Next Article in Journal
Red Upconverter Nanocrystals Functionalized with Verteporfin for Photodynamic Therapy Triggered by Upconversion
Next Article in Special Issue
Neuroprotective Action of Coumarin Derivatives through Activation of TRKB-CREB-BDNF Pathway and Reduction of Caspase Activity in Neuronal Cells Expressing Pro-Aggregated Tau Protein
Previous Article in Journal
Immune Responses to Multi-Frequencies of 1.5 GHz and 4.3 GHz Microwave Exposure in Rats: Transcriptomic and Proteomic Analysis
Previous Article in Special Issue
Brain Metabolic Alterations in Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Allicin and Aged Garlic Extract in Alzheimer’s Disease

1
Department of Chemical, Pharmaceutical and Agricultural Sciences—DOCPAS, University of Ferrara, 44121 Ferrara, Italy
2
Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 6950; https://doi.org/10.3390/ijms23136950
Submission received: 31 March 2022 / Revised: 6 May 2022 / Accepted: 21 June 2022 / Published: 22 June 2022
(This article belongs to the Special Issue Unraveling the Molecular Pathways of Alzheimer's Disease)

Abstract

:
Garlic, Allium sativum, has long been utilized for a number of medicinal purposes around the world, and its medical benefits have been well documented. The health benefits of garlic likely arise from a wide variety of components, possibly working synergistically. Garlic and garlic extracts, especially aged garlic extracts (AGEs), are rich in bioactive compounds, with potent anti-inflammatory, antioxidant and neuroprotective activities. In light of these effects, garlic and its components have been examined in experimental models of Alzheimer’s disease (AD), the most common form of dementia without therapy, and a growing health concern in aging societies. With the aim of offering an updated overview, this paper reviews the chemical composition, metabolism and bioavailability of garlic bioactive compounds. In addition, it provides an overview of signaling mechanisms triggered by garlic derivatives, with a focus on allicin and AGE, to improve learning and memory.

1. Introduction

Botanically, garlic is known as Allium sativum L. (family Liliaceae). The exact origin of the name is unknown, but a relation to the Latin word Olere meaning “to smell” is often pointed out. Garlic grows to approximately 30–90 cm in height in well-fertilized, sandy, and loamy soil during spring and summer [1]. For centuries, common garlic cloves have been widely used as food, as well as seasoning for food [2]. The potency of garlic (Allium sativum) has been acknowledged for approximately 5000 years. In ancient times, garlic was frequently used as a remedy for intestinal disorders, flatulence, worms, respiratory infections, skin diseases, wounds, symptoms of aging and many other ailments, and abundant literature supports the health-promoting effects of garlic and its by-products, which are associated with the bioactive compounds present in their matrix [3,4,5]. It has been used to reduce triglycerides and low-density cholesterol in the human body [6,7], to lower blood pressure [8], for antithrombotic activity [9], and to increase blood insulin levels [10,11]. In recent years, garlic organosulfur derivatives and garlic supplements have been shown to negatively affect the growth of tumor cells and the risk of cancer [12,13,14,15,16,17,18,19]. Antimicrobial, antiviral, and fungitoxicity activities against different pathogens have also been demonstrated [20,21,22,23,24].
Interestingly, the therapeutic potential of garlic extract in treating Alzheimer’s disease (AD) has been examined in different studies [25]. As a result, the goal of this review was to examine recent advances in the antioxidant and neuroprotective properties of garlic extracts and their main components, focusing on allicin and aged garlic extracts (AGEs), with the goal of reporting what has been learned thus far and identifying a potential role for these agents in the treatment of people suffering from cognitive diseases, such as AD.

2. Garlic Bioactive Compounds and Chemistry

2.1. Garlic Proximate Composition

A garlic bulb contains approximately 65–68% water; 28% carbohydrates; 2% protein; 1.2% free amino acids; 1.5% fiber, fatty acids, minerals (high levels of potassium, phosphorus, zinc, sulfur, moderate levels of selenium, calcium, magnesium, manganese and iron, and low levels of sodium), phenolic compounds (gallic acid, rutin, quercetin, ferulic acid, p-coumaric acid, naringenin, apigenin, isorhamnetin and luteolin), vitamin A, C and B-complex; and 2.3% organosulfur compounds (Figure 1) [26].

2.2. Allicin and Organo-Sulfur-Containing Compounds (OSCs)

The numerous biological properties of garlic are mainly attributed to the high contents of bioactive compounds (i.e., organosulfur compounds, phenolic compounds and fructans) [4]. The most significant components, medicinally, are the OSCs (approximately 3–35 mg/g fresh garlic) [27]. OSCs are generally classified into two groups: oil- and water-soluble OSCs (Figure 2). Fresh garlic cloves contain mainly alliin (S-allyl L-cysteine sulfoxide), followed by methiin (S-methylcysteine sulfoxide) and isoalliin, which are formed from the γ -glutamyl cysteine. When garlic is crushed or chewed or cut, alliinase is released, and the conversion of alliin into allicin (allyl 2-propenethiosulfinate) is performed [28]. Therefore, allicin is not found in intact cloves of garlic; both alliin and the enzyme are found in different parts of the bulb or clove [29,30]. Alliin and other cysteine sulfoxides are found in the mesophyll cell, whereas alliinase is localized to a few vascular bundle sheath cells around the veins or phloem. This enzyme is approximately 10 times more abundant in the cloves than in the leaves and represents at least 10% of the total protein in the cloves [31]. The process of allicin production is associated with the defense mechanisms of the plant. Alliinase and alliin form an enzyme-substrate complex in the presence of water, at an optimum temperature of 33 °C and pH of 6.5. Indeed, the enzyme is sensitive to acids, suggesting the employment of enteric-coated formulations of garlic supplements [11,31,32]. Allicin (the most abundant thiosulfinate formed via allinase reactions, approximately 70%) is poorly soluble in water and is responsible for a pungent and unpleasant flavor, but it is very unstable and easily transformed into a high number of products, classified as oil-soluble compounds, mainly including dithiins (formed by the dimerization of thioacrolein created via allicin β-elimination), followed by ajoene, allyl methyl trisulfide (AMTS), diallyl sulfide (DAS), diallyl disulfide (DADS), and diallyl trisulfide (DATS). This breakdown occurs within hours at room temperature and within minutes during cooking [33,34,35,36]. When garlic is extracted in aqueous solvent, γ-glutamyl -S-alk(en)yl-L-cysteines are converted into S-allylcysteine (SAC), S-allylmercaptocysteine (SAMC), metabolites allyl mercaptan (AM) and allyl methyl sulfide (AMS), which are water-soluble organosulfur compounds that are less odorous and more delicate, and have a less characteristic flavor than oil-soluble OSCs. However, although water-soluble OSCs make up a small portion of garlic, they may be considered the main bioactive component in health benefits, such as in cancer prevention and treating AD, as described below [37,38].

2.3. Aged Garlic Extract

There is a well-known and interesting product called aged garlic extract (AGE), which warrants special attention. AGE preparation is usually produced by storing sliced garlic cloves in a non-toxic solvent, usually a mixture of water and ethanol (15–20% ethanol solution in water), which is then aged for more than 20 months at room temperature; then, the extract is filtered and concentrated. This aging process aims to transform the odorous and pungent sulfur compounds to odorless ones. This process is characterized by the transformation of allicin into stable and safe sulfur compounds, in particular, water-soluble organosulfur compounds, including the two major ones, SAC and SAMC, and small amounts of oil-soluble allyl sulfides. The composition of AGE garlic extract has many antioxidants properties and, combined with the high bioavailability of SAC and SAMC which are rapidly absorbed by the intestinal tract, seems to play an important role in the biological effects of garlic [13,36,39,40].

3. Metabolism and Bioavailability

Although there are many studies regarding the sulfur compounds of garlic and its biological properties, we have little information on the metabolism of garlic and sulfur compounds and related bioavailability [36,41]. The study of bioaccessibility and also bioavailability is important to evaluate the health benefits of bioactive compounds [42,43]. Following the chewing and ingestion of garlic, allicin is synthesized and transformed into its metabolites, which are transported through the stomach, intestine and blood to the target tissues (organs), while losses are channeled through the breath, urine and stool [44]. Therefore, considering the oil- and water-soluble derivatives of garlic from different studies, it is interesting to note that allicin, sulfides, ajoene, vinyldithiins and other oil-soluble OSCs cannot be detected in the blood or urine even after the intake of a large amount of garlic, while after the intake of fresh garlic SAC and water-soluble compounds can be detected in the plasma, liver and kidney [45]. The pharmacokinetics of water-soluble OSCs were found to be different from those of oil-soluble ones. Indeed, SAC was rapidly absorbed in the gastrointestinal tract and was identified in blood and its concentration, and other pharmacokinetic parameters were correlated with SAC doses administered to animals [13,46]. The results from different evaluations on the bioavailability and efficacy of SAC indicated that water-soluble garlic compounds seem to play an important role in the biological effects of garlic, in vitro and in vivo.

4. AD

AD represents the most common form of dementia affecting the aging population. It is a progressive neurodegenerative disease affecting memory whose diagnosis is based on cognitive impairment evaluation. Etiology is multifactorial, including both genetic and epigenetic elements. Physical exercise, diet, lifestyle and environmental exposure to heavy metals are crucial factors in its pathogenesis [47,48,49]. From a pathological point of view, AD is characterized by the accumulation of amyloid β-protein (Aβ) plaques and the deposition of hyperphosphorylated tau proteins in the brain, leading to synaptic and neuronal loss [50,51]. Aβ peptide, a 40–42 amino acid residue, is derived from a transmembrane amyloid precursor protein (APP), following cleavage by β- and γ-secretase (BACE1, PS1, PS2 and nicastrin) [52]. After polymerization, the Aβ oligomeric structures devolve into a hazardous molecule, which activates microglia and produces reactive oxygen species (ROS) and inflammatory cytokines, resulting in severe neuronal injury [53,54]. Various amyloid cascade hypothesis-related AD animal models have been developed over the years in order to identify novel therapeutic medications for AD therapy [55,56]. Indeed, it has been shown that the intracerebral administrations of Aβ induces neurodegeneration, as well as deficiencies in learning and memory [55,57]. According to the cholinergic theory, the primary cause of AD is a decrease in acetylcholine (ACh) contents, which are enzymatically destroyed by acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). As a result, these enzymes are important therapeutic strategies for AD. Cholinesterase inhibitors (ChEIs) block these enzymes, resulting in greater ACh levels and a transient alleviation in AD symptoms. Therefore, increasing synaptic ACh concentrations with ChEIs, such as donepezil, rivastigmine and galantamine, is the main current pharmacological therapy for AD patients [58,59]. Unfortunately, these medications, as well as memantine, which decreases glutamate excitotoxicity, only provide symptomatic alleviation and do not slow disease progression; thus, the development of new therapeutic approaches is required. There is currently no conventional medicine for the treatment of AD that is able to modify the pathology. Aducanumab, a novel antibody that targets Aβ protein accumulation, has recently hit the market but its clinical usefulness is still debatable [60,61,62]. In conclusion, as AD is a major health problem, new medications to treat it continue to pique attention.

5. Neuroinflammation

Neuroinflammation is implicated in the pathogenesis of neurodegenerative disorders, such as AD [63]. Microglia are immune cells in the brain that react to tissue injury and healing, and their activation contributes to neuroinflammation and neurodegenerative disorders [64,65,66].
Activated microglia spread and replicate, generating pro-inflammatory cytokines including interleukin 1 (IL-1)β and tumor necrosis factor (TNF)α, as well as oxygen and nitrogen radical species, L-glutamate and prostaglandin E2 [67,68]. In addition, other less recognized neurotoxic molecules released by microglia include Aβ, cathepsin B and D, C-X-C motif chemokine ligand (CXCL)10 and CXCL12 (5–67), high mobility group box, lymphotoxin-α, matrix metalloproteinase (MMP)-2 and MMP-9, platelet-activating factor and prolyl endopeptidase, resulting in neuron death or impairment [69].
Multiple signaling pathways responsible for neuroinflammation have been described [70]. Following the identification of invading pathogens and/or tissue damage by pattern recognition receptors (PRR), innate immune activation in the CNS can be initiated through a variety of routes. Considerable emphasis has been placed on a two-signal theory controlled by Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain-containing protein (nucleotide-binding oligomerization domain-containing protein (NOD)-like receptors (NLRs), the latter forming the inflammasome essential for pro-IL-1 and pro-IL-18 digestion [71]. Several TLRs have been shown to interact with pathogen-associated molecular patterns (PAMPs) expressed on broad microbial subclasses, as well as with endogenous molecules, identified as danger-associated molecular patterns (DAMPs), increasing during cell damage [72]. Signal 1 can be triggered by TLR, through the TLR ligand, and the subsequent recruitment of Myeloid Differentiating factor 88 (MyD88) or via the TNF-α receptor, resulting in the NF-κB-dependent transcriptional activation of pro-IL-1 and pro-IL-18 (Figure 3). TLRs and TNF-α are membrane-spanning receptors, but NLRs are intracellular detectors that oligomerize to generate the inflammasome, a multi-protein complex that requires caspase-1 to convert pro-IL-1 and pro-IL-18 to their mature forms. The NOD signaling, accompanied by proteolytic cleavage via inflammasome activation, is essential for this process and represents signal 2, which can be stimulated by a variety of chemicals, comprising different pore-forming toxins, Aβ, ATP, K+ outflow, silica and uric acid crystals [73,74,75,76] (Figure 3). NLRP3 is the best-described inflammasome of the 22 NLR genes found in humans, and the observation that multiple structurally diverse stimuli are capable of triggering NLRP3 inflammasome activity has led to the notion that NLRP3 detects a general “danger” signal caused by lysosomal dysfunction [76,77]. Other NLRs, on the contrary, react to a more limited stimulus repertoire. IL-1 and IL-18 have been linked to the pathogenesis of a variety of neurodegenerative illnesses, including AD, as well as a number of CNS pathologies.
Alternatively, TLR can interact with MyD88, triggering NF-κB stimulation, thus provoking the secretion of TNF-α and IL-6 [68,78,79,80].
Other signaling pathways linked to neuroinflammation involve cyclooxygenase 1 (COX1) and 2 (COX-II) activation and the related production of prostaglandins by means of ROS activation, with COX I being generally considered as the housekeeping enzyme and COX II being the enzyme responsible for triggering neuroinflammation [81,82].
The PI3K/AKT pathway, including the mammalian target of rapamycin (mTOR), is another important signaling route mediating microglia damage [83,84]. Its activation regulates NF-κB activity in neuroinflammation [85,86]. Furthermore, mitogen-activated protein kinase (MAPK) family kinases, comprising p38 MAPK and stress-activated protein kinases/Jun amino-terminal kinases (SAPK/JNK), play a role in microglia activation, promoting the secretion of proinflammatory cytokines [87].

6. Garlic Bioactive Compounds as Neuroprotective Agents against AD

6.1. Allicin

Recently, the healthy properties of allicin have been widely characterized in terms of neuroprotection for AD therapy [11,88]. A comparison between raw and steamed garlic has been performed in terms of anti-inflammatory activities in LPS-stimulated BV2 microglial cells. It was found that only raw garlic, in which the generation of allicin was preserved, was able to reduce proinflammatory cytokines such as IL-1ꞵ, TNF-α, MCP-1, NO and NF-kB [89]. In particular, an antineuroinflammatory effect was obtained through concentrations of 200 and 400 μM of DATS and DADS, respectively, derived from allicin. Interestingly, the neuroprotective activity of allicin has been investigated in an animal model of AD expressing human double mutant APP and PS1 genes. Allicin induced an improvement in memory by affecting the APP metabolism, with a decrease in Aβ peptide expression, mediated through PS1, PS2, BACE1 and nicastrin reduction. Allicin lowered the elevated levels of oxidative stress (JNK/c-jun-dependent), which are strictly connected to AD pathology, thus reducing mitochondrial dysfunction [90] (Table 1).
Interestingly, it has been reported that JNK/c-jun is associated with APP, Aβ and cognitive decline, suggesting that its blocking may be relevant in the pathogenesis and therapy of AD [40,97,98]. Accordingly, in a previous study, allicin therapy effectively improved age-induced cognitive impairment by increasing the Nrf2 antioxidant signaling pathways, thus protecting the hippocampus against free radical-induced damage [91]. Similarly, in the context of neuroinflammation in depressive-like animal models, allicin reduced microglia activation, and inflammatory cytokines and ROS increased in the hippocampus, while it stimulated SOD activity and Nrf2 signaling. Interestingly, allicin inhibited an overstimulated NLRP3 inflammasome, including a reduction in its components, such as ACS, caspase-1 and IL-1β [92]. Accordingly, in another brain damage-like acute TSCI, causing glial reactivity and neuroinflammation, allicin prevented neuronal injury by limiting oxidative stress, inflammatory response and neuron apoptosis. These events are mediated through Nrf2 pathway activation [93].
The inhibiting action of allicin (IC50 = 62 μM) on enzymes that break down acetylcholine, especially AChE and BChE (from bovine erythrocytes), may also play a role in the favorable effect on cognition [99]. In addition, allicin improved cognition and decreased tau phosphorylation and Aβ42 deposit in the hippocampus through the pERK/Nrf2 antioxidative signaling pathway in a model of endoplasmic reticulum stress-related cognitive deficits, obtained through treating rats with TM [94]. Similarly, allicin had a favorable effect on cognition, learning and memory impairment in an AD mouse model. These benefits may be due to an elevation in SOD and a decrease in malondialdehyde activities. Expressions of Aβ and p38 MAPK were also reduced by allicin in these AD mice [95]. Recently, allicin had a protective effect when administered in rats treated with aluminum chloride and copper sulfate as animal models of AD. Specifically, allicin attenuated Aβ plaque formation, oxidative stress, neuroinflammation and cholinergic neuron damage, decreasing the symptoms typical of AD [96]. In conclusion, by engaging numerous molecular and signaling transduction pathways, allicin may be a candidate molecule for combating neuroinflammation (Figure 4) [88].

6.2. AGE

There is a large body of evidence dating back to the 1990s that reports on the favorable benefits of AGE on memory and neuroprotection (Figure 5) [25,100,101,102].
In particular, in spontaneous senescent mice, the impairment of learning and memory, as well as brain atrophy, was reduced by AGE [103,104] (Table 2).
In addition, in an in vitro cell model of AD represented by Aβ-injured PC12 cells, AGE and one of its major constituents, SAC, reduced ROS production, caspase-3 activation, DNA fragmentation and PARP cleavage, thus protecting against Aβ-induced apoptosis [105,106,109,121,122,123]. In addition, SAC was protective and reduced cell death in organotypic hippocampal slice culture injured with Aβ [107]. AGE and SAC protected neuronal cells from ROS-mediated damage and preserved the levels of the synaptosomal-associated protein of 25kDa (SNAP25) in the presynaptic neuron [108]. Furthermore, SAC reduced neuronal cell death induced by TM, alone and in combination with Aβ, by blocking calpain-, caspase 12- and caspase 3-dependent pathways [110,111]. Accordingly, AGE was able to improve the cognitive functions in both transgenic and Aβ-treated mice. AGE decreased Aβ deposition, thus hampering hippocampal-dependent memory damage [112,113,114]. Further studies indicate that SAC protects against oxidative damage by restoring the levels of antioxidative enzymes in AD animal models obtained through different approaches, such as D-galactose and STZ mice treatment [115,116]. AGE could alleviate LPS-dependent cognitive deficits via a reduction in oxidative stress, neuroinflammation, astrogliosis, and acetylcholinesterase activity [117]. In addition, AGE can ameliorate working and reference memory, by raising glutamate vesicular transporter 1 protein and glutamate decarboxylase levels and hamper the degeneration of cholinergic neurons [118]. The possible pathway responsible for these protective effects of AGE is related to a reduction in microglia activation in the cerebral cortex and hippocampus of Aβ-induced and transgenic AD animal models, with the consequent inhibition of IL-1β and inflammation [112,119,124]. In addition, AGE impaired cognitive damage induced by scopolamine via a reduction in oxidative stress and AchE activity in mice [120].
Other molecules in AGE, on the other hand, might play a role in neuroinflammation, and the mechanism by which AGE modulates neuroinflammation requires further research.

7. Conclusions

Finding a viable molecule for the pharmacological therapy of AD has been one of the most difficult issues in medical research. As most medications aimed at various targets have failed to offer a medical solution, natural products or nutraceutical components, such as garlic, arise as potential protective treatment options. Given that AD is a multifactorial illness, garlic extracts provide the benefit of a multitarget strategy, targeting different biochemical locations in the human brain, as opposed to an individual action, as with most medications used to treat AD. The data given in this review highlight the beneficial antioxidant and neuroprotective anti-inflammatory properties of allicin and AGE contained in garlic extracts. However, these studies are derived from cellular or mouse models, while clinical trials concerning these compounds and AD are not present, suggesting that this evidence should be confirmed in human studies before the beneficial effects of compounds contained in garlic might be translated into therapy. Finally, the importance of the NLRP3 inflammasome pathway in neuroinflammation and neurodegenerative diseases, such as AD, has been established [125], and very recent data on its modulation via allicin have been reported, whereas no evidence has been shown for AGE, implying the need for further characterization.

Author Contributions

Conceptualization and supervision, P.T., S.G., S.M.; writing—original draft preparation, A.T., M.N., M.C.; writing-review and editing P.T., A.C., S.G., S.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by University of Ferrara.

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. Mazza, G.; Oomah, B.D. Garlic constituents and disease prevention. In Herbs, Botanicals & Teas, Functional Foods & Nutraceuticals Series, 1st ed.; Lancaster Publishing Co., Inc.: Lancaster, UK, 2000; Volume 1, pp. 1–16. [Google Scholar]
  2. Brandolini, V.; Tedeschi, P.; Cereti, E.; Maietti, A.; Barile, D.; Coisson, J.D.; Mazzotta, D.; Arlorio, M.; Martelli, A. Chemical and genomic combined approach applied to the characterization and identification of Italian Allium sativum L. J. Agric. Food Chem. 2005, 53, 678–683. [Google Scholar] [CrossRef] [PubMed]
  3. Amagase, H.; Petesch, B.L.; Matsuura, H.; Kasuga, S.; Itakura, Y. Intake of Garlic and Its Bioactive Components. J. Nutr. 2001, 131, 955S–962S. [Google Scholar] [CrossRef] [PubMed]
  4. Ramirez, D.A.; Federici, M.F.; Altamirano, J.C.; Camargo, A.B.; Luco, J.M. Permeability Data of Organosulfur Garlic Compounds Estimated by Immobilized Artificial Membrane Chromatography: Correlation Across Several Biological Barriers. Front. Chem. 2021, 9, 690707. [Google Scholar] [CrossRef] [PubMed]
  5. Subramanian, M.S.; Nandagopal, M.S.; Nordin, S.A.; Karuppiah Thilakavathy, K.; Joseph, N. Prevailing Knowledge on the Bioavailability and Biological Activities of Sulphur Compounds from Alliums: A Potential Drug Candidate. Molecules 2020, 25, 4111. [Google Scholar] [CrossRef] [PubMed]
  6. Adler, A.J.; Holub, B.J. Effect of Garlic and Fish-Oil Supplementation on Serum Lipid and Lipoprotein Concentrations in Hypercholesterolemic Men. Am. J. Clin. Nutr. 1997, 65, 445–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Ried, K.; Toben, C.; Fakler, P. Effect of garlic on serum lipids: An updated meta-analysis. Nutr. Rev. 2013, 71, 282–299. [Google Scholar] [CrossRef] [PubMed]
  8. Gao, X.; Xue, Z.; Ma, Q.; Xing, L.; Santhanam, R.K.; Zhang, M.; Chen, H. Antioxidant and antihypertensive effects of garlic proteine and its hydrolysates and the related mechanism. J. Food Biochem. 2020, 44, 13126. [Google Scholar] [CrossRef]
  9. Liu, W.; Chen, S.; Yu, C.; Li, Y.; Zhang, J.Y. Antihypertensive effects of allicin on spontaneously hypertensive rats via vasorelaxation and hydrogen sulfide mechanisms. Biomed. Pharmacother. 2020, 128, 110240. [Google Scholar]
  10. Toygar, I.; Tureyen, A.; Demir, D.; Cetinkalp, S. Effect of allicin on wound healing: An experimental diabetes model. J. Wound Care 2020, 29, 388–392. [Google Scholar] [CrossRef]
  11. Nadeem, M.S.; Kazmi, I.; Ullah, I.; Muhammad, K.; Anwar, F. Allicin, an Antioxidant and Neuroprotective Agent, Ameliorates Cognitive Impairment. Antioxidants 2022, 11, 87. [Google Scholar] [CrossRef]
  12. Li, W.Q.; Zhang, J.Y.; Ma, J.L.; Li, Z.X.; Zhang, L.; Zhang, Y.; Guo, Y.; Zhou, T.; Li, J.Y.; Shen, L.; et al. Effects of Helicobacter pylori treatment and vitamin and garlic supplementation on gastric cancer incidence and mortality: Follow-up of a randomized intervention trial. BMJ 2019, 366, l5016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zhang, Y.; Liu, X.; Ruan, J.; Zhuang, X.; Zhang, X.; Li, Z. Phytochemicals of garlic: Promising candidates for cancer therapy. Biomed. Pharmacother. 2020, 123, 109730. [Google Scholar] [CrossRef] [PubMed]
  14. Brugnoli, F.; Tedeschi, P.; Grassilli, S.; Maietti, A.; Brandolini, V.; Bertagnolo, V. Ethanol-based garlic extract prevents malignant evolution of non-invasive breast tumor cells induced by moderate hypoxia. Biomed. Pharm. 2021, 142, 112052. [Google Scholar] [CrossRef] [PubMed]
  15. Huang, J.; Yang, B.; Xiang, T.; Peng, W.; Qiu, Z.; Wan, J.; Zhang, L.; Li, H.; Li, H.; Ren, G. Diallyl disulfide inhibits growth and metastatic potential of human triple-negative breast cancer cells through inactivation of the beta-catenin signaling pathway. Mol. Nutr. Food Res. 2015, 59, 1063–1075. [Google Scholar] [CrossRef] [PubMed]
  16. Desai, G.; Schelske-Santos, M.; Nazario, C.M.; Rosario-Rosado, R.V.; Mansilla-Rivera, I.; Ramírez-Marrero, F.; Nie, J.; Myneni, A.A.; Zhang, Z.F.; Freudenheim, J.L.; et al. Onion and Garlic Intake and Breast Cancer, a Case-Control Study in Puerto Rico. Nutr. Cancer 2020, 72, 791–800. [Google Scholar] [CrossRef] [PubMed]
  17. Patiño-Morales, C.C.; Jaime-Cruz, R.; Sánchez-Gómez, C.; Corona, J.C.; Hernández-Cruz, E.Y.; Kalinova-Jelezova, I.; Pedraza-Chaverri, J.; Maldonado, P.D.; Silva-Islas, C.A.; Salazar-García, M. Antitumor Effects of Natural Compounds Derived from Allium sativum on Neuroblastoma: An Overview. Antioxidants 2021, 11, 48. [Google Scholar] [CrossRef]
  18. Juan-García, A.; Agahi, F.; Drakonaki, M.; Tedeschi, P.; Font, G.; Juan, C. Cytoprotection assessment against mycotoxins on HepG2 cells by extracts from Allium sativum L. Food Chem. Toxicol. 2021, 151, 112129. [Google Scholar] [CrossRef]
  19. Agahi, F.; Penalva-Olcina, R.; Font, G.; Juan-García, A.; Juan, C. Effects of Voghiera garlic extracts in neuronal human cell line against zearalenone’s derivates and beauvericin. Food Chem. Toxicol. 2022, 162, 112905. [Google Scholar] [CrossRef]
  20. Tedeschi, P.; Leis, M.; Pezzi, M.; Civolani, S.; Maietti, A.; Brandolini, V. Insecticidal activity and fungitoxicity of plant extracts and components of horseradish (Armoracia rusticana) and garlic (Allium sativum). J. Environ. Sci. Health B 2011, 46, 486–490. [Google Scholar]
  21. Mylona, K.; Garcia-Cela, E.; Sulyok, M.; Medina, A.; Magan, N. Influence of two garlic-derived compounds, propyl propane thiosulfonate (PTS) and propyl propane thiosulfinate (PTSO), on growth and mycotoxin production by Fusarium species in vitro and in stored cereals. Toxins 2019, 11, 495. [Google Scholar] [CrossRef] [Green Version]
  22. Alorainy, M.S. Evaluation of antimicrobial activity of garlic (Allium sativum) against E. coli O 157:H 7. J. Agric. Vet. Sci. 2011, 4, 149–157. [Google Scholar]
  23. Chakraborty, D.; Majumder, A. Garlic (Lahsun)–an immunity booster against SARS-CoV-2. Biotica Res. Today 2020, 2, 755–757. [Google Scholar]
  24. Khubber, S.; Hashemifesharaki, R.; Mohammadi, M.; Gharibzahedi, S.M.T. Garlic (Allium sativum L.): A potential unique therapeutic food rich in organosulfur and flavonoid compounds to fight with COVID-19. Nutr. J. 2020, 19, 124. [Google Scholar] [CrossRef] [PubMed]
  25. Sripanidkulchai, B. Benefits of aged garlic extract on Alzheimer’s disease: Possible mechanisms of action. Exp. Ther. Med. 2020, 19, 1560–1564. [Google Scholar] [CrossRef] [Green Version]
  26. Seki, K.; Ishikawa, J.; Okada, Y. Contribution of 2-Propenesulfenic acid to the Antioxidant activity of allicin. J. Food Sci. 2018, 83, 1265–1270. [Google Scholar] [CrossRef]
  27. Szychowski, K.A.; Rybczynska-Tkaczyk, K.; Gawel-Beben, K.; Swieca, M.; Karas, M.; Jakubczyk, A.; Matysiak, M.; Binduga, U.E.; Gminski, J. Characterization of active compounds of different garlic (Allium sativum L.) cultivars. Pol. J. Food Nutr. Sci. 2018, 68, 73–81. [Google Scholar] [CrossRef]
  28. Cruz-Villalon, G. Synthesis of allicin and purification by solid-phase extraction. Anal. Biochem. 2001, 290, 376–378. [Google Scholar] [CrossRef]
  29. Mansingh, D.P.; Dalpati, N.; Sali, V.K.; Vasanthi, A.H.R. Alliin the precursor of allicin in garlic extract mitigates proliferation of gastric adenocarcinoma cells by modulating apoptosis. Pharmacogn. Mag. 2018, 14, 84–91. [Google Scholar]
  30. Shang, A.; Cao, S.; Xu, X.; Gan, R.; Tang, G.; Corke, H. Bioactive Compounds and Biological Functions of Garlic (Allium sativum L.). Foods 2019, 8, 246. [Google Scholar] [CrossRef] [Green Version]
  31. Lawson, L.D.; Hunsaker, S.M. Allicin Bioavailability and Bioequivalence from Garlic Supplements and Garlic Foods. Nutrients 2018, 10, 812. [Google Scholar] [CrossRef] [Green Version]
  32. Albrecht, F.; Leontiev, R.; Jacob, C.; Slusarenko, A.J. An optimized facile procedure to synthesize and purify allicin. Molecules 2017, 22, 770. [Google Scholar] [CrossRef] [Green Version]
  33. Fujisawa, H.; Suma, K.; Origuchi, K.; Seki, T.; Ariga, T. Thermostability of allicin determined by chemical and biological assays. Biosci. Biotechno. Biochem. 2008, 72, 2877–2883. [Google Scholar] [CrossRef] [PubMed]
  34. Salehi, B.; Zucca, P.; Orhan, I.E.; Azzini, E.; Adetunji, C.O.; Mohammed, S.A.; Ahmad, Z. Allicin and health: A comprehensive review. Trends Food Sci. Technol. 2019, 86, 502–516. [Google Scholar] [CrossRef]
  35. Liu, P.; Weng, R.; Sheng, X.; Wang, X.; Zhang, W.; Qian, Y.; Qiu, J. Profiling of organosulfur compounds and amino acids in garlic from different regions of China. Food Chem. 2020, 305, 125499. [Google Scholar] [CrossRef] [PubMed]
  36. Iciek, M.; Kwiecien, I.; Włodek, L. Biological Properties of Garlic and Garlic-Derived Organosulfur Compounds. Environ. Mol. Mutagen. 2009, 50, 247–265. [Google Scholar] [CrossRef] [PubMed]
  37. Fukushima, S.; Takada, N.; Wanibuchi, H.; Hori, T.; Min, W.; Ogawa, M. Suppression of chemical carcinogenesis by water-soluble organosulfur compounds. J. Nutr. 2001, 131, 1049S–1053S. [Google Scholar] [CrossRef]
  38. Farhat, Z.; Hershberger, P.A.; Freudenheim, J.L.; Mammen, M.J.; Hageman Blair, R.; Aga, D.S.; Mu, L. Types of garlic and their anticancer and antioxidant activity: A review of the epidemiologic and experimental evidence. Eur. J. Nutr. 2021, 60, 3585–3609. [Google Scholar] [CrossRef]
  39. Lanzotti, V.; Scala, F.; Bonanomi, G. Compounds from Allium species with cytotoxic and antimicrobial activity. Phytochem. Rev. 2014, 13, 769–791. [Google Scholar] [CrossRef]
  40. Gourmaud, S.; Paquet, C.; Dumurgier, J.; Pace, C.; Bouras, C.; Gray, F.; Hugon, J. Increased levels of cerebrospinal fluid JNK3 associated with amyloid pathology: Links to cognitive decline. J. Psychiatry Neurosci. 2015, 40, 151–161. [Google Scholar] [CrossRef] [Green Version]
  41. Putnik, P.; Gabrić, D.; Roohinej, S.; Barba, F.J.; Granato, D.; Mallikarjunan, K.; Lorenzo, J.M.; Bursać Kovačević, D. An overview of organosulfur compounds from Allium spp.: From processing and preservation to evaluation of their bioavailability, antimicrobial, and anti-inflammatory properties. Food Chem. 2019, 276, 680–691. [Google Scholar] [CrossRef]
  42. Barba, F.J.; Orlien, V. Processing, bioaccessibility and bioavailability of bioactive sulfur compounds: Facts and gaps. J. Food Compos. Anal. 2017, 61, 1–3. [Google Scholar] [CrossRef]
  43. Barba, F.J.; Mariutti, L.R.B.; Bragagnolo, N.; Mercadante, A.Z.; Barbosa-Cánovas, G.V.; Orlien, V. Bioaccessibility of bioactive compounds from fruits and vegetables after thermal and nonthermal processing. Trends Food Sci. Technol. 2017, 67, 195–206. [Google Scholar] [CrossRef]
  44. Marchese, A.; Barbieri, R.; Sanches-Silva, A.; Daglia, M.; Nabavi, S.F.; Jafari, N.J.; Nabavi, S.M. Antifungal and antibacterial activities of allicin: A review. Trends Food Sci. Technol. 2016, 52, 49–56. [Google Scholar] [CrossRef]
  45. Rahman, M.S. Allicin and other functional active components in garlic: Health benefits and bioavailability. Int. J. Food Prop. 2007, 10, 245–268. [Google Scholar] [CrossRef]
  46. Lawson, L.D.; Gardner, C.D. Composition, stability, and bioavailability of garlic products used in a clinical trial. J. Agric. Food Chem. 2005, 53, 6254–6261. [Google Scholar] [CrossRef] [Green Version]
  47. Lahiri, D.K.; Chen, D.; Ge, Y.W.; Bondy, S.C.; Sharman, E.H. Dietary supplementation with melatonin reduces levels of amyloid beta-peptides in the murine cerebral cortex. J. Pineal Res. 2004, 36, 224–231. [Google Scholar] [CrossRef]
  48. Lahiri, D.K.; Maloney, B.; Zawia, N.H. The LEARn model: An epigenetic explanation for idiopathic neurobiological diseases. Mol. Psychiatry 2009, 14, 992–1003. [Google Scholar] [CrossRef]
  49. Lahiri, D.K.; Maloney, B. The ‘LEARn’ (Latent Early-life Associated Regulation) model integrates environmental risk factors and the developmental basis of Alzheimer’s disease, and proposes remedial steps. Exp. Gerontol. 2010, 45, 291–296. [Google Scholar] [CrossRef] [Green Version]
  50. Price, D.L.; Sisodia, S.S.; Borchelt, D.R. Genetic neurodegenerative diseases: The human illness and transgenic models. Science 1998, 282, 1079–1083. [Google Scholar] [CrossRef]
  51. Varadarajan, S.; Yatin, S.; Aksenova, M.; Butterfield, D.A. Review: Alzheimer’s amyloid beta-peptide-associated free radical oxidative stress and neurotoxicity. J. Struct. Biol. 2000, 130, 184–208. [Google Scholar] [CrossRef]
  52. Vassar, R.; Kovacs, D.M.; Yan, R.; Wong, P.C. The beta-secretase enzyme BACE in health and Alzheimer’s disease: Regulation, cell biology, function, and therapeutic potential. J. Neurosci. 2009, 29, 12787–12794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. El Khoury, J.; Hickman, S.E.; Thomas, C.A.; Cao, L.; Silverstein, S.C.; Loike, J.D. Scavenger receptor-mediated adhesion of microglia to beta-amyloid fibrils. Nature 1996, 382, 716–719. [Google Scholar] [CrossRef] [PubMed]
  54. Coraci, I.S.; Husemann, J.; Berman, J.W.; Hulette, C.; Dufour, J.H.; Campanella, G.K.; Luster, A.D.; Silverstein, S.C.; El-Khoury, J.B. CD36, a class B scavenger receptor, is expressed on microglia in Alzheimer’s disease brains and can mediate production of reactive oxygen species in response to beta-amyloid fibrils. Am. J. Pathol. 2002, 160, 101–112. [Google Scholar] [CrossRef]
  55. Yamada, K.; Nabeshima, T. Animal models of Alzheimer’s disease and evaluation of anti-dementia drugs. Pharmacol. Ther. 2000, 88, 93–113. [Google Scholar] [CrossRef]
  56. Nakai, T.; Yamada, K.; Mizoguchi, H. Alzheimer’s Disease Animal Models: Elucidation of Biomarkers and Therapeutic Approaches for Cognitive Impairment. Int. J. Mol. Sci. 2021, 22, 5549. [Google Scholar] [CrossRef]
  57. Yamada, K.; Ren, X.; Nabeshima, T. Perspectives of pharmaco-therapy in Alzheimer’s disease. Jpn. J. Pharmacol. 1999, 80, 9–14. [Google Scholar]
  58. Park, K.; Kim, E.; Han, H.; Shim, Y.; Kwon, J.; Ku, B.; Park, K.H.; Yi, H.A.; Kim, K.K.; Yang, D.W. Efficacy and Tolerability of Rivastigmine Patch Therapy in Patients with Mild-To-Moderate Alzheimer’s Dementia Associated with Minimal and Moderate Ischemic white Matter Hyperintensities: A Multicenter Prospective Open-Label Clinical Trial. PLoS ONE 2017, 12, e0182123. [Google Scholar] [CrossRef] [Green Version]
  59. Stoiljkovic, M.; Horvath, T.L.; Hajós, M. Therapy for Alzheimer’s disease: Missing targets and functional markers? Ageing Res. Rev. 2021, 68, 101318. [Google Scholar] [CrossRef]
  60. Long, J.M.; Holtzman, D.M. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019, 179, 312–339. [Google Scholar] [CrossRef]
  61. Cummings, J. New Approaches to Symptomatic Treatments for Alzheimer’s Disease. Mol. Neurodegener. 2021, 16, 2. [Google Scholar] [CrossRef]
  62. Whitehouse, P.J.; Saini, V. Making the Case for the Accelerated Withdrawal of Aducanumab. J. Alzheimers Dis. 2022, 87, 1003–1007, Online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  63. Uchida, K. Waste Clearance in the Brain and Neuroinflammation: A Novel Perspective on Biomarker and Drug Target Discovery in Alzheimer’s Disease. Cells 2022, 11, 919. [Google Scholar] [CrossRef] [PubMed]
  64. Walker, D.G. Defining activation states of microglia in human brain tissue: An unresolved issue for Alzheimer’s disease. Neuroimmunol. Neuroinfam. 2020, 7, 194–214. [Google Scholar] [CrossRef]
  65. Leng, F.; Edison, P. Neuroinfammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar] [CrossRef] [PubMed]
  66. McFarland, K.N.; Chakrabarty, P. Microglia in Alzheimer’s Disease: A Key Player in the Transition Between Homeostasis and Pathogenesis. Neurotherapeutics 2022, 19, 186–208. [Google Scholar] [CrossRef]
  67. Neniskyte, U.; Vilalta, A.; Brown, G.C. Tumour necrosis factor alpha-induced neuronal loss is mediated by microglial phagocytosis. FEBS Lett. 2014, 588, 2952–2956. [Google Scholar] [CrossRef] [Green Version]
  68. Wu, L.; Du, L.; Ju, Q.; Chen, Z.; Ma, Y.; Bai, T.; Ji, G.; Wu, Y.; Liu, Z.; Shao, Y.; et al. Silencing TLR4/MyD88/NF-κB signaling pathway alleviated inflammation of corneal epithelial cells infected by ISE. Inflammation 2021, 44, 633–644. [Google Scholar] [CrossRef]
  69. Lindhout, I.A.; Murray, T.E.; Richards, C.M.; Klegeris, A. Potential neurotoxic activity of diverse molecules released by microglia. Neurochem. Int. 2021, 148, 105117. [Google Scholar] [CrossRef]
  70. Shabab, T.; Khanabdali, R.; Moghadamtousi, S.Z.; Kadir, H.A.; Mohan, G. Neuroinflammation pathways: A general review. Int. J. Neurosci. 2017, 127, 624–633. [Google Scholar] [CrossRef]
  71. Hanamsagar, R.; Hanke, M.L.; Kielian, T. Toll-like receptor (TLR) and inflammasome actions in the central nervous system. Trends Immunol. 2012, 33, 333–342. [Google Scholar] [CrossRef] [Green Version]
  72. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef]
  73. Petrilli, V.; Papin, S.; Dostert, C.; Mayor, A.; Martinon, F.; Tschopp, J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ. 2007, 14, 1583–1589. [Google Scholar] [CrossRef]
  74. Dostert, C.; Pétrilli, V.; Van Bruggen, R.; Steele, C.; Mossman, B.T.; Tschopp, J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008, 320, 674–677. [Google Scholar] [CrossRef] [Green Version]
  75. Craven, R.R.; Gao, X.; Allen, I.; Gris, D.; Wardenburg, J.B.; McElvania-TeKippe, E.; Ting, J.P.; Duncan, J.A. Staphylococcus aureus alpha-hemolysin activates the NLRP3-inflammasome in human and mouse monocytic cells. PLoS ONE 2009, 4, e7446. [Google Scholar] [CrossRef]
  76. Schroder, K.; Zhou, R.; Tschopp, J. The NLRP3 inflammasome: A sensor for metabolic danger? Science 2010, 327, 296–300. [Google Scholar] [CrossRef]
  77. Hornung, V.; Bauernfeind, F.; Halle, A.; O Samstad, E.; Kono, H.; Rock, K.L.; Fitzgerald, K.; Latz, E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat. Immunol. 2008, 9, 847–856. [Google Scholar] [CrossRef]
  78. Han, L.P.; Li, C.J.; Sun, B.; Xie, Y.; Guan, Y.; Ma, Z.J.; Chenm, L.M. Protective effects of celastrol on diabetic liver injury via TLR4/MyD88/NF-κB signaling pathway in type 2 diabetic rats. J. Diabetes Res. 2016, 2016, 2641248. [Google Scholar] [CrossRef] [Green Version]
  79. Zhong, Y.; Wu, S.; Yang, Y.; Li, G.Q.; Meng, L.; Zheng, Q.Y.; Li, Y.; Xu, G.L.; Zhang, K.Q.; Peng, K.F. LIGHT aggravates sepsis-associated acute kidney injury via TLR4-MyD88-NF-κB pathway. J. Cell. Mol. Med. 2020, 24, 11936–11948. [Google Scholar] [CrossRef]
  80. El-Sahar, A.E.; Shiha, N.A.; El Sayed, N.S.; Ahmed, L.A. Alogliptin attenuates lipopolysaccharide-induced neuroinflammation in mice through modulation of TLR4/MYD88/NF-κB and miRNA-155/SOCS-1 signaling pathways. Int. J. Neuropsychopharmacol. 2021, 24, 158–169. [Google Scholar] [CrossRef]
  81. Aıd, S.; Bosetti, F. Targeting cyclooxygenases-1 and -2 in neuroinflammation: Therapeutic implications. Biochimie 2011, 93, 46–51. [Google Scholar] [CrossRef] [Green Version]
  82. Ghazanfari, N.; van Waarde, A.; Dierckx, R.A.; Doorduin, J.; de Vries, E.F. Is cyclooxygenase-1 involved in neuroinflammation? J. Neurosci. Res. 2021, 99, 2976–2998. [Google Scholar] [CrossRef]
  83. Xian, M.; Cai, J.; Zheng, K.; Liu, Q.; Liu, Y.; Lin, H.; Liang, S.; Wang, S. Aloe-emodin prevents nerve injury and neuroinflammation caused by ischemic stroke via the PI3K/AKT/mTOR and NF-κB pathway. Food Funct. 2021, 12, 8056–8067. [Google Scholar] [CrossRef]
  84. Zhao, M.; Zhou, A.; Xu, L.; Zhang, X. The role of TLR4-mediated PTEN/PI3K/AKT/NF-κB signaling pathway in neuroinflammation in hippocampal neurons. Neuroscience 2014, 269, 93–101. [Google Scholar] [CrossRef]
  85. Wang, L.; Kou, M.C.; Weng, C.Y.; Hu, L.W.; Wang, Y.J.; Wu, M.J. Arsenic modulates heme oxygenase-1, interleukin-6, and vascular endothelial growth factor expression in endothelial cells: Roles of ROS, NF-κB, and MAPK pathways. Arch. Toxicol. 2012, 86, 879–896. [Google Scholar] [CrossRef]
  86. Srivastava, I.N.; Shperdheja, J.; Baybis, M.; Ferguson, T.; Crino, P.B. mTOR pathway inhibition prevents neuroinflammation and neuronal death in a mouse model of cerebral palsy. Neurobiol. Dis. 2016, 85, 144–154. [Google Scholar] [CrossRef]
  87. Merighi, S.; Bencivenni, S.; Vincenzi, F.; Varani, K.; Borea, P.A.; Gessi, S. A(2B) adenosine receptors stimulate IL-6 production in primary murine microglia through p38 MAPK kinase pathway. Pharmacol. Res. 2017, 117, 9–19. [Google Scholar] [CrossRef]
  88. Mocayar Marón, F.J.; Camargo, A.B.; Manucha, W. Allicin pharmacology: Common molecular mechanisms against neuroinflammation and cardiovascular diseases. Life Sci. 2020, 249, 117513. [Google Scholar] [CrossRef]
  89. Ho, S.; Su, M. Evaluating the anti-neuroinflammatory capacity of raw and steamed garlic as well as five organosulfur compounds. Molecules 2014, 19, 17697–17714. [Google Scholar] [CrossRef] [Green Version]
  90. Zhang, H.; Wang, P.; Xue, Y.; Liu, L.; Li, Z.; Liu, Y. Allicin ameliorates cognitive impairment in APP/PS1 mice via suppressing oxidative stress by blocking JNK signaling pathways. Tissue Cell 2018, 50, 89–95. [Google Scholar] [CrossRef]
  91. Li, X.H.; Li, C.Y.; Lu, J.M.; Tian, R.B.; Wei, J. Allicin ameliorates cognitive deficits ageing-induced learning and memory deficits through enhancing of Nrf2 antioxidant signaling pathways. Neurosci. Lett. 2012, 514, 46–50. [Google Scholar] [CrossRef]
  92. Gao, W.; Wang, W.; Liu, G.; Zhang, J.; Yang, J.; Deng, Z. Allicin attenuated chronic social defeat stress induced depressive-like behaviors through suppression of NLRP3 inflammasome. Metab. Brain Dis. 2019, 34, 319–329. [Google Scholar] [CrossRef]
  93. Lv, R.; Mao, N.; Wu, J.; Lu, C.; Ding, M.; Gu, X.; Wu, Y.; Shi, Z. Neuroprotective effect of allicin in a rat model of acute spinal cord injury. Life Sci. 2015, 143, 114–123. [Google Scholar] [CrossRef]
  94. Zhu, Y.F.; Li, X.H.; Yuan, Z.P.; Li, C.Y.; Tian, R.B.; Jia, W.; Xiao, Z.P. Allicin improves endoplasmic reticulum stress-related cognitive deficits via PERK/Nrf2 antioxidative signaling pathway. Eur. J. Pharmacol. 2015, 762, 239–246. [Google Scholar] [CrossRef]
  95. Li, X.H.; Li, C.Y.; Xiang, Z.G.; Zhong, F.; Chen, Z.Y.; Lu, J.M. Allicin can reduce neuronal death and ameliorate the spatial memory impairment in Alzheimer’s disease models. Neurosciences 2010, 15, 237–243. [Google Scholar]
  96. Kaur, S.; Raj, K.; Gupta, Y.K.; Singh, S. Allicin ameliorates aluminium- and copper-induced cognitive dysfunction in Wistar rats: Relevance to neuro-inflammation, neurotransmitters and Abeta((1–42)) analysis. J. Biol. Inorg. Chem. 2021, 26, 495–510. [Google Scholar] [CrossRef]
  97. Killick, R.; Ribe, E.M.; Al-Shawi, R.; Malik, B.; Hooper, C.; Fernandes, C.; Lovestone, S. Clusterin regulates beta-amyloid toxicity via Dickkopf-1-driven induction of the wnt-PCP-JNK pathway. Mol. Psychiatry 2014, 19, 88–98. [Google Scholar] [CrossRef]
  98. Ahn, J.H.; So, S.P.; Kim, N.Y.; Kim, H.J.; Yoon, S.Y.; Kim, D.H. c-Jun N-terminal Kinase (JNK) induces phosphorylation of amyloid precursor protein (APP) at Thr668, in okadaic acid-induced neurodegeneration. BMB Rep. 2016, 49, 376–381. [Google Scholar] [CrossRef] [Green Version]
  99. Kumar, S. Dual inhibition of acetylcholinesterase and butyrylcholinesterase enzymes by allicin. Indian J. Pharmacol. 2015, 47, 444–446. [Google Scholar] [CrossRef]
  100. Ray, B.; Chauhan, N.B.; Lahiri, D.K. The "aged garlic extract:" (AGE) and one of its active ingredients S-allyl-L-cysteine (SAC) as potential preventive and therapeutic agents for Alzheimer’s disease (AD). Curr. Med. Chem. 2011, 18, 3306–3313. [Google Scholar] [CrossRef]
  101. Qu, Z.; Mossine, V.V.; Cui, J.; Sun, G.Y.; Gu, Z. Protective Effects of AGE and Its Components on Neuroinflammation and Neurodegeneration. Neuromolecular Med. 2016, 18, 474–482. [Google Scholar] [CrossRef]
  102. Ahangar-Sirous, R.; Poudineh, M.; Ansari, A.; Nili, A.; Dana, S.M.M.A.; Nasiri, Z.; Hosseini, Z.; Karami, D.; Mokhtari, M.; Deravi, N. Pharmacotherapeutic Potential of Garlic in Age-Related Neurological Disorders. CNS Neurol. Disord. Drug Targets 2022, 21, 377–398. [Google Scholar] [CrossRef]
  103. Takeda, T.; Hosokawa, M.; Higuchi, K. Senescence-accelerated mouse (SAM): A novel murine model of accelerated senescence. J. Am. Geriatr. Soc. 1991, 39, 911–919. [Google Scholar] [CrossRef]
  104. Nishiyama, N.; Moriguchi, T.; Saito, H. Beneficial effects of aged garlic extract on learning and memory impairment in the senescence-accelerated mouse. Exp. Gerontol. 1997, 32, 149–160. [Google Scholar] [CrossRef]
  105. Griffin, B.; Selassie, M.; Gwebu, E.T. Effect of Aged Garlic Extract on the Cytotoxicity of Alzheimer beta-Amyloid Peptide in Neuronal PC12 Cells. Nutr. Neurosci. 2000, 3, 139–142. [Google Scholar] [CrossRef]
  106. Peng, Q.; Buz’Zard, A.R.; Lau, B.H. Neuroprotective effect of garlic compounds in amyloid-beta peptide-induced apoptosis in vitro. Med. Sci. Monit. 2002, 8, 328–337. [Google Scholar]
  107. Ito, Y.; Ito, M.; Takagi, N.; Saito, H.; Ishige, K. Neurotoxicity induced by amyloid beta-peptide and ibotenic acid in organotypic hippocampal cultures: Protection by S-allyl-L-cysteine, a garlic compound. Brain Res. 2003, 985, 98–107. [Google Scholar] [CrossRef]
  108. Ray, B.; Chauhan, N.B.; Lahiri, D.K. Oxidative insults to neurons and synapse are prevented by aged garlic extract and S-allyl-L-cysteine treatment in the neuronal culture and APP-Tg mouse model. J. Neurochem. 2011, 117, 388–402. [Google Scholar] [CrossRef] [Green Version]
  109. Jeong, J.H.; Jeong, H.R.; Jo, Y.N.; Kim, H.J.; Shin, J.H.; Heo, H.J. Ameliorating effects of aged garlic extracts against Abeta-induced neurotoxicity and cognitive impairment. BMC Complement. Altern. Med. 2013, 13, 268. [Google Scholar] [CrossRef] [Green Version]
  110. Kosuge, Y.; Koen, Y.; Ishige, K.; Minami, K.; Urasawa, H.; Saito, H.; Ito, Y. S-allyl-L-cysteine selectively protects cultured rat hippocampal neurons from amyloid beta-protein- and tunic-amycin-induced neuronal death. Neuroscience 2003, 122, 885–895. [Google Scholar] [CrossRef]
  111. Imai, T.; Kosuge, Y.; Ishige, K.; Ito, Y. Amyloid beta-protein potentiates tunicamycin-induced neuronal death in organotypic hippocampal slice cultures. Neuroscience 2007, 147, 639–651. [Google Scholar] [CrossRef]
  112. Chauhan, N.B. Anti-amyloidogenic effect of Allium sativum in Alzheimer’s transgenic model Tg2576. J. Herb. Pharmacother. 2003, 3, 95–107. [Google Scholar] [CrossRef] [PubMed]
  113. Chauhan, N.B. Effect of aged garlic extract on APP processing and tau phosphorylation in Alzheimer’s transgenic model Tg2576. J. Ethnopharmacol. 2006, 108, 385–394. [Google Scholar] [CrossRef] [PubMed]
  114. Chauhan, N.B.; Sandoval, J. Amelioration of early cognitive deficits by aged garlic extract in Alzheimer’s transgenic mice. Phytother. Res. 2007, 21, 629–640. [Google Scholar] [CrossRef] [PubMed]
  115. Tsai, S.J.; Chiu, C.P.; Yang, H.T.; Yin, M.C. s-Allyl cysteine, s-ethyl cysteine, and spropyl cysteine alleviate beta-amyloid, glycative, and oxidative injury in brain of mice treated by D-galactose. J. Agric. Food Chem. 2011, 59, 6319–6326. [Google Scholar] [CrossRef]
  116. Javed, H.; Khan, M.M.; Khan, A.; Vaibhav, K.; Ahmad, A.; Khuwaja, G.; Ahmed, M.E.; Raza, S.S.; Ashafaq, M.; Tabassum, R.; et al. S-allyl cysteine attenuates oxidative stress associated cognitive impairment and neurodegeneration in mouse model of streptozotocin-induced experimental dementia of Alzheimer’s type. Brain Res. 2011, 1389, 133–142. [Google Scholar] [CrossRef]
  117. Zarezadeh, M.; Baluchnejadmojarad, T.; Kiasalari, Z.; Afshin-Majd, S.; Roghani, M. Garlic active constituent s-allyl cysteine protects against lipopolysaccharide-induced cognitive deficits in the rat: Possible involved mechanisms. Eur. J. Pharmacol. 2017, 795, 13–21. [Google Scholar] [CrossRef]
  118. Thorajak, P.; Pannangrong, W.; Welbat, J.U.; Chaijaroonkhanarak, W.; Sripanidkulchai, K.; Sripanidkulchai, B. Effects of Aged Garlic Extract on Cholinergic, Glutamatergic and GABAergic Systems with Regard to Cognitive Impairment in Aβ-Induced Rats. Nutrients 2017, 9, 686. [Google Scholar] [CrossRef] [Green Version]
  119. Nillert, N.; Pannangrong, W.; Welbat, J.U.; Chaijaroonkhanarak, W.; Sripanidkulchai, K.; Sripanidkulchai, B. Neuroprotective Effects of Aged Garlic Extract on Cognitive Dysfunction and Neuroinflammation Induced by β-Amyloid in Rats. Nutrients 2017, 9, 24. [Google Scholar] [CrossRef] [Green Version]
  120. Li, F.; Kim, M.R. Effect of Aged Garlic Ethyl Acetate Extract on Oxidative Stress and Cholinergic Function of Scopolamine-Induced Cognitive Impairment in Mice. Prev. Nutr. Food Sci. 2019, 24, 165–170. [Google Scholar] [CrossRef]
  121. Borek, C. Antioxidant health effects of aged garlic extract. J. Nutr. 2001, 131, 1010S–1015S. [Google Scholar] [CrossRef] [Green Version]
  122. Borek, C. Garlic reduces dementia and heart-disease risk. J. Nutr. 2006, 136, 810S–812S. [Google Scholar] [CrossRef] [PubMed]
  123. Jackson, R.; McNeil, B.; Taylor, C.; Holl, G.; Ruff, D.; Gwebu, E.T. Effect of aged garlic extract on caspase-3 activity, in vitro. Nutr. Neurosci. 2002, 5, 287–290. [Google Scholar] [CrossRef] [PubMed]
  124. Luo, J.F.; Dong, Y.; Chen, J.Y.; Lu, J.H. The effect and underlying mechanisms of garlic extract against cognitive impairment and Alzheimer’s disease: A systematic review and meta-analysis of experimental animal studies. J. Ethnopharmacol. 2021, 280, 114423. [Google Scholar] [CrossRef] [PubMed]
  125. Merighi, S.; Nigro, M.; Travagli, A.; Pasquini, S.; Borea, P.A.; Varani, K.; Vincenzi, F.; Gessi, S. A2A Adenosine Receptor: A Possible Therapeutic Target for Alzheimer’s Disease by Regulating NLRP3 Inflammasome Activity? Int. J. Mol. Sci. 2022, 23, 5056. [Google Scholar] [CrossRef]
Figure 1. Fresh garlic proximate composition; data are expressed as % w/w.
Figure 1. Fresh garlic proximate composition; data are expressed as % w/w.
Ijms 23 06950 g001
Figure 2. OSCs from garlic. Intact clove garlic contains γ-Glutamyl-S-alk(en)yl-L-cysteines, the primary sulfur compounds, which with hydrolysis or oxidation can be converted into alkyl (en)yl-L-cysteine sulfoxide, commonly called Alliin. When garlic is crushed or cut, the vacuolar enzyme alliinase converts alliin to allicin, poorly soluble in water and responsible for the characteristic pungent flavor of garlic. Allicin is very unstable and rapidly decomposed to form a variety of oil-soluble compounds, including diallyl disulfide (DADS), diallyl sulfide (DAS), diallyl trisulfide (DATS), vinyl dithiin and ajoene, according to different conditions. When garlic is extracted in an aqueous solvent, γ-glutamyl -S-alk(en)yl-L-cysteines can be converted into water-soluble compounds, mainly S-allyl-cysteines (SAC) and S-allylmercaptocysteine (SAMC), which are less odorous than the oil-soluble products but are stable and have important antioxidants and bioactive effects.
Figure 2. OSCs from garlic. Intact clove garlic contains γ-Glutamyl-S-alk(en)yl-L-cysteines, the primary sulfur compounds, which with hydrolysis or oxidation can be converted into alkyl (en)yl-L-cysteine sulfoxide, commonly called Alliin. When garlic is crushed or cut, the vacuolar enzyme alliinase converts alliin to allicin, poorly soluble in water and responsible for the characteristic pungent flavor of garlic. Allicin is very unstable and rapidly decomposed to form a variety of oil-soluble compounds, including diallyl disulfide (DADS), diallyl sulfide (DAS), diallyl trisulfide (DATS), vinyl dithiin and ajoene, according to different conditions. When garlic is extracted in an aqueous solvent, γ-glutamyl -S-alk(en)yl-L-cysteines can be converted into water-soluble compounds, mainly S-allyl-cysteines (SAC) and S-allylmercaptocysteine (SAMC), which are less odorous than the oil-soluble products but are stable and have important antioxidants and bioactive effects.
Ijms 23 06950 g002
Figure 3. The ways through which the NLRP3 inflammasome is activated: The activation of the NLRP3 inflammasome is a complicated regulatory mechanism that requires two triggering steps: priming and activation. TLR4 or TNF-α receptor agonists cause the priming phase (signal 1), which then activates the NF-B pathway by boosting the production of pro-IL-18 and pro-IL-1. The NLRP3 inflammasome is then stimulated (activation step) by numerous triggering events, such as Aβ, ATP, ROS, bacteria, fungi and viruses (signal 2). Then, caspase-1 can induce maturation of IL-18 and IL-1.
Figure 3. The ways through which the NLRP3 inflammasome is activated: The activation of the NLRP3 inflammasome is a complicated regulatory mechanism that requires two triggering steps: priming and activation. TLR4 or TNF-α receptor agonists cause the priming phase (signal 1), which then activates the NF-B pathway by boosting the production of pro-IL-18 and pro-IL-1. The NLRP3 inflammasome is then stimulated (activation step) by numerous triggering events, such as Aβ, ATP, ROS, bacteria, fungi and viruses (signal 2). Then, caspase-1 can induce maturation of IL-18 and IL-1.
Ijms 23 06950 g003
Figure 4. The main events induced by allicin to provide protection in AD.
Figure 4. The main events induced by allicin to provide protection in AD.
Ijms 23 06950 g004
Figure 5. The main events induced by AGE to provide protection in AD.
Figure 5. The main events induced by AGE to provide protection in AD.
Ijms 23 06950 g005
Table 1. Summary of experimental details, including model, chemical concentration and main results of in vitro and in vivo studies of allicin and AD pathology.
Table 1. Summary of experimental details, including model, chemical concentration and main results of in vitro and in vivo studies of allicin and AD pathology.
ReferencesExperimental ModelChemicals
Concentration
Results
[90]APP/PS1 double transgenic mice10 mg/kg/day allicin via intragastric administration on alternate days for 3 months
-
Improves cognition
-
Reduces Aβ expression in the brain
-
Decreases oxidative stress and improves mitochondrial dysfunction by JNK/c-jun
[91]3- and 20-month-old C57BL/6 miceDiet supplemented with
180 mg/kg/day of allicin for 8 consecutive weeks
-
Improves age-induced cognitive impairment by increasing the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factor
-
Recognizes the human Antioxidant Response Element binding site within glutamylcysteine synthetase, and NADPH:quinone oxidoreductase 1 and defends the cell against free radical-induced damage
[92]8-week-old male C57BL/6 J mice (depressive-like model)2, 10, 50 mg/kg allicin once day via intraperitoneal injection
-
Decreases ROS production and microglial activation
-
Upregulates superoxide dismutase (SOD) and Nrf2/HO-1 pathways
-
Attenuates neuronal apoptosis
-
Inhibits NLRP3 inflammasome hyperactivity, ACS, caspase-1, and IL-1β proteins
[93]Rats with acute traumatic spinal cord injury (TSCI)2, 10, 50 mg/kg intraperitoneal injection of allicin for 21 days
-
Induces neuroprotection through antiapoptotic, anti-inflammatory and antioxidant effects
[94]Adult male rats
72 h of lateral ventricular infusion of tunicamycin (TM), an endoplasmic reticulum stress stimulator
(cognitive deficit induction)
Diet supplemented with
180 mg/kg/d of allicin for 16 weeks
-
Decreases tau phosphorylation and Aβ42 deposit in the hippocampus, oxidative stress
-
Increases pERK and Nrf2 expression in the hippocampi
[95]AD mouse model via
injection of Aß(1–42) (1 µL = 4 µg) into the bilateral hippocampi
Allicin via intraperitoneal injection for 14 days 180 mg/kg/day)
-
Prevents learning and memory impairment
-
Increases SOD and decreases ROS
[96]Male Wistar ratsAllicin via intraperitoneal injection (10 and 20 mg/kg) 7 days before metals (aluminum chloride, 200 mg/kg p.o; copper sulfate, 0.5 mg/kg p.o.) administration for 28 days
-
Exhibits neuroprotective effect through antioxidant, anti-inflammatory, neurotransmitter restoration; attenuation of neuroinflammation; and β-amyloid-induced neurotoxicity
Table 2. Summary of experimental details, including model, chemical concentration and main results, of in vitro and in vivo studies of AGE and AD pathology.
Table 2. Summary of experimental details, including model, chemical concentration and main results, of in vitro and in vivo studies of AGE and AD pathology.
ReferencesExperimental ModelChemicals
Concentration
Results
[104]Senescence-Accelerated Mice (SAM)Diet containing 2% (w/w) AGE
-
Provides antiaging effect, increases the survival ratio and ameliorates the memory acquisition deficit and the memory retention impairment
[105]Neuronal PC12 cells treated with NGF for 4 days and injured with Aβ 95 nMCell growth medium containing 0.01% AGE
-
Protects neuronal PC12 cells against Aβ toxicity
[106]Undifferentiated PC12 cells injured with Aβ25-35 40 μMAGE (1–8 mg/mL)
SAC (1–4 mg/mL)
-
Reduces ROS and apoptosis
[107]Hippocampal slice culture injured with Aβ25–35 25 μMSAC (10–100 μM)
-
Protects from cell death induced by Aβ25–35
[108]Neuronal PC12 cells treated with NGF for 12 days and APP-Tg mice0.3% or 1.0% AGE in PC12 cells
diet 2% AGE in mice
-
Protects from ROS-mediated damage
[109]NGF-treated neuronal PC12 cells injured with Aβ25-35 80 μM
ICR mice administered Aβ25-35 via intracerebroventricular injection
AGE 25–200 µg/mL in PC12 cells
Freeze-dried ethyl acetate fraction from AGE at concentrations of 5, 10 and 20 mg/kg in mice
-
Decreases in vitro ROS accumulation
-
Improves cognitive impairment against Aβ-induced neuronal deficit
[110]rat hippocampal neurons
injured with Aβ25–35 5 μM or TM (10 μg/mL)
SAC (1 µM)
-
Decreases neuronal cell death, ROS and caspase 12 induced by Aβ25–35 or TM
[111]rat hippocampal neurons
injured with Aβ25–35 25 μM or TM (20–80 μg/mL)
SAC (100 µM)
-
Blocks Aβ potentiation of TM neurotoxicity
-
Reverses the increase in calpain activity and the active forms of caspase-12 and caspase-3 induced by Aβ + TM
[112]Tg2576 miceAGE (40 mg/kg/d/4 wks)
-
Increases sAPPalpha
-
Decreases Aβ40 and Aβ42 deposition
[113]Tg2576 miceAGE 2%, 20 mg SAC/kg and 20 mg DADS/kg
-
Decreases cerebral plaque, inflammation and TAU-GSK3ꞵ-dependent phosphorylation. The order of ameliorative efficacy
-
is AGE > SAC > DADS
[114]Tg2576 miceDiet 2% AGE for 5 months
-
Prevents deterioration of hippocampal-based memory tasks
[115]C57BL/6 mice treated with D-galactose (AD-like model)SAC (1 g/L into drinking water for 7 weeks)
-
Decreases the production of Aβ and suppresses the expression of APP and BACE1
-
Retaines PKC activity, and the expression of PKC-α and PKC-γ
-
Decreases ROS and protein carbonyl levels and restores brain GPX, SOD and catalase activities
-
Lowers aldose reductase (AR) activity, AR expression, and carboxymethyllysine and pentosidine levels
[116]Intracerebroventricular infusion of streptozotocin (STZ) (model of memory impairment in mice)SAC (30 mg/kg i.p. for 15 days)
-
Prevents increased latency and path length and attenuates oxidative stress induced by STZ
[117]LPS-treated rats (167 μg/kg for 7 days)SAC (25, 50, 100 mg/kg/day p.o. for 7 days)
-
Increases cognition, learning and memory reduced by LPS; increases SOD and GSH
-
Reduces acetylcholinesterase activity NF-κB, TLR4, GFAP and IL-1β, and increases Nrf2
[118]Rats injured with Aβ1–42 1 µg/µL intracerebroventricular infusionAGE (125, 250 and 500 mg/kg p.o. for 65 days)
-
Ameliorates working and reference memory by raising glutamate vesicular transporter 1 protein and glutamate decarboxylase levels
-
Restores cholinergic neuron density reduced by Aβ1–42
[119]Rats injured with Aβ1–42 1 µg/µL intracerebroventricular infusionAGE (125, 250 and 500 mg/kg body weight, p.o., daily for 56 days)
-
Improves short-term recognition memory in cognitively impaired rats
-
Reduces microglial activation and IL-1β
[120]Scopolamine-treated mice (2 mg/kg) injected 30 min before the tests.AGE (25 or 50 mg/kg p.o.)
-
Protects against scopolamine-induced cognitive impairment by decreasing oxidative damage and regulating cholinergic function
-
Increases levels of glutathione, glutathione peroxidase and glutathione reductase, and inhibits lipid peroxidation
-
Attenuates cholinergic degradation by inhibiting acetylcholinesterase activity and increasing choline acetyltransferase activity
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tedeschi, P.; Nigro, M.; Travagli, A.; Catani, M.; Cavazzini, A.; Merighi, S.; Gessi, S. Therapeutic Potential of Allicin and Aged Garlic Extract in Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 6950. https://doi.org/10.3390/ijms23136950

AMA Style

Tedeschi P, Nigro M, Travagli A, Catani M, Cavazzini A, Merighi S, Gessi S. Therapeutic Potential of Allicin and Aged Garlic Extract in Alzheimer’s Disease. International Journal of Molecular Sciences. 2022; 23(13):6950. https://doi.org/10.3390/ijms23136950

Chicago/Turabian Style

Tedeschi, Paola, Manuela Nigro, Alessia Travagli, Martina Catani, Alberto Cavazzini, Stefania Merighi, and Stefania Gessi. 2022. "Therapeutic Potential of Allicin and Aged Garlic Extract in Alzheimer’s Disease" International Journal of Molecular Sciences 23, no. 13: 6950. https://doi.org/10.3390/ijms23136950

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