho, B.A. Bioactivity of essential oils in the control of Alternaria alternata in dragon fruit (Hylocereus undatus Haw.). Ind. Crops Prod. 2017, 97, 101–109. [Google Scholar] [CrossRef]
  52. Mossi, A.J.; Astolfi, V.; Kubiak, G.; Lerin, L.; Zanella, C.; Toniazzo, G.; de Oliveira, D.; Treichel, H.; Devilla, I.A.; Cansian, R. Insecticidal and repellency activity of essential oil of Eucalyptus sp. against Sitophilus zeamais Motschulsky (Coleoptera, Curculionidae). J. Sci. Food Agric. 2011, 91, 273–277. [Google Scholar] [CrossRef]
  53. de Godoi, S.N.; Quatrin, P.M.; Sagrillo, M.R.; Nascimento, K.; Wagner, R.; Klein, B.; Santos, R.C.V.; Ourique, A.F. Evaluation of stability and in vitro security of nanoemulsions containing Eucalyptus globulus oil. BioMed Res. Int. 2017, 2017, 2723418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Galli, G.M.; Roza, L.F.; Santos, R.C.; Quatrin, P.M.; Ourique, A.F.; Klein, B.; Wagner, R.; Baldissera, M.D.; Volpato, A.; Campigotto, G. Low Dose of Nanocapsules Containing Eucalyptus Oil Has Beneficial Repellent Effect against Horn Fly (Diptera: Muscidae). J. Econ. Entomol. 2018, 111, 2983–2987. [Google Scholar] [CrossRef]
  55. Goldbeck, J.C.; do Nascimento, J.E.; Jacob, R.G.; Fiorentini, Â.M.; da Silva, W.P. Bioactivity of essential oils from Eucalyptus globulus and Eucalyptus urograndis against planktonic cells and biofilms of Streptococcus mutans. Ind. Crops Prod. 2014, 60, 304–309. [Google Scholar] [CrossRef]
  56. Tomazoni, E.Z.; Pauletti, G.F.; da Silva Ribeiro, R.T.; Moura, S.; Schwambach, J. In vitro and in vivo activity of essential oils extracted from Eucalyptus staigeriana, Eucalyptus globulus and Cinnamomum camphora against Alternaria solani Sorauer causing early blight in tomato. Sci. Hortic. 2017, 223, 72–77. [Google Scholar] [CrossRef]
  57. Pedrotti, C.; Marcon, Â.R.; Delamare, A.P.L.; Echeverrigaray, S.; da Silva Ribeiro, R.T.; Schwambach, J. Alternative control of grape rots by essential oils of two Eucalyptus species. J. Sci. Food Agric. 2019, 99, 6552–6561. [Google Scholar] [CrossRef]
  58. Tomazoni, E.Z.; Griggio, G.S.; Broilo, E.P.; da Silva Ribeiro, R.T.; Soares, G.L.G.; Schwambach, J. Screening for inhibitory activity of essential oils on fungal tomato pathogen Stemphylium solani Weber. Biocatal. Agric. Biotechnol. 2018, 16, 364–372. [Google Scholar] [CrossRef]
  59. Pansera, M.R.; Conte, R.I.; e Silva, S.M.; Sartori, V.C.; silva Ribeiro, R.T. Strategic control of postharvest decay in peach caused by Monilinia fructicola and Colletotrichum gloeosporioides. Appl. Res. Agrotechnol. 2015, 8, 7–14. [Google Scholar]
  60. Miyamoto, C.; De Sant’anna, J.R.; Franco, C.D.S.; Cunico, M.; Miguel, O.; Côcco, L.; Yamamoto, C.; Corrêa, C.; de Castro-Prado, M. Genotoxic activity of Eucalyptus globulus essential oil in Aspergillus nidulans diploid cells. Folia Microbiol. 2009, 54, 493–498. [Google Scholar] [CrossRef]
  61. Bueno, P.C.P.; Junior, M.G.; Bastos, J.K. A GC-FID validated method for the quality control of Eucalyptus globulus raw material and its pharmaceutical products, and GC-MS fingerprinting of 12 Eucalyptus species. Nat. Prod. Commun. 2014, 9, 1934578X1400901233. [Google Scholar] [CrossRef] [Green Version]
  62. Sameza, M.L.; Boat, M.A.B.; Nguemezi, S.T.; Mabou, L.C.N.; Dongmo, P.M.J.; Boyom, F.F.; Menut, C. Potential use of Eucalyptus globulus essential oil against Phytophthora colocasiae the causal agent of taro leaf blight. Eur. J. Plant Pathol. 2014, 140, 243–250. [Google Scholar] [CrossRef]
  63. Bittner, M.L.; Casanueva, M.E.; Arbert, C.C.; Aguilera, M.A.; Hernández, V.J.; Becerra, J.V. Effects of essential oils from five plant species against the granary weevils Sitophilus zeamais and Acanthoscelides obtectus (Coleoptera). J. Child Chem. Soc. 2008, 53, 1444–1448. [Google Scholar] [CrossRef] [Green Version]
  64. González, A.; Gutiérrez-Cutiño, M.; Moenne, A. Oligo-carrageenan kappa-induced reducing redox status and increase in TRR/TRX activities promote activation and reprogramming of terpenoid metabolism in Eucalyptus trees. Molecules 2014, 19, 7356–7367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Wang, K.; Tang, L.; Zhang, N.; Zhou, Y.; Li, W.; Li, H.; Cheng, D.; Zhang, Z. Repellent and fumigant activities of Eucalyptus globulus and Artemisia carvifolia essential oils against Solenopsis invicta. Bull. Insectol. 2014, 67, 207–211. [Google Scholar]
  66. Liu, Z.; Chen, Z.; Han, F.; Kang, X.; Gu, H.; Yang, L. Microwave-assisted method for simultaneous hydrolysis and extraction in obtaining ellagic acid, gallic acid and essential oil from Eucalyptus globulus leaves using Brönsted acidic ionic liquid [HO3S (CH2) 4mim] HSO4. Ind. Crops Prod. 2016, 81, 152–161. [Google Scholar] [CrossRef]
  67. España, M.D.; Arboleda, J.W.; Ribeiro, J.A.; Abdelnur, P.V.; Guzman, J.D. Eucalyptus leaf byproduct inhibits the anthracnose-causing fungus Colletotrichum gloeosporioides. Ind. Crops Prod. 2017, 108, 793–797. [Google Scholar] [CrossRef]
  68. Cimanga, K.; Kambu, K.; Tona, L.; Apers, S.; De Bruyne, T.; Hermans, N.; Totté, J.; Pieters, L.; Vlietinck, A.J. Correlation between chemical composition and antibacterial activity of essential oils of some aromatic medicinal plants growing in the Democratic Republic of Congo. J. Ethnopharmacol. 2002, 79, 213–220. [Google Scholar] [CrossRef]
  69. Sacchetti, G.; Maietti, S.; Muzzoli, M.; Scaglianti, M.; Manfredini, S.; Radice, M.; Bruni, R. Comparative evaluation of 11 essential oils of different origin as functional antioxidants, antiradicals and antimicrobials in foods. Food Chem. 2005, 91, 621–632. [Google Scholar] [CrossRef]
  70. Elzayyat, E.; Elleboudy, N.; Moustafa, A.; Ammar, A. Insecticidal, Oxidative, and Genotoxic Activities of Syzygium aromaticum and Eucalyptus globulus on Culex pipiens Adults and Larvae. Türkiye Parazitolojii Derg. 2018, 42, 213. [Google Scholar] [CrossRef]
  71. Yones, D.A.; Bakir, H.Y.; Bayoumi, S.A. Chemical composition and efficacy of some selected plant oils against Pediculus humanus capitis in vitro. Parasitol. Res. 2016, 115, 3209–3218. [Google Scholar] [CrossRef]
  72. Mekonnen, A.; Yitayew, B.; Tesema, A.; Taddese, S. In vitro antimicrobial activity of essential oil of Thymus schimperi, Matricaria chamomilla, Eucalyptus globulus, and Rosmarinus officinalis. Int. J. Microbiol. 2016, 2016, 9545693. [Google Scholar] [CrossRef] [Green Version]
  73. Dagne, E.; Bisrat, D.; Alemayehu, M.; Worku, T. Essential oils of twelve Eucalyptus species from Ethiopia. J. Essent. Oil Res. 2000, 12, 467–470. [Google Scholar] [CrossRef]
  74. Rossi, P.-G.; Berti, L.; Panighi, J.; Luciani, A.; Maury, J.; Muselli, A.; Serra, D.d.R.; Gonny, M.; Bolla, J.-M. Antibacterial action of essential oils from Corsica. J. Essent. Oil Res. 2007, 19, 176–182. [Google Scholar] [CrossRef]
  75. Mulyaningsih, S.; Sporer, F.; Reichling, J.; Wink, M. Antibacterial activity of essential oils from Eucalyptus and of selected components against multidrug-resistant bacterial pathogens. Pharm. Biol. 2011, 49, 893–899. [Google Scholar] [CrossRef] [PubMed]
  76. Vivekanandhan, P.; Usha-Raja-Nanthini, A.; Valli, G.; Subramanian Shivakumar, M. Comparative efficacy of Eucalyptus globulus (Labill) hydrodistilled essential oil and temephos as mosquito larvicide. Nat. Prod. Res. 2020, 34, 2626–2629. [Google Scholar] [CrossRef] [PubMed]
  77. Yadav, M.; Jindal, D.K.; Parle, M.; Kumar, A.; Dhingra, S. Targeting oxidative stress, acetylcholinesterase, proinflammatory cytokine, dopamine and GABA by eucalyptus oil (Eucalyptus globulus) to alleviate ketamine-induced psychosis in rats. Inflammopharmacology 2019, 27, 301–311. [Google Scholar] [CrossRef]
  78. Pandey, A.; Chattopadhyay, P.; Banerjee, S.; Pakshirajan, K.; Singh, L. Antitermitic activity of plant essential oils and their major constituents against termite Odontotermes assamensis Holmgren (Isoptera: Termitidae) of North East India. Int. Biodeterior. Biodegrad. 2012, 75, 63–67. [Google Scholar] [CrossRef]
  79. Joshi, A.; Sharma, A.; Bachheti, R.; Pandey, D. A Comparative study of the chemical composition of the essential oil from Eucalyptus globulus growing in Dehradun (India) and around the World. Orient. J. Chem. 2016, 32, 331–340. [Google Scholar] [CrossRef] [Green Version]
  80. Pant, M.; Dubey, S.; Patanjali, P.; Naik, S.; Sharma, S. Insecticidal activity of eucalyptus oil nanoemulsion with karanja and jatropha aqueous filtrates. Int. Biodeterior. Biodegrad. 2014, 91, 119–127. [Google Scholar] [CrossRef]
  81. Kumar, P.; Mishra, S.; Malik, A.; Satya, S. Compositional analysis and insecticidal activity of Eucalyptus globulus (family: Myrtaceae) essential oil against housefly (Musca domestica). Acta Trop. 2012, 122, 212–218. [Google Scholar] [CrossRef]
  82. Tyagi, A.K.; Malik, A. Antimicrobial potential and chemical composition of Eucalyptus globulus oil in liquid and vapour phase against food spoilage microorganisms. Food Chem. 2011, 126, 228–235. [Google Scholar] [CrossRef]
  83. Gupta, S.; Bhagat, M.; Sudan, R.; Dogra, S.; Jamwal, R. Comparative Chemoprofiling and Biological Potential of Three Eucalyptus Species Growing in Jammu and Kashmir. J. Essent. Oil Bear. Plants 2015, 18, 409–415. [Google Scholar] [CrossRef]
  84. Mousavi, S.M.; Wilson, G.; Raftos, D.; Mirzargar, S.S.; Omidbaigi, R. Antibacterial activities of a new combination of essential oils against marine bacteria. Aquac. Int. 2011, 19, 205–214. [Google Scholar] [CrossRef]
  85. Azadbakht, E.; Maghsoudlou, Y.; Khomiri, M.; Kashiri, M. Development and structural characterization of chitosan films containing Eucalyptus globulus essential oil: Potential as an antimicrobial carrier for packaging of sliced sausage. Food Packag. Shelf Life 2018, 17, 65–72. [Google Scholar] [CrossRef]
  86. Laquale, S.; Candido, V.; Avato, P.; Argentieri, M.; d’Addabbo, T. Essential oils as soil biofumigants for the control of the root-knot nematode Meloidogyne incognita on tomato. Ann. Appl. Biol. 2015, 167, 217–224. [Google Scholar] [CrossRef]
  87. Nardoni, S.; Giovanelli, S.; Pistelli, L.; Mugnaini, L.; Profili, G.; Pisseri, F.; Mancianti, F. In vitro activity of twenty commercially available, plant-derived essential oils against selected dermatophyte species. Nat. Prod. Commun. 2015, 10, 1473–1478. [Google Scholar] [CrossRef] [Green Version]
  88. Nardoni, S.; Ebani, V.V.; D’Ascenzi, C.; Pistelli, L.; Mancianti, F. Sensitivity of entomopathogenic fungi and bacteria to plants secondary metabolites, for an alternative control of Rhipicephalus (Boophilus) microplus in cattle. Front. Pharmacol. 2018, 9, 937. [Google Scholar] [CrossRef]
  89. Ebani, V.V.; Najar, B.; Bertelloni, F.; Pistelli, L.; Mancianti, F.; Nardoni, S. Chemical Composition and In Vitro Antimicrobial Efficacy of Sixteen Essential Oils against Escherichia coli and Aspergillus fumigatus Isolated from Poultry. Vet. Sci. 2018, 5, 62. [Google Scholar] [CrossRef] [Green Version]
  90. Fratini, F.; Casella, S.; Leonardi, M.; Pisseri, F.; Ebani, V.V.; Pistelli, L.; Pistelli, L. Antibacterial activity of essential oils, their blends and mixtures of their main constituents against some strains supporting livestock mastitis. Fitoterapia 2014, 96, 1–7. [Google Scholar] [CrossRef]
  91. Tampieri, M.; Galuppi, R.; Carelle, M.; Macchioni, F.; Cioni, P.; Morelli, I. Effect of selected essential oils and pure compounds on Saprolegnia parasitica. Pharm. Biol. 2003, 41, 584–591. [Google Scholar] [CrossRef] [Green Version]
  92. Tampieri, M.P.; Galuppi, R.; Macchioni, F.; Carelle, M.S.; Falcioni, L.; Cioni, P.L.; Morelli, I. The inhibition of Candida albicans by selected essential oils and their major components. Mycopathologia 2005, 159, 339–345. [Google Scholar] [CrossRef]
  93. Perrucci, S. Acaricidal activity of some essential oils and their constituents against Tyrophagus longior, a mite of stored food. J. Food Prot. 1995, 58, 560–563. [Google Scholar] [CrossRef] [PubMed]
  94. Della Porta, G.; Porcedda, S.; Marongiu, B.; Reverchon, E. Isolation of eucalyptus oil by supercritical fluid extraction. Flavour Frag J. 1999, 14, 214–218. [Google Scholar] [CrossRef]
  95. Karemu, C.K.; Ndung’u, M.W.; Githua, M. Repellent effects of essential oils from selected eucalyptus species and their major constituents against Sitophilus zeamais (Coleoptera: Curculionidae). Int. J. Trop. Insect Sci. 2013, 33, 188–194. [Google Scholar] [CrossRef]
  96. Damjanović-Vratnica, B.; Đakov, T.; Šuković, D.; Damjanović, J. Antimicrobial effect of essential oil isolated from Eucalyptus globulus Labill. from Montenegro. Czech. J. Food Sci. 2011, 29, 277–284. [Google Scholar] [CrossRef] [Green Version]
  97. Chebli, B.; Hmamouchi, M.; Achouri, M.; Hassani, L.I. Composition and in vitro fungitoxic activity of 19 essential oils against two post-harvest pathogens. J. Essent. Oil Res. 2004, 16, 507–511. [Google Scholar] [CrossRef]
  98. Khaddor, M.; Lamarti, A.; Tantaoui-Elaraki, A.; Ezziyyani, M.; Castillo, M.-E.C.; Badoc, A. Antifungal activity of three essential oils on growth and toxigenesis of Penicillium aurantiogriseum and Penicillium viridicatum. J. Essent. Oil Res. 2006, 18, 586–589. [Google Scholar] [CrossRef]
  99. Aazza, S.; Lyoussi, B.; Megias, C.; Cortes-Giraldo, I.; Vioque, J.; Figueiredo, A.C.; Miguel, M.G. Anti-oxidant, anti-inflammatory and anti-proliferative activities of Moroccan commercial essential oils. Nat. Prod. Commun. 2014, 9, 587–594. [Google Scholar] [CrossRef] [Green Version]
  100. Ghaffar, A.; Yameen, M.; Kiran, S.; Kamal, S.; Jalal, F.; Munir, B.; Saleem, S.; Rafiq, N.; Ahmad, A.; Saba, I. Chemical composition and in-vitro evaluation of the antimicrobial and antioxidant activities of essential oils extracted from seven Eucalyptus species. Molecules 2015, 20, 20487–20498. [Google Scholar] [CrossRef] [Green Version]
  101. Iqbal, Z.; Hussain, I.; Hussain, A.; Ashraf, M.Y. Genetic variability to essential oil contents and composition in five species of Eucalyptus. Pak. J. Bot. 2003, 35, 843–852. [Google Scholar]
  102. Silvestre, A.J.D.; Cavaleiro, J.A.S.; Delmond, B.; Filliatre, C.; Bourgeois, G. The Essential Oil of Eucalyptus globulus Labill. from Portugal. Flavour Frag J. 1994, 9, 51–53. [Google Scholar] [CrossRef]
  103. Topiar, M.; Sajfrtova, M.; Pavela, R.; Machalova, Z. Comparison of fractionation techniques of CO2 extracts from Eucalyptus globulus-Composition and insecticidal activity. J. Supercrit Fluid 2015, 97, 202–210. [Google Scholar] [CrossRef]
  104. Císarová, M.; Tančinová, D.; Medo, J.; Kačániová, M. The in vitro effect of selected essential oils on the growth and mycotoxin production of Aspergillus species. J. Environ. Sci. Health Part B 2016, 51, 668–674. [Google Scholar] [CrossRef]
  105. Combrinck, S.; Regnier, T.; Kamatou, G. In vitro activity of eighteen essential oils and some major components against common postharvest fungal pathogens of fruit. Ind. Crops Prod. 2011, 33, 344–349. [Google Scholar] [CrossRef]
  106. Diánez, F.; Santos, M.; Parra, C.; Navarro, M.; Blanco, R.; Gea, F. Screening of antifungal activity of 12 essential oils against eight pathogenic fungi of vegetables and mushroom. Lett. Appl. Microbiol. 2018, 67, 400–410. [Google Scholar] [CrossRef]
  107. Luís, Â.; Duarte, A.; Gominho, J.; Domingues, F.; Duarte, A.P. Chemical composition, antioxidant, antibacterial and anti-quorum sensing activities of Eucalyptus globulus and Eucalyptus radiata essential oils. Ind. Crops Prod. 2016, 79, 274–282. [Google Scholar] [CrossRef]
  108. Jesser, E.N.; Werdin-González, J.O.; Murray, A.P.; Ferrero, A.A. Efficacy of essential oils to control the Indian meal moth, Plodia interpunctella (Hübner)(Lepidoptera: Pyralidae). J. Asia-Pac. Entomol. 2017, 20, 1122–1129. [Google Scholar] [CrossRef] [Green Version]
  109. Tadtong, S.; Kamkaen, N.; Watthanachaiyingcharoen, R.; Ruangrungsi, N. Chemical components of four essential oils in aromatherapy recipe. Nat. Prod. Commun. 2015, 10, 1934578X1501000673. [Google Scholar] [CrossRef] [Green Version]
  110. Soonwera, M.; Wongnet, O.; Sittichok, S. Ovicidal effect of essential oils from Zingiberaceae plants and Eucalytus globulus on eggs of head lice, Pediculus humanus capitis De Geer. Phytomedicine 2018, 47, 93–104. [Google Scholar] [CrossRef]
  111. Ghrab, S.; Balme, S.; Cretin, M.; Bouaziz, S.; Benzina, M. Adsorption of terpenes from Eucalyptus globulus onto modified beidellite. Appl. Clay Sci. 2018, 156, 169–177. [Google Scholar] [CrossRef]
  112. Elaissi, A.; Medini, H.; Khouja, M.L.; Simmonds, M.; Lynen, F.; Farhat, F.; Chemli, R.; Harzallah-Skhiri, F. Variation in volatile leaf oils of five Eucalyptus species harvested from Jbel Abderrahman Arboreta (Tunisia). Chem. Biodivers. 2011, 8, 352–361. [Google Scholar] [CrossRef]
  113. Jerbi, A.; Derbali, A.; Elfeki, A.; Kammoun, M. Essential oil composition and biological activities of Eucalyptus globulus leaves extracts from Tunisia. J. Essent. Oil Bear. Plants 2017, 20, 438–448. [Google Scholar] [CrossRef]
  114. Slimane, B.B.; Ezzine, O.; Dhahri, S.; Jamaa, M.L.B. Essential oils from two Eucalyptus from Tunisia and their insecticidal action on Orgyia trigotephras (Lepidotera, Lymantriidae). Biol. Res. 2014, 47, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Hafsa, J.; ali Smach, M.; Khedher, M.R.B.; Charfeddine, B.; Limem, K.; Majdoub, H.; Rouatbi, S. Physical, antioxidant and antimicrobial properties of chitosan films containing Eucalyptus globulus essential oil. LWT-Food Sci. Technol. 2016, 68, 356–364. [Google Scholar] [CrossRef]
  116. Wei, A.; Shibamoto, T. Antioxidant/lipoxygenase inhibitory activities and chemical compositions of selected essential oils. J. Agric. Food Chem. 2010, 58, 7218–7225. [Google Scholar] [CrossRef]
  117. Swanson, B.G. Tannins and Polyphenols. In Encyclopedia of Food Sciences and Nutrition, 2nd ed.; Caballero, B., Ed.; Academic Press: Cambridge, MA, USA, 2003; pp. 5729–5733. [Google Scholar]
  118. Boulekbache-Makhlouf, L.; Meudec, E.; Mazauric, J.P.; Madani, K.; Cheynier, V. Qualitative and Semi-quantitative Analysis of Phenolics in Eucalyptus globulus Leaves by High-performance Liquid Chromatography Coupled with Diode Array Detection and Electrospray Ionisation Mass Spectrometry. Phytochem. Anal. 2013, 24, 162–170. [Google Scholar] [CrossRef]
  119. Santos, S.A.; Freire, C.S.; Domingues, M.R.; Silvestre, A.J.; Pascoal Neto, C. Characterization of phenolic components in polar extracts of Eucalyptus globulus Labill. bark by high-performance liquid chromatography-mass spectrometry. J. Agric. Food Chem. 2011, 59, 9386–9393. [Google Scholar] [CrossRef] [PubMed]
  120. Harborne, J.B. Phytochemical Methods: A Guide to Modern Techniques of Plant. Analysis, 3rd ed.; Chapman and Hall: London, UK, 1998. [Google Scholar]
  121. Dezsi, S.; Badarau, A.S.; Bischin, C.; Vodnar, D.C.; Silaghi-Dumitrescu, R.; Gheldiu, A.M.; Mocan, A.; Vlase, L. Antimicrobial and antioxidant activities and phenolic profile of Eucalyptus globulus Labill. and Corymbia ficifolia (F. Muell.) K.D. Hill & L.A.S. Johnson leaves. Molecules 2015, 20, 4720–4734. [Google Scholar] [PubMed] [Green Version]
  122. Salgado, P.; Márquez, K.; Rubilar, O.; Contreras, D.; Vidal, G. The effect of phenolic compounds on the green synthesis of iron nanoparticles (Fe x O y-NPs) with photocatalytic activity. Appl. Nanosci. 2019, 9, 371–385. [Google Scholar] [CrossRef]
  123. Ji, Y.E.; Sun, X.; Kim, M.K.; Li, W.Y.; Lee, S.W.; Koppula, S.; Yu, S.H.; Kim, H.B.; Kang, T.B.; Lee, K.H. Eucalyptus globulus Inhibits Inflammasome-Activated Pro-Inflammatory Responses and Ameliorate Monosodium Urate-Induced Peritonitis in Murine Experimental Model. Am. J. Chin. Med. 2018, 46, 423–433. [Google Scholar] [CrossRef] [PubMed]
  124. Ghareeb, M.A.; Sobeh, M.; El-Maadawy, W.H.; Mohammed, H.S.; Khalil, H.; Botros, S.; Wink, M. Chemical Profiling of Polyphenolics in Eucalyptus globulus and Evaluation of Its Hepato-Renal Protective Potential against Cyclophosphamide Induced Toxicity in Mice. Antioxidants 2019, 8, 415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Proestos, C.; Komaitis, M. Analysis of naturally occurring phenolic compounds in aromatic plants by RP-HPLC coupled to diode array detector (DAD) and GC-MS after silylation. Foods 2013, 2, 90–99. [Google Scholar] [CrossRef] [PubMed]
  126. Nile, S.H.; Keum, Y.S. Chemical composition, antioxidant, anti-inflammatory and antitumor activities of Eucalyptus globulus Labill. Indian J. Exp. Biol. 2018, 56, 734–742. [Google Scholar]
  127. Bajpai, M.; Pande, A.; Tewari, S.; Prakash, D. Phenolic contents and antioxidant activity of some food and medicinal plants. Int. J. Food Sci. Nutr. 2005, 56, 287–291. [Google Scholar] [CrossRef] [PubMed]
  128. Almeida, I.F.; Fernandes, E.; Lima, J.L.; Valentão, P.; Andrade, P.B.; Seabra, R.M.; Costa, P.; Bahia, M. Oxygen and nitrogen reactive species are effectively scavenged by Eucalyptus globulus leaf water extract. J. Med. Food 2009, 12, 175–183. [Google Scholar] [CrossRef]
  129. Puig, C.G.; Reigosa, M.J.; Valentao, P.; Andrade, P.B.; Pedrol, N. Unravelling the bioherbicide potential of Eucalyptus globulus Labill: Biochemistry and effects of its aqueous extract. PLoS ONE 2018, 13, e0192872. [Google Scholar] [CrossRef] [Green Version]
  130. Park, B.; Hwang, E.; Seo, S.A.; Cho, J.G.; Yang, J.E.; Yi, T.H. Eucalyptus globulus extract protects against UVB-induced photoaging by enhancing collagen synthesis via regulation of TGF-beta/Smad signals and attenuation of AP-1. Arch. Biochem. Biophys. 2018, 637, 31–39. [Google Scholar] [CrossRef]
  131. Hardy, J.; Allsop, D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol. Sci. 1991, 12, 383–388. [Google Scholar] [CrossRef]
  132. Vassar, R.; Bennett, B.D.; Babu-Khan, S.; Kahn, S.; Mendiaz, E.A.; Denis, P.; Teplow, D.B.; Ross, S.; Amarante, P.; Loeloff, R.; et al. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science 1999, 286, 735–741. [Google Scholar] [CrossRef] [Green Version]
  133. Lin, X.; Koelsch, G.; Wu, S.; Downs, D.; Dashti, A.; Tang, J. Human aspartic protease memapsin 2 cleaves the beta-secretase site of beta-amyloid precursor protein. Proc. Natl. Acad. Sci. USA 2000, 97, 1456–1460. [Google Scholar] [CrossRef] [Green Version]
  134. Leissring, M.A.; Murphy, M.P.; Mead, T.R.; Akbari, Y.; Sugarman, M.C.; Jannatipour, M.; Anliker, B.; Muller, U.; Saftig, P.; De Strooper, B.; et al. A physiologic signaling role for the gamma -secretase-derived intracellular fragment of APP. Proc. Natl. Acad. Sci. USA 2002, 99, 4697–4702. [Google Scholar] [CrossRef] [Green Version]
  135. Cole, S.L.; Vassar, R. The Alzheimer’s disease beta-secretase enzyme, BACE1. Mol. Neurodegener. 2007, 2, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Farzan, M.; Schnitzler, C.E.; Vasilieva, N.; Leung, D.; Choe, H. BACE2, a beta-secretase homolog, cleaves at the beta site and within the amyloid-beta region of the amyloid-beta precursor protein. Proc. Natl. Acad. Sci. USA 2000, 97, 9712–9717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Dovey, H.F.; John, V.; Anderson, J.P.; Chen, L.Z.; de Saint Andrieu, P.; Fang, L.Y.; Freedman, S.B.; Folmer, B.; Goldbach, E.; Holsztynska, E.J.; et al. Functional gamma-secretase inhibitors reduce beta-amyloid peptide levels in brain. J. Neurochem. 2001, 76, 173–181. [Google Scholar] [CrossRef]
  138. Citron, M. Alzheimer’s disease: Strategies for disease modification. Nat. Rev. Drug Discov. 2010, 9, 387–398. [Google Scholar] [CrossRef]
  139. De Strooper, B. Lessons from a failed gamma-secretase Alzheimer trial. Cell 2014, 159, 721–726. [Google Scholar] [CrossRef] [Green Version]
  140. Oehlrich, D.; Berthelot, D.J.; Gijsen, H.J. gamma-Secretase modulators as potential disease modifying anti-Alzheimer’s drugs. J. Med. Chem 2011, 54, 669–698. [Google Scholar] [CrossRef]
  141. Menting, K.W.; Claassen, J.A. beta-secretase inhibitor; a promising novel therapeutic drug in Alzheimer’s disease. Front. Aging Neurosci. 2014, 6, 165. [Google Scholar] [CrossRef] [Green Version]
  142. Mangialasche, F.; Solomon, A.; Winblad, B.; Mecocci, P.; Kivipelto, M. Alzheimer’s disease: Clinical trials and drug development. Lancet Neurol. 2010, 9, 702–716. [Google Scholar] [CrossRef]
  143. Kadavath, H.; Hofele, R.V.; Biernat, J.; Kumar, S.; Tepper, K.; Urlaub, H.; Mandelkow, E.; Zweckstetter, M. Tau stabilizes microtubules by binding at the interface between tubulin heterodimers. Proc. Natl. Acad. Sci. USA 2015, 112, 7501–7506. [Google Scholar] [CrossRef] [Green Version]
  144. Simic, G.; Babic Leko, M.; Wray, S.; Harrington, C.; Delalle, I.; Jovanov-Milosevic, N.; Bazadona, D.; Buee, L.; de Silva, R.; Di Giovanni, G.; et al. Tau Protein Hyperphosphorylation and Aggregation in Alzheimer’s Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules 2016, 6, 6. [Google Scholar] [CrossRef] [Green Version]
  145. Grill, J.D.; Cummings, J.L. Current therapeutic targets for the treatment of Alzheimer’s disease. Expert Rev. Neurother. 2010, 10, 711–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Liu, S.L.; Wang, C.; Jiang, T.; Tan, L.; Xing, A.; Yu, J.T. The Role of Cdk5 in Alzheimer’s Disease. Mol. Neurobiol. 2016, 53, 4328–4342. [Google Scholar] [CrossRef] [PubMed]
  147. Hernandez, F.; Gomez de Barreda, E.; Fuster-Matanzo, A.; Lucas, J.J.; Avila, J. GSK3: A possible link between beta amyloid peptide and tau protein. Exp. Neurol. 2010, 223, 322–325. [Google Scholar] [CrossRef] [PubMed]
  148. Pei, J.J.; Tanaka, T.; Tung, Y.C.; Braak, E.; Iqbal, K.; Grundke-Iqbal, I. Distribution, levels, and activity of glycogen synthase kinase-3 in the Alzheimer disease brain. J. Neuropathol. Exp. Neurol. 1997, 56, 70–78. [Google Scholar] [CrossRef] [Green Version]
  149. Phiel, C.J.; Wilson, C.A.; Lee, V.M.; Klein, P.S. GSK-3alpha regulates production of Alzheimer’s disease amyloid-beta peptides. Nature 2003, 423, 435–439. [Google Scholar] [CrossRef] [PubMed]
  150. Kwak, H.M.; Jeon, S.Y.; Sohng, B.H.; Kim, J.G.; Lee, J.M.; Lee, K.B.; Jeong, H.H.; Hur, J.M.; Kang, Y.H.; Song, K.S. beta-Secretase (BACE1) inhibitors from pomegranate (Punica granatum) husk. Arch. Pharm Res. 2005, 28, 1328–1332. [Google Scholar] [CrossRef] [PubMed]
  151. Feng, Y.; Yang, S.G.; Du, X.T.; Zhang, X.; Sun, X.X.; Zhao, M.; Sun, G.Y.; Liu, R.T. Ellagic acid promotes Abeta42 fibrillization and inhibits Abeta42-induced neurotoxicity. Biochem. Biophys. Res. Commun. 2009, 390, 1250–1254. [Google Scholar] [CrossRef]
  152. Shen, Y.C.; Juan, C.W.; Lin, C.S.; Chen, C.C.; Chang, C.L. Neuroprotective Effect of Terminalia Chebula Extracts and Ellagic Acid in Pc12 Cells. Afr J. Tradit. Complement. Altern. Med. 2017, 14, 22–30. [Google Scholar] [CrossRef] [Green Version]
  153. Jha, A.B.; Panchal, S.S.; Shah, A. Ellagic acid: Insights into its neuroprotective and cognitive enhancement effects in sporadic Alzheimer’s disease. Pharm. Biochem. Behav. 2018, 175, 33–46. [Google Scholar] [CrossRef]
  154. Zhong, L.; Liu, H.; Zhang, W.; Liu, X.; Jiang, B.; Fei, H.; Sun, Z. Ellagic acid ameliorates learning and memory impairment in APP/PS1 transgenic mice via inhibition of beta-amyloid production and tau hyperphosphorylation. Exp. Med. 2018, 16, 4951–4958. [Google Scholar]
  155. Jimenez-Aliaga, K.; Bermejo-Bescos, P.; Benedi, J.; Martin-Aragon, S. Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci. 2011, 89, 939–945. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, S.W.; Wang, Y.J.; Su, Y.J.; Zhou, W.W.; Yang, S.G.; Zhang, R.; Zhao, M.; Li, Y.N.; Zhang, Z.P.; Zhan, D.W.; et al. Rutin inhibits beta-amyloid aggregation and cytotoxicity, attenuates oxidative stress, and decreases the production of nitric oxide and proinflammatory cytokines. Neurotoxicology 2012, 33, 482–490. [Google Scholar] [CrossRef] [PubMed]
  157. Kim, H.; Park, B.S.; Lee, K.G.; Choi, C.Y.; Jang, S.S.; Kim, Y.H.; Lee, S.E. Effects of naturally occurring compounds on fibril formation and oxidative stress of beta-amyloid. J. Agric. Food Chem. 2005, 53, 8537–8541. [Google Scholar] [CrossRef]
  158. Ono, K.; Yoshiike, Y.; Takashima, A.; Hasegawa, K.; Naiki, H.; Yamada, M. Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: Implications for the prevention and therapeutics of Alzheimer’s disease. J. Neurochem. 2003, 87, 172–181. [Google Scholar] [CrossRef] [PubMed]
  159. Descamps, O.; Spilman, P.; Zhang, Q.; Libeu, C.P.; Poksay, K.; Gorostiza, O.; Campagna, J.; Jagodzinska, B.; Bredesen, D.E.; John, V. AβPP-selective BACE inhibitors (ASBI): Novel class of therapeutic agents for Alzheimer’s disease. J. Alzheimer’s Dis. JAD 2013, 37, 343. [Google Scholar] [CrossRef] [Green Version]
  160. Omar, S.H.; Scott, C.J.; Hamlin, A.S.; Obied, H.K. Biophenols: Enzymes (beta-secretase, Cholinesterases, histone deacetylase and tyrosinase) inhibitors from olive (Olea europaea L.). Fitoterapia 2018, 128, 118–129. [Google Scholar] [CrossRef]
  161. Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Flavonols and flavones as BACE-1 inhibitors: Structure–activity relationship in cell-free, cell-based and in silico studies reveal novel pharmacophore features. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2008, 1780, 819–825. [Google Scholar] [CrossRef]
  162. Paris, D.; Mathura, V.; Ait-Ghezala, G.; Beaulieu-Abdelahad, D.; Patel, N.; Bachmeier, C.; Mullan, M. Flavonoids lower Alzheimer’s Aβ production via an NFκB dependent mechanism. Bioinformation 2011, 6, 229. [Google Scholar] [CrossRef]
  163. Espargaro, A.; Ginex, T.; Vadell, M.D.; Busquets, M.A.; Estelrich, J.; Munoz-Torrero, D.; Luque, F.J.; Sabate, R. Combined in Vitro Cell-Based/in Silico Screening of Naturally Occurring Flavonoids and Phenolic Compounds as Potential Anti-Alzheimer Drugs. J. Nat. Prod. 2017, 80, 278–289. [Google Scholar] [CrossRef]
  164. Jiang, W.; Luo, T.; Li, S.; Zhou, Y.; Shen, X.Y.; He, F.; Xu, J.; Wang, H.Q. Quercetin Protects against Okadaic Acid-Induced Injury via MAPK and PI3K/Akt/GSK3beta Signaling Pathways in HT22 Hippocampal Neurons. PLoS ONE 2016, 11, e0152371. [Google Scholar]
  165. Shen, X.Y.; Luo, T.; Li, S.; Ting, O.Y.; He, F.; Xu, J.; Wang, H.Q. Quercetin inhibits okadaic acid-induced tau protein hyperphosphorylation through the Ca2+-calpain-p25-CDK5 pathway in HT22 cells. Int. J. Mol. Med. 2018, 41, 1138–1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Yuan, Z.; Luan, G.; Wang, Z.; Hao, X.; Li, J.; Suo, Y.; Li, G.; Wang, H. Flavonoids from Potentilla parvifolia Fisch. and Their Neuroprotective Effects in Human Neuroblastoma SH-SY 5Y Cells in vitro. Chem. Biodivers. 2017, 14, e1600487. [Google Scholar] [CrossRef] [PubMed]
  167. Chen, J.; Deng, X.; Liu, N.; Li, M.; Liu, B.; Fu, Q.; Qu, R.; Ma, S. Quercetin attenuates tau hyperphosphorylation and improves cognitive disorder via suppression of ER stress in a manner dependent on AMPK pathway. J. Funct. Foods 2016, 22, 463–476. [Google Scholar] [CrossRef]
  168. Jagota, S.; Rajadas, J. Effect of phenolic compounds against Aβ aggregation and Aβ-induced toxicity in transgenic C. elegans. Neurochem. Res. 2012, 37, 40–48. [Google Scholar] [CrossRef] [PubMed]
  169. Regitz, C.; Dussling, L.M.; Wenzel, U. Amyloid-beta (Abeta(1)(-)(4)(2))-induced paralysis in Caenorhabditis elegans is inhibited by the polyphenol quercetin through activation of protein degradation pathways. Mol. Nutr. Food Res. 2014, 58, 1931–1940. [Google Scholar] [CrossRef]
  170. Wang, D.M.; Li, S.Q.; Wu, W.L.; Zhu, X.Y.; Wang, Y.; Yuan, H.Y. Effects of long-term treatment with quercetin on cognition and mitochondrial function in a mouse model of Alzheimer’s disease. Neurochem. Res. 2014, 39, 1533–1543. [Google Scholar] [CrossRef]
  171. Sabogal-Guaqueta, A.M.; Munoz-Manco, J.I.; Ramirez-Pineda, J.R.; Lamprea-Rodriguez, M.; Osorio, E.; Cardona-Gomez, G.P. The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 2015, 93, 134–145. [Google Scholar] [CrossRef] [Green Version]
  172. Paula, P.C.; Angelica Maria, S.G.; Luis, C.H.; Gloria Patricia, C.G. Preventive Effect of Quercetin in a Triple Transgenic Alzheimer’s Disease Mice Model. Molecules 2019, 24, 2287. [Google Scholar] [CrossRef] [Green Version]
  173. Vargas-Restrepo, F.; Sabogal-Guaqueta, A.M.; Cardona-Gomez, G.P. Quercetin ameliorates inflammation in CA1 hippocampal region in aged triple transgenic Alzheimer s disease mice model. Biomedica 2018, 38, 69–76. [Google Scholar] [CrossRef]
  174. Zhang, X.; Hu, J.; Zhong, L.; Wang, N.; Yang, L.; Liu, C.C.; Li, H.; Wang, X.; Zhou, Y.; Zhang, Y.; et al. Quercetin stabilizes apolipoprotein E and reduces brain Abeta levels in amyloid model mice. Neuropharmacology 2016, 108, 179–192. [Google Scholar] [CrossRef]
  175. Lu, Y.; Liu, Q.; Yu, Q. Quercetin enrich diet during the early-middle not middle-late stage of alzheimer’s disease ameliorates cognitive dysfunction. Am. J. Transl. Res. 2018, 10, 1237. [Google Scholar] [PubMed]
  176. Woo, M.; Noh, J.S.; Cho, E.J.; Song, Y.O. Bioactive Compounds of Kimchi Inhibit Apoptosis by Attenuating Endoplasmic Reticulum Stress in the Brain of Amyloid beta-Injected Mice. J. Agric. Food Chem. 2018, 66, 4883–4890. [Google Scholar] [CrossRef] [PubMed]
  177. Li, Y.; Tian, Q.; Li, Z.; Dang, M.; Lin, Y.; Hou, X. Activation of Nrf2 signaling by sitagliptin and quercetin combination against beta-amyloid induced Alzheimer’s disease in rats. Drug Dev. Res. 2019, 80, 837–845. [Google Scholar] [CrossRef] [PubMed]
  178. Xu, P.-X.; Wang, S.-W.; Yu, X.-L.; Su, Y.-J.; Wang, T.; Zhou, W.-W.; Zhang, H.; Wang, Y.-J.; Liu, R.-T. Rutin improves spatial memory in Alzheimer’s disease transgenic mice by reducing Aβ oligomer level and attenuating oxidative stress and neuroinflammation. Behav. Brain Res. 2014, 264, 173–180. [Google Scholar] [CrossRef] [PubMed]
  179. Lovell, M.A.; Markesbery, W.R. Oxidative DNA damage in mild cognitive impairment and late-stage Alzheimer’s disease. Nucleic Acids Res. 2007, 35, 7497–7504. [Google Scholar] [CrossRef] [Green Version]
  180. Nunomura, A.; Castellani, R.J.; Zhu, X.; Moreira, P.I.; Perry, G.; Smith, M.A. Involvement of oxidative stress in Alzheimer disease. J. Neuropathol. Exp. Neurol. 2006, 65, 631–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Zhao, Y.; Zhao, B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxid Med. Cell Longev. 2013, 2013, 316523. [Google Scholar] [CrossRef] [Green Version]
  182. Mizuno, D.; Konoha-Mizuno, K.; Mori, M.; Yamazaki, K.; Haneda, T.; Koyama, H.; Kawahara, M. An In Vitro System Comprising Immortalized Hypothalamic Neuronal Cells (GT1-7 Cells) for Evaluation of the Neuroendocrine Effects of Essential Oils. Evid.-Based Complement. Altern. Med. 2015, 2015, 343942. [Google Scholar] [CrossRef] [Green Version]
  183. Ryu, S.; Park, H.; Seol, G.H.; Choi, I.Y. 1,8-Cineole ameliorates oxygen-glucose deprivation/reoxygenation-induced ischaemic injury by reducing oxidative stress in rat cortical neuron/glia. J. Pharm. Pharm. 2014, 66, 1818–1826. [Google Scholar] [CrossRef]
  184. Khan, A.; Vaibhav, K.; Javed, H.; Tabassum, R.; Ahmed, M.E.; Khan, M.M.; Khan, M.B.; Shrivastava, P.; Islam, F.; Siddiqui, M.S.; et al. 1,8-cineole (eucalyptol) mitigates inflammation in amyloid Beta toxicated PC12 cells: Relevance to Alzheimer’s disease. Neurochem. Res. 2014, 39, 344–352. [Google Scholar] [CrossRef]
  185. Lee, G.Y.; Lee, C.; Park, G.H.; Jang, J.H. Amelioration of Scopolamine-Induced Learning and Memory Impairment by alpha-Pinene in C57BL/6 Mice. Evid.-Based Complement. Altern. Med. 2017, 2017, 4926815. [Google Scholar]
  186. Branca, C.; Ferreira, E.; Nguyen, T.V.; Doyle, K.; Caccamo, A.; Oddo, S. Genetic reduction of Nrf2 exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. Hum. Mol. Genet. 2017, 26, 4823–4835. [Google Scholar] [CrossRef] [PubMed]
  187. Bahn, G.; Jo, D.G. Therapeutic Approaches to Alzheimer’s Disease Through Modulation of NRF2. Neuromol. Med. 2019, 21, 1–11. [Google Scholar] [CrossRef] [PubMed]
  188. Kabiraj, P.; Marin, J.E.; Varela-Ramirez, A.; Zubia, E.; Narayan, M. Ellagic acid mitigates SNO-PDI induced aggregation of Parkinsonian biomarkers. ACS Chem. Neurosci. 2014, 5, 1209–1220. [Google Scholar] [CrossRef]
  189. Kiasalari, Z.; Heydarifard, R.; Khalili, M.; Afshin-Majd, S.; Baluchnejadmojarad, T.; Zahedi, E.; Sanaierad, A.; Roghani, M. Ellagic acid ameliorates learning and memory deficits in a rat model of Alzheimer’s disease: An exploration of underlying mechanisms. Psychopharmacology 2017, 234, 1841–1852. [Google Scholar] [CrossRef]
  190. Kumar, M.; Bansal, N. Ellagic acid prevents dementia through modulation of PI3-kinase-endothelial nitric oxide synthase signalling in streptozotocin-treated rats. Naunyn-Schmiedebergs Arch. Pharm. 2018, 391, 987–1001. [Google Scholar] [CrossRef]
  191. Bansal, N.; Yadav, P.; Kumar, M. Ellagic Acid Administration Negated the Development of Streptozotocin-Induced Memory Deficit in Rats. Drug Res. 2017, 67, 425–431. [Google Scholar] [CrossRef]
  192. Uzar, E.; Alp, H.; Cevik, M.U.; Firat, U.; Evliyaoglu, O.; Tufek, A.; Altun, Y. Ellagic acid attenuates oxidative stress on brain and sciatic nerve and improves histopathology of brain in streptozotocin-induced diabetic rats. Neurol. Sci. 2012, 33, 567–574. [Google Scholar] [CrossRef]
  193. Choi, S.M.; Kim, B.C.; Cho, Y.H.; Choi, K.H.; Chang, J.; Park, M.S.; Kim, M.K.; Cho, K.H.; Kim, J.K. Effects of Flavonoid Compounds on beta-amyloid-peptide-induced Neuronal Death in Cultured Mouse Cortical Neurons. Chonnam Med. J. 2014, 50, 45–51. [Google Scholar] [CrossRef] [Green Version]
  194. Yu, X.L.; Li, Y.N.; Zhang, H.; Su, Y.J.; Zhou, W.W.; Zhang, Z.P.; Wang, S.W.; Xu, P.X.; Wang, Y.J.; Liu, R.T. Rutin inhibits amylin-induced neurocytotoxicity and oxidative stress. Food Funct. 2015, 6, 3296–3306. [Google Scholar] [CrossRef]
  195. Heo, H.J.; Lee, C.Y. Protective effects of quercetin and vitamin C against oxidative stress-induced neurodegeneration. J. Agric. Food Chem. 2004, 52, 7514–7517. [Google Scholar] [CrossRef] [PubMed]
  196. Ansari, M.A.; Abdul, H.M.; Joshi, G.; Opii, W.O.; Butterfield, D.A. Protective effect of quercetin in primary neurons against Abeta(1-42): Relevance to Alzheimer’s disease. J. Nutr. Biochem. 2009, 20, 269–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Godoy, J.A.; Lindsay, C.B.; Quintanilla, R.A.; Carvajal, F.J.; Cerpa, W.; Inestrosa, N.C. Quercetin Exerts Differential Neuroprotective Effects Against H2O2 and Abeta Aggregates in Hippocampal Neurons: The Role of Mitochondria. Mol. Neurobiol. 2017, 54, 7116–7128. [Google Scholar] [CrossRef]
  198. Lee, M.; McGeer, E.G.; McGeer, P.L. Quercetin, not caffeine, is a major neuroprotective component in coffee. Neurobiol. Aging 2016, 46, 113–123. [Google Scholar] [CrossRef]
  199. Javed, H.; Khan, M.M.; Ahmad, A.; Vaibhav, K.; Ahmad, M.E.; Khan, A.; Ashafaq, M.; Islam, F.; Siddiqui, M.S.; Safhi, M.M.; et al. Rutin prevents cognitive impairments by ameliorating oxidative stress and neuroinflammation in rat model of sporadic dementia of Alzheimer type. Neuroscience 2012, 210, 340–352. [Google Scholar] [CrossRef]
  200. Choi, J.Y.; Lee, J.M.; Lee, D.G.; Cho, S.; Yoon, Y.H.; Cho, E.J.; Lee, S. The n-Butanol Fraction and Rutin from Tartary Buckwheat Improve Cognition and Memory in an In Vivo Model of Amyloid-beta-Induced Alzheimer’s Disease. J. Med. Food 2015, 18, 631–641. [Google Scholar] [CrossRef] [PubMed]
  201. Qu, J.; Zhou, Q.; Du, Y.; Zhang, W.; Bai, M.; Zhang, Z.; Xi, Y.; Li, Z.; Miao, J. Rutin protects against cognitive deficits and brain damage in rats with chronic cerebral hypoperfusion. Br. J. Pharm. 2014, 171, 3702–3715. [Google Scholar] [CrossRef] [Green Version]
  202. Ramalingayya, G.V.; Cheruku, S.P.; Nayak, P.G.; Kishore, A.; Shenoy, R.; Rao, C.M.; Krishnadas, N. Rutin protects against neuronal damage in vitro and ameliorates doxorubicin-induced memory deficits in vivo in Wistar rats. Drug Des. Devel. 2017, 11, 1011–1026. [Google Scholar] [CrossRef] [Green Version]
  203. Tota, S.; Awasthi, H.; Kamat, P.K.; Nath, C.; Hanif, K. Protective effect of quercetin against intracerebral streptozotocin induced reduction in cerebral blood flow and impairment of memory in mice. Behav. Brain Res. 2010, 209, 73–79. [Google Scholar] [CrossRef]
  204. Maciel, R.M.; Carvalho, F.B.; Olabiyi, A.A.; Schmatz, R.; Gutierres, J.M.; Stefanello, N.; Zanini, D.; Rosa, M.M.; Andrade, C.M.; Rubin, M.A.; et al. Neuroprotective effects of quercetin on memory and anxiogenic-like behavior in diabetic rats: Role of ectonucleotidases and acetylcholinesterase activities. Biomed. Pharm. 2016, 84, 559–568. [Google Scholar] [CrossRef]
  205. Choi, G.N.; Kim, J.H.; Kwak, J.H.; Jeong, C.-H.; Jeong, H.R.; Lee, U.; Heo, H.J. Effect of quercetin on learning and memory performance in ICR mice under neurotoxic trimethyltin exposure. Food Chem. 2012, 132, 1019–1024. [Google Scholar] [CrossRef]
  206. Kim, J.H.; Lee, J.; Lee, S.; Cho, E.J. Quercetin and quercetin-3-β-d-glucoside improve cognitive and memory function in Alzheimer’s disease mouse. Appl. Biol. Chem. 2016, 59, 721–728. [Google Scholar] [CrossRef]
  207. Khan, A.; Ali, T.; Rehman, S.U.; Khan, M.S.; Alam, S.I.; Ikram, M.; Muhammad, T.; Saeed, K.; Badshah, H.; Kim, M.O. Neuroprotective Effect of Quercetin Against the Detrimental Effects of LPS in the Adult Mouse Brain. Front. Pharm. 2018, 9, 1383. [Google Scholar] [CrossRef] [PubMed]
  208. Moghbelinejad, S.; Nassiri-Asl, M.; Farivar, T.N.; Abbasi, E.; Sheikhi, M.; Taghiloo, M.; Farsad, F.; Samimi, A.; Hajiali, F. Rutin activates the MAPK pathway and BDNF gene expression on beta-amyloid induced neurotoxicity in rats. Toxicol Lett. 2014, 224, 108–113. [Google Scholar] [CrossRef]
  209. Liu, R.; Zhang, T.T.; Zhou, D.; Bai, X.Y.; Zhou, W.L.; Huang, C.; Song, J.K.; Meng, F.R.; Wu, C.X.; Li, L.; et al. Quercetin protects against the Abeta(25-35)-induced amnesic injury: Involvement of inactivation of rage-mediated pathway and conservation of the NVU. Neuropharmacology 2013, 67, 419–431. [Google Scholar] [CrossRef]
  210. Sen, N. ER Stress, CREB, and Memory: A Tangled Emerging Link in Disease. Neuroscientist 2019, 25, 420–433. [Google Scholar] [CrossRef]
  211. Placido, A.I.; Pereira, C.M.; Duarte, A.I.; Candeias, E.; Correia, S.C.; Carvalho, C.; Cardoso, S.; Oliveira, C.R.; Moreira, P.I. Modulation of endoplasmic reticulum stress: An opportunity to prevent neurodegeneration? CNS Neurol. Disord. Drug Targets 2015, 14, 518–533. [Google Scholar] [CrossRef]
  212. Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086. [Google Scholar] [CrossRef] [Green Version]
  213. Lee, A.H.; Iwakoshi, N.N.; Glimcher, L.H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol. Cell Biol. 2003, 23, 7448–7459. [Google Scholar] [CrossRef] [Green Version]
  214. Hetz, C.; Chevet, E.; Oakes, S.A. Proteostasis control by the unfolded protein response. Nat. Cell Biol. 2015, 17, 829–838. [Google Scholar] [CrossRef] [Green Version]
  215. Harding, H.P.; Zhang, Y.; Bertolotti, A.; Zeng, H.; Ron, D. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol. Cell 2000, 5, 897–904. [Google Scholar] [CrossRef]
  216. Harding, H.P.; Zhang, Y.; Zeng, H.; Novoa, I.; Lu, P.D.; Calfon, M.; Sadri, N.; Yun, C.; Popko, B.; Paules, R.; et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 2003, 11, 619–633. [Google Scholar] [CrossRef]
  217. Tabas, I.; Ron, D. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat. Cell Biol. 2011, 13, 184–190. [Google Scholar] [CrossRef] [PubMed]
  218. Tsaytler, P.; Bertolotti, A. Exploiting the selectivity of protein phosphatase 1 for pharmacological intervention. FEBS J. 2013, 280, 766–770. [Google Scholar] [CrossRef]
  219. Hayakawa, M.; Itoh, M.; Ohta, K.; Li, S.; Ueda, M.; Wang, M.X.; Nishida, E.; Islam, S.; Suzuki, C.; Ohzawa, K.; et al. Quercetin reduces eIF2alpha phosphorylation by GADD34 induction. Neurobiol. Aging 2015, 36, 2509–2518. [Google Scholar] [CrossRef] [PubMed]
  220. 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]
  221. Kim, Y.S.; Joh, T.H. Microglia, major player in the brain inflammation: Their roles in the pathogenesis of Parkinson’s disease. Exp. Mol. Med. 2006, 38, 333–347. [Google Scholar] [CrossRef] [Green Version]
  222. Goldgaber, D.; Harris, H.W.; Hla, T.; Maciag, T.; Donnelly, R.J.; Jacobsen, J.S.; Vitek, M.P.; Gajdusek, D.C. Interleukin 1 regulates synthesis of amyloid beta-protein precursor mRNA in human endothelial cells. Proc. Natl. Acad. Sci. USA 1989, 86, 7606–7610. [Google Scholar] [CrossRef] [Green Version]
  223. Quintanilla, R.A.; Orellana, D.I.; Gonzalez-Billault, C.; Maccioni, R.B. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp. Cell Res. 2004, 295, 245–257. [Google Scholar] [CrossRef]
  224. 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. Alzheimers Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef]
  225. Griffin, W.S.; Stanley, L.C.; Ling, C.; White, L.; MacLeod, V.; Perrot, L.J.; White, C.L., 3rd; Araoz, C. Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc. Natl. Acad. Sci. USA 1989, 86, 7611–7615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Rogers, J.; Luber-Narod, J.; Styren, S.D.; Civin, W.H. Expression of immune system-associated antigens by cells of the human central nervous system: Relationship to the pathology of Alzheimer’s disease. Neurobiol. Aging 1988, 9, 339–349. [Google Scholar] [CrossRef]
  227. Gasparini, L.; Ongini, E.; Wenk, G. Non-steroidal anti-inflammatory drugs (NSAIDs) in Alzheimer’s disease: Old and new mechanisms of action. J. Neurochem. 2004, 91, 521–536. [Google Scholar] [CrossRef] [PubMed]
  228. Breitner, J.C. Inflammatory processes and antiinflammatory drugs in Alzheimer’s disease: A current appraisal. Neurobiol. Aging 1996, 17, 789–794. [Google Scholar] [CrossRef]
  229. Akhtar, M.A.; Raju, R.; Beattie, K.D.; Bodkin, F.; Munch, G. Medicinal Plants of the Australian Aboriginal Dharawal People Exhibiting Anti-Inflammatory Activity. Evid.-Based Complement. Altern. Med. 2016, 2016, 2935403. [Google Scholar] [CrossRef]
  230. Rojanathammanee, L.; Puig, K.L.; Combs, C.K. Pomegranate polyphenols and extract inhibit nuclear factor of activated T-cell activity and microglial activation in vitro and in a transgenic mouse model of Alzheimer disease. J. Nutr. 2013, 143, 597–605. [Google Scholar] [CrossRef] [Green Version]
  231. Bispo da Silva, A.; Cerqueira Coelho, P.L.; Alves Oliveira Amparo, J.; Alves de Almeida Carneiro, M.M.; Pereira Borges, J.M.; Dos Santos Souza, C.; Dias Costa, M.F.; Mecha, M.; Guaza Rodriguez, C.; Amaral da Silva, V.D.; et al. The flavonoid rutin modulates microglial/macrophage activation to a CD150/CD206 M2 phenotype. Chem. Biol. Interact. 2017, 274, 89–99. [Google Scholar] [CrossRef]
  232. Lerner, A.G.; Upton, J.-P.; Praveen, P.; Ghosh, R.; Nakagawa, Y.; Igbaria, A.; Shen, S.; Nguyen, V.; Backes, B.J.; Heiman, M. IRE1α induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell Metab. 2012, 16, 250–264. [Google Scholar] [CrossRef] [Green Version]
  233. Jung, S.H.; Murphy, E.A.; McClellan, J.L.; Carmichael, M.D.; Davis, J.M. The dietary flavonoid quercetin decreases neuroinflammation in a mouse model of Alzheimer’s disease. FASEB J. 2010, 24, 604–617. [Google Scholar] [CrossRef]
  234. 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]
  235. Pappas, B.A.; Bayley, P.J.; Bui, B.K.; Hansen, L.A.; Thal, L.J. Choline acetyltransferase activity and cognitive domain scores of Alzheimer’s patients. Neurobiol. Aging 2000, 21, 11–17. [Google Scholar] [CrossRef]
  236. Upadhyaya, P.; Seth, V.; Ahmad, M. Therapy of Alzheimer’s disease: An update. Afr. J. Pharm. Pharm. 2010, 4, 408–421. [Google Scholar]
  237. McGleenon, B.M.; Dynan, K.B.; Passmore, A.P. Acetylcholinesterase inhibitors in Alzheimer’s disease. Br. J. Clin. Pharm. 1999, 48, 471–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Burcul, F.; Blazevic, I.; Radan, M.; Politeo, O. Terpenes, Phenylpropanoids, Sulfur and Other Essential Oil Constituents as Inhibitors of Cholinesterases. Curr. Med. Chem. 2020, 27, 4297–4343. [Google Scholar] [CrossRef] [PubMed]
  239. Kaufmann, D.; Dogra, A.K.; Wink, M. Myrtenal inhibits acetylcholinesterase, a known Alzheimer target. J. Pharm. Pharm. 2011, 63, 1368–1371. [Google Scholar] [CrossRef] [PubMed]
  240. Ademosun, A.O.; Oboh, G.; Bello, F.; Ayeni, P.O. Antioxidative Properties and Effect of Quercetin and Its Glycosylated Form (Rutin) on Acetylcholinesterase and Butyrylcholinesterase Activities. J. Evid.-Based Complement. Altern. Med. 2016, 21, NP11–NP17. [Google Scholar] [CrossRef]
  241. Vijayakumar, S.; Manogar, P.; Prabhu, S.; Sanjeevkumar Singh, R.A. Novel ligand-based docking; molecular dynamic simulations; and absorption, distribution, metabolism, and excretion approach to analyzing potential acetylcholinesterase inhibitors for Alzheimer’s disease. J. Pharm. Anal. 2018, 8, 413–420. [Google Scholar] [CrossRef]
  242. Orhan, I.; Kartal, M.; Tosun, F.; Şener, B. Screening of various phenolic acids and flavonoid derivatives for their anticholinesterase potential. Z. Für Nat. C 2007, 62, 829–832. [Google Scholar] [CrossRef]
  243. Khan, M.T.H.; Orhan, I.; Şenol, F.; Kartal, M.; Şener, B.; Dvorská, M.; Šmejkal, K.; Šlapetová, T. Cholinesterase inhibitory activities of some flavonoid derivatives and chosen xanthone and their molecular docking studies. Chem.-Biol. Interact. 2009, 181, 383–389. [Google Scholar] [CrossRef]
  244. Jung, M.; Park, M. Acetylcholinesterase inhibition by flavonoids from Agrimonia pilosa. Molecules 2007, 12, 2130–2139. [Google Scholar] [CrossRef] [Green Version]
  245. Balkis, A.; Tran, K.; Lee, Y.Z.; Ng, K. Screening flavonoids for inhibition of acetylcholinesterase identified baicalein as the most potent inhibitor. Int. J. Biol. 2015, 7, 26. [Google Scholar] [CrossRef] [Green Version]
  246. Olennikov, D.N.; Kashchenko, N.I.; Chirikova, N.K.; Akobirshoeva, A.; Zilfikarov, I.N.; Vennos, C. Isorhamnetin and quercetin derivatives as anti-acetylcholinesterase principles of marigold (Calendula officinalis) flowers and preparations. Int. J. Mol. Sci. 2017, 18, 1685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Islam, M.R.; Zaman, A.; Jahan, I.; Chakravorty, R.; Chakraborty, S. In silico QSAR analysis of quercetin reveals its potential as therapeutic drug for Alzheimer’s disease. J. Young Pharm. 2013, 5, 173–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Zhao, Y.; Dou, J.; Wu, T.; Aisa, H.A. Investigating the antioxidant and acetylcholinesterase inhibition activities of Gossypium herbaceam. Molecules 2013, 18, 951–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Kumar, A.; Mehta, V.; Raj, U.; Varadwaj, P.K.; Udayabanu, M.; Yennamalli, R.M.; Singh, T.R. Computational and In-Vitro Validation of Natural Molecules as Potential Acetylcholinesterase Inhibitors and Neuroprotective Agents. Curr. Alzheimer Res. 2019, 16, 116–127. [Google Scholar] [CrossRef] [PubMed]
  250. Lanctôt, K.L.; Herrmann, N.; Mazzotta, P.; Khan, L.R.; Ingber, N. GABAergic Function in Alzheimer’s Disease: Evidence for Dysfunction and Potential as a Therapeutic Target for the Treatment of Behavioural and Psychological Symptoms of Dementia. Can. J. Psychiatry 2004, 49, 439–453. [Google Scholar] [CrossRef] [Green Version]
  251. Bai, X.; Edden, R.A.; Gao, F.; Wang, G.; Wu, L.; Zhao, B.; Wang, M.; Chan, Q.; Chen, W.; Barker, P.B. Decreased gamma-aminobutyric acid levels in the parietal region of patients with Alzheimer’s disease. J. Magn. Reson. Imaging 2015, 41, 1326–1331. [Google Scholar] [CrossRef] [Green Version]
  252. Hanyu, H.; Kume, K.; Sato, T.; Hirao, K.; Kanetaka, H.; Sakurai, H.; Iwamoto, T. Regional differences in cortical benzodiazepine receptors of Alzheimer, vascular, and mixed dementia patients. J. Neurol. Sci. 2012, 323, 71–76. [Google Scholar] [CrossRef]
  253. Pappata, S.; Varrone, A.; Vicidomini, C.; Milan, G.; De Falco, C.; Sansone, V.; Iavarone, A.; Comerci, M.; Lore, E.; Panico, M.R.; et al. SPECT imaging of GABA(A)/benzodiazepine receptors and cerebral perfusion in mild cognitive impairment. Eur. J. Nucl. Med. Mol. Imaging 2010, 37, 1156–1163. [Google Scholar] [CrossRef]
  254. Luchetti, S.; Bossers, K.; Van de Bilt, S.; Agrapart, V.; Morales, R.R.; Frajese, G.V.; Swaab, D.F. Neurosteroid biosynthetic pathways changes in prefrontal cortex in Alzheimer’s disease. Neurobiol. Aging 2011, 32, 1964–1976. [Google Scholar] [CrossRef]
  255. Lee, B.Y.; Ban, J.Y.; Seong, Y.H. Chronic stimulation of GABAA receptor with muscimol reduces amyloid beta protein (25-35)-induced neurotoxicity in cultured rat cortical cells. Neurosci. Res. 2005, 52, 347–356. [Google Scholar] [CrossRef] [PubMed]
  256. Yang, H.; Woo, J.; Pae, A.N.; Um, M.Y.; Cho, N.C.; Park, K.D.; Yoon, M.; Kim, J.; Lee, C.J.; Cho, S. alpha-Pinene, a Major Constituent of Pine Tree Oils, Enhances Non-Rapid Eye Movement Sleep in Mice through GABAA-benzodiazepine Receptors. Mol. Pharm. 2016, 90, 530–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Geddes, J.W.; Chang-Chui, H.; Cooper, S.M.; Lott, I.T.; Cotman, C.W. Density and distribution of NMDA receptors in the human hippocampus in Alzheimer’s disease. Brain Res. 1986, 399, 156–161. [Google Scholar] [CrossRef]
  258. Arundine, M.; Tymianski, M. Molecular mechanisms of calcium-dependent neurodegeneration in excitotoxicity. Cell Calcium. 2003, 34, 325–337. [Google Scholar] [CrossRef]
  259. Olney, J.W.; Wozniak, D.F.; Farber, N.B. Excitotoxic neurodegeneration in Alzheimer disease. New hypothesis and new therapeutic strategies. Arch. Neurol. 1997, 54, 1234–1240. [Google Scholar] [CrossRef] [PubMed]
  260. Lipton, S.A. Paradigm shift in neuroprotection by NMDA receptor blockade: Memantine and beyond. Nat. Rev. Drug Discov. 2006, 5, 160–170. [Google Scholar] [CrossRef] [PubMed]
  261. Avram, S.; Mernea, M.; Bagci, E.; Hritcu, L.; Borcan, L.C.; Mihailescu, D.F. Advanced Structure-activity Relationships Applied to Mentha spicata L. Subsp. spicata Essential Oil Compounds as AChE and NMDA Ligands, in Comparison with Donepezil, Galantamine and Memantine—New Approach in Brain Disorders Pharmacology. CNS Neurol. Disord. Drug Targets 2017, 16, 800–811. [Google Scholar] [CrossRef]
  262. Schliebs, R.; Arendt, T. The cholinergic system in aging and neuronal degeneration. Behav. Brain Res. 2011, 221, 555–563. [Google Scholar] [CrossRef]
  263. Stevens, R. Scopolamine impairs spatial maze performance in rats. Physiol. Behav. 1981, 27, 385–386. [Google Scholar] [CrossRef]
  264. Dumas, J.A.; Newhouse, P.A. The cholinergic hypothesis of cognitive aging revisited again: Cholinergic functional compensation. Pharm. Biochem. Behav. 2011, 99, 254–261. [Google Scholar] [CrossRef] [Green Version]
  265. Mansouri, M.T.; Farbood, Y.; Naghizadeh, B.; Shabani, S.; Mirshekar, M.A.; Sarkaki, A. Beneficial effects of ellagic acid against animal models of scopolamine- and diazepam-induced cognitive impairments. Pharm. Biol. 2016, 54, 1947–1953. [Google Scholar] [CrossRef] [PubMed]
  266. Richetti, S.K.; Blank, M.; Capiotti, K.M.; Piato, A.L.; Bogo, M.R.; Vianna, M.R.; Bonan, C.D. Quercetin and rutin prevent scopolamine-induced memory impairment in zebrafish. Behav. Brain Res. 2011, 217, 10–15. [Google Scholar] [CrossRef] [PubMed]
  267. Ramalingayya, G.V.; Nampoothiri, M.; Nayak, P.G.; Kishore, A.; Shenoy, R.R.; Mallikarjuna Rao, C.; Nandakumar, K. Naringin and Rutin Alleviates Episodic Memory Deficits in Two Differentially Challenged Object Recognition Tasks. Pharm. Mag. 2016, 12, S63–S70. [Google Scholar]
  268. Patil, C.S.; Singh, V.P.; Satyanarayan, P.S.; Jain, N.K.; Singh, A.; Kulkarni, S.K. Protective effect of flavonoids against aging- and lipopolysaccharide-induced cognitive impairment in mice. Pharmacology 2003, 69, 59–67. [Google Scholar] [CrossRef]
  269. Ashrafpour, M.; Parsaei, S.; Sepehri, H. Quercetin improved spatial memory dysfunctions in rat model of intracerebroventricular streptozotocin-induced sporadic Alzheimer’sdisease. Natl. J. Physiol. Pharm. Pharmacol. 2015, 5, 411–415. [Google Scholar] [CrossRef] [Green Version]
  270. Li, Y.L.; Guo, H.; Zhao, Y.Q.; Li, A.F.; Ren, Y.Q.; Zhang, J.W. Quercetin protects neuronal cells from oxidative stress and cognitive degradation induced by amyloid beta-peptide treatment. Mol. Med. Rep. 2017, 16, 1573–1577. [Google Scholar] [CrossRef] [Green Version]
  271. Karimipour, M.; Rahbarghazi, R.; Tayefi, H.; Shimia, M.; Ghanadian, M.; Mahmoudi, J.; Bagheri, H.S. Quercetin promotes learning and memory performance concomitantly with neural stem/progenitor cell proliferation and neurogenesis in the adult rat dentate gyrus. Int. J. Dev. Neurosci. 2019, 74, 18–26. [Google Scholar] [CrossRef]
  272. Nakagawa, T.; Itoh, M.; Ohta, K.; Hayashi, Y.; Hayakawa, M.; Yamada, Y.; Akanabe, H.; Chikaishi, T.; Nakagawa, K.; Itoh, Y.; et al. Improvement of memory recall by quercetin in rodent contextual fear conditioning and human early-stage Alzheimer’s disease patients. Neuroreport 2016, 27, 671–676. [Google Scholar] [CrossRef]
Figure 1. Major compounds found in essential oils and phenolic extracts obtained from E. globulus leaves.
Figure 1. Major compounds found in essential oils and phenolic extracts obtained from E. globulus leaves.
Ijms 23 08812 g001
Figure 2. Effect of compounds obtained from E. globulus leaves in the amyloidogenic pathway and in the formation of amyloid-β (Aβ) in AD. The amyloidogenic pathway is initiated with the enzymatic breakdown of amyloid precursor protein (APP) by β-secretase enzyme followed by catalytic cleavage of APP by γ-secretase to originate non-soluble protein or Aβ. Aβ oligomerization and accumulation leads to synaptic dysfunction and neurodegeneration.
Figure 2. Effect of compounds obtained from E. globulus leaves in the amyloidogenic pathway and in the formation of amyloid-β (Aβ) in AD. The amyloidogenic pathway is initiated with the enzymatic breakdown of amyloid precursor protein (APP) by β-secretase enzyme followed by catalytic cleavage of APP by γ-secretase to originate non-soluble protein or Aβ. Aβ oligomerization and accumulation leads to synaptic dysfunction and neurodegeneration.
Ijms 23 08812 g002
Figure 3. Effect of compounds obtained from E. globulus leaves on tau aggregation and formation of neurofibrillary tangles (NFTs) in AD. Irregular phosphorylation of tau proteins destabilizes microtubules, leading to the formation of insoluble tau oligomers, which then accumulate to generate protomers. Then, two twisted protomers originate paired helical filaments, which after aggregation lead to the formation of NFTs. These intracellular structures are involved in synaptic and neuronal dysfunction, thus contributing to cognitive decline in AD.
Figure 3. Effect of compounds obtained from E. globulus leaves on tau aggregation and formation of neurofibrillary tangles (NFTs) in AD. Irregular phosphorylation of tau proteins destabilizes microtubules, leading to the formation of insoluble tau oligomers, which then accumulate to generate protomers. Then, two twisted protomers originate paired helical filaments, which after aggregation lead to the formation of NFTs. These intracellular structures are involved in synaptic and neuronal dysfunction, thus contributing to cognitive decline in AD.
Ijms 23 08812 g003
Figure 4. Effect of compounds obtained from E. globulus leaves in oxidative stress and mitochondrial damage in AD. Aβ oligomers can insert the plasma membrane originating pores by which Ca2+ pass into the cytoplasm. Aβ can also interact with metal ions (Fe2+ and Cu+) to generate reactive oxygen species (ROS), which cause membrane lipid peroxidation. As consequence, the membrane turns depolarized, and voltage-dependent Ca2+ channels (VDCC) and glutamate receptor-associated channels (in particular NMDAR, N-methyl-D-aspartate receptor) open increasing cytoplasmic Ca2+ content. Additionally, Aβ overproduction can cause mitochondrial damage, which culminates in ROS accumulation and ATP depletion that can impair axonal transport consequently originating abnormal mitochondrial dynamics and promoting neurotransmission deficits. ATP depletion can also lead to ionic alterations in the cytosol due to dysfunction of ATP-dependent ion channels. Moreover, ROS accumulation affects the mitochondrial permeability transition pore (MPTP), which further potentiates mitochondrial damage due to Ca2+ overload and inhibition of the electron transport chain. ROS increase also promotes damage to proteins, namely DNA and RNA.
Figure 4. Effect of compounds obtained from E. globulus leaves in oxidative stress and mitochondrial damage in AD. Aβ oligomers can insert the plasma membrane originating pores by which Ca2+ pass into the cytoplasm. Aβ can also interact with metal ions (Fe2+ and Cu+) to generate reactive oxygen species (ROS), which cause membrane lipid peroxidation. As consequence, the membrane turns depolarized, and voltage-dependent Ca2+ channels (VDCC) and glutamate receptor-associated channels (in particular NMDAR, N-methyl-D-aspartate receptor) open increasing cytoplasmic Ca2+ content. Additionally, Aβ overproduction can cause mitochondrial damage, which culminates in ROS accumulation and ATP depletion that can impair axonal transport consequently originating abnormal mitochondrial dynamics and promoting neurotransmission deficits. ATP depletion can also lead to ionic alterations in the cytosol due to dysfunction of ATP-dependent ion channels. Moreover, ROS accumulation affects the mitochondrial permeability transition pore (MPTP), which further potentiates mitochondrial damage due to Ca2+ overload and inhibition of the electron transport chain. ROS increase also promotes damage to proteins, namely DNA and RNA.
Ijms 23 08812 g004
Figure 5. Effect of compounds obtained from E. globulus leaves on neuroinflammation in AD. A vicious circle between Aβ and tau accumulation in the brain, microglia activation, and release of pro-inflammatory cytokines culminates in neuronal death in AD.
Figure 5. Effect of compounds obtained from E. globulus leaves on neuroinflammation in AD. A vicious circle between Aβ and tau accumulation in the brain, microglia activation, and release of pro-inflammatory cytokines culminates in neuronal death in AD.
Ijms 23 08812 g005
Figure 6. Effect of compounds obtained from E. globulus leaves on cholinesterase activity in AD. Synthesis of acetylcholine (ACh) neurotransmitter from acetyl coenzyme A (Acetyl CoA) and choline (Ch) occurs by the action of the enzyme choline acetyltransferase (ChAT) in the presynaptic terminal. Acetylcholine is released in the synaptic cleft, where it can activate both muscarinic (mAChR) and nicotinic (nAChR) receptors. Acetylcholinesterase (AChE) or butyrylcholinesterase (BChE) break acetylcholine into choline and acetate. ACh levels are low in AD brains and cholinergic neurotransmission in impaired. AChE and BChE inhibitors correct these deficits increasing the amount of ACh that remains in the synaptic cleft and interacts with postsynaptic receptors.
Figure 6. Effect of compounds obtained from E. globulus leaves on cholinesterase activity in AD. Synthesis of acetylcholine (ACh) neurotransmitter from acetyl coenzyme A (Acetyl CoA) and choline (Ch) occurs by the action of the enzyme choline acetyltransferase (ChAT) in the presynaptic terminal. Acetylcholine is released in the synaptic cleft, where it can activate both muscarinic (mAChR) and nicotinic (nAChR) receptors. Acetylcholinesterase (AChE) or butyrylcholinesterase (BChE) break acetylcholine into choline and acetate. ACh levels are low in AD brains and cholinergic neurotransmission in impaired. AChE and BChE inhibitors correct these deficits increasing the amount of ACh that remains in the synaptic cleft and interacts with postsynaptic receptors.
Ijms 23 08812 g006
Figure 7. Effect of compounds obtained from E. globulus leaves on inhibitory (A) and excitatory (B) synapses in AD. (A) The inhibitory γ-aminobutyric acid (GABA) synapse. GABA is synthesized from glutamate by the glutamic acid decarboxylase enzymes in the presynaptic terminal of GABAergic neuron. The vesicular GABA transporter packs GABA into vesicles, which, after release in the synaptic cleft, binds GABAA receptors localized on the postsynaptic neuron. The reuptake of GABA into the presynaptic axon stops the GABA action in the synapse. GABA levels are significantly reduced in AD patients as well as the GABAA receptor density. (B) The excitatory glutamate synapse. Glutamine is converted to glutamate via glutaminase in the presynaptic terminal of glutamatergic neuron, and the vesicular glutamate transporter packs glutamate into vesicles. After glutamate release in the synaptic cleft, it acts on glutamate receptors localized on the postsynaptic neuron. The excitatory amino acid transporters (EAATs) present in nearby astrocytes clear the glutamate from the synaptic cleft. Glutamate is converted to glutamine via glutamine synthetase in astrocytes before being transported to presynaptic neurons. In AD, Aβ oligomers affect extrasynaptic N-methyl-D-aspartate (NMDA) receptors enriched in NR2B subunits, leading to an excessive activation and consequently to an excess of Ca2+ accumulation in the post-synaptic cell.
Figure 7. Effect of compounds obtained from E. globulus leaves on inhibitory (A) and excitatory (B) synapses in AD. (A) The inhibitory γ-aminobutyric acid (GABA) synapse. GABA is synthesized from glutamate by the glutamic acid decarboxylase enzymes in the presynaptic terminal of GABAergic neuron. The vesicular GABA transporter packs GABA into vesicles, which, after release in the synaptic cleft, binds GABAA receptors localized on the postsynaptic neuron. The reuptake of GABA into the presynaptic axon stops the GABA action in the synapse. GABA levels are significantly reduced in AD patients as well as the GABAA receptor density. (B) The excitatory glutamate synapse. Glutamine is converted to glutamate via glutaminase in the presynaptic terminal of glutamatergic neuron, and the vesicular glutamate transporter packs glutamate into vesicles. After glutamate release in the synaptic cleft, it acts on glutamate receptors localized on the postsynaptic neuron. The excitatory amino acid transporters (EAATs) present in nearby astrocytes clear the glutamate from the synaptic cleft. Glutamate is converted to glutamine via glutamine synthetase in astrocytes before being transported to presynaptic neurons. In AD, Aβ oligomers affect extrasynaptic N-methyl-D-aspartate (NMDA) receptors enriched in NR2B subunits, leading to an excessive activation and consequently to an excess of Ca2+ accumulation in the post-synaptic cell.
Ijms 23 08812 g007
Figure 8. Effect of compounds obtained from E. globulus leaves in the AD-associated memory and learning impairment.
Figure 8. Effect of compounds obtained from E. globulus leaves in the AD-associated memory and learning impairment.
Ijms 23 08812 g008
Table 1. Chemical components present at ≥2%, of the essential oils from Eucalyptus globulus leaves.
Table 1. Chemical components present at ≥2%, of the essential oils from Eucalyptus globulus leaves.
Origin Compounds Source Extraction Yield (%) References
Algeria 1,8-cineole (78.5 %), o-cymene (2.2%) Fresh leaves Steam distillation 0.96 [28]
1,8-cineole (71.3 %), α-pinene (8.8%), trans-pinocarveol (3.3%), limonene (2.7%), α-terpineol (2.7%) Fresh leaves Steam distillation 1.10 [32]
1,8-cineole (55.3%), isovaleraldehyde (10.0%), spathulenol (7.4%), α-terpineol (5.5%), α-pinene (4.6%) Dry leaves Hydrodistillation 2.53 [33]
1,8-cineole (51.1%), α-pinene (24.6%), trans-pinocarveol (10.0%), globulol (2.8%) Fresh leaves Steam distillation 0.96 [34]
1,8-cineole (48.6%), globulol (10.9%), trans-pinocarveol (10.7%), α-terpineol (6.6%), aromadendrene (4.6%) Fresh leaves Hydrodistillation 2.50 [35]
1,8-cineole (47.1%), globulol (8.7%), α-pinene (7.7%), α-terpinene (3.6%), p-cymene (3.5%), terpineol (2.4%) Dry leaves Steam distillation - [36]
γ-terpinene (94.5%), 1,8-cineole (3.2%) Dry leaves Hydrodistillation 3.50 [30]
Argentina 1,8-cineole (98.9%) Commercial - - [20]
1,8-cineole (90.7%), α-pinene (4.1%) Fresh leaves Hydrodistillation 2.68 [37]
a 1,8-cineole (77.9%), α-terpineol (6.0%), α-pinene (5.8%), γ-terpinene (4.8%), p-cymene (2.3%) Fresh leaves Hydrodistillation 2.25 [38,39,40]
b 1,8-cineole (76.7%), α-pinene (11.1%), α-terpineol acetate (4.0%) 1.66
1,8-cineole (76.7%), limonene (18.9%) Leaves Hydrodistillation - [41]
Australia 1,8-cineole (90.0%), α-pinene (2.2%) Commercial - - [24]
1,8-cineole (86.3%) Commercial - - [42]
1,8-cineole (79.4%), α-pinene (3.7%), α-terpineol (3.0%) Commercial - - [43]
1,8-cineole (77.0%), limonene (7.5%), p-cymene (5.5%), γ-terpinene (5.3%) Commercial - - [44]
1,8-cineole (64.4%), limonene (5%), α-pinene (3.8%) Commercial - - [45]
1,8-cineole (51.0%), α-pinene (16.7%), limonene (6.2%), globulol (7.3%) Leaves Hydrodistillation - [46]
Belgium 1,8-cineole (80.4%), limonene, (7.5%), γ-terpinene (3.7%), p-cymene (2.5%) Commercial - - [47]
Brazil 1,8-cineole (90.0%) Commercial - - [48]
1,8-cineole (90.0%), tricyclene (3.0%) Commercial - - [23]
1,8-cineole (83.9%), limonene (8.2%), α-pinene (4.2%), o-cymene (3.0%) Commercial - - [49]
1,8-cineole (83.7%), limonene (6.4%), p-cymene (5.4%), α-pinene (4.6%) Fresh leaves Hydrodistillation - [50]
1,8-cineole (78.9%), limonene (8.5%), p-cymene (5.7%), α-pinene (3.6%) Commercial - - [51]
1,8-cineole (77.5%), α-pinene (14.2%) Dry leaves Hydrodistillation 3.10 [52]
1,8-cineole (75.7%), p-cymene (7.5%), α-pinene (7.3%),
limonene (6.4%)
Commercial - - [53,54]
1,8-cineole (71.0%), α-pinene (8.3%), α-guaiene (4.8%), globulol (3.5%), cis-verbenol (2.7%) Dry leaves Steam distillation 1.33 [55]
1,8-cineole (69.3%), camphene (9.4%), α-pinene (7.5%), α-terpineol (5.1%), globulol (2.7%) Dry leaves Hydrodistillation 1.60 [56]
1,8-cineole (68.3%), α-pinene (16.2%), α-terpineol (6.4%), limonene (3.0%) Dry leaves Steam distillation - [57]
1,8-cineole (64.3%), α-pinene (8.9%), α-terpineol (7.2%), globulol (4.8%) Dry leaves Hydrodistillation 1.60 [58]
1,8-cineole (61.3%), camphenene (12.6%), α-pinene (5.8%), limonene (4.1%), vidiflorol (3.1%), aromadrendene (2.8%) Dry leaves Hydrodistillation 1.50 [59]
1,8-cineole (49.0%), camphenene (8.9%), globulol (7.0%), aromadendrene (2.3%), α-terpineol (2.0%) Dry leaves Hydrodistillation 0.60 [60]
1,8-cineole (44.7%), α-pinene (14.3%), globulol (9.2%), aromadendrene (7.3%), p-cymene (4.7%) Dry leaves Hydrodistillation - [61]
Cameroon 1.8-cineole (26.4%), α-pinene (14.1%), p-cymene (10.2%), β-ionone epoxyde (7.0%), p-menthen-8-ol (6.5%) Fresh leaves Steam distillation 1.00 [62]
Chile 1,8-cineole (82.6%), α-pinene (9.5%), m-mentha-6.8-diene (4.7%) Leaves Hydrodistillation - [63]
1,8-cineole (76.0%), α-pinene (7.4%), aromadendrene (3.6%), silvestrene (2.8%), sabinene (2.0%) Fresh leaves Hydrodistillation - [64]
China 1,8-cineole (94.3%) Commercial - - [65]
1,8-cineole (39.2%), α-terpineol acetate (13.8%), α-terpineol (11.3%), α-pinene (11.3%), endo-borneol (5.4%) Dry leaves Hydrodistillation - [66]
Columbia 1,8-cineole (52.3%), α-pinene (15.3%), α-terpineol (9.8%), globulol (7.6%) Fresh leaves Hydrodistillation 1.50 [67]
Democratic Republic of the Congo 1,8-cineole (44.3%), camphene (23.1%), α-pinene (9.3%), globulol (7.3%), limonene (5.1%) Fresh leaves Hydrodistillation 1.87 [68]
Ecuador 1,8-cineole (52.6%), α-pinene (20.0%), α-phellandrene (6.2%), α-terpinyl acetate (3.7%) Commercial - - [69]
Egypt 1,8-cineole (46.8%), limonene (9.6%), tolueno (8.6%), o-cymene (6.5%), fenchene (6.3%) Dry leaves Hydrodistillation - [70]
1,8-cineole (21.4%), o-cymene (21.4%), α-pinene (6.7%), spathulenol (6.3%), 4-terpineol (3.9%) Fresh leaves Hydrodistillation 0.40 [71]
Ethiopia 1,8-cineole (81.6%), α-pinene (2.8%), cuminaldehyde (2.8%), trans-caryophyllene (2.5%) Fresh leaves Hydrodistillation - [26]
1,8-cineole (63.0%), α-pinene (16.1%), camphor (3.4%) Fresh leaves Hydrodistillation - [72]
1,8-cineole (57.5%), α-pinene (15.2%), limonene (7.8%), α-terpinyl acetate (5.3%), α-terpineol (2.0%) Fresh leaves Hydrodistillation 1.10 [73]
France 1,8-cineole (57.9%), α-pinene (13.9%), globulol (3.6%), p-cymene (3.3%), trans-pinocarveol (2.8%) Commercial - - [74]
Germany 1,8-cineole (86.5%), α-pinene (4.7%), γ-terpinene (2.6%) Commercial - - [75]
India 1,8-cineole (85.0%), α-pinene (3.0%) Commercial - [25]
1,8-cineole (71.7%), α-pinene (9.14%), α-terpineol acetate (3.6%), alloaromadendrene (2.4%), α-terpineol (2.2%) Dry leaves Hydrodistillation - [76]
1,8-cineole (71.6%), 3-carene (15.1%), cis-ocimene (6.2%) Commercial - - [77]
1,8-cineole (68.8%), α-pinene (2.8%), p-cymene (2.1%) Commercial - - [78]
1,8-cineole (54.8%), β-pinene (18.5%), α-pinene (11.5%), β-eudesmol (4.7%), α-phellandrene (2.1%), Fresh leaves Hydrodistillation 1.10 [79]
1,8-cineole (66.3%), cis-ocimene (21.3%), α-terpinyl acetate (3.4%), aromadendrene (2.9%), globulol (1.4%) Commercial - - [80]
1,8-cineole (33.6%), α-pinene (14.2%), limonene (10.1%), α-terpinolene (6%), α-terpineol (4.7%) Commercial - - [81]
1,8-cineole (45.4%), limonene (17.8%), p-cymene (9.5%), γ-terpinene (8.8%), α-pinene (4.2%) Commercial - - [82]
p-cymene (31.9%), 1,8-cineole (17.5%), α-pinene (17.2%), α-terpinene (8.9%), β-pinene (7.5%) Fresh leaves Hydrodistillation 0.90 [83]
cymene (26.4%), β-pinene (15.2%), eudesmol (11.4%), α-pinene (10.6%), 1-phellandrene (10.3%) Dry leaves Hydrodistillation 2.00 [31]
Iran 1,8-cineole (88.0%), α-pinene (2.2%) Dry leaves Hydrodistillation - [84]
1,8-cineole (58.1%), α-phellandrene (6.0%), neo-isodihydrocarveol (3.6%), α-pinene (3.3%), α-eudesmol (3.2%) Commercial - - [85]
Italy 1,8-cineole (95.5%), α-pinene (2.5%) Commercial - - [22]
1,8-cineole (91.5%), p-cymene (3.1%), α-pinene (2.7%) Commercial - - [86]
1,8-cineole (89.8%), p-cymene (6.7%), α-pinene (2.0%) Commercial - - [87,88,89]
1,8-cineole (84.9%), α-pinene (5.6%), p-cymene (5.3%) Commercial - - [90]
1,8-cineole (81.4%), limonene (7.0%) Commercial - - [91,92]
1,8-cineole (76.0%), α-pinene (6.6%), limonene (5.7%), α-terpineol (3.1%) Commercial - - [93]
1,8-cineole (48.2%), aromadendrene (13.7%), guaiol (7.6%), α-pinene (6.9%), p-mentha-1,3,5-triene (3.8%) Dry leaves Hydrodistillation 2.00 [94]
Kenya 1,8-cineole (79.6%), α-pinene (6.9%), α-terpineol (3.8%), limonene (2.7%) Fresh leaves Hydrodistillation - [26]
1,8-cineole (17.2%), α-pinene (7.1%), spathulenol (6.5%), cryptone (5.4%), isoborneol (2.5%) Fresh leaves Steam distillation - [95]
Montenegro 1,8-cineole (85.8%), α-pinene (7.2%) Dry leaves Hydrodistillation 1.80 [96]
Morocco 1,8-cineole (80.0%), limonene (6.7%), p-cymene (5.1%), γ-terpinene (3.9%) Dry leaves Steam distillation 2.70 [27]
1,8-cineole (70.6%), α-pinene (12.9%) Dry leaves Hydrodistillation 0.60 [97]
1,8-cineole (29.5%), p-cymene (11.5%), α-terpineol (5.2%) Dry leaves Hydrodistillation 1.20 [98]
p-cymene (37.8%), 1,8-cineole (29.3%), limonene (26.1%), α-pinene (3.5%) Commercial - - [99]
Pakistan 1,8-cineole (56.5%), limonene (28.0%), α-pinene (4.2%) α-terpineol, (4.0%), globulol (2.4%) Fresh leaves Hydrodistillation 1.89 [100]
β-phellandrene (32.1%), 1,8-cineole (26.6%), α-pinene (16.8%), p-cymene (8.9%), Δ3-carene (8.1%) Fresh leaves Hydrodistillation 1.10 [101]
Portugal 1,8-cineole (74.6%), α-pinene (12.9%), metileugenol (3.5%), globulol (3.2%), terpinen-4-ol (2.0%) Dry leaves Hydrodistillation - [29]
1,8-cineole (62.5%), α-pinene (18.5%), limonene (4.0%) aromadendrene (3.1%), δ-cadinene (2.9%) Fresh leaves Hydrodistillation 1.90–2.70 [102]
1,8-cineole (36.7%), β-pinene (9.3%), aromedendrene (6.3%), globulol (5.1%), trans-pinocarveol (2.5%) Dry leaves Hydrodistillation 2.67 [103]
Slovakia 1,8-cineole (70.0%), limonene (12.0%), α-pinene (9.0%) Leaves Hydrodistillation - [104]
South Africa 1,8-cineole (80.8%), limonene (8.0%), γ-terpinene (2.8%) Commercial - - [105]
Spain 1,8-cineole (84.3%), cymene (7.5%), γ-terpinene (3.5%) Commercial - - [106]
1,8-cineole (63.8%), α-pinene (16.1%), aromadendrene (3.7%), o-cymene (2.4%) Commercial - - [107]
Switzerland 1,8-cineole (88.0%), p-cymene (6.7%), γ-terpinene (3.5%) Commercial - - [108]
Thailand 1,8-cineole (82.6%), limonene (7.4%), o-cymene (4.3%), γ-terpinene (2.7%) Commercial - - [109]
1,8-cineole (48.5%), α-pinene (20.6%), β-pinene (15.5%), terpineol (15.4%) Fresh leaves Hydrodistillation - [110]
Tunisia 1,8-cineole (95.6%) Commercial - - [21]
1,8-cineole (62.8%), 4-methyl-2-pentyl acetate (22.3%), α-pinene (8.8%), caryophyllene (2.5%), β-humulene (2.4%) Commercial - - [111]
1,8-cineole (53.8%), α-pinene (12.1%), globulol (4.5%), trans-pinocarveol (3.7%), aromadendrene (3.4%) Dry leaves Hydrodistillation 3.80 [112]
1,8-cineole (48.2%), α-pinene (16.1%), γ-terpinene (8.9%), p-cymene (8.8%), globulol (3.8%) Fresh leaves Hydrodistillation 0.74 [113]
1,8-cineole (43.2%), α-pinene (13.6%), aromadendrene (10.1%), 4-carene (6.9%), β-cymene (4.0%) Dry leaves Hydrodistillation 1.25 [114]
p-cymene (18.2%), methyl eugenol (8.8%), terpinenol (8.5%), s-methyl 3-methylbutanethioate (7.3%), γ-terpinene (5.1%) Fresh leaves Steam distillation - [115]
USA 1,8-cineole (90.0%), p-cymene (3.7%), α-pinene (3.5%) Commercial - - [116]
aE. globulus ssp. maidenii; b E. globulus ssp. globulus.
Table 2. Phenolic compounds from Eucalyptus globulus leaves extracts.
Table 2. Phenolic compounds from Eucalyptus globulus leaves extracts.
Origin Compounds *
mg/100 g Plant Material
Total Phenolic Content
mg GAE/g
Source Extraction Yield
g/100 g Plant Material
Reference
Algeria Sideroxylonal (1902.39), ellagic acid (284.30), methylellagic acid hexose (174.88), eucalbanine (113.13), quercetin 3-O-rhamnoside (108.43) - Dry leaves 70% Acetone and 0.5% acetic acid 24.70 [118]
Australia Hyperoside (66.64), quercetin (28.78), myricetin (9.23), rutin (4.87), isoquercetin (3.90) 235.87 a Dry leaves 70% Ethanol at 60 °C - [121]
Chile Luteolin (260.00), quercetin (250.00), morin (170.00), sinapic acid (170.00), ellagic acid (60.00) 0.043 b Fresh leaves Methanol - [64]
Gallic acid (2175.00), gentisic acid (1358.33), rutin (456.83), caffeic acid (351.67), 3,4-dihydroxybenzoic acid (34.33) 54.02 c Dry leaves Water at 100 °C [122]
China Rutin, isorhamnetin-hexoside, isorhamnetin-rhamnoside - Dry leaves Methanol at 45 °C 10.50 [123]
Egypt Isorhamnetin 3-O-beta-D-glucuronoside, galloyl cypellocarpin B, cypellocarpin C, methyl gallate, valoneoyl-digalloyl-glucopyranose - Dry leaves Methanol 14.67 [124]
Greece p-Coumaric acid (6.60), quercetin (2.50), rutin (1.80), gallic acid (1.50) - Dry leaves 62.5% Methanol and HCl at 90 °C - [125]
India Gallic acid (8.62), ellagic acid (6.58), vanillic acid (4.89), p-hydroxybenzoic acid (4.36), syringic acid (3.86) 242.50 c Dry leaves Methanol - [126]
Gallic acid (5.36), ellagic acid (4.20), p-hydroxybenzoic acid (3.55), vanillic acid (2.56), syringic acid (2.45) 156.30 c Chloroform
Gallic acid (3.08), p-hydroxybenzoic acid (2.10), syringic acid (1.24), ellagic acid (0.86) 98.70 c Hexane
Rutin (113.20), quercetin (44.00), ferulic acid (6.66), gallic acid (3.00), caffeic acid (1.40) 40.10 c Dry leaves 80% Methanol and 5.5% HCl at 85 °C - [127]
Lithuania Chlorogenic acid, phlorizin, rutin, quinic acid, isoquercetin - Dry leaves 70% Methanol - [13]
Chlorogenic acid, phlorizin, rutin, quinic acid, isoquercetin - 70% Acetone
Chlorogenic acid, phlorizin, quinic acid, quercetin, apigenin - 70% Ethanol
Portugal Flavonol glycoside (234.52), chlorogenic acid (106.91), rutin (105.71), ellagic acid (63.81), quercetin (57.38) 311.00 c Dry leaves Water at 40 °C 23.80 [128]
Spain Hyperoside (29.09), chlorogenic acid (17.54), rutin (16.64), quercetin (6.30), p-coumaric acid derivative (2.40) - Dry leaves Water 9.69 [129]
USA Gallic acid (132.90) - Dry leaves 50% Methanol 30.00 [130]
* Only the five major compounds from highest to lowest concentration are shown in the table; a value expressed in dry weight plant material; b value expressed in fresh weight plant material; c value expressed in extract.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Moreira, P.; Matos, P.; Figueirinha, A.; Salgueiro, L.; Batista, M.T.; Branco, P.C.; Cruz, M.T.; Pereira, C.F. Forest Biomass as a Promising Source of Bioactive Essential Oil and Phenolic Compounds for Alzheimer’s Disease Therapy. Int. J. Mol. Sci. 2022, 23, 8812. https://doi.org/10.3390/ijms23158812

AMA Style

Moreira P, Matos P, Figueirinha A, Salgueiro L, Batista MT, Branco PC, Cruz MT, Pereira CF. Forest Biomass as a Promising Source of Bioactive Essential Oil and Phenolic Compounds for Alzheimer’s Disease Therapy. International Journal of Molecular Sciences. 2022; 23(15):8812. https://doi.org/10.3390/ijms23158812

Chicago/Turabian Style

Moreira, Patrícia, Patrícia Matos, Artur Figueirinha, Lígia Salgueiro, Maria Teresa Batista, Pedro Costa Branco, Maria Teresa Cruz, and Cláudia Fragão Pereira. 2022. "Forest Biomass as a Promising Source of Bioactive Essential Oil and Phenolic Compounds for Alzheimer’s Disease Therapy" International Journal of Molecular Sciences 23, no. 15: 8812. https://doi.org/10.3390/ijms23158812

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop