Next Article in Journal
Impact of Lung-Related Polygenic Risk Scores on Chronic Obstructive Pulmonary Disease Risk and Their Interaction with w-3 Fatty Acid Intake in Middle-Aged and Elderly Individuals
Previous Article in Journal
Hepatic and Skeletal Muscle Autophagy Marker Levels in Rat Models of Prenatal and Postnatal Protein Restriction
Previous Article in Special Issue
Taxifolin Suppresses Inflammatory Responses of High-Glucose-Stimulated Mouse Microglia by Attenuating the TXNIP–NLRP3 Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Triterpenoid Lupeol as an Antioxidant and Anti-Neuroinflammatory Agent: Impacts on Oxidative Stress in Alzheimer’s Disease

1
Division of Life Sciences and Applied Life Science (BK21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Republic of Korea
2
Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
3
Alz-Dementia Korea Co., Jinju 52828, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this manuscript.
Nutrients 2023, 15(13), 3059; https://doi.org/10.3390/nu15133059
Submission received: 1 June 2023 / Revised: 3 July 2023 / Accepted: 4 July 2023 / Published: 7 July 2023

Abstract

:
Alzheimer’s disease (AD) is the most common neurodegenerative disease illustrated by neuronal dysfunctions, leading to memory weaknesses and personality changes mostly in the aged population worldwide. The exact cause of AD is unclear, but numerous studies have addressed the involvement of oxidative stress (OS), induced by reactive oxygen species (ROS), to be one of the leading causes in developing AD. OS dysregulates the cellular homeostasis, causing abnormal protein and lipid metabolism. Nutrition plays a pivotal role in modulating the antioxidant system and decreases the neuronal ROS level, thus playing an important therapeutic role in neurodegenerative diseases, especially in AD. Hence, medicinal herbs and their extracts have received global attention as a commercial source of antioxidants Lupeol. Lupeol is a pentacyclic triterpenoid and has many biological functions. It is available in fruits, vegetables, and medicinal plants. It has shown effective antioxidant and anti-inflammatory properties, and higher blood–brain barrier permeability. Also, the binding and inhibitory potentials of Lupeol have been investigated and proved to be effective against certain receptor proteins and enzymes in AD studies by computational molecular docking approaches. Therefore, AD-related research has gained interest in investigating the therapeutic effects of Lupeol. However, despite its beneficial effects in AD, there is still a lack of research in Lupeol. Hence, we compiled in this analysis all preclinical research that looked at Lupeol as an antioxidant and anti-inflammatory agent for AD.

1. Introduction

Alzheimer’s disease (AD) is an irreversible [1] neural malfunction, leading to changes in personality, cognition, and memory in older people over time [2]. Additionally, globally, AD causes a significant impact not only to the patient and their caretakers, but also to society and the economy [3]. The main pathology of AD is the accumulation of amyloid-beta (Aβ) peptides and hyperphosphorylated tau proteins [4]. Furthermore, other factors such as abnormalities in cholinergic neurons, inflammatory cascades, and oxidative stress (OS) are contributing factors to the initiation and propagation of AD pathology [5]. OS is known to disturb the cellular metabolic system of biomolecules such as proteins, lipids, and nucleic acids, creating a load of cellular dysfunction [6]. However, there is no cure for AD and the available treatments resulted in limited effectiveness [7]. Therefore, the preventive potential of nutrition is one of the major and leading possible solutions in ameliorating AD pathology [8]. For example, a balanced diet containing antioxidants has been reported to exhibit neuroprotective properties, by eliminating the reactive oxygen species (ROS), in AD [9,10]. Triterpenoids are reported to have promising health effects over the past decades and play a versatile role in different types of chronic disorders [11], including neuronal dysfunction such as AD [12]. Particularly, Lupeol, a pentacyclic triterpenoid, has numerous biochemical properties and it is available in a wide range of vegetables and fruits such as cucumber, white cabbage, strawberry, and mango [13,14], through the mevalonate pathway [11]. Also, it has been shown to have antioxidant and neuroprotective properties in various animal mice models by inhibiting mitochondrial ROS and reducing the oxidative burden in AD-like diseases [15].
Despite its neuroprotective properties, Lupeol has not received as much attention as other bioactive compounds; hence, the aim of this review is to draw attention to its role as an antioxidant and anti-inflammatory agent and its potential to treat AD. It is hoped that knowledge of the connection between OS and neurodegenerative diseases like AD with Lupeol may lead to the development of real molecular targets and new medications with the potential to be instrumental in treating diseases like AD.

2. Oxidative Stress and Alzheimer’s Disease (AD)

Oxidative burden is caused by a homeostatic imbalance between the ROS and antioxidant defense mechanism, which has been shown to be involved in AD pathology [16]. The important sources of cellular signaling molecules in normal homeostatic states are superoxide (O2), hydroxyl (-OH), peroxyl (ROO), hydrogen peroxide (H2O2), and organic peroxides (R-O-O-R′), whereas abnormal accumulation may result in excessive ROS production that causes irreparable damage to the biomolecules [17]. The mitochondrion is known as the powerhouse of the cell [18] because it has the metabolic potency to synthesize adenosine triphosphate (ATP) [19] through oxidative phosphorylation, which involves the transfer of electrons from high-energy substrates to oxygen via the electron transport chain [20]. ATP is the source of energy to drive and store a variety of vital activities at the cellular level [21]. Mitochondria are stress sensitive [22], where ROS are created as a result of abnormalities in several enzymes, including α-ketoglutarate dehydrogenase complex (KGDHC) [23] and pyruvate dehydrogenase complex (PDHC) [24], which play a pivotal role in denaturing their metabolic system [25,26]. Additionally, the degradation of the promoter of the encoding gene of mitochondrial synthase enzyme (due to oxidative damage) leads to the excessive generation of free radicals, causing mitochondrial abnormalities and ATP depletion [27].
The development of AD was observed in the APP/PS1 mouse model, where the mitochondrial ROS were reported to be involved in AD pathology [28]. Similarly, other studies have reported the involvement of mitochondrial ROS in AD pathology by inducing an oxidative burden in the brain, leading to synaptic damage and cognition [29]. Furthermore, the enhanced ROS-induced oxidative damages including lipid peroxidation, protein oxidative damage, and glycoxidation was observed in AD patients [30]. Moreover, evidence from an in vitro study also confirmed ROS the leading agent of neuronal dysfunction, including AD pathology [31]. Additionally, the evidence of the progression of AD pathology due to OS was further confirmed by an in vivo study where the administration of a nanoparticle-based approach in mice exhibited the inhibition of the neuronal ROS [32]. Overall, these findings reveal that ROS triggers an imbalance in the antioxidant system which is the causal pathway for the progression of AD.

2.1. Contributory Sources of Oxidative Stress in AD

As OS is a major hallmark of AD like neurodegeneration [33], studies have reported other factors were also involved in boosting the ROS level, e.g., abnormal Aβ deposition, glial cell activation, homeostatic imbalance of nutrients (trace minerals zinc, iron, and copper) [34], altered cellular signaling pathways, [35] and abnormal nuclear factor erythroid 2-related factor (Nrf2) signaling [36].

2.1.1. Accumulation of Abnormal Amyloid Beta

1–42, which is obtained as a result of the proteolytic cleavage of amyloid precursor protein (APP) by different secretases (amyloidogenic pathway), is reported to be involved in causing oxidative burden in AD [15]. The abnormal Aβ peptides causes OS, which has been shown to initiate calcium dyshomeostasis in mice brains, activating N-methyl-D-aspartate (NMDA) receptors [37]. The excessive accumulation of free radicals, due to the abnormal Aβ metabolism, damage numerous biomolecules including proteins, unsaturated fatty acids, and mitochondrial DNA. This can lead to toxicity in different cellular signaling pathways such as pathogen invading defense system, gene transcriptional regulation and other enzymatic activities, which are the basic etiological factors in AD [38].

2.1.2. Activation of Glial Cells

Microglia and astrocytes are the basic safeguards for the central nervous system (CNS), as they facilitate the clearance of Aβ in the brain and maintain the permeability of the blood–brain barrier (BBB) [39,40]. Upon the over-activation of glial cells, through OS because of abnormal deposited Aβ protein or lipopolysaccharides (LPS), the inflammatory cytokines and mediators including interleukin (IL)-1β, IL-6, and interferons (IFNs), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumor necrosis factor alpha (TNF-α), and nitric oxide synthase (NOS) are released. These inflammatory cytokines and mediators initiate neuroinflammatory cascades, neuronal dysfunction, and synaptic and memory abnormalities which have been shown to be linked with progression towards AD pathology [15,41]. Similar activation of the glial cells was reported to be involved in inflammatory cascade by using APP/PS1 mice and an AD brain, where the enhanced expression of IL-1β and IL-6 was observed [42].

2.1.3. Abnormal Cellular Pathways

An important consequence of the loss of nerve cells is altered neurotransmission and OS is one of the major players in abnormal neurotransmission [43], as numerous enzymatic metabolic dysfunctions and their abnormal activation were reported to be provoked by OS in different signaling pathways. For example, the stress-activated protein kinase (SAPK) pathways play a pivotal role in mediating the stress signals into the nucleus. Therefore, these pathways initiate different kinds of activated protein signaling such as ROS/protein kinase C (PKC)-dependent NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways that are reported to be involved in the accumulation of abnormal Aβ peptides [44]. Additionally, other studies also highlighted the toxic role of OS that are enhancing the expression of c-Jun N-terminal kinase (JNK)/p38 levels, related to the deposition of neuritic plaques and neurofibrillary tangles in AD [45]. To prevent neural dysfunction, the activation of endogenous antioxidant mediators is crucial and, in AD, the inflammatory and oxidative hypotheses appears to play a significant role [46]. The electron transport chain’s byproducts, such as hydrogen peroxide radicals, superoxide radicals, and hydroxyl radicals, are mostly where the oxidative pathways pass through [47] and nuclear factor erythroid 2-related factor 2 (Nrf2) appears to operate as an upstream mediator to control these pathways/mediators. In turn, antioxidant response elements (ARE) and Kelch-like ECH-associated protein-1 (Keap1) tightly regulate Nrf2 [48,49]. As a result of Keap1 alteration brought on by oxidative stress, the Keap1/Nrf2 connection is broken and Nrf2 degradation is prevented. This causes Nrf2 to translocate into the nucleus, bind to ARE, and then activate antioxidant enzymes. The production of inflammatory mediators such as IL-1, IL-6, TNFs are also boosted by increasing the levels of oxidative stress, phosphatidylinositol 3-kinases (PI3K)/protein kinase B (PKB)/mammalian target of rapamycin (mTOR) [50,51]. Lastly, pro-inflammatory cytokines generated during inflammation cause synapse loss and neuronal damage that contributes to the course of AD [36].

3. Bioactive Compounds and Their Role in AD as Antioxidants

A healthy lifestyle through balanced diet, which contains various bioactive compounds, plays a pivotal role in reducing the risk of AD dementia [52]. Due to the lack of sufficient knowledge, the jury is out on these compounds, their availability in food, chemical forms, and their role in neuroprotection. However, some of these natural compounds and their roles as antioxidants in neuroprotection have been reported in some observational epidemiological studies and experimental research, annotating the proper molecular signaling mechanism and role in neuroprotection, especially in AD [53,54].
Antioxidants have been reported to be involved in homeostatic balance in different biological activities. These natural or synthetic antioxidants include glutathione peroxidase, Catalase (CAT), glutathione reductase, Superoxide Dismutase (SOD), Nicotinamide Adenine Dinucleotide Phosphate (NADPH), vitamin C, mannitol, bilirubin, and Glutathione (GSH), β-carotene, which play a major role in ROS reduction in AD [9]. Numerous natural antioxidants that are available in food, such as Lupeol from tomato [15], catechins, and theaflavins from tea, and curcumin from turmeric are the major focus of researchers due to their wide range of biological functions, including neuroprotection in AD [55]. For example, mitochondrial ROS generate free radicals which are counteracted by the natural antioxidant defense system. However, these natural antioxidant defense mechanisms fail in the event of excessive oxidative damage because the metabolic machinery in the cell is unresponsive. To cope with this damage, researchers are investigating different types of strategies, e.g., the use of natural bioactive compounds extracted from different medicinal plants that are used as antioxidants because of their easy accessibility, cost-effectiveness, and versatile bioactivities, such as scavenging or nearly inhibiting ROS and free radicals in different health issues, including AD [56,57].

4. Lupeol as an Antioxidant and Anti-Inflammatory Agent (Neuroprotective Features)

4.1. Antioxidant Potentials of Lupeol

Numerous naturally occurring plants, including onion, oranges, apple, and tea, have been used to extract bioactive compounds that are used as antioxidants to treat neuronal dysfunction, out of which triterpenoids are playing a pivotal role in different types of chronic disorders, including neurodegenerative diseases such as AD, Huntington’s disease (HD), and Parkinson’s disease (PD) [11,58]. These compounds are metabolic derivatives of the oligomeric isopentenyl pyrophosphate (phytochemicals). Lupeol is a pentacyclic triterpenoid and has many biological functions. It is available in fruits (e.g., mango, fig, strawberry, and red grapes), vegetables (e.g., white cabbage, pepper, cucumber, and tomato), and numerous other medicinal plants [13,14]. Lupeol has shown beneficial effects against numerous health issues, including anti-cancer, anti-microbial [59], anti-diabetic [60], cardio [61], and hepatoprotection [62] (Table 1).
Evidence has declared the beneficial antioxidant features of Lupeol in streptozotocin (STZ)-induced hyperglycemic rats’ model, where the expression of Superoxide Dismutase 2 (SOD-2) and Heme Oxygenase-1 (HO-1) were noticeably increased by the treatment of Lupeol [13]. A study was conducted where the antioxidant effect of Lupeol was investigated in the streptozotocin (STZ) and aluminum chloride (AlCl3)-induced male Sprague–Dawley rat model. The improved antioxidants in cortex cerebellum, such as, CAT, SOD, thiobarbituric acid reactive substances (TBARS) and GSH were found [60]. The antioxidant potentials of Lupeol and its derivatives (isolated from the stem bark of crateva nurvala) were further confirmed in triton-induced hyperlipidemic adult male rats of the Charles Forest strain, where the reduced superoxide anions and hydroxyl free radicals were found by the administration of Lupeol and chalcone (derivatives of Lupeol) [63]. Furthermore, Santiago et al. also confirmed the antioxidant nature of Lupeol (extracted from Ficus pseudo Palma Blanco) (Moraceae) against nitric oxide (NO), hydroxyl and superoxide radical scavenging potentials [64]. Similarly, Lupeol was demonstrated in diabetic rats to play a role in hepatic glucose metabolism. An enhanced and improved liver glucose level was found, including antioxidant functions [65]. Similarly, a study was carried out by Sunitha et al. to find the antioxidant capability of Lupeol and its chalcone Lupeol Lineolate by oral administration on the hepatotoxicity in the rat model. Significant improvement was observed in antioxidant level in the liver [66].The fetal cardiotoxicity induced by oxidative stress was inhibited by Lupeol, its ester, and Lupeol linoleate administration in cyclophosphamide-treated experimental rats, where the antioxidant potency was also exhibited by Lupeol [67]. Reduced oxidative abnormalities and improved enzymatic SOD, GPx and non-enzymatic GSH, ascorbic acid, vitamin E and antioxidants by the treatment of Lupeol and its linoleate ester derivative were reported in the early stage of hypercholesterolemic atherosclerosis in rats [68].

Role of Lupeol as an Antioxidant in Neuroprotection

The antioxidant and neuroprotective activities of Lupeol have been well reported in cognitive deficit, and neurochemical and biochemical abnormalities in rats as well [69] (Table 1; Figure 1). The nano-based delivery approach was used, where the antioxidant and neuroprotective potentials of Lupeol were investigated in an ischemic brain [70]. Since it is a naturally abundant triterpenoid, its medicinal outcomes were well studied [71]. For example, one study has shown a free radical scavenging activity of Lupeol since it has the potentials to donate electron/hydrogen in its structure [72]. The antioxidant potentials of Lupeol (oral administration of Lupeol at a dose of 50 mg/kg for two weeks) are well observed in Aβ-induced neuronal dysfunction in a mouse model, where Lupeol was observed to be involved in reducing oxidative stress and memory impairments by enhanced expression of Nrf2, HO-1 level [15]. Similarly, traumatically brain-injured mice were treated with Lupeol (50 mg/kg/day/mice/p.o.) to check their antioxidant potentials, where the oxidative stress and ROS level were observed to be reduced by Lupeol treatment [73]. Furthermore, Lupeol was treated against a STZ+AlCl3-induced diabetic and AD rat model to observe its antioxidant features and a significant improvement was found in cognition and memory impairments [60]. A study was conducted where Lupeol was investigated for its antioxidant and neuroprotective potentials against Aβ1–42-induced oxidative burden and neurodegeneration in mice model. The expression level of some of the markers related to oxidative stress such as Nrf-2 and HO-1 were found to be upregulated by the treatment of Lupeol (at a dose of 50 mg/kg) [15]. A similar study was carried out by Zhang et al. in middle cerebral artery occlusion (MCAO) ischemic rats, where oxidative stress and the neuroprotection of Lupeol (6 mg/mL dissolved in olive oil) was observed, while a significant improvement in activated Nrf2 and inhibited phosphorylated p38 MAPK was found [74].
Table 1. Antioxidant features of Lupeol.
Table 1. Antioxidant features of Lupeol.
SourceMechanismModelReference
Crataegus oxyacantha↓NF-κB, Vegf-A, IL-6,
↑FGF-2,TGF-β1, ↑collagen III, ↑IL-10
Streptozotocin-induced hyperglycemic rats[13]
Mango Pulp, Egg, Plant, Cucumber, and Soybean↑NO, ↓Mg2+,Ca2+
↓endonuclease III
Escherichia coli[59]
Hedera Nepalensis crude extract (HNC)↑CAT, SOD
↓GSH, dopamine, serotonin
In vivo STZ + AlCl3-induced diabetic AD[60]
Medicinal Plants↓NRCMs, ANP, ↓BNP, ↓β-MHC, NF-κB p65In vivo and in vitro cardiac hypertrophy in neonatal rat’s cardiomyocytes (NRCMs)[61]
Vegetables, Edible Fruits↓IκBαIn vivo dextran sulfate sodium (DSS)-induced acute colitis[75]
Fruits, Vegetables↓TGFβ1, ↑Nrf2In vivo LPS/D-galactosamine(D-GalN)-induced liver injury[62]
Stem Bark of C. Nurvala↓TC, PLTAGIn vivo triton-WR 1339-induced hyperlipidemia.[63]
Ficus pseudopalma Blanco (Moraceae)↓NOEthanolic leaf extract of F. pseudopalma[64]
Cassia Fistula↓MDA
↑SOD, GSH, CAT
In vivo anti-diabetic study l[76]
Banana Flower↑SOD, CAT, GPx
↑GSH, VitC, VitE
In vivo hyperglycemic model[65]
Medicinal Plants↑SOD, GST, G6PD, GSH, ↑GPX, γ-GTIn vivo chronic cadmium exposure in kidney[77]
Stem Bark of C. nurala↑SOD, CAT, GPx, ↑G6PD, GST, GR, ϒ-GTIn vivo cadmium-induced hepatotoxicity[66]
Mango Pulp Extract (MPE)↓ROS,
↑Cu, Zn-SOD, CAT, GR and GST
In vivo androgen-induced oxidative stress in prostate.[78]
Crataeva Nurvala Stem Bark↑GSH, Vit C, Vit EIn vivo CP-induced cardiotoxicity[67]
Crataeva Nurvala Buch-Ham (Capparidaceae)↓TC, TG, LDL VLDL
↑HDL
In vivo hypercholesterolemic atherosclerosis[68]
Betula Alnoides↓AChE, MDA, nitrite,
↑GSH
In vivo amyloid beta-induced neuronal damage[69]
Bombax Ceiba↑Tgel
↓rhodamine-B, 5,6-carboxyfluorescein
Molecular modeling studies, X-ray diffraction data and FTIR studies[71]
Crateva Adansonii Oliv. (Capparidaceae)↓MDA,
↑CAT, GSH
In vivo CCl4-induced hepatotoxicity[72]
Fruits, Vegetables, and Herbs↓Aβ, ↓NOS2
↑Nrf2, HO-1
In vivo Aβ-induced AD[15]
Vegetables, Fruits↑Nrf2, HO-1In vivo traumatic brain injury[73]
Vegetables and Fruits↑Nrf2, ↓P38In vivo ischemic toxicity[74]
Abbreviations: Nuclear Factor-κB (p-NF-κB); Vascular Endothelial Growth Factor-A (Vegf-A); Fibroblast Growth Factor-2 (FGF-2); Transforming Growth Factor Beta-1 (TGF-β1); Interleukin-6 (IL-6); Nitric Oxide (NO); Magnesium (Mg2); Calcium (Ca2); Catalase (CAT); Superoxide Dismutase (SOD); Glutathione (GSH); Neonatal Rat Cardiomyocytes (NRCMs); Atrial Natriuretic Peptide (ANP); Brain Natriuretic Peptide (BNP), β-myosin Heavy Chain (β-MHC); Nuclear Factor of Kappa Light Polypeptide Gene Enhancer in B-cells Inhibitor, alpha (IκBα); Transforming Growth Factor Beta 1 (TGFβ1); Nuclear Factor Erythroid 2–related Factor 2 (Nrf2); Glutathione (GSH); Glutathione S Transferase (GST); Nitric Oxide (NO); Malondialdehyde (MDA); Total Cholesterol (TC); Glutathione Peroxidase (GPx); Glucose-6-Phosphate Dehydrogenase (G6PD); Glutathione Reductase (GR); Copper (Cu); Zinc (Zn); Low-Density Lipoprotein (LDL); Haemoxygenase (HO-1); Very-Low-Density Lipoprotein (VLDL); High-Density Lipoprotein HDL, Acetylcholinesterase (AChE); Gel-to-sol Transition Temperature (Tgel), Amyloid Beta (Aβ); Beta-secretase 1 (BACE1); Glial Fibrillary Acidic Protein (GFAP); Ionized Calcium-Binding Adapter Molecule (1Iba-1); Cyclooxygenase-2 (COX-2); Caspase-3 (Casp-3), Bcl-2-Associated X Protein (Bax); Cytochrome C (Cyt C). The symbols ↓ and ↑ are representing the expression level of proteins to be decreased and increased respectively.

4.2. Anti-Inflammatory Potential of Lupeol

Chronic inflammation produces excessive cytokines and chemokines that develop a chronic pathological condition [79]. Lupeol has shown anti-inflammatory effects in numerous pathological insults [61,80], including neuronal inflammation [74] (Table 2). The anti-inflammatory potentials of Lupeol were observed in skin wound healing in rats by the modulation of NF-kB and Ki-67 [81]. Lupeol treatment was also reported to be involved in anti-inflammatory activities of allergic airways in a murine model [82]. To confirm the anti-inflammatory potentials of Lupeol, a study was performed where the activation of inflammatory markers toll-like receptor-4 (TLR4) and NF-κB were found in an osteoarthritis rat model, while Lupeol treatment attenuated these inflammatory agents and exhibited its anti-inflammatory effects [83]. The inflammation induced by LPS in retinal pigment epithelium cells (ARPE-19) was recovered by the intravitreal injection of Lupeol (100 µM) in non-infectious uveitis rats [84]. Moreover, the intraperitoneal administration of Lupeol (10, 25, or 50 mg/kg) was found to be effective against inflammatory cytokines such as tumor TNF-α, IL-1, and IL-6, in cerulein-induced acute pancreatitis in mice [85]. Lupeol (50, 100 mg/kg) was used to alleviate the inflammation brought due to the activation of TLR4 in a mouse model of viral myocarditis induced by coxsackie virus B3 (CVB3) [86]. Moreover, a study was conducted where the inhibitory and anti-inflammatory effects of Lupeol were observed in inflammatory bowel disease by inhibiting M1 and boosting M2 macrophages. The reduced expression of the pro-inflammatory cytokines, including IL-12, IL6, IL-1β and TNF-α, and an enhanced production of IL-10, an anti-inflammatory cytokine, were found by the oral administration of Lupeol (50 mg/kg, q.d.) [87]. In vivo and in silico approaches were used to show the anti-inflammatory scope of Lupeol isolated from Indian traditional plant Crateva adansonii, where its inhibitory and anti-inflammatory effects against key molecules of inflammation such as MPO, PGE2, and eight pro-inflammatory cytokines were found to be effective [88]. However, since in silico studies take the form of computational analysis, they need to be validated through an in vitro/vivo model. Lastly, Lupeol (10 μg/ear) was used to investigate its anti-inflammatory activities in a mouse model of skin inflammation [89]. The acute ear edema was reduced by 51 ± 7% using Lupeol (10 μg/ear).

Anti-Neuroinflammatory Features

The neuroprotective and anti-neuroinflammatory features of Lupeol were well studied against LPS-induced neuroinflammation via the p38/JNK pathway in mouse brain [41]. Similarly, the anti-inflammatory activity of Lupeol (extracted from constituents of Pyrus pyrifolia fruit) on the LPS-induced nitric oxide (NO) production and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in macrophages and microglia were confirmed in an in vitro work, where the protein expression level of iNOS and COX-2 were increased due to NO induction, while Lupeol treatment (in a dose-dependent manner over a concentration range from 2.5 to 10 µg/mL) significantly inhibited the iNOS and COX-2 expression [90]. This finding suggests that Lupeol treatment is able to inhibit the NO level in macrophages and microglia via different mechanisms in neurodegenerative disease to reduce neuronal inflammation, including AD. The activation of stress-activated protein kinases, including p38-MAPK and JNK were induced by LPS and generate oxidative burden. Moreover, the phosphorylated p38 and JNK further activate the transcription factor activator protein (AP)-1, leading to the initiation of numerous other inflammatory genes such as IL-1, IL-2, CD40, TNF-α, and c-Jun, ultimately causing cell death. Lupeol administration brought a significant reduction in p-p38 and p-JNK protein levels [41]. Similarly, the anti-inflammatory and neuroprotective effects of Lupeol in LPS-induced neuronal inflammation in primary cerebellar cultures were also investigated, and RT-qPCR analysis exhibited the downregulation of the mRNA expression for TNF-α-, iNOS- and NLRP3-like proinflammatory markers, while a gradual reduction in the production of NO was also observed [91]. Lupeol (200–500 ng) extracted from Celastrus paniculatus (10 mg/mL), commonly called black oil plant, was used in the treatment of Parkinson’s disease (PD), which exhibited differential neuroprotective and protein-aggregation-mitigating effects in C. elegans. Later, the highest percentage of neuroprotection, including an improved nervous system performance and a reduction in the level of symptoms of PD, was observed [92]. Moreover, the anti-inflammatory and neuroprotective features of Lupeol in traumatic brain injury were also reported via the Nrf2/HO-1 pathway in a mouse model [73]. The in vitro inhibition of monoamine oxidase A and B (MAO-A and -B) in mouse macrophages exhibited the neuroprotective and anti-inflammatory potentials of Lupeol [93]. Similarly, the anti-inflammatory potentials of Lupeol (6 mg/mL) were further investigated in a middle cerebral artery-induced cerebral ischemia in rats involving Nrf2 and P38 MAPK modulation [74]. In this study, they also studied the cell viability assay to know about the toxicity features of this compound. The inflammatory effects produced due to the LPS-induced NO were investigated by the treatment of Lupeol (isolated from Pueraria lobata roots) in LPS-stimulated RAW 264.7 cells, where Lupeol was found to induce anti-inflammatory effects [94]. Lupeol (12.5, 25, 50, 100 mg/kg) reduced cerebral ischemia–reperfusion damage in relation to PI3K/Akt pathway modulation [95]. The better BACE1 binding affinity and BACE1 inhibitory potentials of Lupeol (obtained from Leea indica, and Pueraria lobata roots) as compared to ursolic acid [96] and quercetin [97] were observed.
In short, in accordance with the all these findings regarding the inhibitory role of Lupeol in different inflammatory signaling pathways, including the neuro- inflammation and neuroprotective mechanism in different neurodegenerations, it can be expected that a new window of observational study and research can be opened to investigate the neuroprotective and anti-inflammatory potentials of Lupeol for the inhibition of numerous neuronal diseases (Table 2).
Table 2. Anti-(neuro)inflammatory potentials of Lupeol.
Table 2. Anti-(neuro)inflammatory potentials of Lupeol.
SourceMechanismModelReference
Edible Plants, such as Olive, Fig, Mango, Carrot, Soybean↓NF-kB,
↑Ki-67
Skin wound healing in rats[81]
Stembark of D. Ferruginea Benth.↓IL-4, IL-5, IL-13In vivo animal model of allergic airway inflammation[82]
SHM Herbs↓TLR-4, NF-ĸB, IL-1In vivo rats with osteoarthritis[83]
Maytenus Salicifolia Reissek↓IL-6, IL-8In vitro rodent model of pan uveitis[84]
Vegetable Oils, Cereals, Fruits↓TNF-α, IL-1β, IL-6In vitro cerulein-induced acute pancreatitis in mice[85]
Vegetables, Fruits↓TLR4, TNF-α, IL-1βIn vivo mouse model of viral myocarditis induced by coxsackie Virus B3 (CVB3)[86]
White Cabbage, Pepper, Cucumber, Tomato, and Fruits such as Olive↓IL-12, IL6, IL-1β, TNF-α, CD86
↑IL-10, ↑CD206
In vivo and in vitro DSS-induced colitis mouse model[87]
Crateva Adansonii Leaf Extracts↓TNF-α, IL-1, IL-6In vivo and in silico approaches in rats[88]
Crude extract of Cariniana domestica Fruit Peels (CdE), ↓MPO activity
↓Ear edema, inflammatory cell infiltration
Mouse model of skin inflammation[89]
Pyrus Pyrifolia↓iNOS, COX2LPS-activated RAW 264.7 and BV2 cell lines[90]
Olives, Mangos
and Strawberries
↓Bax, Cyt-C, caspase 9
↓caspase 3, TNF-α, iNOS, IL-1β
↓ p-JNK, P38
In vivo LPS-induced neuronal dysfunctions[41]
Olea Europaea (Olive), Mangifera Indica(Mango)↓TNF, iNOS and NLRP3
↓IL-6 mRNA
In vitro LPS-induced neurodegeneration[91]
Celastrus paniculatus (CP)↓α-synIn vivo C. elegans PD model[92]
Vegetables↑Nrf2/HO-1
↓Iba-1, GFAP
↓p-NFkB, TNFα, COX-2, IL-1
↓Casp3, Cyt-C, BAX/BCL2
In vivo TBI-induced neurodegeneration in male wildtype C57BL/6 N mice[73]
Vitex Grandifolia↓ iNOS, NFkBIn vitro MAO-A and B-induced neuronal dysfunction and neuroprotection in mouse macrophages[93]
Peppers, Tomatoes, Olive Oil↑PI3K/AktIn vivo cerebral ischemia–reperfusion injury in rat model[95]
Vegetables and Fruits↑Nrf2,
↓BAX/Bcl-2, caspase-3
↓p38 MAPK
In vivo middle cerebral artery occlusion (MCAO) followed by reperfusion (MCAO/R)-induced cerebral ischemia.[74]
Medicinal Plants↓Antioxidant ActivitiesIn vivo middle cerebral artery occlusion (MCAO)-induced ischemic stroke(nano approach)[70]
Pueraria Lobata
Roots
↓NO, iNOS, COX-2
↓ ROS
In vitro study of RAW 264.7 murine macrophages[94]
Peppers, Tomatoes, Olive oil, Figs↑p-PDK1, p-Akt, pc-Raf, p BAD, ↓Casp-3In vivo cerebral ischemia–reperfusion injury in rats[95]
Abbreviations: Nuclear factor-κB (p-NF-κB); Interleukin-1 beta (IL-1β); Toll-like receptors (TLR-4), Tumor necrosis factor alpha (TNF-α); Cluster of Differentiation 86 (CD86); Myeloperoxidase (MPO); Inducible nitric oxide synthase (iNOS); Phosphorylated Cyclooxygenase-2 (COX-2); Bcl-2 Associated X-protein (BAX); Phosphorylated c-Jun N-terminal Kinase (p-JNK); NLR Family Pyrin Domain Containing 3 (NLRP3); Cytochrome C; Phosphoinositide 3-kinases (PI3K); Alpha-synuclein (α-syn); Interleukin 6 (IL-6); Nuclear factor erythroid 2-related factor 2 (Nrf2); Heme oxygenase 1 (HO-1); p38 mitogen-activated protein kinases (p38); Glial fibrillary acidic protein (GFAP); Ionized calcium-binding adapter molecule 1 (IBA1); Reactive oxygen species (ROS), Phosphoinositide-dependent protein kinase 1 (PDK1), Phosphorylated-protein kinase p-Akt, proapoptotic BH3-only protein BAD. The symbols ↓ and ↑ are representing the expression level of proteins to be decreased and increased respectively.

5. Neuroprotective Role of Lupeol in AD

Lupeol has shown neuroprotective effects in numerous neurological diseases, including AD (Table 3). Research has shown that Lupeol reduced the Aβ-induced oxidative burden, while increasing the level of Nrf2 and HO-1 in the cortex and hippocampus of Aβ-induced mice [15]. Glutamate is claimed to be involved in the initiation of apoptosis by stimulating abnormal redox-induced oxidative stress, and the reduction of glutathione levels and Aβ which leads to neuronal ROS generation [98]. Therefore, an in vitro study investigated glutamate and Aβ-induced OS-mediated neuronal toxicity, where Lupeol, extracted from Rhinacanthus nasutus, was effective against glutamate and Aβ-induced neurotoxicity in HT-22 mouse hippocampal cells [98]. Similarly, the neuroprotective potentials of Lupeol were investigated against Aβ-induced neurotoxicity, where behavioral and memory impairments, enhanced oxidative burdens, a reduction in antioxidants enzymatic system, and an increased expression level of proinflammatory cytokines were found in Wistar rat brains, while Lupeol (25, 50, and 100 mg/kg/day per orally) was found to be effective against all these parameters [69]. Moreover, the neuroprotective and antioxidant features of Lupeol were reported against STZ+AlCl3 in diabetic-induced AD in a rat model, where the reduced blood glucose level, rising level of CAT, SOD and lower GSH level were observed by the oral administration of Lupeol (10 mg/kg) [60]. Although the neurotransmitter acetylcholine (ACh) is degraded at the cholinergic synaptic position, that results in the generation of the acetyl group and choline by acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). This reduced level of AChE is found to be involved in originating numerous biochemical changes, including neuronal dysfunctions in AD individuals. For this reason, the inhibitory potentials of Lupeol against AChE and BChE was investigated using the docking procedures using the Genetic Optimization for Ligand Docking suit v5.4.1 [99]. Moreover, the potential of Lupeol in AlCl3-induced neurotoxicity in Wistar rats was observed, where the elevated levels of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) and degenerative changes in the hippocampal brain region in AD were inhibited by treatment with Lupeol (25 and 50 mg/kg, p.o.) [100]. Moreover, BACE1 (beta-site amyloid precursor protein (APP) cleaving enzyme 1), a member of the pepsin family of aspartyl proteases, was first identified in 1991 [99]. BACE1 is abundantly expressed in several types of neuronal cells and is concentrated in neurons, oligodendrocytes, and astrocytes in the brain [101]. Previous studies have focused on its functions as the secretase that causes the synthesis of amyloid beta, which is shown in Alzheimer’s disease [102]. The inhibitory and strong binding potentials of Lupeol (obtained from Lea indica) against BACE1 was observed through the application of molecular docking and molecular dynamics-based approaches, where the binding mechanism of Lupeol and ursolic acid with BACE1 was compared by the help of induced fit docking and classical molecular dynamics, including the steered molecular dynamics mechanism. Lupeol produced a higher binding free energy (211.87 kJ/mol) than ursolic acid (−50.23 kJ/mol), showing that Lupeol binds to the BACE1 enzyme significantly stronger than ursolic acid [96]. Furthermore, in this study, the classical and steered dynamics also revealed the favorable hydrophobic interactions between the Lupeol and the residues of the flap or catalytic dyad of BACE1. Moreover, in silico docking of mangrove legends against the AD receptor protein also confirmed the binding affinity of Lupeol with Aβ and acetylcholinesterase inhibition [103]. In this study, the binding strength, hydrogen bond length, and components of amino acid and cluster-like features were observed. However, since only the in-silico approach was used in this study, in vitro or in vivo confirmation is needed to validate the in silico model. Additionally, the inhibition of BACE1 through Lupeol was explored by comparative molecular studies, where neuroprotective inhibitory capacities of Lupeol against BACE1 were found in AD by applying the enzyme kinetics study [97]. The low inhibition constant (K) value of 1.43 μmol/L indicates the binding possibilities of Lupeol with BACE1 and the inhibitory features of Lupeol.

6. Pharmacokinetics of Lupeol

The Pharmacokinetic parameters of a certain compound play an important role in clinical and preclinical research to develop a drug through which the theoretical details are provided for showing the use of drugs, estimate of drugs, mechanism of action and to discover the fresh tactics to change the natural products, and plan the drug transfer system [105]. Some important characteristics of the target drug includes absorption, distribution, metabolism, and excretion (ADME) inside the body. Due to the widespread pharmacological functions of Lupeol, it is likely to be a potential therapeutic agent for different kinds of disorders [106] (Table 4). These ADME features (such as drug likeness (DL) 0.78, Caco-2 1.46, and oral bioavailability (OB) 12.12%) of Lupeol were further investigated in the traditional Chinese medicine systems pharmacology database and analysis platform (TCMSP), backing its drug resemblance [107]. Furthermore, the pharmacokinetic parameters, binding to protein, and drug relations of Lupeol were investigated in both rat and mice models [108,109]. The systemic bioavailability of Lupeol was evaluated in female mice of the CD-1 strain where Lupeol was administered orally at 200 mg/kg, using the UPLCAPCI+-MS/MS procedure. In this study, different pharmacokinetic parameters were evaluated by using the mono-compartmental model, where higher values of both the AUC (area under the curve) and Cmax (maximum plasma concentration) were noticed in solid–lipid nanoparticles (SLNs). The results revealed the elimination of Lupeol through feces with a maximum elimination time of 12 h, having a value of 163.28 ± 9.83 ng/mg. Additionally, the absorption of Lupeol by animals was also better [109]. The quantification of Lupeol in rats’ plasma was investigated through LC-MS/MS method, where the mean pharmacokinetic parameters of Lupeol were measured after the intravenous (i.v) and oral injection of 1 mg/kg and 30 mg/kg doses. The AUC0-t (h × ng/mL), Cmax (ng/mL), Kel (h− 1), Tmax (h) and T1/2 (h) were calculated after the administration of 1 mg/kg dose of Lupeol, such as 21,584.53, 12,485.69, 21,866.18, 0.14, 0.08, and 4.95, respectively, while 30 mg/kg dose of Lupeol oral administration the AUC0-t (h × ng/mL), Cmax (ng/mL), AUC0-t (h × ng/mL), Kel (h − 1), Tmax (h) and T1/2 (h) were measured as, 2190.49, 133.33, 2727.52, 0.08, 4.67, and 8.66, respectively. It was concluded from the oral administration route of Lupeol that the bioavailability of Lupeol orally is less than 1% [110]. However, steps should be taken to develop strategies through which the solubility and oral availability of Lupeol can be improved. Further, the effects of solid lipid nanoparticles (SLN) on the bioavailability of Lupeol (extracted from Ficus religiosa L.) was investigated, where 50 mg/kg of Lupeol was administered orally and the various pharmacokinetic parameters were observed, where it was noticed that all the investigated parameters were improved by the oral treatment of SLN-loaded Lupeol at 50 mg/kg [108]. The PEGylated liposomes loaded with Lupeol were also reported by Jun Zhang et al. to have the potential to improve the pharmacokinetic parameters of Lupeol via solving the bioavailability and hydrophilicity-like parameters [111]. In this study, the intravenous administration of Lupeol (10 mg/kg) was performed in Sprague Dawley rats, where the different pharmacokinetic parameters were measured and the AUC (area under the plasma concentration–time curve), MRT (mean residence time) and t1/2 values were improved.
Due to insufficient research on the metabolic mechanism of Lupeol, there is a scarcity in pharmacokinetic literature of this compound. As Lupeol has a poor water solubility (PubChem Compound Summary for CID 259846, Lupeol) remedies are needed on how to increase its solubility in water and, also, drug loading should be taken into consideration while designing the formulations for the method of administration in a traditional mode. Strategies are needed to conduct more pharmacokinetic studies on Lupeol so that its viability in clinical purposes may be assessed.

Basic Challenges and Prospects to Boost the Pharmacokinetics and Pharmacodynamics of Lupeol

Despite the limited therapeutic uses of Lupeol because of some basic limitations, including its low solubility, bioavailability and drug delivery, which were reported in [111], numerous novel advances of targeted drug transfer and advanced techniques were also introduced regarding this compound, such as Lupeol-loaded PEGylated liposomes [111], NF-kB-PLGA nanoparticles that were loaded with Lupeol [109], Lupeol-entangled chitosan–gelatin hydrogel films [112], gold nanoparticles [113], and solid–lipid nanoparticles (SLNs) [108]. Briefly, the latest remedies are expected to develop novel methods by which all the designed parameters of Lupeol can be boosted, including solubility, cell proliferation and its effectiveness against neurodegeneration, especially AD.

7. Safety and Toxicity Contour of Lupeol

Lupeol has been investigated in different animal and clinical studies which reported the beneficial effects, with no toxic effects [114]. The oral administration of Lupeol in a dose of 2 mg/kg had no lethal effects over mice and rats [115]. Similarly, Lupeol (50 mg/kg/day/mice/p.o.) treatment attenuated the activation of glial cells and oxidative-stress-mediated neuronal dysfunctions in a mouse model of traumatic brain injury and did not cause any harmful effects [73]. Moreover, the neurotropic and anti-inflammatory role of Lupeol(0.1 μM) was demonstrated in LPS-induced neuronal inflammation in primary cerebellar cultures, and induced neuroprotection related to altered response of astrocytes and expression of neurotrophic and inflammatory factors, where no toxic outcomes of lupeol were found [91]. Furthermore, the antioxidant activities of Lupeol against glutamate and Aβ toxicity were also reported in an in vitro model with beneficial outcomes [98]. A clinical study performed in carcinoma patients [116] also exhibited the effective beneficial capabilities of Lupeol. Meanwhile, a pilot study was performed for the treatment of actinic keratoses using birch bark extract (containing Lupeol and other compounds), where twenty-eight patients of actinic keratoses were observed in a non-randomized pilot study for two months of clinical observation, and the birch-bark-containing Lupeol was able to clear 75% of lesions in patients [117]. Similarly, a double-blind randomized placebo-controlled phase II clinical trial was observed in children having the health issue of bedwetting (enuresis), which is upsetting and stressful for the child’s life. Lupeol (stem bark extract standardized at 1.5%) was able to give the best health outcomes, including reduced frequency of nocturia, safety, quality of life, and daytime incontinency [118]. Overall, all these observational studies have provided evidence that the administration of Lupeol has no serious health concerns. Furthermore, these studies may open a new window for Lupeol in clinical studies, especially in the neuroprotection of AD.

8. Conclusions, Limitations and Future Remedies

To conclude, many mechanisms such as OS, aberrant Aβ-deposition, mitochondrial ROS, and gliosis contribute to neurodegeneration. OS is one of the most promising targets of researchers in today’s time to find the inhibitory therapeutic pathways for neuronal dysfunctions, including AD. Associated with synthetic compounds are limited health outcomes and higher toxicity profiles [119] in various diseases, including neurodegenerative disorders, where their neuroprotective potentials are exhibited in animal models, but fail in clinical trials [119]. Therefore, natural compounds may be preferred over synthetic compounds. Herein, we contend that the targeting of OS with Lupeol as a natural antioxidant therapeutic agent opens a reasonable window for reducing or alleviating neuronal dysfunctions. The ADME (pharmacokinetic parameters) analysis of Lupeol also suggest its therapeutic potential. A nano-based approach is also used to enhance the potentials of Lupeol in neuroprotection [70]. According to Jose David Sachez et al., 98% of the bioactive compounds have negligible BBB permeability [120], leading to their beneficial health outcomes being limited. The BBB permeability of Lupeol (isolated from Crataegus oxyacantha) was found to be effective. The anticholinesterase functions of this compound were also investigated via the molecular docking approaches [104]. However, due to lack of interest and inaccessibility to Lupeol, its pharmacokinetics, discrete molecular structural features, binding energy level, and effective dose concentration, its metabolism in CNS-like features is still missing, resulting in limited or failed clinical trials. Therefore, through extensive research analysis, the future therapeutic window for Lupeol as a neuroprotective agent can be opened, especially in AD pathogenesis.

Author Contributions

Review design, J.S.P., I.U.R. and K.C.; literature revision, data collection, and data analysis, I.U.R.; drafting manuscript, J.S.P. and I.U.R.; revising manuscript content, R.A. and H.J.L.; approving final version of manuscript, M.O.K., J.S.P., I.U.R. and K.C. who take responsibility for the integrity of the data analysis. All authors have read and agreed to the published version of the manuscript.

Funding

This review article was supported by the Neurological Disorder Research Program of the National Research Foundation (NRF), funded by the Korean Government (MSIT), grant number 2020M3E5D9080660.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Abubakar, M.B.; Sanusi, K.O.; Ugusman, A.; Mohamed, W.; Kamal, H.; Ibrahim, N.H.; Khoo, C.S.; Kumar, J. Alzheimer’s Disease: An Update and Insights Into Pathophysiology. Front. Aging Neurosci. 2022, 14, 742408. [Google Scholar] [CrossRef] [PubMed]
  2. Ball, S.L.; Holland, A.J.; Hon, J.; Huppert, F.A.; Treppner, P.; Watson, P.C. Personality and behaviour changes mark the early stages of Alzheimer’s disease in adults with Down’s syndrome: Findings from a prospective population-based study. Int. J. Geriatr. Psychiatry A J. Psychiatry Late Life Allied Sci. 2006, 21, 661–673. [Google Scholar] [CrossRef] [PubMed]
  3. Ayton, S.; Bush, A.I. β-amyloid: The known unknowns. Ageing Res. Rev. 2021, 65, 101212. [Google Scholar] [CrossRef] [PubMed]
  4. Nisbet, R.M.; Gotz, J. Amyloid-beta and Tau in Alzheimer’s Disease: Novel Pathomechanisms and Non-Pharmacological Treatment Strategies. J. Alzheimer’s Dis. 2018, 64, S517–S527. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, Z.R.; Huang, J.B.; Yang, S.L.; Hong, F.F. Role of Cholinergic Signaling in Alzheimer’s Disease. Molecules 2022, 27, 1816. [Google Scholar] [CrossRef] [PubMed]
  6. Ju, Y.; Tam, K.Y. Pathological mechanisms and therapeutic strategies for Alzheimer’s disease. Neural Regen. Res. 2022, 17, 543–549. [Google Scholar] [CrossRef]
  7. Ullah, R.; Park, T.J.; Huang, X.; Kim, M.O. Abnormal amyloid beta metabolism in systemic abnormalities and Alzheimer’s pathology: Insights and therapeutic approaches from periphery. Ageing Res. Rev. 2021, 71, 101451. [Google Scholar] [CrossRef]
  8. Roman, G.C.; Jackson, R.E.; Gadhia, R.; Roman, A.N.; Reis, J. Mediterranean diet: The role of long-chain omega-3 fatty acids in fish; polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine; probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Rev. Neurol. 2019, 175, 724–741. [Google Scholar] [CrossRef]
  9. Pritam, P.; Deka, R.; Bhardwaj, A.; Srivastava, R.; Kumar, D.; Jha, A.K.; Jha, N.K.; Villa, C.; Jha, S.K. Antioxidants in Alzheimer’s Disease: Current Therapeutic Significance and Future Prospects. Biology 2022, 11, 212. [Google Scholar] [CrossRef]
  10. Morris, M.C.; Tangney, C.C.; Wang, Y.; Sacks, F.M.; Bennett, D.A.; Aggarwal, N.T. MIND diet associated with reduced incidence of Alzheimer’s disease. Alzheimer’s Dement. 2015, 11, 1007–1014. [Google Scholar] [CrossRef] [Green Version]
  11. Sohag, A.A.M.; Hossain, M.T.; Rahaman, M.A.; Rahman, P.; Hasan, M.S.; Das, R.C.; Khan, M.K.; Sikder, M.H.; Alam, M.; Uddin, M.J.; et al. Molecular pharmacology and therapeutic advances of the pentacyclic triterpene Lupeol. Phytomedicine 2022, 99, 154012. [Google Scholar] [CrossRef]
  12. Kessas, K.; Chouari, Z.; Ghzaiel, I.; Zarrouk, A.; Ksila, M.; Ghrairi, T.; El Midaoui, A.; Lizard, G.; Kharoubi, O. Role of Bioactive Compounds in the Regulation of Mitochondrial Dysfunctions in Brain and Age-Related Neurodegenerative Diseases. Cells 2022, 11, 257. [Google Scholar] [CrossRef]
  13. Beserra, F.P.; Vieira, A.J.; Gushiken, L.F.S.; de Souza, E.O.; Hussni, M.F.; Hussni, C.A.; Nobrega, R.H.; Martinez, E.R.M.; Jackson, C.J.; de Azevedo Maia, G.L.; et al. Lupeol, a Dietary Triterpene, Enhances Wound Healing in Streptozotocin-Induced Hyperglycemic Rats with Modulatory Effects on Inflammation, Oxidative Stress, and Angiogenesis. Oxidative Med. Cell. Longev. 2019, 2019, 3182627. [Google Scholar] [CrossRef] [PubMed]
  14. Hajialyani, M.; Hosein Farzaei, M.; Echeverria, J.; Nabavi, S.M.; Uriarte, E.; Sobarzo-Sanchez, E. Hesperidin as a Neuroprotective Agent: A Review of Animal and Clinical Evidence. Molecules 2019, 24, 648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ahmad, R.; Khan, A.; Lee, H.J.; Ur Rehman, I.; Khan, I.; Alam, S.I.; Kim, M.O. Lupeol, a Plant-Derived Triterpenoid, Protects Mice Brains against Abeta-Induced Oxidative Stress and Neurodegeneration. Biomedicines 2020, 8, 380. [Google Scholar] [CrossRef] [PubMed]
  16. Martins, R.N.; Villemagne, V.; Sohrabi, H.R.; Chatterjee, P.; Shah, T.M.; Verdile, G.; Fraser, P.; Taddei, K.; Gupta, V.B.; Rainey-Smith, S.R.; et al. Alzheimer’s Disease: A Journey from Amyloid Peptides and Oxidative Stress, to Biomarker Technologies and Disease Prevention Strategies-Gains from AIBL and DIAN Cohort Studies. J. Alzheimer’s Dis. 2018, 62, 965–992. [Google Scholar] [CrossRef] [Green Version]
  17. Thorpe, G.W.; Reodica, M.; Davies, M.J.; Heeren, G.; Jarolim, S.; Pillay, B.; Breitenbach, M.; Higgins, V.J.; Dawes, I.W. Superoxide radicals have a protective role during H2O2 stress. Mol. Biol. Cell 2013, 24, 2876–2884. [Google Scholar] [CrossRef]
  18. Guerra, F.; Arbini, A.A.; Moro, L. Mitochondria and cancer chemoresistance. Biochim. Biophys. Acta Bioenerg. 2017, 1858, 686–699. [Google Scholar] [CrossRef]
  19. Benador, I.Y.; Veliova, M.; Mahdaviani, K.; Petcherski, A.; Wikstrom, J.D.; Assali, E.A.; Acin-Perez, R.; Shum, M.; Oliveira, M.F.; Cinti, S.; et al. Mitochondria Bound to Lipid Droplets Have Unique Bioenergetics, Composition, and Dynamics that Support Lipid Droplet Expansion. Cell Metab. 2018, 27, 869–885.e6. [Google Scholar] [CrossRef] [Green Version]
  20. Kerr, J.S.; Adriaanse, B.A.; Greig, N.H.; Mattson, M.P.; Cader, M.Z.; Bohr, V.A.; Fang, E.F. Mitophagy and Alzheimer’s Disease: Cellular and Molecular Mechanisms. Trends Neurosci. 2017, 40, 151–166. [Google Scholar] [CrossRef] [Green Version]
  21. Maltzman, J.S. Mitochondria-More than just ATP for CTLs. Sci. Immunol. 2021, 6, eabn0249. [Google Scholar] [CrossRef]
  22. Yuzefovych, L.V.; Musiyenko, S.I.; Wilson, G.L.; Rachek, L.I. Mitochondrial DNA damage and dysfunction, and oxidative stress are associated with endoplasmic reticulum stress, protein degradation and apoptosis in high fat diet-induced insulin resistance mice. PLoS ONE 2013, 8, e54059. [Google Scholar] [CrossRef] [PubMed]
  23. Gibson, G.E.; Park, L.C.; Sheu, K.F.; Blass, J.P.; Calingasan, N.Y. The alpha-ketoglutarate dehydrogenase complex in neurodegeneration. Neurochem. Int. 2000, 36, 97–112. [Google Scholar] [CrossRef] [PubMed]
  24. Geng, X.; Elmadhoun, O.; Peng, C.; Ji, X.; Hafeez, A.; Liu, Z.; Du, H.; Rafols, J.A.; Ding, Y. Ethanol and normobaric oxygen: Novel approach in modulating pyruvate dehydrogenase complex after severe transient and permanent ischemic stroke. Stroke 2015, 46, 492–499. [Google Scholar] [CrossRef] [Green Version]
  25. Gibson, G.E.; Blass, J.P.; Beal, M.F.; Bunik, V. The alpha-ketoglutarate-dehydrogenase complex: A mediator between mitochondria and oxidative stress in neurodegeneration. Mol. Neurobiol. 2005, 31, 43–63. [Google Scholar] [CrossRef] [PubMed]
  26. Mailloux, R.J.; Young, A.; O’Brien, M.; Gill, R.M. Simultaneous Measurement of Superoxide/Hydrogen Peroxide and NADH Production by Flavin-containing Mitochondrial Dehydrogenases. J. Vis. Exp. 2018, 24, 56975. [Google Scholar] [CrossRef]
  27. Misrani, A.; Tabassum, S.; Yang, L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 617588. [Google Scholar] [CrossRef] [PubMed]
  28. Dixit, S.; Fessel, J.P.; Harrison, F.E. Mitochondrial dysfunction in the APP/PSEN1 mouse model of Alzheimer’s disease and a novel protective role for ascorbate. Free Radic. Biol. Med. 2017, 112, 515–523. [Google Scholar] [CrossRef]
  29. Cai, Q.; Tammineni, P. Mitochondrial Aspects of Synaptic Dysfunction in Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 57, 1087–1103. [Google Scholar] [CrossRef]
  30. Barbagallo, M.; Marotta, F.; Dominguez, L.J. Oxidative stress in patients with Alzheimer’s disease: Effect of extracts of fermented papaya powder. Mediat. Inflamm. 2015, 2015, 624801. [Google Scholar] [CrossRef] [Green Version]
  31. Ji, Z.H.; Zhao, H.; Liu, C.; Yu, X.Y. In-vitro neuroprotective effect and mechanism of 2beta-hydroxy-delta-cadinol against amyloid beta-induced neuronal apoptosis. Neuroreport 2020, 31, 245–250. [Google Scholar] [CrossRef] [PubMed]
  32. Dhas, N.; Mehta, T. Cationic biopolymer functionalized nanoparticles encapsulating lutein to attenuate oxidative stress in effective treatment of Alzheimer’s disease: A non-invasive approach. Int. J. Pharm. 2020, 586, 119553. [Google Scholar] [CrossRef] [PubMed]
  33. Yadav, U.C.; Rani, V.; Deep, G.; Singh, R.K.; Palle, K. Oxidative Stress in Metabolic Disorders: Pathogenesis, Prevention, and Therapeutics. Oxidative Med. Cell. Longev. 2016, 2016, 9137629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ayton, S.; Lei, P.; Bush, A.I. Metallostasis in Alzheimer’s disease. Free Radic. Biol. Med. 2013, 62, 76–89. [Google Scholar] [CrossRef] [PubMed]
  35. Martinez-Cue, C.; Rueda, N. Signalling Pathways Implicated in Alzheimer’s Disease Neurodegeneration in Individuals with and without Down Syndrome. Int. J. Mol. Sci. 2020, 21, 6906. [Google Scholar] [CrossRef] [PubMed]
  36. Prasad, K.N. Simultaneous activation of Nrf2 and elevation of antioxidant compounds for reducing oxidative stress and chronic inflammation in human Alzheimer’s disease. Mech. Ageing Dev. 2016, 153, 41–47. [Google Scholar] [CrossRef]
  37. Arbel-Ornath, M.; Hudry, E.; Boivin, J.R.; Hashimoto, T.; Takeda, S.; Kuchibhotla, K.V.; Hou, S.; Lattarulo, C.R.; Belcher, A.M.; Shakerdge, N.; et al. Soluble oligomeric amyloid-beta induces calcium dyshomeostasis that precedes synapse loss in the living mouse brain. Mol. Neurodegener. 2017, 12, 27. [Google Scholar] [CrossRef] [Green Version]
  38. Selvakumar, G.P.; Ahmed, M.E.; Raikwar, S.P.; Thangavel, R.; Kempuraj, D.; Dubova, I.; Saeed, D.; Zahoor, H.; Premkumar, K.; Zaheer, S.; et al. CRISPR/Cas9 Editing of Glia Maturation Factor Regulates Mitochondrial Dynamics by Attenuation of the NRF2/HO-1 Dependent Ferritin Activation in Glial Cells. J. Neuroimmune Pharmacol. 2019, 14, 537–550. [Google Scholar] [CrossRef]
  39. Reich, N.; Holscher, C. The neuroprotective effects of glucagon-like peptide 1 in Alzheimer’s and Parkinson’s disease: An in-depth review. Front. Neurosci. 2022, 16, 970925. [Google Scholar] [CrossRef]
  40. Fakhoury, M. Microglia and Astrocytes in Alzheimer’s Disease: Implications for Therapy. Curr. Neuropharmacol. 2018, 16, 508–518. [Google Scholar] [CrossRef]
  41. Badshah, H.; Ali, T.; Shafiq-ur, R.; Faiz-ul, A.; Ullah, F.; Kim, T.H.; Kim, M.O. Protective Effect of Lupeol Against Lipopolysaccharide-Induced Neuroinflammation via the p38/c-Jun N-Terminal Kinase Pathway in the Adult Mouse Brain. J. Neuroimmune Pharmacol. 2016, 11, 48–60. [Google Scholar] [CrossRef] [PubMed]
  42. Lopez-Rodriguez, A.B.; Hennessy, E.; Murray, C.L.; Nazmi, A.; Delaney, H.J.; Healy, D.; Fagan, S.G.; Rooney, M.; Stewart, E.; Lewis, A.; et al. Acute systemic inflammation exacerbates neuroinflammation in Alzheimer’s disease: IL-1beta drives amplified responses in primed astrocytes and neuronal network dysfunction. Alzheimer’s Dement. 2021, 17, 1735–1755. [Google Scholar] [CrossRef] [PubMed]
  43. Tonnies, E.; Trushina, E. Oxidative Stress, Synaptic Dysfunction, and Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 57, 1105–1121. [Google Scholar] [CrossRef] [Green Version]
  44. Song, X.M.; Yu, Q.; Dong, X.; Yang, H.O.; Zeng, K.W.; Li, J.; Tu, P.F. Aldose reductase inhibitors attenuate beta-amyloid-induced TNF-alpha production in microlgia via ROS-PKC-mediated NF-kappaB and MAPK pathways. Int. Immunopharmacol. 2017, 50, 30–37. [Google Scholar] [CrossRef]
  45. Ferrer, I.; Blanco, R.; Carmona, M.; Puig, B. Phosphorylated mitogen-activated protein kinase (MAPK/ERK-P), protein kinase of 38 kDa (p38-P), stress-activated protein kinase (SAPK/JNK-P), and calcium/calmodulin-dependent kinase II (CaM kinase II) are differentially expressed in tau deposits in neurons and glial cells in tauopathies. J. Neural Transm. 2001, 108, 1397–1415. [Google Scholar] [CrossRef] [PubMed]
  46. Skibinski, G.; Hwang, V.; Ando, D.M.; Daub, A.; Lee, A.K.; Ravisankar, A.; Modan, S.; Finucane, M.M.; Shaby, B.A.; Finkbeiner, S. Nrf2 mitigates LRRK2-and α-synuclein–induced neurodegeneration by modulating proteostasis. Proc. Natl. Acad. Sci. USA 2017, 114, 1165–1170. [Google Scholar] [CrossRef]
  47. Teixeira, J.P.; de Castro, A.A.; Soares, F.V.; da Cunha, E.F.F.; Ramalho, T.C. Future Therapeutic Perspectives into the Alzheimer’s Disease Targeting the Oxidative Stress Hypothesis. Molecules 2019, 24, 4410. [Google Scholar] [CrossRef] [Green Version]
  48. McMahon, M.; Lamont, D.J.; Beattie, K.A.; Hayes, J.D. Keap1 perceives stress via three sensors for the endogenous signaling molecules nitric oxide, zinc, and alkenals. Proc. Natl. Acad. Sci. USA 2010, 107, 18838–18843. [Google Scholar] [CrossRef]
  49. Kobayashi, M.; Li, L.; Iwamoto, N.; Nakajima-Takagi, Y.; Kaneko, H.; Nakayama, Y.; Eguchi, M.; Wada, Y.; Kumagai, Y.; Yamamoto, M. The antioxidant defense system Keap1-Nrf2 comprises a multiple sensing mechanism for responding to a wide range of chemical compounds. Mol. Cell. Biol. 2009, 29, 493–502. [Google Scholar] [CrossRef] [Green Version]
  50. Heras-Sandoval, D.; Avila-Muñoz, E.; Arias, C. The phosphatidylinositol 3-kinase/mTor pathway as a therapeutic target for brain aging and neurodegeneration. Pharmaceuticals 2011, 4, 1070–1087. [Google Scholar] [CrossRef] [Green Version]
  51. Xu, F.; Na, L.; Li, Y.; Chen, L. Roles of the PI3K/AKT/mTOR signalling pathways in neurodegenerative diseases and tumours. Cell Biosci. 2020, 10, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Morris, M.C.; Tangney, C.C.; Wang, Y.; Sacks, F.M.; Barnes, L.L.; Bennett, D.A.; Aggarwal, N.T. MIND diet slows cognitive decline with aging. Alzheimer’s Dement. 2015, 11, 1015–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Matsuyama, K.; Yamamoto, Y.; Sora, I. Effect of Feru-guard 100M on amyloid-beta deposition in individuals with mild cognitive impairment. Psychogeriatrics 2020, 20, 726–736. [Google Scholar] [CrossRef] [PubMed]
  54. Kotyczka, C.; Boettler, U.; Lang, R.; Stiebitz, H.; Bytof, G.; Lantz, I.; Hofmann, T.; Marko, D.; Somoza, V. Dark roast coffee is more effective than light roast coffee in reducing body weight, and in restoring red blood cell vitamin E and glutathione concentrations in healthy volunteers. Mol. Nutr. Food Res. 2011, 55, 1582–1586. [Google Scholar] [CrossRef] [PubMed]
  55. Ratheesh, G.; Tian, L.; Venugopal, J.R.; Ezhilarasu, H.; Sadiq, A.; Fan, T.-P.; Ramakrishna, S. Role of medicinal plants in neurodegenerative diseases. Biomanuf. Rev. 2017, 2, 1–16. [Google Scholar] [CrossRef] [Green Version]
  56. Mancuso, C.; Bates, T.E.; Butterfield, D.A.; Calafato, S.; Cornelius, C.; Lorenzo, A.D.; Dinkova Kostova, A.T.; Calabrese, V. Natural antioxidants in Alzheimer’s disease. Expert Opin. Investig. Drugs 2007, 16, 1921–1931. [Google Scholar] [CrossRef]
  57. Frank, B.; Gupta, S. A review of antioxidants and Alzheimer’s disease. Ann. Clin. Psychiatry 2005, 17, 269–286. [Google Scholar] [CrossRef]
  58. Dash, R.; Jahan, I.; Ali, M.C.; Mitra, S.; Munni, Y.A.; Timalsina, B.; Hannan, M.A.; Moon, I.S. Potential roles of natural products in the targeting of proteinopathic neurodegenerative diseases. Neurochem. Int. 2021, 145, 105011. [Google Scholar] [CrossRef]
  59. Kim, H.; Lee, D.G. Lupeol-induced nitric oxide elicits apoptosis-like death within Escherichia coli in a DNA fragmentation-independent manner. Biochem. J. 2021, 478, 855–869. [Google Scholar] [CrossRef]
  60. Hashmi, W.J.; Ismail, H.; Mehmood, F.; Mirza, B. Neuroprotective, antidiabetic and antioxidant effect of Hedera nepalensis and Lupeol against STZ + AlCl3 induced rats model. Daru 2018, 26, 179–190. [Google Scholar] [CrossRef]
  61. Li, D.; Guo, Y.Y.; Cen, X.F.; Qiu, H.L.; Chen, S.; Zeng, X.F.; Zeng, Q.; Xu, M.; Tang, Q.Z. Lupeol protects against cardiac hypertrophy via TLR4-PI3K-Akt-NF-kappaB pathways. Acta Pharmacol. Sin. 2022, 43, 1989–2002. [Google Scholar] [CrossRef] [PubMed]
  62. Huang, S.; Mo, C.; Zeng, T.; Lai, Y.; Zhou, C.; Xie, S.; Chen, L.; Wang, Y.; Chen, Y.; Huang, S.; et al. Lupeol ameliorates LPS/D-GalN induced acute hepatic damage by suppressing inflammation and oxidative stress through TGFbeta1-Nrf2 signal pathway. Aging 2021, 13, 6592–6605. [Google Scholar] [CrossRef] [PubMed]
  63. Srivastava, S.; Sonkar, R.; Mishra, S.K.; Tiwari, A.; Balaramnavar, V.M.; Mir, S.; Bhatia, G.; Saxena, A.K.; Lakshmi, V. Antidyslipidemic and antioxidant effects of novel Lupeol-derived chalcones. Lipids 2013, 48, 1017–1027. [Google Scholar] [CrossRef]
  64. Santiago, L.A.; Mayor, A.B. Lupeol: An antioxidant triterpene in Ficus pseudopalma Blanco (Moraceae). Asian Pac. J. Trop. Biomed. 2014, 4, 109–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ramu, R.; S. Shirahatti, P.; Zameer, F.; Lakkappa Dhananjaya, B.; MN, N.P. Assessment of In Vivo Antidiabetic Properties of Umbelliferone and Lupeol Constituents of Banana (Musa sp. var. Nanjangud Rasa Bale) Flower in Hyperglycaemic Rodent Model. PLoS ONE 2016, 11, e0151135. [Google Scholar] [CrossRef] [Green Version]
  66. Sunitha, S.; Nagaraj, M.; Varalakshmi, P. Hepatoprotective effect of Lupeol and Lupeol linoleate on tissue antioxidant defence system in cadmium-induced hepatotoxicity in rats. Fitoterapia 2001, 72, 516–523. [Google Scholar] [CrossRef]
  67. Sudharsan, P.T.; Mythili, Y.; Selvakumar, E.; Varalakshmi, P. Cardioprotective effect of pentacyclic triterpene, Lupeol and its ester on cyclophosphamide-induced oxidative stress. Hum. Exp. Toxicol. 2005, 24, 313–318. [Google Scholar] [CrossRef]
  68. Sudhahar, V.; Kumar, S.A.; Varalakshmi, P. Role of Lupeol and Lupeol linoleate on lipemic-oxidative stress in experimental hypercholesterolemia. Life Sci. 2006, 78, 1329–1335. [Google Scholar] [CrossRef]
  69. Kaundal, M.; Akhtar, M.; Deshmukh, R. Lupeol Isolated from Betula alnoides Ameliorates Amyloid Beta Induced Neuronal Damage via Targeting Various Pathological Events and Alteration in Neurotransmitter Levels in Rat’s Brain. J. Neurol. Neurosci. 2017, 8, 195. [Google Scholar] [CrossRef] [Green Version]
  70. Zhang, S.; Peng, B.; Chen, Z.; Yu, J.; Deng, G.; Bao, Y.; Ma, C.; Du, F.; Sheu, W.C.; Kimberly, W.T.; et al. Brain-targeting, acid-responsive antioxidant nanoparticles for stroke treatment and drug delivery. Bioact. Mater. 2022, 16, 57–65. [Google Scholar] [CrossRef]
  71. Panja, S.K.; Patra, S.; Bag, B.G. Self-assembly of the monohydroxy triterpenoid Lupeol yielding nano-fibers, sheets and gel: Environmental and drug delivery applications. RSC Adv. 2021, 11, 33500–33510. [Google Scholar] [CrossRef] [PubMed]
  72. Tchimene, M.K.; Nwaehujor, C.O.; Ezenwali, M.; Okoli, C.C.; Iwu, M.M. Free radical scavenging activity of Lupeol isolated from the methanol leaf extract of Crateva adansonii Oliv. (Capparidaceae). Int. J. Pharmacogn. Phytochem. Res. 2016, 8, 419–426. [Google Scholar]
  73. Ahmad, R.; Khan, A.; Rehman, I.U.; Lee, H.J.; Khan, I.; Kim, M.O. Lupeol Treatment Attenuates Activation of Glial Cells and Oxidative-Stress-Mediated Neuropathology in Mouse Model of Traumatic Brain Injury. Int. J. Mol. Sci. 2022, 23, 6086. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, Z.; Xu, C.; Hao, J.; Zhang, M.; Wang, Z.; Yin, T.; Lin, K.; Liu, W.; Jiang, Q.; Li, Z.; et al. Beneficial consequences of Lupeol on middle cerebral artery-induced cerebral ischemia in the rat involves Nrf2 and P38 MAPK modulation. Metab. Brain Dis. 2020, 35, 841–848. [Google Scholar] [CrossRef]
  75. Lee, C.; Lee, J.W.; Seo, J.Y.; Hwang, S.W.; Im, J.P.; Kim, J.S. Lupeol inhibits LPS-induced NF-kappa B signaling in intestinal epithelial cells and macrophages, and attenuates acute and chronic murine colitis. Life Sci. 2016, 146, 100–108. [Google Scholar] [CrossRef] [PubMed]
  76. Malik, A.; Jamil, U.; Butt, T.T.; Waquar, S.; Gan, S.H.; Shafique, H.; Jafar, T.H. In silico and in vitro studies of Lupeol and iso-orientin as potential antidiabetic agents in a rat model. Drug Des. Devel Ther. 2019, 13, 1501–1513. [Google Scholar] [CrossRef] [Green Version]
  77. Nagaraj, M.; Sunitha, S.; Varalakshmi, P. Effect of Lupeol, a pentacyclic triterpene, on the lipid peroxidation and antioxidant status in rat kidney after chronic cadmium exposure. J. Appl. Toxicol. 2000, 20, 413–417. [Google Scholar] [CrossRef]
  78. Prasad, S.; Kalra, N.; Singh, M.; Shukla, Y. Protective effects of Lupeol and mango extract against androgen induced oxidative stress in Swiss albino mice. Asian J. Androl. 2008, 10, 313–318. [Google Scholar] [CrossRef]
  79. Zhong, J.; Shi, G. Editorial: Regulation of Inflammation in Chronic Disease. Front. Immunol. 2019, 10, 737. [Google Scholar] [CrossRef] [Green Version]
  80. Das, A.K.; Hossain, U.; Ghosh, S.; Biswas, S.; Mandal, M.; Mandal, B.; Brahmachari, G.; Bagchi, A.; Sil, P.C. Amelioration of oxidative stress mediated inflammation and apoptosis in pancreatic islets by Lupeol in STZ-induced hyperglycaemic mice. Life Sci. 2022, 305, 120769. [Google Scholar] [CrossRef]
  81. Pereira Beserra, F.; Sergio Gushiken, L.F.; Vieira, A.J.; Augusto Bergamo, D.; Luisa Bergamo, P.; Oliveira de Souza, M.; Alberto Hussni, C.; Kiomi Takahira, R.; Henrique Nobrega, R.; Monteiro Martinez, E.R.; et al. From Inflammation to Cutaneous Repair: Topical Application of Lupeol Improves Skin Wound Healing in Rats by Modulating the Cytokine Levels, NF-kappaB, Ki-67, Growth Factor Expression, and Distribution of Collagen Fibers. Int. J. Mol. Sci. 2020, 21, 4952. [Google Scholar] [CrossRef]
  82. Vasconcelos, J.F.; Teixeira, M.M.; Barbosa-Filho, J.M.; Lucio, A.S.; Almeida, J.R.; de Queiroz, L.P.; Ribeiro-Dos-Santos, R.; Soares, M.B. The triterpenoid lupeol attenuates allergic airway inflammation in a murine model. Int. Immunopharmacol. 2008, 8, 1216–1221. [Google Scholar] [CrossRef] [PubMed]
  83. Ma, X.; Hao, C.; Zhang, Z.; Jiang, H.; Zhang, W.; Huang, J.; Chen, X.; Yang, W. Shenjinhuoxue Mixture Attenuates Inflammation, Pain, and Cartilage Degeneration by Inhibiting TLR-4 and NF-kappaB Activation in Rats with Osteoarthritis: A Synergistic Combination of Multitarget Active Phytochemicals. Oxidative Med. Cell. Longev. 2021, 2021, 4190098. [Google Scholar] [CrossRef] [PubMed]
  84. Toledo, C.R.; Paiva, M.R.B.; Castro, B.F.M.; Pereira, V.V.; Cenachi, S.P.F.; Vasconcelos-Santos, D.V.; Fialho, S.L.; Silva-Cunha, A. Intravitreal lupeol: A new potential therapeutic strategy for noninfectious uveitis. Biomed. Pharmacother. 2021, 143, 112145. [Google Scholar] [CrossRef]
  85. Kim, M.J.; Bae, G.S.; Choi, S.B.; Jo, I.J.; Kim, D.G.; Shin, J.Y.; Lee, S.K.; Kim, M.J.; Song, H.J.; Park, S.J. Lupeol Protects Against Cerulein-Induced Acute Pancreatitis in Mice. Phytother. Res. 2015, 29, 1634–1639. [Google Scholar] [CrossRef] [PubMed]
  86. Xu, M.; Li, X.; Song, L.; Tao, C.; Fang, J.; Tao, L. Lupeol alleviates coxsackievirus B3-induced viral myocarditis in mice via downregulating toll-like receptor 4. J. Int. Med. Res. 2020, 48, 300060520910908. [Google Scholar] [CrossRef] [Green Version]
  87. Zhu, Y.; Li, X.; Chen, J.; Chen, T.; Shi, Z.; Lei, M.; Zhang, Y.; Bai, P.; Li, Y.; Fei, X. The pentacyclic triterpene Lupeol switches M1 macrophages to M2 and ameliorates experimental inflammatory bowel disease. Int. Immunopharmacol. 2016, 30, 74–84. [Google Scholar] [CrossRef]
  88. Rathinavel, T.; Ammashi, S.; Shanmugam, G. Analgesic and anti-inflammatory potential of Lupeol isolated from Indian traditional medicinal plant Crateva adansonii screened through in vivo and in silico approaches. J. Genet. Eng. Biotechnol. 2021, 19, 62. [Google Scholar] [CrossRef]
  89. Milani, G.B.; Camponogara, C.; Piana, M.; Silva, C.R.; Oliveira, S.M. Cariniana domestica fruit peels present topical anti-inflammatory efficacy in a mouse model of skin inflammation. Naunyn Schmiedeberg’s Arch. Pharmacol. 2019, 392, 513–528. [Google Scholar] [CrossRef]
  90. Yoo, J.-H.; Yang, K.-S. Constituents of Pyrus pyrifolia with inhibitory activity on the NO production and the expression of iNOS and COX-2 in macrophages and microglia. Nat. Prod. Sci. 2012, 18, 183–189. [Google Scholar]
  91. Oliveira-Junior, M.S.; Pereira, E.P.; de Amorim, V.C.M.; Reis, L.T.C.; do Nascimento, R.P.; da Silva, V.D.A.; Costa, S.L. Lupeol inhibits LPS-induced neuroinflammation in cerebellar cultures and induces neuroprotection associated to the modulation of astrocyte response and expression of neurotrophic and inflammatory factors. Int. Immunopharmacol. 2019, 70, 302–312. [Google Scholar] [CrossRef]
  92. Anjaneyulu, J.; R, V.; Godbole, A. Differential effect of Ayurvedic nootropics on C. elegans models of Parkinson’s disease. J. Ayurveda Integr. Med. 2020, 11, 440–447. [Google Scholar] [CrossRef]
  93. Bello, O.M.; Ogbesejana, A.B.; Adetunji, C.O.; Oguntoye, S.O. Flavonoids Isolated from Vitex grandifolia, an Underutilized Vegetable, Exert Monoamine A & B Inhibitory and Anti-inflammatory Effects and Their Structure-activity Relationship. Turk. J. Pharm. Sci. 2019, 16, 437–443. [Google Scholar] [CrossRef]
  94. Jin, S.E.; Son, Y.K.; Min, B.S.; Jung, H.A.; Choi, J.S. Anti-inflammatory and antioxidant activities of constituents isolated from Pueraria lobata roots. Arch. Pharm. Res. 2012, 35, 823–837. [Google Scholar] [CrossRef]
  95. Wang, Z.; Han, Y.; Tian, S.; Bao, J.; Wang, Y.; Jiao, J. Lupeol Alleviates Cerebral Ischemia-Reperfusion Injury in Correlation with Modulation of PI3K/Akt Pathway. Neuropsychiatr. Dis. Treat. 2020, 16, 1381–1390. [Google Scholar] [CrossRef]
  96. Hosen, S.M.Z.; Rubayed, M.; Dash, R.; Junaid, M.; Mitra, S.; Alam, M.S.; Dey, R. Prospecting and Structural Insight into the Binding of Novel Plant-Derived Molecules of Leea indica as Inhibitors of BACE1. Curr. Pharm. Des. 2018, 24, 3972–3979. [Google Scholar] [CrossRef] [Green Version]
  97. Koirala, P.; Seong, S.H.; Jung, H.A.; Choi, J.S. Comparative molecular docking studies of Lupeol and lupenone isolated from Pueraria lobata that inhibits BACE1: Probable remedies for Alzheimer’s disease. Asian Pac. J. Trop. Med. 2017, 10, 1117–1122. [Google Scholar] [CrossRef]
  98. Brimson, J.M.; Brimson, S.J.; Brimson, C.A.; Rakkhitawatthana, V.; Tencomnao, T. Rhinacanthus nasutus extracts prevent glutamate and amyloid-beta neurotoxicity in HT-22 mouse hippocampal cells: Possible active compounds include Lupeol, stigmasterol and beta-sitosterol. Int. J. Mol. Sci. 2012, 13, 5074–5097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Hrabinova, M.; Pejchal, J.; Kucera, T.; Jun, D.; Schmidt, M.; Soukup, O. Is It the Twilight of BACE1 Inhibitors? Curr. Neuropharmacol. 2021, 19, 61–77. [Google Scholar] [CrossRef] [PubMed]
  100. Deshmukh, R.; Sharma, K. P1-082: Neuroprotective potential of Lupeol against aluminium chloride–induced learning and memory deficit in rats: Possible role of hippocampal neurochemistry and neuroinflammatory mechanisms. Alzheimer’s Dement. 2018, 14, P302. [Google Scholar] [CrossRef]
  101. Taylor, H.A.; Przemylska, L.; Clavane, E.M.; Meakin, P.J. BACE1: More than just a beta-secretase. Obes. Rev. 2022, 23, e13430. [Google Scholar] [CrossRef]
  102. Koelsch, G. BACE1 Function and Inhibition: Implications of Intervention in the Amyloid Pathway of Alzheimer’s Disease Pathology. Molecules 2017, 22, 1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Manigandan, V.; Ramanathan, T. In silico docking of mangrove derived ligands against Alzheimer’s receptor proteins. Curr. Res. Neurosci. 2014, 4, 18–24. [Google Scholar] [CrossRef]
  104. Ali, M.; Muhammad, S.; Shah, M.R.; Khan, A.; Rashid, U.; Farooq, U.; Ullah, F.; Sadiq, A.; Ayaz, M.; Ali, M.; et al. Neurologically Potent Molecules from Crataegus oxyacantha; Isolation, Anticholinesterase Inhibition, and Molecular Docking. Front. Pharmacol. 2017, 8, 327. [Google Scholar] [CrossRef]
  105. Cohen, A. Pharmacokinetic and pharmacodynamic data to be derived from early-phase drug development: Designing informative human pharmacology studies. Clin. Pharmacokinet. 2008, 47, 373–381. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, K.; Zhang, X.; Xie, L.; Deng, M.; Chen, H.; Song, J.; Long, J.; Li, X.; Luo, J. Lupeol and its derivatives as anticancer and anti-inflammatory agents: Molecular mechanisms and therapeutic efficacy. Pharmacol. Res. 2021, 164, 105373. [Google Scholar] [CrossRef] [PubMed]
  107. Ru, J.; Li, P.; Wang, J.; Zhou, W.; Li, B.; Huang, C.; Li, P.; Guo, Z.; Tao, W.; Yang, Y.; et al. TCMSP: A database of systems pharmacology for drug discovery from herbal medicines. J. Cheminform. 2014, 6, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Priyanka, K.; Kosuru, R.; Sharma, R.P.; Sahu, P.L.; Singh, S. Assessment of pharmacokinetic parameters of Lupeol in Ficus religiosa L. extract after oral administration of suspension and solid lipid nanoparticles to Wistar rats. J. Drug Deliv. Sci. Technol. 2017, 41, 58–67. [Google Scholar] [CrossRef]
  109. Chairez-Ramirez, M.H.; Gallegos-Infante, J.A.; Moreno-Jimenez, M.R.; Gonzalez-Laredo, R.F.; Rocha-Guzman, N.E. Absorption and distribution of lupeol in CD-1 mice evaluated by UPLC-APCI+-MS/MS. Biomed. Chromatogr. 2019, 33, e4432. [Google Scholar] [CrossRef]
  110. Khatal, L.; More, H. Development and validation of a liquid chromatography-tandem mass spectrometry method for quantification of Lupeol in plasma and its application to pharmacokinetic study in rats. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2019, 1121, 58–65. [Google Scholar] [CrossRef]
  111. Zhang, J.; Liang, H.; Yao, H.; Qiu, Z.; Chen, X.; Hu, X.; Hu, J.; Zheng, G. The preparation, characterization of Lupeol PEGylated liposome and its functional evaluation in vitro as well as pharmacokinetics in rats. Drug Dev. Ind. Pharm. 2019, 45, 1052–1060. [Google Scholar] [CrossRef]
  112. Papi Reddy, K.; Singh, A.B.; Puri, A.; Srivastava, A.K.; Narender, T. Synthesis of novel triterpenoid (Lupeol) derivatives and their in vivo antihyperglycemic and antidyslipidemic activity. Bioorg. Med. Chem. Lett. 2009, 19, 4463–4466. [Google Scholar] [CrossRef] [PubMed]
  113. Bociort, F.; Crisan, C.N.; Dragoi, R.; Heghes, A.; Szuhanek, C.; Radulescu, M.; Berceanu-Vaduva, D.; Tischer, A.; Motoc, A. Green and Synthetic Metallic Nanoparticles-Obtaining, Characterization and Biological Evaluation in Association with Lupeol. Rev. De. Chim. 2020, 71, 299–304. [Google Scholar] [CrossRef]
  114. Patocka, J. Biologically active pentacyclic triterpenes and their current medicine signification. J. Appl. Biomed. 2003, 1, 7–12. [Google Scholar] [CrossRef] [Green Version]
  115. Geetha, T.; Varalakshmi, P. Anti-inflammatory activity of lupeol and lupeol linoleate in adjuvant-induced arthritis. Fitoterapia 1998, 69, 13–19. [Google Scholar]
  116. Yang, X.; Feng, Y.; Liu, Y.; Ye, X.; Ji, X.; Sun, L.; Gao, F.; Zhang, Q.; Li, Y.; Zhu, B.; et al. Fuzheng Jiedu Xiaoji formulation inhibits hepatocellular carcinoma progression in patients by targeting the AKT/CyclinD1/p21/p27 pathway. Phytomedicine 2021, 87, 153575. [Google Scholar] [CrossRef]
  117. Huyke, C.; Laszczyk, M.; Scheffler, A.; Ernst, R.; Schempp, C.M. [Treatment of actinic keratoses with birch bark extract: A pilot study]. J. Dtsch. Dermatol. Ges. 2006, 4, 132–136. [Google Scholar] [CrossRef]
  118. Schloss, J.; Ryan, K.; Reid, R.; Steel, A. A randomised, double-blind, placebo-controlled clinical trial assessing the efficacy of bedtime buddy(R) for the treatment of nocturnal enuresis in children. BMC Pediatr. 2019, 19, 421. [Google Scholar] [CrossRef]
  119. Nogueira, C.W.; Barbosa, N.V.; Rocha, J.B.T. Toxicology and pharmacology of synthetic organoselenium compounds: An update. Arch. Toxicol. 2021, 95, 1179–1226. [Google Scholar] [CrossRef]
  120. Sanchez-Martinez, J.D.; Bueno, M.; Alvarez-Rivera, G.; Tudela, J.; Ibanez, E.; Cifuentes, A. In vitro neuroprotective potential of terpenes from industrial orange juice by-products. Food Funct. 2021, 12, 302–314. [Google Scholar] [CrossRef]
Figure 1. Neuroprotective effects of Lupeol against oxidative stress in AD pathogenesis. The activations of KGDHC and PDHC due to the excessive burden of free radicals are inducing oxidative stress, which is leading to the downregulation of the antioxidant defense system. Also, the overburden of AChE/BuChase and abnormal deposition of amyloid beta(Aβ) protein, due to the cleavage of amyloid precursor protein (APP), are inducing an elevated ROS level, where the inflammatory cascade is initiated due to activation of inflammatory cytokines that are proceeded by neuronal inflammation, initiating AD pathogenesis. Lupeol treatment annotated at different doses at different intervals of time was able to improve memory and cognition by inhibiting the over burden of oxidative stress and neuroinflammation.
Figure 1. Neuroprotective effects of Lupeol against oxidative stress in AD pathogenesis. The activations of KGDHC and PDHC due to the excessive burden of free radicals are inducing oxidative stress, which is leading to the downregulation of the antioxidant defense system. Also, the overburden of AChE/BuChase and abnormal deposition of amyloid beta(Aβ) protein, due to the cleavage of amyloid precursor protein (APP), are inducing an elevated ROS level, where the inflammatory cascade is initiated due to activation of inflammatory cytokines that are proceeded by neuronal inflammation, initiating AD pathogenesis. Lupeol treatment annotated at different doses at different intervals of time was able to improve memory and cognition by inhibiting the over burden of oxidative stress and neuroinflammation.
Nutrients 15 03059 g001
Table 3. Effects of Lupeol on the treatment of Alzheimer’s disease.
Table 3. Effects of Lupeol on the treatment of Alzheimer’s disease.
SourceMechanismModelReference
Fruits, Vegetables, and
Herbs
↓Aβ, BACE1, GFAP, Iba-1
↓p-NFkB, TNF-α, NOS2
↑Nrf2, HO-1
In vivo Aβ-induced AD mouse model[15]
Pueraria Lobata Roots↓Aβ, BACE1Comparative molecular
Docking in AD studies
[97]
Rhinacanthus Nasutus↓Glutamate and Aβ
↓ ROS
In vitro glutamate and Aβ-induced AD Mouse hippocampal HT-22 cell lines[98]
Betula Alnoides↓Aβ, TNFα, IL-1β, IL-6In vivo Aβ-induced AD Male Wistar Rats[69]
Hedera Nepalensis↓SOD, CAT and GSHIn vivo STZ-+ALCL3-induced diabetes-mediated AD[60]
Leea Indica↓BACE1Molecular docking and molecular dynamic based approaches in AD model[96]
Crataegus Oxyacantha↓AChE, BuChaseUsing in vitro experimental results and the docking score in AChE-induced AD model[104]
Desmodium
Triquetrum
↓iNOS
↓IL-1b, IL-6, TNF-α
↓AChE
In vivo aluminum chloride (AlCl3)-induced neurotoxicity in AD Wistar Rats[100]
Mangroves↓Aβ, AChEMolecular docking AD model[103]
Abbreviations: Amyloid Beta (Aβ); Beta-secretase 1 (BACE1); Glial fibrillary acidic protein (GFAP); Ionized calcium-binding adapter molecule 1 (IBA1); Phosphorylated Nuclear factor-κB (p-NF-κB); Tumor necrosis factor alpha (TNF-α); Nitric Oxide Synthase 2 (NOS2); Nuclear factor erythroid 2–related factor 2 (Nrf2); Heme oxygenase 1 (HO-1); Reactive oxygen species (ROS); Interleukin-1 beta (IL-1β); Interleukin 6 (IL-6); Superoxide dismutase (SOD); Chloramphenicol acetyltransferase (CAT); Glutathione (GSH); Acetylcholinesterase (AChE); Butyrylcholinesterase (BuChase); Inducible nitric oxide synthase (iNOS). The symbols ↓ and ↑ are representing the expression level of proteins to be decreased and increased respectively.
Table 4. Evaluated pharmacokinetic parameters of Lupeol in different models.
Table 4. Evaluated pharmacokinetic parameters of Lupeol in different models.
Pharmacokinetics ParametersAnalytical Method/Animal ModelReference
AUC: 9.2-folds ↑Cmax: 3.9-folds ↑T1/2: 15.3 ± 1.3 in SLN
SLN enhanced AUC and Cmax and prolonged T1/2
Solid–lipid nanoparticle (SLNs) loaded with Lupeol in Rats[108]
Tmax: 6.444 ± 0.851 h;
Cmax: 8.071 ± 2.930 μg/mL.
Post-administration times:
stomach, 137.25 ± 19.94 ng/mg and small intestine, 99.00 ± 12.99 ng/mg.
Excretion: fecal; T1/2e: 12 h post-administration (163.28 ± 9.83 μg/mg). F: 0.645 ± 0.0581
UPLC-APCI+-MS/MS in CD-1 strain of Mice[109]
1 mg/kg dose of Lupeol i.v. administration.
AUC0-t (h × ng/mL); 21,584.53
Cmax (ng/mL); 12,485.69
Kel (h − 1); 21,866.18
Tmax (h); 0.14, 0.08
T1/2 (h); 4.95
30 mg/kg dose of Lupeol orally administration.
AUC0-t (h × ng/mL); 2190.49
Cmax (ng/mL); 133.33,
Kel (h − 1); 2727.52
Tmax (h); 0.08, 4.67
T1/2 (h); 8.66
LC–MS/MS Wistar Rat plasma[110]
AUC: 3.2 times higher after IV. MRT: 2.5× and t1/2: 4.1×Lupeol-loaded PEGylated liposomes in Rat Model[111]
Abbreviations: AUC (area under the plasma concentration–time curve); Cmax (plasma concentration); SLN (solid liquid nanoparticle); Tmax (maximum time); Kel (apparent elimination rate constant). ↑—increase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Park, J.S.; Rehman, I.U.; Choe, K.; Ahmad, R.; Lee, H.J.; Kim, M.O. A Triterpenoid Lupeol as an Antioxidant and Anti-Neuroinflammatory Agent: Impacts on Oxidative Stress in Alzheimer’s Disease. Nutrients 2023, 15, 3059. https://doi.org/10.3390/nu15133059

AMA Style

Park JS, Rehman IU, Choe K, Ahmad R, Lee HJ, Kim MO. A Triterpenoid Lupeol as an Antioxidant and Anti-Neuroinflammatory Agent: Impacts on Oxidative Stress in Alzheimer’s Disease. Nutrients. 2023; 15(13):3059. https://doi.org/10.3390/nu15133059

Chicago/Turabian Style

Park, Jun Sung, Inayat Ur Rehman, Kyonghwan Choe, Riaz Ahmad, Hyeon Jin Lee, and Myeong Ok Kim. 2023. "A Triterpenoid Lupeol as an Antioxidant and Anti-Neuroinflammatory Agent: Impacts on Oxidative Stress in Alzheimer’s Disease" Nutrients 15, no. 13: 3059. https://doi.org/10.3390/nu15133059

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