Abstract
Glutamate, as an essential neurotransmitter, has been thought to have different roles in the central nervous system (CNS), including nerve regeneration, synaptogenesis, and neurogenesis. Excessive glutamate causes an up-regulation of the multiple signaling pathways, including phosphoinositide-3 kinase/protein kinase B (PI3K/Akt), Akt/mammalian target of rapamycin (mTOR) protein, mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK)1/2, and autophagy that are involved in neurodegenerative diseases pathophysiology. There are numerous findings on curcumin, astaxanthin, thymoquinone, and berberine, as natural products, which have outstanding effects in cell signaling far beyond their anti-oxidant activity, considering as a potential therapeutic target for glutamate excitotoxicity. Herein, we address the role of glutamate as a potential target in neurodegenerative diseases and discuss the protective effects of certain phytochemicals on glutamate-induced neurotoxicity.
Key Words: Excitotoxicity, Glutamate, Neurotransmitter, Neurodegenerative- disorders, Phytochemicals
Introduction
Glutamate, as an excitatory neurotransmitter of central nervous system (CNS), plays a crucial role in memory, synaptic plasticity, learning, motor function, and neural transmission. Also, it has been proposed that glutamate can modulate nerve regeneration, tumor development, synaptogenesis, neurogenesis, and apoptosis (1). Excessive glutamate, as a critical pathogenic event, causes brain disorders such as Alzheimer’s disease (AD), and Parkinson’s disease (PD) (2, 3).
It has been hypothesized that elevated glutamate is mediated by the over-induction of glutamate receptors, leading to increased calcium (Ca2+) influx. Another hypothesis is reactive oxygen species (ROS) elevation, depletion of glutathione content, and accumulation of hydrogen peroxide (4). Also, excessive glutamate causes up-regulation of the phosphoinositide-3 kinase/protein kinase B (PI3K/Akt), Akt/mammalian target of rapamycin (mTOR) protein, mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK)1/2, and autophagy cascades that are involved in neurodegenerative diseases pathophysiology, as well (3, 5).
Plants and plant extracts consist of many components that are additively or even synergistically thought to function on several molecular targets. Nowadays, phytochemicals are applied in neurodegenerative disorders because of their active components that have anti-oxidant properties (6-11). Hence, in the current review, we elaborate on the role of glutamate as a promising target in neurodegenerative diseases and discuss the protective effects of some phytochemicals on glutamate-induced neurotoxicity.
Glutamate signaling pathway in neurodegenerative disorders
Glutamate receptors are classified into two major classes, metabotropic and ionotropic receptors. Ionotropic glutamate receptors (iGluRs) are divided into N-methyl-D aspartic acid (NMDA, high calcium conductivity), kainite (mediate sodium influx), and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA, mediate sodium influx). In contrast, metabotropic glutamate receptors (mGluRs) are G-protein-coupled receptors (GPCRs) that initiate intracellular cascades, prompting the modification of ion channels (12, 13).
The levels of glutamate are low in the extracellular spaces in the cerebrum because of three major carriers: glutamate transporter 1 (GLT1, expressed in astrocytic cells), excitatory amino acid carrier 1 (EAAC1, manifested into the brain), and glutamate/aspartate transporter (GLAST, expressed in glial cells) (14).
Neurodegenerative diseases are related to the glutamatergic pathway, prompting the activation of nitric oxide synthase, impairment of cellular calcium homeostasis, ROS generation, and apoptosis. Recently, further mechanistic details identified with glutamate overstimulation have been detailed in neurodegenerative diseases (15). Changes in the enactment pattern of NMDA receptors (NMDARs), as the fundamental receptor involved in neurodegenerative diseases, at various cellular locations have been indicated as crucial in initiating pathways driven to neuroprotection versus neuro-destruction. For this reason, evidence suggests that glutamatergic excitotoxicity is interceded by the activation of extrasynaptic relative to synaptic NMDARs. Various investigations have shown that high enactment of extrasynaptic pathways explicitly promote apoptotic signal transduction cascades, causing neuronal cell death, while the actuation of synaptic NMDARs has a neuroprotective role via Ca2+-mediated signal transduction pathways advancing neuronal survival (Figure 1) (16, 17). Mitochondria, as a cytoplasmic organelle and the first modulator of ROS, control the production of ATP, intracellular Ca2+, and free-radical scavenging. Neuronal cell death is specifically intervened by the Ca2+ entry over NMDARs. Hence, mitochondrial dysfunction, particularly in excitable cells, is in charge of expanding ROS production, and consequently, oxidative damage, promoting neurodegenerative disorders (18, 19).
Figure 1.
Glutamatergic transmission in the brain. Because of the high release of glutamate from the presynaptic axon, the glutamate receptor (mainly NMDA) is activated, and the raised levels of Ca2+ are gathered in the mitochondria. Ca2+ overload results in mitochondrial dysfunction. Mitochondrial changes as higher ROS development, adjusted redox potential, and the release of apoptotic mediators, and ATP synthesis failure can promote modified cellular homeostasis causing swelling and cellular disturbance
Glu: Glutamate; NMDA: N-Methyl-d-aspartic acid; EAAC: Excitatory amino acid carrier; ROS: Reactive oxygen species
Phytochemicals as promising compounds against glutamate-induced excitotoxicity
Ethnopharmacological studies have provided data identifying potential new medications taken from plant sources (9). In traditional medicine, various therapeutic plants and natural products have been utilized to treat neurodegenerative disorders (20-22). For instance, many drugs that are accessible in medicine were initially derived from plants, including anti-cholinesterase alkaloids isolated from plants, which have been explored for their likely effects on AD (7, 23). As shown in Table 1, we summarized the potential protective results of specific medicinal herbs against glutamate toxicity.
Table 1.
The protective effects of some of medicinal plants on glutamate-induced neurotoxicity
| Medicinal plant | Family | Type of study | Protocol | Findings | Reference (s) |
|---|---|---|---|---|---|
| Acanthus ebracteatus | Acanthaceae | In vitro | Treatment of HT22 cells with different concentrations of extract (3-50 µg/ml) and glutamate (5 mM) for 24 hours | 1) Reduction of cell death via attenuation of ROS generation, nuclear AIF translocation 2) Activation of the Nrf2/ARE pathway |
(24) |
| Alpinia oxyphylla | Zingiberaceae | In vitro | Co-treatment of primary cultured mouse cortical neurons with the extract (80-240 µg/ml) and glutamate (30 µM) | 1) An increase in the cell viability 2) A reduction in the number of apoptotic cells 3) Decrease in the intensity of DNA fragmentation. |
(25) |
| Amburana cearensis | Fabaceae | In vitro | Pretreatment of PC12 cells with glutamate at a dose of 1 mM, then exposed to different doses of the extract (0.1-1000 µg/ml) | Anti-oxidant activity | (26) |
| Aronia melanocarpa | Rosaceae | In vitro | Co-treatment of HT22 cells with the extract at doses of 10 and 100 µg/ml and glutamate (2 mM) for 24 hours | 1) Reduction in ROS level 2) A decrease in intracellular Ca2+ 3) Increase in anti-oxidant enzymes |
(27) |
| Boswellia serrata | Burseraceae | In vitro | Co-treatment of N2a and PC12 cells with the extract (25-100 μg/ml) and glutamate (8 mM) | Amelioration of the oxidative stress and the resultant apoptosis | (28) |
| Calendula officinalis | Asteraceae | In vivo | The rats received oral extracts at doses of 100 and 200 mg/kg, 1 hour after monosodium glutamate injection for seven days. | Improvement in oxidative stress, hippocampal damage, and behavioral changes | (29) |
| Citrus aurantium | Rutaceae | In vitro | Pretreatment of PC12 cells with the extract alone (6 to 200 µg/ml) for 2 hours and then incubation with glutamate (8 mM) for 24 hours | 1) Reduction in ROS level 2) Reduction in MDA level 3) Reduction in apoptotic cells |
(30) |
| Cymbopogon citratus and Ferula assafoetida | Poaceae and Apiaceae | In vitro | Cerebellar granule neurons were treated with extract at a dose of 100 μg/ml before, after, and during exposure to 30 μM of glutamate. | Reduction in cell death and apoptosis | (31) |
| Ferula gummosa | Apiaceae | In vitro | Pretreatment of PC12 and N2a cells with different concentrations of extract (25 to 200 μg/ml) for 2 hours, then incubation with glutamate (8 mM) for 24 hours | 1) Reduction in ROS level 2) Reduction in MDA level 3) Reduction in apoptotic cells |
(32) |
| GLGZD (extracts of Zingiber officinale, Trichosanthis Radix, Paeonia lactiflora, Ramulus Cinnamomi, Fructus Jujubae, Roscoe and Glycyrrhiza) | - | In vitro | Co-treatment of BV-2 microglial cells with different concentrations of extract (125 to 1000 μg/ml) and glutamate (30 mM) for 24 hours | 1) Down-regulation of Bax/Bcl-2 ratio 2) Inhibition of caspase-3 expression |
(33) |
| Glycine max (soybean) | Fabaceae | In vitro | Incubation of cortical cell cultures with the indicated compound for one hour and then exposed to 100 μM glutamate for 24 hours | Inhibition of glutamate-induced toxicity via the neuroprotective effects of triterpene glycosides | (34) |
| Polygonum multiflorum Thunb | Polygonaceae | In vitro | Pretreatment of HT22 cells with various concentrations of the extract (0.1 to 10 µg/ml) for 24 hours and then exposed to glutamate (5 mM) for 24 hours | 1) Inhibition of glutamate-induced oxidative neuronal death 2) Prevention of ERK and p38 activation |
(35) |
| Reseda luteola | Resedaceae | In vitro | Co-treatment of HT22 cells with 20 μg/ml of the extract and 5 mM of glutamate for 24 hours | Enhancement of anti-oxidant systems, such as HO-1, peroxiredoxin, catalase, and NQO1 gene | (36) |
| Rheum turkestanicum | Polygonaceae | In vitro | Pretreatment of PC12 and N2a cells with various concentrations of the extract (6-200 µg/ml), then incubation with glutamate | Reduction in cell death, apoptosis, lipid peroxidation, and ROS generation | (37) |
| Rhinacanthus nasutus | Acanthaceae | In vitro | Treatment of HT-22 cells with extract (0.1, 1 and 10 µg/ml) and glutamate (5 mM) for 18 hours | Anti-oxidant activities | (38) |
| Saussurea pulvinata Maximo | Compositae | In vitro | Pretreatment of PC12 cells with different concentration of the extract (10−7, 3×10−7, or 10−6 M) for 1 hour, and then exposing to glutamate (5 mM) for 24 hours | 1) Anti-oxidative effects 2) Anti-apoptotic properties |
(39) |
| Scrophularia genus | Scrophulariaceae | In vitro | Exposing cerebellar granule neurons to 125 μM of glutamate for 12 hours following 24 hours of incubation with extract (10 mcg/ml) | Reduction of oxidative stress | (40) |
| Solanum torvum | Solanaceae | In vivo | Mice received the oral extract (100 and 300 mg/kg) at doses of monosodium glutamate (1000 mg/kg) for 14 days | Improvement in behavioral tests decreased lipid-peroxidation, while increased anti-oxidant content such as catalase, SOD and glutathione | (41) |
| Uncaria sinensis | Rubiaceae | In vitro | 1) Pretreatment of primary cultured cortical neurons with extract (1 and 5 µg/ml), then exposing to glutamate (200 μM) 2) Incubation of cerebellar granule cells with different concentrations (3 to 300 µM) of testing materials with glutamate (100 µM) |
1) Inhibition of death receptor 4 2) Expression of anti-apoptotic proteins XIAP and Bcl-2 3) Inhibition of Ca2+ influx |
(42, 43) |
| Withania somnifera | Solanaceae | In vitro | Pretreatment of rat glioma (C6) and human neuroblastoma (IMR-32) cells with the extract (0.05% and 0.1%), then exposing to glutamate (0.06 mM-10 mM) | Inhibition of glutamate-induced cell death | (44) |
ROS: Reactive oxygen species; AIF: Apoptosis-inducing factor; Nrf2: Nuclear factor erythroid 2-related factor 2; ARE: Anti-oxidant responsive element; MDA: Malondialdehyde; ERK: Extracellular-signal-regulated kinase; HO-1: Heme oxygenase-1; SOD: Superoxide dismutase; XIAP: X-linked inhibitor of apoptosis
Different studies have proposed that active compounds isolated from plants conceivably postpone neurodegeneration and improve memory and cognitive function through their anti-inflammatory and anti-oxidant activities (24, 25). Hence, in this section, we reviewed the protective impacts of some of the phytochemicals that have been utilized in glutamate-induced neurotoxicity.
Astaxanthin
Astaxanthin (AST), a carotenoid compound, is used as a dietary supplement intended for human, animal, and aquaculture consumption (26). In vivo and in vitro studies have reported anti-carcinogenic and anti-inflammatory activity, as well as neuroprotective, and powerful anti-oxidative effects of AST (27-29). Research has shown that AST elevated the expression of heme oxygenase-1 (HO-1) and reduced oxidative stress in SH-SY5Y neuroblastoma cells (30). In another study, mouse neuronal cells (HT22) were exposed to different concentrations of AST (1.25–5 µM) and then incubated with glutamate at a dose of 5 mM. The results have shown that AST reduced glutamate toxicity via reduction of cell death, decreased the level of lactate dehydrogenase (LDH), and attenuated the expression of caspase-3/-8/-9 activity, leading to mitochondrial dysfunction by modulating the Akt/glycogen synthase kinase (GSK)-3β signaling pathway in HT22 cells (31). Also, in a different study, the neuroprotective roles and probable mechanisms of AST against glutamate-induced neurotoxicity in PC12 cells were explored. Inhibition of neuronal cell death (reducing the elevation of caspase-3 activation and Bax/Bcl-2 ratio), decreased Ca2+ influx, and the down-regulation of ROS-associated NF-κB and MAPK pathways were the main possible mechanisms of AST action (53).
Berberine
Berberine, as an isoquinoline alkaloid, is found in various plants, especially in Berberis (54). This natural compound has numerous pharmacological effects (55), such as anti-fungal (56), anti-convulsant (57), anti-tumor (58), anti-viral (59), anti-inflammatory (60), reduction of ischemic brain damage (61), and anti-oxidative activities (62). Also, different studies have reported its neuroprotective effects against neurological diseases, such as anxiety, brain stroke, mental depression, and AD (63). Pharmacokinetic properties have revealed that berberine rapidly passes through the blood brain barrier, while slowly eliminating (64). Sadeghnia et al. investigated the protective effect of berberine (50–1000 µM) against glutamate excitotoxicity in PC12 and N2a cells. It was found that berberine reduced ROS, malondialdehyde (MDA), DNA fragmentation, and increased superoxide dismutase (SOD) activity. Also, berberine attenuated the level of caspase-3 and Bax/Bcl-2 ratio (65). In agreement with their results, Lin et al. have shown that berberine prevented glutamate release from rat’s cortical synaptosomes by the inhibition of presynaptic Cav2.1 channels and the ERK/synapsin I signaling pathway (66). Furthermore, a recent study has shown that berberine (25 μg/ml) improved viability, axonal outgrowth, and transport in calyculin A-injured N2a cells. The neuroprotective effect of this compound was related to anti-oxidant activity, decreasing of MDA, increasing of SOD, and inhibiting the hyper-phosphorylation of Tau protein, and neurofilaments (67). Besides, berberine possesses neuroprotective effects through anti-oxidant defense stimulation, oxygen consumption inhibition, and mitochondrial membrane potential (MMP) elevation (68). Preclinical studies have demonstrated that berberine has shown important memory improvement activities with several mechanisms, including anti-inflammatory, anti-amyloid, anti-oxidant, and cholinesterase inhibitory activities (69). Based on these findings, berberine could be a useful agent to improve AD and PD by modulating oxidative stress (70).
Casuarinin
Casuarinin, an ellagitannin, exists in the pericarp of pomegranates (Punica granatum). In a study by Song et al., casuarinin reduced glutamate-induced HT22 murine hippocampal neuronal cell death by inhibiting ROS production, reducing chromatin condensation, and inhibiting oxidative stress-mediated MAPK phosphorylation (71).
Chebulinic acid
Chebulinic acid isolated from the extracts of the Terminalia chebula attenuates glutamate-induced HT22 cell death by inhibiting calcium influx, oxidative stress, and MAPKs phosphorylation (72).
Cinnamaldehyde
Cinnamaldehyde is an important component of cinnamon oil that is obtained from the stem bark of Cinnamomum cassia (73). It has various pharmacological properties, such as anti-oxidant (74), anti-bacterial (75), anti-inflammatory (76), and anti-tumor (77) activities. Also, cinnamaldehyde, as a potential modulator of dopaminergic neurons in the substantia nigra, has beneficial effects in neurotoxin-induced disorders, including PD (78).
Suppression of NMDA receptors may be relevant to cinnamaldehyde neuroprotective effects against amyloid-β-induced neurotoxicity (79). In a study, the protective effects of cinnamaldehyde (5, 10, 20 μM) on glutamate-induced excitotoxicity were examined in PC12 cells. Glutamate at a concentration of 4 mM caused the generation of ROS and reduced the level of glutathione (GSH) and SOD activity. Pretreatment of the cells with cinnamaldehyde reduced cell death, ROS production, the activity of caspase -3/-9, and the release of cytochrome C, and modulated the expression of the Bcl-2 family (80).
Curcumin
Over the past ten years, the impact of curcumin on various diseases was reported to have hepatoprotective, cardioprotective, anti-carcinogenic, thrombo-suppressive, anti-arthritis, and anti-infective activities (81). Lately, curcumin, as a significant constituent of Curcuma longa, has been reported to have anti-excitotoxicity activity, as well (82). A major enthusiasm has been created due mainly to the lack of toxicity and low cost of curcumin in several preclinical trials for neurodegenerative diseases (83). Several studies have demonstrated that curcumin counteracts the depolarization-evoked release of glutamate by reducing voltage-dependent Ca2+ entering from nerve terminals in rat (84). Also, curcumin protects neurons from excitotoxicity caused by glutamate through the AKAP79-PKA membrane-anchored interaction network and decreasing mGluR5 and NMDA expression (85, 86). Wang et al. have found that curcumin protects the cerebral cortical neurons of rats against glutamate excitotoxicity by increasing brain-derived neurotrophic factor (BDNF) level and activating tropomyosin receptor kinase B (TrkB) (87). Chang et al. have reported that curcumin has protective effects in PC12 cells against the glutamate-induced neurotoxicity through glutathione-dependent nitric oxide- ROS pathway and the mitochondria-dependent nitric oxide- ROS pathway (88). Furthermore, Suh et al. have shown that by blocking MAPK signals, curcumin alleviates glutamate-induced HT22 cell death (89). The results of an encouraging study have shown that curcumin attenuated the glutamate-induced neurotoxicity by inhibiting endoplasmic reticulum stress-associated TXNIP/NLRP3 inflammasome activation in a manner dependent on 5’ AMP-activated protein kinase (90). Hence, curcumin could be a therapeutic agent in the treatment of neurotoxic situations.
Ginkgolide K
Ginkgolide K, as an active compound isolated from leaves of the ginkgo Biloba, inhibits the release of beta-glucuronides from platelet (a platelet-activating factor antagonist) and ameliorates neurotoxicity induced by cerebral ischemia (91, 92). Various doses of ginkgolide K (10, 50, 100 mM) were added to PC12 cells with glutamate (10 mM) for 12 hrs. Glutamate has been shown to increase the concentration of Ca2+ influx, MDA, Bax/Bcl-2 ratio, LDH, caspase-3 protein, the release of cytochrome C, and ROS production. Also, it leads to a decrease in SOD, cell viability, MMP, and GSH peroxidase activity. The outcomes revealed that ginkgolide K attenuated the toxicity of glutamate in PC12 cells by various mechanisms, such as preventing Ca2+ influx, ROS production, and apoptosis reduction (93).
Huperzine A
Huperzine A, a naturally occurring sesquiterpene alkaloid found in Huperzia serrata, has been investigated as a promising treatment for neurological conditions such as AD. The protective effects of huperzine A against oxidative glutamate toxicity in mouse-derived hippocampal HT22 cells were evaluated to explore its probable mechanisms. It was shown that huperzine A attenuated oxidative glutamate excitotoxicity in murine hippocampal HT22 cells via activating the BDNF/TrkB-dependent PI3K/Akt/mTOR signaling pathway (94).
Paeoniflorin
The primary active component of the aqueous extract of Radix Paeoniae alba is paeoniflorin (PF). This ingredient, as a monoterpene glycoside, can enhance memory and learning and has anti-oxidant and sedative properties (95, 96). It has been revealed that PF (0.1, 1.0, and 10 μM) reduce glutamate toxicity in PC12 cells, in a dose-dependent manner. Its protective effect was attributed to the inhibition of apoptosis via regulation of Bax/Bcl-2 signaling and MMP (97). In agreement with this finding, Mao et al. hypothesized that the possible protective mechanisms of PF are through anti-oxidant mechanisms and Ca2+ antagonism in PC12 cells (98). Besides, pretreatment of PC12 cells with PF (100, 200, and 300 μM) inhibited the cytotoxicity of glutamate (15 mM) by increasing Bcl-2, Bcl-xL, reducing Bax and Bad expression, and preventing caspase-3/-9 activity (99). So, PF seems to be a potential natural compound for the treatment of neurodegenerative disorders.
Naringenin
Naringenin, as an anti-oxidant bioflavonoid isolated from Dracocephalum rupestre, is found in a variety of fruits and herbs (100). In a study, the neuroprotective impacts of naringenin on excitotoxicity incited by glutamate in primary hippocampal neurons of neonatal mice were investigated. Naringenin directed ERK1/2 and Akt phosphorylation and decreased the degeneration of dendrites because of glutamate exposure in cultured hippocampal neurons. Besides, naringenin actuated the BDNF and other neuroprotective cytokines and notably improved the survival rates of the neurons 24 hrs following glutamate exposure (101).
Protopanaxadiol
Protopanaxadiol (PPD), characterizing a group of ginsenosides, is found in ginseng (Panax ginseng) and notoginseng (Panax pseudo ginseng). Bak et al. demonstrated the neuroprotective effects of 20(S)-PPD (10 µM) against excitotoxicity induced by glutamate (5 mM) in PC12 cells. PPD has been shown to prevent glutamate-induced apoptosis via improvement of mitochondrial function and anti-oxidant activity (102).
Tanshinone
Tanshinone, isolated from Salvia miltiorrhiza, has been proposed to have cytotoxicity as well as anti-inflammatory and anti-oxidative effects on a variety of cells and modulates breast cancer metastasis by regulating adhesion molecules (103, 104). Tanshinone is divided into three classes: dihydrotanshinone, tanshinone I, or tanshinone IIA. In Li et al. study, it was shown that tanshinone IIA mitigates glutamate-induced oxidative toxicity through the suppression of mitochondrial dysfunction and inhibition of MAPK activation in neuroblastoma cells (105).
Thymoquinone
Thymoquinone, as volatile oil, is abundantly found in Nigella sativa (10). It has different therapeutic effects, such as anti-oxidant, anti-cancer, anti-inflammatory, and neuroprotective actions (106-111). Also, the neuroprotective effect of thymoquinone has been demonstrated against neurotoxic agents such as amyloid-β and α-synuclein peptides (109, 111). The SH-SY5Y cells were exposed to different concentrations of thymoquinone (0.1–3 µM) for 18 hours, and then glutamate (8 mM) was added for 8 hours. Findings have shown that glutamate increased cell death, ROS production, and disturbance of the mitochondrial and apoptosis pathway by increasing caspase-9 and Bax expression, and reducing Bcl-2, while thymoquinone attenuated glutamate toxicity via reduction of ROS generation and apoptosis (112). Besides, in a placebo-controlled clinical trial, Nigella sativa seed capsule (500 mg, twice daily for nine weeks) was shown to improve cognition, memory, and attention in elderly volunteers (113). Hence, Nigella sativa, especially its constituent, Thymoquinone, could be a promising neuromodulator agent for the treatment of neurodegenerative diseases.
The pharmacological mechanisms of the discussed- phytochemicals against glutamate-excitotoxicity is demonstrated in Table 2.
Table 2.
Structure and pharmacological properties of specific phytochemicals against glutamate-induced neurotoxicity
ROS: Reactive oxygen species; MDA: Malondialdehyde; SOD: Superoxide dismutase; ERK: Extracellular signal-regulated kinase; LDH: Lactate dehydrogenase; Akt: Protein kinase B; GSK-3β: Glycogen synthase kinase-3 beta; MAPK: Mitogen-activated protein kinase; NF-κB: Nuclear factor-kappa B; NMDA: N-methyl-D-aspartic acid; mGluR: Metabotropic glutamate receptor; BDNF: Brain-derived neurotrophic factor; :TrkB Tropomyosin receptor kinase B; PI3K: Phosphoinositide 3-kinase; mTOR: Mammalian target of rapamycin
Conclusion
The idea of excitotoxicity as a vital mechanism to comprehend neurotoxicity has made significant progress in determining its role in the pathogenesis of neurodegenerative disorders. It is currently accepted that glutamate is the major excitatory neurotransmitter, and when its level elevates over its threshold value, it leads to excitotoxicity. In this review, we concentrated on different induced neurotoxicities in various studies (in vitro and in vivo) and investigated the protective effects of plants and their constituents on induced toxicity in neural cells. Because of the limited studies available that evaluate their neuroprotective activities, neuroprotective properties of natural compounds, especially curcumin, cinnamaldehyde, thymoquinone, and astaxanthin, need to be discussed more. Most of these promising phytochemicals could play their neuroprotective roles through a reduction in neuronal cell death, restoration of GSH, a decrease in MDA levels, preventing caspases activity, modulation of oxidative stress and protection of neurons against ROS as anti-oxidant activities, reduction of Ca2+, and anti-inflammatory activities. Hence, our research strongly indicates the potential for the use of phytochemicals as pharmacological targets in neurotoxic situations in the future.
Acknowledgment
The authors would like to thank the Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran. The authors received no specific funding for this work.
Conflicts of Interest
The authors declare that there are no conflicts of interest.
References
- 1.Haroon E, Miller AH, Sanacora G. Inflammation, glutamate, and glia: a trio of trouble in mood disorders. Neuropsychopharmacol. 2017;42:193–215. doi: 10.1038/npp.2016.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Obrenovitch T, Urenjak J, Zilkha E, Jay T. Excitotoxicity in neurological disorders-the glutamate paradox. Int J Dev Neurosci. 2000;18:281–287. doi: 10.1016/s0736-5748(99)00096-9. [DOI] [PubMed] [Google Scholar]
- 3.Buckingham SC, Campbell SL, Haas BR, Montana V, Robel S, Ogunrinu T, et al. Glutamate release by primary brain tumors induces epileptic activity. Nat Med. 2011;17:1269–1274. doi: 10.1038/nm.2453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog Neurobiol. 2014;115:157–188. doi: 10.1016/j.pneurobio.2013.11.006. [DOI] [PubMed] [Google Scholar]
- 5.Sontheimer H. A role for glutamate in growth and invasion of primary brain tumors. J Neurochem. 2008;105:287–295. doi: 10.1111/j.1471-4159.2008.05301.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Al-Snafi AE. Therapeutic properties of medicinal plants: a review of medicinal plants with central nervous effects (part 1) Int J of Pharmacology & Toxicol. 2015;5:177–192. [Google Scholar]
- 7.Afshari AR, Sadeghnia HR, Mollazadeh H. A review on potential mechanisms of Terminalia chebula in Alzheimer’s disease. Adv Pharmacol Sci. 2016;2016:8964849. doi: 10.1155/2016/8964849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Boroushaki MT, Mollazadeh H, Afshari AR. Pomegranate seed oil: A comprehensive review on its therapeutic effects. Int J Pharm Sci Rev Res. 2016;7:430–442. [Google Scholar]
- 9.Sadeghnia HR, Jamshidi R, Afshari AR, Mollazadeh H, Forouzanfar F, Rakhshandeh H. Terminalia chebula attenuates quinolinate-induced oxidative PC12 and OLN-93 cell death. Mult Scler Relat Disord. 2017;14:60–67. doi: 10.1016/j.msard.2017.03.012. [DOI] [PubMed] [Google Scholar]
- 10.Mollazadeh H, Afshari AR, Hosseinzadeh H. Review on the potential therapeutic roles of Nigella sativa in the treatment of patients with Cancer: involvement of apoptosis:-Black cumin and cancer. J Pharmacopuncture. 2017;20:158–172. doi: 10.3831/KPI.2017.20.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Afshari AR, Roshan MK, Soukhtanloo M, Ghorbani A, Rahmani F, Jalili-Nik M, et al. Cytotoxic effects of auraptene against a human malignant glioblastoma cell line. Avicenna J Phytomed. 2019;9:334–346. [PMC free article] [PubMed] [Google Scholar]
- 12.Crupi R, Impellizzeri D, Cuzzocrea S. Role of Metabotropic Glutamate Receptors in Neurological Disorders. Front Mol Neurosci. 2019:12. doi: 10.3389/fnmol.2019.00020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Carvalho TG, Alves-Silva J, de Souza JM, Real AL, Doria JG, Vieira EL, et al. Metabotropic glutamate receptor 5 ablation accelerates age-related neurodegeneration and neuroinflammation. Neurochem Int. 2019;126:218–228. doi: 10.1016/j.neuint.2019.03.020. [DOI] [PubMed] [Google Scholar]
- 14.Palazzo E, Neugebauer V, Maione S. Metabotropic Glutamate Receptors and Neurological/Psychiatric Disorders. Front Mol Neurosci. 2019;12 doi: 10.3389/fnmol.2019.00067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lewerenz J, Maher P. Chronic glutamate toxicity in neurodegenerative diseases—what is the evidence? Front Neurosci. 2015;9:469. doi: 10.3389/fnins.2015.00469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wang R, Reddy PH. Role of glutamate and NMDA receptors in Alzheimer’s disease. J Alzheimers Dis. 2017;57:1041–1048. doi: 10.3233/JAD-160763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhang Y, Li P, Feng J, Wu M. Dysfunction of NMDA receptors in Alzheimer’s disease. Neurol Sci. 2016;37:1039–1047. doi: 10.1007/s10072-016-2546-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Nicholls DG, Budd SL. Mitochondria and neuronal survival. Physiol Rev. 2000;80:315–360. doi: 10.1152/physrev.2000.80.1.315. [DOI] [PubMed] [Google Scholar]
- 19.Ward MW, Rego AC, Frenguelli BG, Nicholls DG. Mitochondrial membrane potential and glutamate excitotoxicity in cultured cerebellar granule cells. J Neurosci. 2000;20:7208–7219. doi: 10.1523/JNEUROSCI.20-19-07208.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rehman MU, Wali AF, Ahmad A, Shakeel S, Rasool S, Ali R, et al. Neuroprotective Strategies for Neurological Disorders by Natural Products: An update. Curr Neuropharmacol. 2019;17:247–267. doi: 10.2174/1570159X16666180911124605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hügel HM, Lingham AR, Jackson N, Rook T. Dietary Directions Against Dementia Disorders. Pathology, Prevention and Therapeutics of Neurodegenerative Disease. Springer; 2019. pp. 265–278. [Google Scholar]
- 22.Wang Z, He C, Shi J. Natural Products for the Treatment of Neurodegenerative Diseases. Curr Med Chem. 2019 doi: 10.2174/0929867326666190527120614. [DOI] [PubMed] [Google Scholar]
- 23.Ansari N, Khodagholi F. Natural products as promising drug candidates for the treatment of Alzheimer’s disease: molecular mechanism aspect. Curr Neuropharmacol. 2013;11:414–429. doi: 10.2174/1570159X11311040005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mattson MP. Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol. 2000;1:120–129. doi: 10.1038/35040009. [DOI] [PubMed] [Google Scholar]
- 25.Mattson MP. Glutamate and neurotrophic factors in neuronal plasticity and disease. Ann N Y Acad Sci. 2008;1144:97. doi: 10.1196/annals.1418.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hussein G, Sankawa U, Goto H, Matsumoto K, Watanabe H. Astaxanthin, a carotenoid with potential in human health and nutrition. J Nat Prod. 2006;69:443–449. doi: 10.1021/np050354+. [DOI] [PubMed] [Google Scholar]
- 27.Wu Q, Zhang X-S, Wang H-D, Zhang X, Yu Q, Li W, et al. Astaxanthin activates nuclear factor erythroid-related factor 2 and the antioxidant responsive element (Nrf2-ARE) pathway in the brain after subarachnoid hemorrhage in rats and attenuates early brain injury. Mar Drugs. 2014;12:6125–6141. doi: 10.3390/md12126125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Barros MP, Poppe SC, Bondan EF. Neuroprotective properties of the marine carotenoid astaxanthin and omega-3 fatty acids, and perspectives for the natural combination of both in krill oil. Nutrients. 2014;6:1293–1317. doi: 10.3390/nu6031293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Niwano T, Terazawa S, Nakajima H, Wakabayashi Y, Imokawa G. Astaxanthin and withaferin A block paracrine cytokine interactions between UVB-exposed human keratinocytes and human melanocytes via the attenuation of endothelin-1 secretion and its downstream intracellular signaling. Cytokine. 2015;73:184–197. doi: 10.1016/j.cyto.2015.02.006. [DOI] [PubMed] [Google Scholar]
- 30.Wang H-Q, Sun X-B, Xu Y-X, Zhao H, Zhu Q-Y, Zhu C-Q. Astaxanthin upregulates heme oxygenase-1 expression through ERK1/2 pathway and its protective effect against beta-amyloid-induced cytotoxicity in SH-SY5Y cells. Brain Res. 2010;1360:159–167. doi: 10.1016/j.brainres.2010.08.100. [DOI] [PubMed] [Google Scholar]
- 31.Paw I, Carpenter RC, Watabe K, Debinski W, Lo HW. Mechanisms regulating glioma invasion. Cancer letters. 2015;362:1–7. doi: 10.1016/j.canlet.2015.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Prasansuklab A, Tencomnao T. Acanthus ebracteatus leaf extract provides neuronal cell protection against oxidative stress injury induced by glutamate. BMC Complement Altern Med. 2018;18:278. doi: 10.1186/s12906-018-2340-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yu X, Ceballos YW, Zhao H, Xu Z. Neuroprotective effect of Alpinia oxyphylla extract against glutamate-induced apoptosis in cultured mouse cortical neurons. Neurosci Res Commun. 2003;33:105–103. [Google Scholar]
- 34.Pereira EP, Braga-de-Souza S, Santos CC, Santos LO, Cerqueira MD, Ribeiro PR, et al. Amburana cearensis seed extracts protect PC-12 cells against toxicity induced by glutamate. Rev Bras Farmacogn. 2017;27:199–205. [Google Scholar]
- 35.Lee HY, Weon JB, Ryu G, Yang WS, Kim NY, Kim MK, et al. Neuroprotective effect of Aronia melanocarpa extract against glutamate-induced oxidative stress in HT22 cells. BMC Complement Altern Med. 2017;17:207. doi: 10.1186/s12906-017-1716-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rajabian A, Boroushaki MT, Hayatdavoudi P, Sadeghnia HR. Boswellia serrata protects against glutamate-induced oxidative stress and apoptosis in PC12 and N2a cells. DNA Cell Biol. 2016;35:666–679. doi: 10.1089/dna.2016.3332. [DOI] [PubMed] [Google Scholar]
- 37.Shivasharan B, Nagakannan P, Thippeswamy B, Veerapur V. Protective effect of Calendula officinalis L flowers against monosodium glutamate induced oxidative stress and excitotoxic brain damage in rats. Indian J Clin Biochem. 2013;28:292–298. doi: 10.1007/s12291-012-0256-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hosseini A, Rajabian A. Protective effect of Rheum turkestanikum root against doxorubicin-induced toxicity in H9c2 cells. Revist Brasi Farmacogn. 2016;26:347–351. [Google Scholar]
- 39.Tayeboon GS, Tavakoli F, Hassani S, Khanavi M, Sabzevari O, Ostad SN. Effects of Cymbopogon citratus and Ferula assa-foetida extracts on glutamate-induced neurotoxicity. In Vitro Cell Dev Biol Anim. 2013;49:706–715. doi: 10.1007/s11626-013-9656-7. [DOI] [PubMed] [Google Scholar]
- 40.Sadeghnia HR, Rajabian A, Ghorbani A, Moradzadeh M, Hosseini A. Effects of standardized extract of Ferula gummosa root on glutamate-induced neurotoxicity. Folia Neuropathol. 2017;55:340–346. doi: 10.5114/fn.2017.72399. [DOI] [PubMed] [Google Scholar]
- 41.Li Z, Hu H, Lin R, Mao J, Zhu X, Hong Z, et al. Neuroprotective effects of Gua Lou Gui Zhi decoction against glutamate-induced apoptosis in BV-2 cells. Int J Mol Med. 2014;33:597–604. doi: 10.3892/ijmm.2013.1612. [DOI] [PubMed] [Google Scholar]
- 42.Moon H-I, Lee J-H. Neuroprotective effects of triterpene glycosides from glycine max against glutamate induced toxicity in primary cultured rat cortical cells. Int J Mol Sci. 2012;13:9642–9648. doi: 10.3390/ijms13089642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kim HN, Kim YR, Jang JY, Choi YW, Baek JU, Hong JW, et al. Neuroprotective effects of Polygonum multiflorum extract against glutamate-induced oxidative toxicity in HT22 hippocampal cells. J Ethnopharmacol. 2013;150:108–115. doi: 10.1016/j.jep.2013.08.014. [DOI] [PubMed] [Google Scholar]
- 44.Kim SS, Seo JY, Lim SS, Suh HJ, Kim L, Kim J-S. Neuroprotective effect of Reseda luteola L extract in a mouse neuronal cell model. Food Sci Biotechnol. 2015;24:333–339. [Google Scholar]
- 45.Rajabian A, Sadeghnia H-R, Moradzadeh M, Hosseini A. Rheum turkestanicum reduces glutamate toxicity in PC12 and N2a cell lines. Folia Neuropathol. 2018;56:354–361. doi: 10.5114/fn.2018.80869. [DOI] [PubMed] [Google Scholar]
- 46.Brimson JM, Brimson SJ, Brimson CA, Rakkhitawatthana V, Tencomnao T. Rhinacanthus nasutus extracts prevent glutamate and amyloid-β neurotoxicity in HT-22 mouse hippocampal cells: possible active compounds include lupeol, stigmasterol and β-sitosterol. Int J Mol Sci. 2012;13:5074–5097. doi: 10.3390/ijms13045074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang C-J, Hu C-P, Xu K-P, Yuan Q, Li F-S, Zou H, et al. Protective effect of selaginellin on glutamate-induced cytotoxicity and apoptosis in differentiated PC12 cells Naunyn Schmiedebergs. Arch Pharmacol. 2010;381:73–81. doi: 10.1007/s00210-009-0470-4. [DOI] [PubMed] [Google Scholar]
- 48.Salavati P, Ramezani M, Monsef-Esfahani HR, Hajiagha R, Parsa M, Tavajohi S, et al. Neuroprotective effect of total and sequential extract of Scrophularia striata Boissin rat cerebellar granule neurons following glutamate-induced neurotoxicity: an in-vitro study. Iran J Pharm Res. 2013;12:389–394. [PMC free article] [PubMed] [Google Scholar]
- 49.Mohan M, Gangurde SK, Kadam V. Protective effect of Solanum torvum on monosodium glutamate-induced neurotoxicity in mice. Indian Nat Prod Resour. 2018;8:351–359. [Google Scholar]
- 50.Jang JY, Kim HN, Kim YR, Hong JW, Choi YW, Choi YH et al. Hexane extract from Uncaria sinensis exhibits anti-apoptotic properties against glutamate-induced neurotoxicity in primary cultured cortical neurons. Int J Mol Med. 2012;30:1465–1472. doi: 10.3892/ijmm.2012.1134. [DOI] [PubMed] [Google Scholar]
- 51.Shimada Y, Goto H, Kogure T, Shibahara N, Sakakibara I, Sasaki H, et al. Protective effect of phenolic compounds isolated from the hooks and stems of Uncaria sinensis on glutamate-induced neuronal death. American J Chinese Med. 2001;29:173–180. doi: 10.1142/S0192415X01000198. [DOI] [PubMed] [Google Scholar]
- 52.Kataria H, Wadhwa R, Kaul SC, Kaur G. Water extract from the leaves of Withania somnifera protect RA differentiated C6 and IMR-32 cells against glutamate-induced excitotoxicity. PLoS One. 2012;7:e37080. doi: 10.1371/journal.pone.0037080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang Y, Wang W, Hao C, Mao X, Zhang L. Astaxanthin protects PC12 cells from glutamate-induced neurotoxicity through multiple signaling pathways. J Funct Foods. 2015;16:137–151. [Google Scholar]
- 54.Ahmed T, Abdollahi M, Daglia M, Nabavi SF, Nabavi SM. Berberine and neurodegeneration: A review of literature. Pharmacol Rep. 2015;67:970–979. doi: 10.1016/j.pharep.2015.03.002. [DOI] [PubMed] [Google Scholar]
- 55.Imenshahidi M, Hosseinzadeh H. Berberis vulgaris and berberine: an update review. Phytother Res. 2016;30:1745–1764. doi: 10.1002/ptr.5693. [DOI] [PubMed] [Google Scholar]
- 56.Sarma B, Pandey V, Mishra G, Singh U. Antifungal activity of berberine iodide, a constituent ofFumaria indica. Folia Microbiol. 1999;44:164–166. [Google Scholar]
- 57.Bhutada P, Mundhada Y, Bansod K, Dixit P, Umathe S, Mundhada D. Anticonvulsant activity of berberine, an isoquinoline alkaloid in mice. Epilepsy Behav. 2010;18:207–210. doi: 10.1016/j.yebeh.2010.03.007. [DOI] [PubMed] [Google Scholar]
- 58.Katiyar SK, Meeran SM, Katiyar N, Akhtar S. p53 cooperates berberine-induced growth inhibition and apoptosis of non-small cell human lung cancer cells in vitro and tumor xenograft growth in vivo. Mol Carcinog. 2009;48:24–37. doi: 10.1002/mc.20453. [DOI] [PubMed] [Google Scholar]
- 59.Hayashi K, Minoda K, Nagaoka Y, Hayashi T, Uesato S. Antiviral activity of berberine and related compounds against human cytomegalovirus. Bioorg Med Chem Lett. 2007;17:1562–1564. doi: 10.1016/j.bmcl.2006.12.085. [DOI] [PubMed] [Google Scholar]
- 60.Kuo C-L, Chi C-W, Liu T-Y. The anti-inflammatory potential of berberine in vitro and in vivo. Cancer letters. 2004;203:127–137. doi: 10.1016/j.canlet.2003.09.002. [DOI] [PubMed] [Google Scholar]
- 61.Wang F, Zhao G, Cheng L, Zhou H-Y, Fu L-Y, Yao W-X. Effects of berberine on potassium currents in acutely isolated CA1 pyramidal neurons of rat hippocampus. Brain Res. 2004;999:91–97. doi: 10.1016/j.brainres.2003.11.036. [DOI] [PubMed] [Google Scholar]
- 62.Punitha ISR, Shirwaikar A, Shirwaikar A. Antidiabetic activity of benzyl tetra isoquinoline alkaloid berberine in streptozotocin-nicotinamide induced type 2 diabetic rats. Diabetol Croat. 2005;34:117–128. [Google Scholar]
- 63.Pires ENS, Frozza RL, Hoppe JB, de Melo Menezes B, Salbego CG. Berberine was neuroprotective against an in vitro model of brain ischemia: survival and apoptosis pathways involved. Brain Res. 2014;1557:26–33. doi: 10.1016/j.brainres.2014.02.021. [DOI] [PubMed] [Google Scholar]
- 64.Kulkarni S, Dhir A. Berberine: a plant alkaloid with therapeutic potential for central nervous system disorders. Phytother Res. 2010;24:317–324. doi: 10.1002/ptr.2968. [DOI] [PubMed] [Google Scholar]
- 65.Sadeghnia HR, Kolangikhah M, Asadpour E, Forouzanfar F, Hosseinzadeh H. Berberine protects against glutamate-induced oxidative stress and apoptosis in PC12 and N2a cells. Iran J Basic Med Sci. 2017;20:594. doi: 10.22038/IJBMS.2017.8847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lin TY, Lin YW, Lu CW, Huang SK, Wang SJ. Berberine inhibits the release of glutamate in nerve terminals from rat cerebral cortex. PLoS One. 2013;8:e67215. doi: 10.1371/journal.pone.0067215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Liu X, Zhou J, Abid MDN, Yan H, Huang H, Wan L, et al. Berberine attenuates axonal transport impairment and axonopathy induced by calyculin a in N2a cells. PloS One. 2014;9:e93974. doi: 10.1371/journal.pone.0093974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Moneim AEA. Mercury-induced neurotoxicity and neuroprotective effects of berberine. Neural Regen Res. 2015;10:881–882. doi: 10.4103/1673-5374.158336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yuan NN, Cai CZ, Wu MY, Su HX, Li M, Lu JH. Neuroprotective effects of berberine in animal models of Alzheimer’s disease: a systematic review of pre-clinical studies. BMC Complement Altern Med. 2019;19:109. doi: 10.1186/s12906-019-2510-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Campisi A, Acquaviva R, Mastrojeni S, Raciti G, Vanella A, De Pasquale R, et al. Effect of berberine and Berberis aetnensis C alkaloid extract on glutamate-evoked tissue transglutaminase up-regulation in astroglial cell cultures. Phytother Res. 2011;25:816–820. doi: 10.1002/ptr.3340. [DOI] [PubMed] [Google Scholar]
- 71.Song JH, Kang KS, Choi YK. Protective effect of casuarinin against glutamate-induced apoptosis in HT22 cells through inhibition of oxidative stress-mediated MAPK phosphorylation. Bioorg Med Chem Lett. 2017;27:5109–5113. doi: 10.1016/j.bmcl.2017.10.075. [DOI] [PubMed] [Google Scholar]
- 72.Song JH, Shin MS, Hwang GS, Oh ST, Hwang JJ, Kang KS. Chebulinic acid attenuates glutamate-induced HT22 cell death by inhibiting oxidative stress, calcium influx and MAPKs phosphorylation. Bioorg Med Chem Lett. 2018;28:249–253. doi: 10.1016/j.bmcl.2017.12.062. [DOI] [PubMed] [Google Scholar]
- 73.Wijesekera R. Historical overview of the cinnamon industry. CRC Crit Rev Food Sci Nutr. 1978;10:1–30. doi: 10.1080/10408397809527243. [DOI] [PubMed] [Google Scholar]
- 74.Singh G, Maurya S, Catalan CA. A comparison of chemical, antioxidant and antimicrobial studies of cinnamon leaf and bark volatile oils, oleoresins and their constituents. Food Chem Toxicol. 2007;45:1650–1661. doi: 10.1016/j.fct.2007.02.031. [DOI] [PubMed] [Google Scholar]
- 75.Chang S-T, Chen P-F, Chang S-C. Antibacterial activity of leaf essential oils and their constituents from Cinnamomum osmophloeum. J Ethnopharmacol. 2001;77:123–127. doi: 10.1016/s0378-8741(01)00273-2. [DOI] [PubMed] [Google Scholar]
- 76.Kim DH, Kim CH, Kim M-S, Kim JY, Jung KJ, Chung JH, et al. Suppression of age-related inflammatory NF-κB activation by cinnamaldehyde. Biogerontology. 2007;8:545–554. doi: 10.1007/s10522-007-9098-2. [DOI] [PubMed] [Google Scholar]
- 77.Ka H, Park HJ, Jung HJ, Choi JW, Cho KS, Ha J, et al. Cinnamaldehyde induces apoptosis by ROS-mediated mitochondrial permeability transition in human promyelocytic leukemia HL-60 cells. Cancer Lett. 2003;196:143–152. doi: 10.1016/s0304-3835(03)00238-6. [DOI] [PubMed] [Google Scholar]
- 78.Mehraein F, Zamani M, Negahdar F, Shojaee A. Cinnamaldehyde attenuates dopaminergic neuronal loss in substantia nigra and induces midbrain catalase activity in a mouse model of Parkinson’s disease. J Basic Clin Pathophysiol. 2018;6:9–16. [Google Scholar]
- 79.Emamghoreishi M, Farrokhi MR, Amiri A, Keshavarz M. The neuroprotective mechanism of cinnamaldehyde against amyloid-β in neuronal SHSY5Y cell line: The role of N-methyl-D-aspartate, ryanodine, and adenosine receptors and glycogen synthase kinase-3β. Avicenna J phytomed. 2019;9:271. [PMC free article] [PubMed] [Google Scholar]
- 80.Lv C, Yuan X, Zeng HW, Liu RH, Zhang WD. Protective effect of cinnamaldehyde against glutamate-induced oxidative stress and apoptosis in PC12 cells. Eur J Pharmacol. 2017;815:487–494. doi: 10.1016/j.ejphar.2017.09.009. [DOI] [PubMed] [Google Scholar]
- 81.Shehzad A, Islam SU, Lee YS. Curcumin and Inflammatory Brain Diseases. Curcumin for Neurological and Psychiatric Disorders. Elsevier; 2019. pp. 437–458. [Google Scholar]
- 82.Sundar Dhilip Kumar S, Houreld NN, Abrahamse H. Therapeutic potential and recent advances of curcumin in the treatment of aging-associated diseases. Molecules. 2018;23:835. doi: 10.3390/molecules23040835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rahmani AH, Alsahli MA, Aly SM, Khan MA, Aldebasi YH. Role of curcumin in disease prevention and treatment. Adv Biomed Res. 2018;7:38. doi: 10.4103/abr.abr_147_16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lin TY, Lu CW, Wang C-C, Wang Y-C, Wang S-J. Curcumin inhibits glutamate release in nerve terminals from rat prefrontal cortex: possible relevance to its antidepressant mechanism. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35:1785–1793. doi: 10.1016/j.pnpbp.2011.06.012. [DOI] [PubMed] [Google Scholar]
- 85.Chen K, An Y, Tie L, Pan Y, Li X. Curcumin protects neurons from glutamate-induced excitotoxicity by membrane anchored AKAP79-PKA interaction network. Evid Based Complement Alternat Med. 2015:2015. doi: 10.1155/2015/706207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Khalil RM, Khedr NF. Curcumin protects against monosodium glutamate neurotoxicity and decreasing NMDA2B and mGluR5 expression in rat hippocampus. Neurosignals. 2016;24:81–87. doi: 10.1159/000442614. [DOI] [PubMed] [Google Scholar]
- 87.Wang R, Li Y-B, Li Y-H, Xu Y, Wu H-l, Li X-J. Curcumin protects against glutamate excitotoxicity in rat cerebral cortical neurons by increasing brain-derived neurotrophic factor level and activating TrkB. Brain Res. 2008;1210:84–91. doi: 10.1016/j.brainres.2008.01.104. [DOI] [PubMed] [Google Scholar]
- 88.Chang C-H, Chen H-X, Yü G, Peng C-C, Peng RY. Curcumin-protected PC12 cells against glutamate-induced oxidative toxicity. Food Technol Biotechnol. 2014;52:468–478. doi: 10.17113/ftb.52.04.14.3622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Suh HW, Kang S, Kwon KS. Curcumin attenuates glutamate-induced HT22 cell death by suppressing MAP kinase signaling. Mol Cell Biochem. 2007;298:187–194. doi: 10.1007/s11010-006-9365-6. [DOI] [PubMed] [Google Scholar]
- 90.Li Y, Li J, Li S, Li Y, Wang X, Liu B, et al. Curcumin attenuates glutamate neurotoxicity in the hippocampus by suppression of ER stress-associated TXNIP/NLRP3 inflammasome activation in a manner dependent on AMPK. Toxicol Appl Pharmacol. 2015;286:53–63. doi: 10.1016/j.taap.2015.03.010. [DOI] [PubMed] [Google Scholar]
- 91.Lou F, Ling Y, Tang Y, Wang Y. Isolation, purification and identification of ginkgo terpene lactones. Chin J Nat Med. 2004;2:11–15. [Google Scholar]
- 92.Vitolo O, Gong B, Cao Z, Ishii H, Jaracz S, Nakanishi K, et al. Protection against β-amyloid induced abnormal synaptic function and cell death by Ginkgolide J. Neurobiol Aging. 2009;30:257–265. doi: 10.1016/j.neurobiolaging.2007.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ma S, Liu H, Jiao H, Wang L, Chen L, Liang J, et al. Neuroprotective effect of ginkgolide K on glutamate-induced cytotoxicity in PC 12 cells via inhibition of ROS generation and Ca 2+ influx. Neurotoxicology. 2012;33:59–69. doi: 10.1016/j.neuro.2011.11.003. [DOI] [PubMed] [Google Scholar]
- 94.Mao XY, Zhou HH, Li X, Liu ZQ. Huperzine A alleviates oxidative glutamate toxicity in hippocampal HT22 cells via activating BDNF/TrkB-dependent PI3K/Akt/mTOR signaling pathway. Cell Mol Neurobiol. 2016;36:915–925. doi: 10.1007/s10571-015-0276-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Liu J, Jin DZ, Xiao L, Zhu XZ. Paeoniflorin attenuates chronic cerebral hypoperfusion-induced learning dysfunction and brain damage in rats. Brain Res. 2006;1089:162–170. doi: 10.1016/j.brainres.2006.02.115. [DOI] [PubMed] [Google Scholar]
- 96.Yu HY, Liu MG, Liu DN, Shang GW, Wang Y, Qi C, et al. Antinociceptive effects of systemic paeoniflorin on bee venom-induced various ‘phenotypes’ of nociception and hypersensitivity. Pharmacol Biochem Behav. 2007;88:131–140. doi: 10.1016/j.pbb.2007.07.013. [DOI] [PubMed] [Google Scholar]
- 97.Sun R, Wang K, Wu D, Li X, Ou Y. Protective effect of paeoniflorin against glutamate-induced neurotoxicity in PC12 cells via Bcl-2/Bax signal pathway. Folia Neuropathol. 2012;50:270–276. doi: 10.5114/fn.2012.30527. [DOI] [PubMed] [Google Scholar]
- 98.Mao QQ, Zhong XM, Feng CR, Pan AJ, Li ZY, Huang Z. Protective effects of paeoniflorin against glutamate-induced neurotoxicity in PC12 cells via antioxidant mechanisms and Ca2+ antagonism. Cell Mol Neurobiol. 2010;30:1059–1066. doi: 10.1007/s10571-010-9537-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Chen A, Wang H, Zhang Y, Wang X, Yu L, Xu W, et al. Paeoniflorin exerts neuroprotective effects against glutamateinduced PC12 cellular cytotoxicity by inhibiting apoptosis. Int J Mol Med. 2017;40:825–833. doi: 10.3892/ijmm.2017.3076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Han X, Pan J, Ren D, Cheng Y, Fan P, Lou H. Naringenin-7-O-glucoside protects against doxorubicin-induced toxicity in H9c2 cardiomyocytes by induction of endogenous antioxidant enzymes. Food Chemical Toxicol. 2008;46:3140–3146. doi: 10.1016/j.fct.2008.06.086. [DOI] [PubMed] [Google Scholar]
- 101.Xu XH, Ma CM, Han YZ, Li Y, Liu C, Duan ZH, et al. Protective effect of naringenin on glutamate-induced neurotoxicity in cultured hippocampal cells. Arch Biol Sci. 2015;67:639–646. [Google Scholar]
- 102.Bak D-H, Kim HD, Kim YO, Park CG, Han S-Y, Kim J-J. Neuroprotective effects of 20 (S)-protopanaxadiol against glutamate-induced mitochondrial dysfunction in PC12 cells. Int J Mol Med. 2016;37:378–386. doi: 10.3892/ijmm.2015.2440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Wang X, Wei Y, Yuan S, Liu G, Lu Y, Zhang J, et al. Potential anticancer activity of tanshinone IIA against human breast cancer. Int J Cancer. 2005;116:799–807. doi: 10.1002/ijc.20880. [DOI] [PubMed] [Google Scholar]
- 104.Xu S, Liu P. Tanshinone II-A: new perspectives for old remedies. Taylor & Francis. 2013 doi: 10.1517/13543776.2013.743995. [DOI] [PubMed] [Google Scholar]
- 105.Li H, Han W, Wang H, Ding F, Xiao L, Shi R, et al. Tanshinone IIA inhibits glutamate-induced oxidative toxicity through prevention of mitochondrial dysfunction and suppression of MAPK activation in SH-SY5Y human neuroblastoma cells. Oxid Med Cell Longev. 2017:2017. doi: 10.1155/2017/4517486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Gali-Muhtasib H, Roessner A, Schneider-Stock R. Thymoquinone: a promising anti-cancer drug from natural sources. Int J Biochem Cell Biol. 2006;38:1249–1253. doi: 10.1016/j.biocel.2005.10.009. [DOI] [PubMed] [Google Scholar]
- 107.Mansour MA, Nagi MN, El-Khatib AS, Al-Bekairi AM. Effects of thymoquinone on antioxidant enzyme activities, lipid peroxidation and DT-diaphorase in different tissues of mice: a possible mechanism of action. Cell Biochem Funct. 2002;20:143–151. doi: 10.1002/cbf.968. [DOI] [PubMed] [Google Scholar]
- 108.Woo CC, Kumar AP, Sethi G, Tan KHB. Thymoquinone: potential cure for inflammatory disorders and cancer. Biochem Pharmacol. 2012;83:443–451. doi: 10.1016/j.bcp.2011.09.029. [DOI] [PubMed] [Google Scholar]
- 109.Khan A, Vaibhav K, Javed H, Khan MM, Tabassum R, Ahmed ME, et al. Attenuation of Aβ-induced neurotoxicity by thymoquinone via inhibition of mitochondrial dysfunction and oxidative stress. Mol Cell Biochem. 2012;369:55–65. doi: 10.1007/s11010-012-1368-x. [DOI] [PubMed] [Google Scholar]
- 110.Genrikhs EE, Stelmashook EV, Popova OV, Kapay NA, Korshunova GA, Sumbatyan NV, et al. Mitochondria-targeted antioxidant SkQT1 decreases trauma-induced neurological deficit in rat and prevents amyloid-β-induced impairment of long-term potentiation in rat hippocampal slices. J Drug Target. 2015;23:347–352. doi: 10.3109/1061186X.2014.997736. [DOI] [PubMed] [Google Scholar]
- 111.Alhebshi A, Gotoh M, Suzuki I. Thymoquinone protects cultured rat primary neurons against amyloid β-induced neurotoxicity. Biochem Biophys Res Commun. 2013;433:362–367. doi: 10.1016/j.bbrc.2012.11.139. [DOI] [PubMed] [Google Scholar]
- 112.Al Mamun A, Hashimoto M, Katakura M, Hossain S, Shido O. Neuroprotective effect of thymoquinone against glutamate-induced toxicity in SH-SY5Y cells. Current Topics in Nutraceuticals Research. 2015;13:143. [Google Scholar]
- 113.Sayeed MSB, Asaduzzaman M, Morshed H, Hossain MM, Kadir MF, Rahman MR. The effect of Nigella sativa Linn seed on memory, attention and cognition in healthy human volunteers. J Ethnopharmacol. 2013;148:780–786. doi: 10.1016/j.jep.2013.05.004. [DOI] [PubMed] [Google Scholar]
- 114.Wen X, Huang A, Hu J, Zhong Z, Liu Y, Li Z, et al. Neuroprotective effect of astaxanthin against glutamate-induced cytotoxicity in HT22 cells: Involvement of the Akt/GSK-3β pathway. Neuroscience. 2015;303:558–568. doi: 10.1016/j.neuroscience.2015.07.034. [DOI] [PubMed] [Google Scholar]
- 115.Lin TY, Lu CW, Wang SJ. Astaxanthin inhibits glutamate release in rat cerebral cortex nerve terminals via suppression of voltage-dependent Ca2+ entry and mitogen-activated protein kinase signaling pathway. J Agric Food Chem. 2010;58:8271–8278. doi: 10.1021/jf101689t. [DOI] [PubMed] [Google Scholar]
- 116.Lin X, Zhao Y, Li S. Astaxanthin attenuates glutamate-induced apoptosis via inhibition of calcium influx and endoplasmic reticulum stress. Eur J Pharmacol. 2017;806:43–51. doi: 10.1016/j.ejphar.2017.04.008. [DOI] [PubMed] [Google Scholar]
- 117.Kysenius K, Brunello CA, Huttunen HJ. Mitochondria and NMDA receptor-dependent toxicity of berberine sensitizes neurons to glutamate and rotenone injury. PloS One. 2014;9:e107129. doi: 10.1371/journal.pone.0107129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Biradar SP, Tamboli AS, Khandare RV, Pawar PK. Chebulinic acid and Boeravinone B act as anti-aging and anti-apoptosis phyto-molecules during oxidative stress. Mitochondrion. 2019;46:236–246. doi: 10.1016/j.mito.2018.07.003. [DOI] [PubMed] [Google Scholar]
- 119.Yokoyama T, Ohbuchi T, Saito T, Sudo Y, Fujihara H, Minami K, et al. Allyl isothiocyanates and cinnamaldehyde potentiate miniature excitatory postsynaptic inputs in the supraoptic nucleus in rats. Eur J Pharmacol. 2011;655:31–37. doi: 10.1016/j.ejphar.2011.01.011. [DOI] [PubMed] [Google Scholar]
- 120.Ma S, Liu H, Jiao H, Wang L, Chen L, Liang J, et al. Neuroprotective effect of ginkgolide K on glutamate-induced cytotoxicity in PC 12 cells via inhibition of ROS generation and Ca2+ influx. Neurotoxicology. 2012;33:59–69. doi: 10.1016/j.neuro.2011.11.003. [DOI] [PubMed] [Google Scholar]
- 121.Zhou X, Wang H-Y, Wu B, Cheng C-Y, Xiao W, Wang Z-Z, et al. Ginkgolide K attenuates neuronal injury after ischemic stroke by inhibiting mitochondrial fission and GSK-3β-dependent increases in mitochondrial membrane permeability. Oncotarget. 2017;8:44682. doi: 10.18632/oncotarget.17967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ved HS, Koenig ML, Dave JR, Doctor BP. Huperzine A, a potential therapeutic agent for dementia, reduces neuronal cell death caused by glutamate. Neuroreport. 1997;8:963–967. doi: 10.1097/00001756-199703030-00029. [DOI] [PubMed] [Google Scholar]
- 123.Chen A, Wang H, Zhang Y, Wang X, Yu L, Xu W, et al. Paeoniflorin exerts neuroprotective effects against glutamate-induced PC12 cellular cytotoxicity by inhibiting apoptosis. Int J Mol Med. 2017;40:825–833. doi: 10.3892/ijmm.2017.3076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Lu C, Dong L, Lv J, Wang Y, Fan B, Wang F, et al. 20 (S)-protopanaxadiol (PPD) alleviates scopolamine-induced memory impairment via regulation of cholinergic and antioxidant systems, and expression of Egr-1, c-Fos and c-Jun in mice. Chem Biol Interact. 2018;279:64–72. doi: 10.1016/j.cbi.2017.11.008. [DOI] [PubMed] [Google Scholar]
- 125.Lin TY, Lu CW, Huang SK, Wang S-J. Tanshinone IIA, a constituent of Danshen, inhibits the release of glutamate in rat cerebrocortical nerve terminals. J Ethnopharmacol. 2013;147:488–496. doi: 10.1016/j.jep.2013.03.045. [DOI] [PubMed] [Google Scholar]
- 126.Zhang M, Qian Y. Protection of Tanshinone II A on cellular damage induced by glutamate and β-amyloid protein. Chinese J Gerontol. 2009;29:2465–2468. [Google Scholar]
- 127.Fouad IA, Sharaf NM, Abdelghany RM, El Sayed NSED. Neuromodulatory effect of thymoquinone in attenuating glutamate-mediated neurotoxicity targeting the amyloidogenic and apoptotic pathways. Front Neurol. 2018;9:236. doi: 10.3389/fneur.2018.00236. [DOI] [PMC free article] [PubMed] [Google Scholar]


