Next Article in Journal
Comparative Anti-Infectious Bronchitis Virus (IBV) Activity of (-)-Pinene: Effect on Nucleocapsid (N) Protein
Previous Article in Journal
Convenient Synthesis of 3,4-Dichloro-5-hydroxy-2(5H)-Furanone Glycoconjugates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inhibitors of Microglial Neurotoxicity: Focus on Natural Products

1
Department of Biotechnology, Konkuk University, Chungju 380-701, Korea
2
Department of Pharmacology, Brain Science and Engineering Institute, CMRI, Kyungpook National University School of Medicine, Daegu 700-422, Korea
*
Authors to whom correspondence should be addressed.
Molecules 2011, 16(2), 1021-1043; https://doi.org/10.3390/molecules16021021
Submission received: 7 December 2010 / Revised: 2 January 2011 / Accepted: 17 January 2011 / Published: 25 January 2011
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
Microglial cells play a dual role in the central nervous system as they have both neurotoxic and neuroprotective effects. Uncontrolled and excessive activation of microglia often contributes to inflammation-mediated neurodegeneration. Recently, much attention has been paid to therapeutic strategies aimed at inhibiting neurotoxic microglial activation. Pharmacological inhibitors of microglial activation are emerging as a result of such endeavors. In this review, natural products-based inhibitors of microglial activation will be reviewed. Potential neuroprotective activity of these compounds will also be discussed. Future works should focus on the discovery of novel drug targets that specifically mediate microglial neurotoxicity rather than neuroprotection. Development of new drugs based on these targets may require a better understanding of microglial biology and neuroinflammation at the molecular, cellular, and systems levels.

1. Introduction

Inflammation in the brain and the rest of the central nervous system (CNS) is a key factor in neurodegenerative diseases. Multiple lines of evidence suggest that microglia, the resident immune cells of the CNS, play a critical role in inflammation-mediated neurodegeneration. Normally, microglia cells in their resting state vigilantly monitor the health of neurons and have a benevolent effect. In the event of brain damage or infection, microglia cells become activated and may secrete a variety of inflammatory mediators and neurotoxic factors. In this condition, activated microglia cells trigger and maintain an inflammatory response, deluging neurons with a whole host of inflammatory mediators that may ultimately lead to neuronal cell death. Neurodegenerative CNS diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), tauopathies, and age-related macular degeneration (ARMD), are all associated with chronic neuroinflammation and elevated levels of several cytokines. Microglial activation and chronic inflammation thereafter is the starting point for elevated levels of a wide array of potentially neurotoxic molecules including pro-inflammatory cytokines, proteinases, and reactive oxygen species (ROS) [1,2,3,4,5,6], which are believed to contribute to neurodegenerative processes [7,8]. Several methods have become available for identifying activated microglia, and their presence has been demonstrated in a variety of neuroinflammatory/neurodegenerative diseases such as AD, PD, ALS and multiple sclerosis (MS) [9,10,11]. This participation of activated microglia and the release of neurotoxic products in the demise of neurons have now been postulated in most, if not all, neurodegenerative diseases.
A corollary of neuroinflammation proposes that suppression of microglial production of neurotoxic mediators will result in neuroprotection. This heightens the interest in the rapid discovery of neuroinflammation-targeted therapeutics. Efforts include the isolation of natural products and their active components and the screening of existing CNS drugs approved for other uses, as well as the development of novel synthetic compounds that selectively downregulate neuroinflammatory responses. Here, we review recent literature on natural products-based inhibitors of microglia-mediated neurotoxicity.

2. Natural Products and Their Active Constituents as Inhibitors of Microglia-Mediated Neurotoxicity

In recent years, many herbal plants and their active components have emerged and have been subjects of extensive research. These medications have been validated by traditional usage and are time-tested, as compared to modern day trendy supplements. The traditional herbal medicines with dependable ethnopharmacological properties have recently been demonstrated to possess neurotrophic and neuroprotective abilities, which can be useful in preventing various forms of neuronal cell loss in neurodegenerative and neuroinflammatory diseases. During the last two decades, several ingredients from natural products have been tested for their inhibitory actions on neuroinflammation and used as aids to improve memory, treat neurodegenerative diseases or create favorable effects on the CNS. Numerous botanicals have been shown to provide anti-inflammatory and antioxidant activity, which may protect the brain from inflammatory damage. Increasing evidence suggests that traditional herbal extracts possess neuroprotective benefits through distinct and multiple mechanisms, including anti-inflammation [12,13,14,15,16,17]. Natural compounds specifically aimed at blocking microglial activation may be more efficacious at ameliorating microglia-associated neurodegenerative and neuroinflammatory diseases. In the following sections, we will focus on popular and important natural products and their active constituents as well as their anti-inflammatory activities based on inhibiting microglial activation.

3. Ginsenosides from Panax ginseng

Ginseng, part of the Araliaceae family and species in the genus Panax, is found throughout the world. The name Panax means ‘‘all healing,” which describes the traditional belief that ginseng has properties to heal all aspects of the body. Ginseng is prepared and used in several ways: as fresh ginseng (sliced and eaten, or brewed in a tea), white ginseng (peeled and dried), or red ginseng (peeled, steamed, and dried). Traditional medicine suggests that red ginseng is the most potent, but modern research has shown that all forms have many beneficial properties [18,19,20]. Mechanisms include inhibition of DNA damage [21], induction of cancer cell apoptosis [22], and inhibition of cell proliferation [23]. Ginseng has been proved to possess potent chemotherapeutic effects. Ginseng and its active components significantly decrease several types of cancers in the pharynx, stomach, liver, pancreas, and colon [24,25].
There is abundant evidence that ginseng has potent effects on key players in the inflammatory cascade. Ginseng extracts and total saponins significantly suppress NF-κB and MAP kinase activities, which are upstream signaling molecules in inflammation. The ginsenosides Rh2, Rh3 and compound K significantly inhibit lipopolysaccharide (LPS)-induced inducible nitric oxide synthase (iNOS) and cytokine expressions, thereby proving their beneficial effect in various neuroinflammatory diseases. Another recent study revealed that American ginseng selectively inhibits the expression of iNOS via suppression of the STAT cascade in inflamed macrophages [26]. Ginseng also inhibits the LPS-induced tumor necrosis factor alpha (TNF-α) and other pro-inflammatory cytokines produced by cultured macrophages [27]. Recent studies by Park et al. showed the beneficial effect of ginseng extract and total saponins on microglial activation in co-cultured murine BV-2 microglia and B35 rat neuroblastoma cells [28].
The single compound ginsenoside Rg3 (Figure 1A) inhibited phorbol ester–induced cyclooxygenase-2 (COX-2) and NF-κB induction [29]. Studies also revealed that ginsan, a polysaccharide extracted from P. ginseng, inhibits the p38 MAP kinase pathway and NF-κB in vitro, and the pro-inflammatory cytokines in vivo [30]. BST204, a fermented ginseng extract, can inhibit iNOS expression and subsequent nitric oxide (NO) production from LPS-stimulated RAW264.7 murine macrophages. Ginsenoside Rg3 was also reported to attenuate neuroinflammation in the co-culture of mouse primary dopaminergic neurons and glia. The anti-inflammatory effects of ginsenoside were demonstrated by a reduction in NO formation and PGE2 synthesis and involve interference with iNOS and COX-2 expression, making the molecule useful in treating or preventing various neuroinflammatory diseases including PD [31]. Various forms of ginseng have ubiquitous properties to stop inflammation via microglia-mediated mechanisms. Furthermore, intravenous infusion of ginsenoside derivative Rb1 prevented the ischemic neuronal death, and it passed through blood-brain barrier [32]. This natural herb is therefore one of the most important sources for treating various neuroinflammatory diseases.

4. Curcumin from Curcuma longa

Curcumin, a yellow pigment compound (Figure 1B), in the widely used spice turmeric, exerts anti-inflammatory activity. Curcumin antagonizes many steps in the inflammatory cascade, including activator protein-1 transcription, activation of nuclear factor-κB, iNOS, and JNK [33,34]. Curcumin seems to be relatively safe, even in clinical trials for prevention of relapse of ulcerative colitis [35]. Recent reports indicate that curcumin protects pre-oligodendrocytes from activated microglia in vitro and in vivo. Curcumin significantly inhibits the apoptosis of pre-oligodendrocytes and expression of either iNOS or NOX in the LPS-activated microglia. In in vivo studies, curcumin decreases activated microglia and inhibits microglial expression of iNOS and translocation of p67phox and gp91phox to microglial cell membranes in neonatal rat brains following LPS injection [36]. Studies have also demonstrated the beneficial effects of curcumin on oxidative damage and amyloid β pathology in a transgenic mouse model of AD. The authors reported that low and high doses of curcumin significantly lowered oxidized proteins and interleukin-1 beta (IL-1β), a pro-inflammatory cytokine elevated in thebrains of these mice [37]. In microglia (rat primary microglia and murine BV-2 microglial cells), curcumin effectively suppressed the ganglioside-, LPS-, or IFN-γ-stimulated induction of COX-2 and iNOS, important enzymes that mediate inflammatory processes. The suppressive effect of curcumin is thought to involve regulating the JAK-STAT inflammatory signaling inactivated microglia [38]. A recent report by Wang et al. revealed that curcumin reduces the amyloid-β-stimulated inflammatory responses in primary astrocytes. The deleterious effects of amyloid-β, such as increased expression of COX-2 and glial fibrillary acidic protein and decreased peroxisome proliferator-activated receptor gamma (PPARgamma), were attenuated by pretreatment with curcumin [39].
Accumulating cell culture and animal model data show that curcumin is a strong candidate for use in the prevention or treatment of major disabling age-related neurodegenerative diseases like AD, PD, and stroke [40]. Curcumin administration has been reported to attenuate cognitive deficits, neuroinflammation, and plaque pathology in AD models [41,42,43]. It was also reported that curcumin crosses the blood-brain barrier in an animal model of Alzheimer’s disease when injected peripherally [43]. Further, the outstanding safety profile of curcumin and its pleiotropic actions with potential for neuroprotective efficacy including anti-inflammatory, antioxidant, and anti-protein-aggregate activities achieved at sub-micromolar levels make this compound a potential therapeutic agent in treating neuroinflammatory diseases.

5. Epigallocatechin-3-Gallate (EGCG) from Camellia sinensis

One of the greatest achievements in liquid nutrition has come from green tea, a popular drink made from the dried leaves of Camellia sinensis (L). It is well known that green tea, one of the oldest beverages in the world, has several beneficial effects in many diseases including cardiovascular disorders, obesity and cancer, in addition to slowing down the process of aging. Therefore, the effect of green tea in the prevention and alleviation of neurodegenerative diseases has been of particular interest in recent years.
The most active component of green tea that shows therapeutic effects is (–)-epigallocatechin-3-gallate (EGCG) (Figure 1C). The green tea polyphenol EGCG inhibits the production of numerous inflammatory mediators, including TNF-α, IL-1β and IL-6 [44]. Furthermore, green tea crosses the blood brain barrier, reduces inflammation, provides antioxidant activity, and reduces neural cell death [45]. EGCG is reported to be significantly more potent than other known antioxidants such as vitamin C and vitamin E in scavenging free radicals [46,47,48]. The neuroprotective effect of EGCG could be achieved through complementary mechanisms involving the down-regulation of pro-apoptotic genes [49,50], the influence of amyloid precursor protein (APP) processing by the elevation of α-secretase activity [51], inhibition of β-secretase activity [52], promotion of cell survival, defense against anti-inflammation [53], oxidative stress [54], scavenging of ROS [55,56], iron chelation, and stabilization of mitochondrial function [57].
Aktas and group reported in 2004 that EGCG mediates NF-κB inhibition and exerts neuroprotection in autoimmune encephalomyelitis. The authors suggest that EGCG is capable of protecting against neuronal injury in brain tissue induced by N-methyl-D-aspartate or TRAIL and of directly blocking the formation of neurotoxic ROS in neurons [58]. Another report by Li et al. indicated that EGCG inhibits LPS-induced microglial activation and protects against inflammation-mediated dopaminergic neuronal injury. The authors suggest that EGCG potently inhibits LPS-activated microglial secretion of NO and TNF-α through the down-regulation of iNOS and TNF-α gene expression. In addition, EGCG exerts significant protection against microglial activation-induced neuronal injury both in the human dopaminergic cell line SH-SY5Y and in primary rat mesencephalic cultures [53]. The antioxidative activity of green tea has also been verified in human clinical trials [59]. Based on the above mentioned reports, green tea, in particular EGCG could be a promising candidate for treating or preventing various neurodegenerative and neuroinflammatory diseases.

6. Resveratrol

Resveratrol (trans-3,4,5-trihydroxystilbene), a natural polyphenol (Figure 1D), is a phytoalexin present in grapes and various medicinal plants [60]. Studies have documented that resveratrol exhibits a wide range of biological and pharmacological activities, such as anti-carcinogenesis, cardiovascular protection, and anti-inflammatory effects [61,62]. Resveratrol can attenuate the activation of immune cells and the subsequent synthesis and release of pro-inflammatory mediators through the inhibition of transcriptional factors such as NF-κB and activator protein-1 (AP-1) [63]. Earlier published data revealed that resveratrol potently inhibits the production of NO and TNF-α by LPS-activated microglia and suppresses LPS-induced degradation of I-kappa B alpha (IκB-α), expression of iNOS and phosphorylation of p38 MAPKs in N9 microglial cells [64]. Recently, Meng et al. evaluated the effects of 21 resveratrol derivatives on LPS-induced NO production in microglia and their structure–activity relationships. The authors reported the potent inhibitory effects of selected compounds on microglial activation, suggesting their potential use for treatment of neurodegenerative diseases accompanied by microglial activation [65]. Moreover, the neuroprotective effects of resveratrol have been extensively studied in several cell lines and primary microglial cultures [64,65,66,67]. Resveratrol has been shown to inhibit the activation of microglia and reduce the production of pro-inflammatory factors through intracellular cascades of signaling pathways such as MAPKs, phosphoinositide3-kinase (PI3-K)/Akt, and glycogen synthase kinase-3β (GSK-3β) pathways. Resveratrol was also neuroprotective by inhibiting the activity of NADPH oxidase signaling pathway. In addition, resveratrol can cross the blood–brain barrier and modulate some of the symptoms of debilitating neurological disorders, such as ischemia [68], PD [69], AD [70,71] and Huntington’s disease [72].
Figure 1. The molecular structure of ginsenoside Rg3 (A), curcumin (B), epigallocatechin-3-gallate (C) and resveratrol (D).
Figure 1. The molecular structure of ginsenoside Rg3 (A), curcumin (B), epigallocatechin-3-gallate (C) and resveratrol (D).
Molecules 16 01021 g001
In vitro studies have also proved the beneficial effect of resveratrol in various neuronal and microglial cells such as PC12, SH-SY5Y, BV-2 and N9 cells [73,74,75]. It has been noted that resveratrol prevents both Aβ- and MPP+-induced PC12 cell death by inhibiting ROS production and caspase-3 and Bax activities, and by up-regulating Bcl-2 activity [73,75]. In human neuroblastoma SH-SY5Y cells, excess dopamine-induced cell death could be inhibited by resveratrol through ameliorating intracellular oxidative stress and enhancing the activity of Bcl-2, thus disrupting the apoptotic machinery [76]. Several studies in the N9 microglial cell line have indicated that resveratrol attenuates LPS-induced phosphorylation of p38 MAPK and degradation of IκB-α, thus reducing the production of NO and TNF-α [64,67].
These studies add to our understanding of the potential mechanisms of resveratrol-mediated neuro-protection against various neurotoxicants through its antioxidant and anti-inflammatory properties. Further, as resveratrol crosses the blood–brain barrier and has a good safety profile, this compound can be an ideal candidate for treating neuroinflammatory and neurodegenerative diseases.

7. Gastrodin from Gastrodia Elata

Gastrodia elata Blume (GE) has been used as a traditional herb and has been considered one of the most important medicinal plants in Oriental countries for centuries. GE has been used for a variety of conditions such as treatment of headaches, dizziness, vertigo, and convulsive illnesses such as epilepsy and tetanus [77]. GE reduces oxygen free radicals [78], protects against neuronal damage [79,80,81], and exhibits anxiolytic-like effects via the GABAergic nervous system [82]. GE inhibits NO production and expression of iNOS and COX-2 upon stimulation by LPS in RAW264.7 macrophages [83].
Another study indicated that the ethyl ether fraction of GE dramatically protects neuronal cells from amyloid-β peptide-induced cell death in vitro [38], which is closely associated with AD, and has a protective effect against neuronal damage following global ischemia in gerbils [38]. Methanol extract of GE reduces neuronal nitric oxide synthase activity, microglial activation and apoptosis in the kainic acid-treated rat hippocampus [84]. GE also prevents serum-deprived apoptosis of rat pheochromocytoma cells through activation of the serine/threonine kinase-dependent pathway and suppression of c-Jun NH2-terminal kinase (JNK) [85]. Studies conducted in our lab revealed that GE effectively attenuates cytotoxicity and improves cell viability in MPP+-induced toxicity in SH-SY5Y cells. GE effectively inhibits both the increased production of ROS and the increase in Bax/Bcl-2 ratio, as well as caspase-3 cleavage and PARP proteolysis. The protective effect may be ascribed to its significant anti-oxidative and anti-apoptotic properties [79].
The major active components in GE are phenolic compounds, gastrodin, p-hydroxybenzyl alcohol, vanillyl alcohol, 4-hydroxybenzaldehyde, vanillin, succinic acid, organic acids, glucose, and β-sitosterol [78,86,87,88]. These components play various roles in the treatment of diseases. Recently, GE was reported as an important medical food, and some of the active constituents possess anti-atherosclerotic properties [89,90], anti-angiogenic activity [83], and anti-tumor activity [91]. Its beneficial effects in the CNS were also documented [80,82,92,93,94,95,96].
Regarding its neuroprotective effects, gastrodin (Figure 2A) exerts beneficial actions making it useful for treatment of dizziness, epilepsy, stroke and dementia [78,97]. Gastrodin and para-hydroxybenzyl alcohol facilitate memory consolidation and retrieval as shown in the passive avoidance task test in rats [92]. The aqueous extract of GE improves D-galaxies-induced memory impairment in mice and performance on a step-down, passive-avoidance task in senescent mice. Based on these reports, GE could be further developed as a potential therapeutic candidate for treating microglia activation-mediated neuroinflammatory and neurodegenerative diseases.

8. Gingerol from Zingiber officinale

Ginger, the rhizome of the plant Zingiber officinale, has a long history of medicinal use. In traditional oriental medicine, ginger has been used to treat a wide range of ailments including stomach aches, diarrhea, nausea, asthma, respiratory disorders, toothache, gingivitis, and arthritis [98,99,100]. Several studies have shown that ginger inhibits pro-inflammatory cytokines, including IL-1β, IL-2, IL-12, TNF-α, and interferon (IFN)-gamma [101]. Ginger also has been shown to decrease synthesis of pro-inflammatory prostaglandins and leukotrienes via inhibition of COX-2 and 5-lipoxygenase (5-LOX) enzymes, which are the targets for numerous anti-inflammatory pharmaceuticals.
Grzanna et al. tested the effects of a ginger extract on THP-1 monocytic cells to determine whether it can block the induction of pro-inflammatory cytokines in these cells stimulated with LPS. The results of this study suggest that the anti-inflammatory properties of the ginger extract may provide beneficial effects similar to those of currently used COX inhibitors [102].
Recently, Jung et al. reported that the hexane fraction of Zingiberis Rhizoma Crudus extract inhibits the production of nitric oxide and pro-inflammatory cytokines in LPS-stimulated BV-2 microglial cells via the NF-κB pathway [103]. The authors indicated that ginger hexane extract significantly inhibited the excessive production of NO, PGE2, TNF-α, and IL-1β in LPS-stimulated BV-2 cells. Ginger extract also attenuated the mRNA expressions and protein levels of iNOS, COX-2, and pro-inflammatory cytokines. The molecular mechanisms that underlie ginger hexane extract-mediated attenuation of neuroinflammation were related to the inhibition of the phosphorylation of three mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinases 1 and 2 (ERK1/2), p38 MAPK, and c-Jun N-terminal kinase (JNK), and the activation of NF-κB [103].
6-Gingerol (Figure 2B), one of the active ingredients of ginger, has been reported to impart ginger with its anti-inflammatory properties. The 6-gingerol inhibited the production of pro-inflammatory cytokines from LPS-stimulated macrophages, and inhibited COX-2 expression by blocking the activation of p38 MAP kinase and NF-κB in phorbol ester-stimulated mouse skin [104,105]. Data indicate that several doses of 6-gingerol selectively inhibit production of pro-inflammatory cytokines such as TNF-α, IL-1, and IL-12 by murine peritoneal macrophages in the presence of LPS stimulation. The authors also revealed that 6-gingerol does not affect antigen presenting cell (APC) function or cell surface expression of MHC II and co-stimulatory molecules [105]. These remarkable beneficial properties of ginger and 6-gingerol and the lack of gastrointestinal and renal side effects distinguish it from other NSAIDS. Considering the broad spectrum of ginger’s anti-inflammatory actions and its safety record in clinical trials, it is likely to be a valuable dietary supplement in the treatment of neurodegenerative and neuroinflammatory diseases. However, the ability of gingerol to cross blood-brain barrier has not yet been explicitly demonstrated and needs further investigation.

9. Obovatol from Magnolia Obovata

Obovatol is the main component of leaves of Magnolia obovata, a medicinal plant that has long been used as a folk remedy in East Asia. Previous studies showed various biological activities of obovatol such as anti-bacterial, anti-tumor, and anti-platelet effects [106,107,108]. The anti-inflammatory activity of obovatol was also reported in LPS-stimulated mouse macrophage RAW264.7 cells [109]. With respect to CNS-related activity, obovatol reportedly has anxiolytic effects that are mediated by GABA-benzodiazepine receptors-activated Cl- channel opening [110]. Most recently, the effects of obovatol on microglial activation and neuroinflammation were examined using cultured cells and a mouse model of neuroinflammation [111]. The study showed that obovatol inhibited inflammatory activation of microglia in vitro and neuroinflammation in vivo, and the compound exerted protective effects against microglia-mediated neurotoxicity. However, no direct evidence regarding its ability to cross blood-brain barrier has been reported. Based on the affinity chromatography followed by liquid chromatography and tandem mass spectrometric analysis, the authors identified peroxiredoxin 2 (Prx2) as the molecular target of obovatol, implicating microglial redox regulation in the inflammatory neurodegeneration. The report also suggested that Prx2 is a novel drug target that can be exploited for the therapeutic modulation of microglia activation and neuroinflammation.
Figure 2. The molecular structure of gastrodin (A), 6-gingerol (B) and inflexin (C).
Figure 2. The molecular structure of gastrodin (A), 6-gingerol (B) and inflexin (C).
Molecules 16 01021 g002

10. Other Natural Products and Components

Inflexin is a compound derived from the natural plant Isodon excisus (Max.) Kudo (Labiateae), which is a perennial herb distributed widely in Korea, China, and Japan (Figure 2C). The aerial parts of this plant have been used for detoxification and treatment of gastrointestinal disorders [112], anorexia, indigestion, stomach ache, inflammation, and esophageal carcinoma [113]. Studies from our lab and others revealed that inflexin has the potential to inhibit LPS-induced NF-κB activation in RAW264.7 macrophage cells [114] and in LPS-treated BV-2 microglial cells [115]. In the later study from our laboratory, inflexin significantly inhibited the release of nitric oxide (NO). Both the mRNA and the protein levels of iNOS were decreased in a concentration-dependent manner. Inflexin also inhibited the expression of COX-2, and effectively reduced the LPS-induced expression of pro-inflammatory cytokines in a dose-dependent manner. Furthermore, inflexin inhibited the degradation of IκB-α and the activation of NF-κB p65 subunit and Akt [115]. Due to its potent beneficial action in the inhibition of microglial activation, this compound could give rise to stable derivatives for the treatment of microglia-mediated neuroinflammation and could be developed as a potent therapeutic agent in treating various neuroinflammatory diseases.
Piper kadsura Ohwi (Piperaceae) is a medicinal vine-like plant distributed in the coastal forest regions of Korea, Japan, Taiwan, and China where it covers rocks and trees. The stem part of this plant, known as haifengteng, is widely used in the herbal medicinal prescriptions for the treatment of asthma and arthritic conditions. Recently, the neolignans (Figure 3A and Figure 3B) isolated from Piper kadsura were shown to possess strong anti-neuroinflammatory activity as demonstrated by a reduction in nitric oxide (NO) production in LPS-activated BV-2 microglial cells [116].
Another natural product Ganoderma lucidum (GL) protects against dopaminergic neuron degeneration through inhibition of microglial activation. GL extracts significantly prevent the production of microglia-derived pro-inflammatory and cytotoxic factors (nitric oxide, TNF-α, and IL-1β), and down-regulate the TNF-α and IL-1β gene expressions at the mRNA level as well, indicating that GL may be a promising agent for the treatment of neuroinflammatory diseases [117].
Blueberries are flowering plants of the genus Vaccinium. Blueberries have a diverse range of micronutrients, with notably high levels of the essential dietary mineral manganese, vitamin B6, vitamin C, vitamin K and dietary fiber. Blueberries contain anthocyanins, other antioxidant pigments, and various phytochemicals that may reduce the risks of inflammatory disease and certain cancers [118,119]. Recently, blueberry has been reported to oppose β-amyloid peptide-induced microglial activation via inhibition of p44/42 mitogen-activation protein kinase in murine primary microglial culture. Also, blueberry significantly enhances microglial clearance of Aβ1–42, inhibits aggregation of Aβ1–42, and suppresses microglial activation, all via suppression of the p44/42 MAPK pathway [120].
Berberine (Figure 3C), one of the major constituents of the Chinese herb Rhizoma coptidis, suppresses neuroinflammatory responses through AMP-activated protein kinase activation in BV-2 microglial cells [121].
Epimedium brevicornum Maxim (Berberidaceae) is an important traditional herbal medicine originally used in ancient China for its tonic effect. Icariin (ICA, C33H40O15, MW: 676.65; Figure 3D), with the chemical name 2-(4′-methoxylphenyl)-3-rhamnosido-5-hydroxyl-7-glucosido-8-(3′-methyl-2-butylenyl)-4-chromanone, is a major component isolated from E. brevicornum. As a highly interesting natural flavonoid compound for drug development, icariin has a broad spectrum of established pharmacological functions, including antioxidant [122], immunoregulatory [123], and antidepressant-like effects [23], stimulation of angiogenesis [124], and induction of cardiomyocyte differentiation [125]. Recent studies have reported that icariin inhibits the inflammatory response by decreasing the production of TNF-α, IL-6, NO and adhesion molecules (CD11b) both in the RAW264.7 macrophage cell line and in mouse serum [126]. In addition, icariin also exhibits an anti-inflammatory effect on LPS-treated murine chondrocytes through inhibition of NO and MMP synthesis [127]. A recent report by Zheng et al. revealed that Icariin attenuates LPS-induced microglial activation by inhibiting TAK1/IKK/NF-κB and JNK/p38 MAPK pathways. These findings provide mechanistic insights into the suppressive effect of icariin on LPS-induced neuroinflammatory response in microglia, and emphasize the neuroprotective effect and therapeutic potential of icariin in neuroinflammatory diseases [128].
Figure 3. The molecular structure of neolignans (A and B), berberine (C) and icariin (D).
Figure 3. The molecular structure of neolignans (A and B), berberine (C) and icariin (D).
Molecules 16 01021 g003
Isodon japonicus (Burm.) Hara (Lamiaceae) is a perennial plant with wide distribution in Korea, China, and Japan. The aerial parts of I. japonicus have commonly been used as traditional folk medicines for treatment of gastrointestinal disorders, tumors, and inflammatory disease [129]. Sequential column chromatography of I. japonicus has allowed isolation of a new ent-kaurane type diterpene, isodojaponin D (19-hydroxy-1α, 6-diacetoxy- 6, 7-seco-ent-kaur-16-en-15-one-7, 20-olide; Figure 4A) [130]. Earlier reports indicated that isodojaponin D has neuroprotective effects against β-amyloid-induced toxicity [116]. Recently, isodojaponin D was reported to inhibit LPS-induced microglial activation [131]. Isodojaponin D significantly decreased the LPS-induced production of COX-2 and iNOS. In addition, LPS-induced pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α, were also decreased through NF-κB and MAPK signaling pathways. The reports suggest that isodojaponin D could play a beneficial role in the treatment of neurodegenerative and neuro-inflammatory diseases.
Furthermore, the anti-inflammatory constituents tetrandrine and fangchinoline (Figure 4B) found in Stephania tetrandra have been shown to decrease IL-1β, IL-6, IL-8 and TNF-α [132], and to decrease leukotriene and prostaglandin generation [133]. More importantly, tetrandrine has been shown to inhibit the production of TNF-α and IL-6 by microglial cells [134], which damage nerve cells. Similarly, Urtica dioica, also known as “stinging nettle”, reduces IL-1β, IL-2, IFN- , and TNF-α [135,136]. Supplementation of stinging nettle in humans has been shown to decrease LPS induction of inflammatory mediators, triggering an 80% reduction in TNF-α and a 99% reduction in IL-1β [137].
Several flavonoids derived from plant origin such as wogonin, tectorigenin, tectoridin, apigenin, luteolin, and fisetin modulate microglial activation at the cellular level and exert their anti-neuroinflammatory properties, suggesting their importance as a potent therapeutic agent for treating neuroinflammatory diseases [14,138,139,140,141,142]. In particular, fisetin (3,3,4,7-tetrahydroxy flavone), a naturally occurring flavonoid commonly found in strawberries and other fruits and vegetables, inhibits neurotoxic microglial activation. Studies have also demonstrated that fisetin exhibits a wide variety of biological activities, including anti-cancer [143], neurotrophic [144], anti-oxidant [145], and anti-inflammatory effects in mast cells [146]. Along with the anti-inflammatory effects of fisetin in peripheral mast cells, its anti-inflammatory effect in brain microglia has recently been investigated [147]. Fisetin inhibits the production of inflammatory mediators, and suppresses NF-κB and p38 mitogen-activated protein kinase activation in LPS-stimulated mouse microglia cells. In addition, the flavonoid exhibits a neuroprotective effect by attenuating microglial neurotoxicity in the microglia-neuron co-culture model.
Figure 4. The molecular structure of isodojaponin D (A), fangchinoline and tetrandrine (B).
Figure 4. The molecular structure of isodojaponin D (A), fangchinoline and tetrandrine (B).
Molecules 16 01021 g004

11. Conclusions

A growing body of evidence indicates that neuroinflammation may contribute to the neurodegenerative process. It is believed that activated microglia cells, which compose the majority of this inflammatory response, contribute to the neurodegenerative process. Suppression of microglia activation may provide an effective therapeutic intervention that alleviates the progression of the neurodegenerative diseases. Recently, the natural products and their components have received considerable attention as alternative candidates for therapeutic purposes. They have a reputation for being safe, inexpensive, and readily available. However, only a few substances have been studied in depth, and much of the work is in the preliminary stages. It remains be adequately studied clinically whether the therapeutic effects indicated by the experimental settings are similar in humans. If these substances have benefits for humans that are reflected in the laboratory studies, then moderate doses consumed regularly over years might be able to prevent or at least slow the development of the disease. Continued investigation of the mechanisms underlying microglial activation, regulation of neuroinflammation, and the modulatory role of natural products and their components could not only lead to the discovery of novel neuroprotective agents, but also help us to understand the complex pathophysiology of neurodegenerative diseases.

Acknowledgements

We apologize to authors whose works are not cited here due to space limitations. This research was financially supported by the Ministry of Education, Science Technology (MEST) and Korea Institute for Advancement of Technology (KIAT) through the Human Resource Training Project for Regional Innovation. This work was also supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the MEST (2010-0029460).

References

  1. Boje, K.M.; Arora, P.K. Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death. Brain Res. 1992, 587, 250–256. [Google Scholar] [CrossRef]
  2. Chao, C.C.; Hu, S.; Ehrlich, L.; Peterson, P.K. Interleukin-1 and tumor necrosis factor-alpha synergistically mediate neurotoxicity: involvement of nitric oxide and of N-methyl-D-aspartate receptors. Brain Behav. Immun. 1995, 9, 355–365. [Google Scholar] [CrossRef]
  3. Chao, C.C.; Hu, S.; Molitor, T.W.; Shaskan, E.G.; Peterson, P.K. Activated microglia mediate neuronal cell injury via a nitric oxide mechanism. J. Immunol. 1992, 149, 2736–2741. [Google Scholar]
  4. Jeohn, G.H.; Kong, L.Y.; Wilson, B.; Hudson, P.; Hong, J.S. Synergistic neurotoxic effects of combined treatments with cytokines in murine primary mixed neuron/glia cultures. J. Neuroimmunol. 1998, 85, 1–10. [Google Scholar] [CrossRef]
  5. McGuire, S.O.; Ling, Z.D.; Lipton, J.W.; Sortwell, C.E.; Collier, T.J.; Carvey, P.M. Tumor necrosis factor alpha is toxic to embryonic mesencephalic dopamine neurons. Exp. Neurol. 2001, 169, 219–230. [Google Scholar] [CrossRef]
  6. Xie, Z.; Wei, M.; Morgan, T.E.; Fabrizio, P.; Han, D.; Finch, C.E.; Longo, V.D. Peroxynitrite mediates neurotoxicity of amyloid beta-peptide1-42- and lipopolysaccharide-activated microglia. J. Neurosci. 2002, 22, 3484–3492. [Google Scholar]
  7. Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef]
  8. Ransohoff, R.M.; Perry, V.H. Microglial physiology: unique stimuli, specialized responses. Annu. Rev. Immunol. 2009, 27, 119–145. [Google Scholar] [CrossRef]
  9. Cagnin, A.; Myers, R.; Gunn, R.N.; Lawrence, A.D.; Stevens, T.; Kreutzberg, G.W.; Jones, T.; Banati, R.B. In vivo visualization of activated glia by [11C] (R)-PK11195-PET following herpes encephalitis reveals projected neuronal damage beyond the primary focal lesion. Brain 2001, 124, 2014–2027. [Google Scholar] [CrossRef]
  10. Chen, M.K.; Baidoo, K.; Verina, T.; Guilarte, T.R. Peripheral benzodiazepine receptor imaging in CNS demyelination: functional implications of anatomical and cellular localization. Brain 2004, 127, 1379–1392. [Google Scholar] [CrossRef]
  11. Turner, M.R.; Cagnin, A.; Turkheimer, F.E.; Miller, C.C.; Shaw, C.E.; Brooks, D.J.; Leigh, P.N.; Banati, R.B. Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: an [11C](R)-PK11195 positron emission tomography study. Neurobiol. Dis. 2004, 15, 601–609. [Google Scholar] [CrossRef]
  12. Chen, L.W.; Wang, Y.Q.; Wei, L.C.; Shi, M.; Chan, Y.S. Chinese herbs and herbal extracts for neuroprotection of dopaminergic neurons and potential therapeutic treatment of Parkinson's disease. CNS Neurol. Disord. Drug Targets 2007, 6, 273–281. [Google Scholar] [CrossRef]
  13. Ho, Y.S.; Yu, M.S.; Lai, C.S.; So, K.F.; Yuen, W.H.; Chang, R.C. Characterizing the neuroprotective effects of alkaline extract of Lycium barbarum on beta-amyloid peptide neurotoxicity. Brain Res. 2007, 1158, 123–134. [Google Scholar] [CrossRef]
  14. Lee, H.; Kim, Y.O.; Kim, H.; Kim, S.Y.; Noh, H.S.; Kang, S.S.; Cho, G.J.; Choi, W.S.; Suk, K. Flavonoid wogonin from medicinal herb is neuroprotective by inhibiting inflammatory activation of microglia. FASEB J. 2003, 17, 1943–1944. [Google Scholar]
  15. Li, F.Q.; Wang, T.; Pei, Z.; Liu, B.; Hong, J.S. Inhibition of microglial activation by the herbal flavonoid baicalein attenuates inflammation-mediated degeneration of dopaminergic neurons. J. Neural Transm. 2005, 112, 331–347. [Google Scholar] [CrossRef]
  16. Shimazawa, M.; Chikamatsu, S.; Morimoto, N.; Mishima, S.; Nagai, H.; Hara, H. Neuroprotection by Brazilian Green Propolis against In vitro and In vivo Ischemic Neuronal Damage. Evid. Based Complement. Alternat. Med. 2005, 2, 201–207. [Google Scholar] [CrossRef]
  17. Suk, K.; Lee, H.; Kang, S.S.; Cho, G.J.; Choi, W.S. Flavonoid baicalein attenuates activation-induced cell death of brain microglia. J. Pharmacol. Exp. Ther. 2003, 305, 638–645. [Google Scholar] [CrossRef]
  18. Corbit, R.M.; Ferreira, J.F.; Ebbs, S.D.; Murphy, L.L. Simplified extraction of ginsenosides from American ginseng (Panax quinquefolius L.) for high-performance liquid chromatography-ultraviolet analysis. J. Agric. Food Chem. 2005, 53, 9867–9873. [Google Scholar] [CrossRef]
  19. Kitts, D.D.; Wijewickreme, A.N.; Hu, C. Antioxidant properties of a North American ginseng extract. Mol. Cell. Biochem. 2000, 203, 1–10. [Google Scholar] [CrossRef]
  20. Yun, T.K. Panax ginseng--a non-organ-specific cancer preventive? Lancet Oncol. 2001, 2, 49–55. [Google Scholar] [CrossRef]
  21. Park, S.; Yeo, M.; Jin, J.H.; Lee, K.M.; Jung, J.Y.; Choue, R.; Cho, S.W.; Hahm, K.B. Rescue of Helicobacter pylori-induced cytotoxicity by red ginseng. Dig. Dis. Sci. 2005, 50, 1218–1227. [Google Scholar] [CrossRef]
  22. Volate, S.R.; Davenport, D.M.; Muga, S.J.; Wargovich, M.J. Modulation of aberrant crypt foci and apoptosis by dietary herbal supplements (quercetin, curcumin, silymarin, ginseng and rutin). Carcinogenesis 2005, 26, 1450–1456. [Google Scholar] [CrossRef]
  23. Kang, K.A.; Kim, Y.W.; Kim, S.U.; Chae, S.; Koh, Y.S.; Kim, H.S.; Choo, M.K.; Kim, D.H.; Hyun, J.W. G1 phase arrest of the cell cycle by a ginseng metabolite, compound K, in U937 human monocytic leukamia cells. Arch. Pharm. Res. 2005, 28, 685–690. [Google Scholar] [CrossRef]
  24. Kakizoe, T. Asian studies of cancer chemoprevention: latest clinical results. Eur. J. Cancer 2000, 36, 1303–1309. [Google Scholar] [CrossRef]
  25. Shin, H.R.; Kim, J.Y.; Yun, T.K.; Morgan, G.; Vainio, H. The cancer-preventive potential of Panax ginseng: a review of human and experimental evidence. Cancer Causes Control 2000, 11, 565–576. [Google Scholar] [CrossRef]
  26. Ichikawa, T.; Li, J.; Nagarkatti, P.; Nagarkatti, M.; Hofseth, L.J.; Windust, A.; Cui, T. American ginseng preferentially suppresses STAT/iNOS signaling in activated macrophages. J. Ethnopharmacol. 2009, 125, 145–150. [Google Scholar] [CrossRef]
  27. Rhule, A.; Navarro, S.; Smith, J.R.; Shepherd, D.M. Panax notoginseng attenuates LPS-induced pro-inflammatory mediators in RAW264.7 cells. J. Ethnopharmacol. 2006, 106, 121–128. [Google Scholar] [CrossRef]
  28. Park, J.S.; Park, E.M.; Kim, D.H.; Jung, K.; Jung, J.S.; Lee, E.J.; Hyun, J.W.; Kang, J.L.; Kim, H.S. Anti-inflammatory mechanism of ginseng saponins in activated microglia. J. Neuroimmunol. 2009, 209, 40–49. [Google Scholar] [CrossRef]
  29. Keum, Y.S.; Han, S.S.; Chun, K.S.; Park, K.K.; Park, J.H.; Lee, S.K.; Surh, Y.J. Inhibitory effects of the ginsenoside Rg3 on phorbol ester-induced cyclooxygenase-2 expression, NF-kappaB activation and tumor promotion. Mutat. Res. 2003, 523-524, 75–85. [Google Scholar] [CrossRef]
  30. Ahn, J.Y.; Choi, I.S.; Shim, J.Y.; Yun, E.K.; Yun, Y.S.; Jeong, G.; Song, J.Y. The immunomodulator ginsan induces resistance to experimental sepsis by inhibiting Toll-like receptor-mediated inflammatory signals. Eur. J. Immunol. 2006, 36, 37–45. [Google Scholar] [CrossRef]
  31. Lin, W.M.; Zhang, Y.M.; Moldzio, R.; Rausch, W.D. Ginsenoside Rd attenuates neuroinflammation of dopaminergic cells in culture. J. Neural Transm. Suppl. 2007, 105–112. [Google Scholar]
  32. Zhang, B.; Hata, R.; Zhu, P.; Sato, K.; Wen, T.C.; Yang, L.; Fujita, H.; Mitsuda, N.; Tanaka, J.; Samukawa, K.; Maeda, N.; Sakanaka, M. Prevention of ischemic neuronal death by intravenous infusion of a ginseng saponin, ginsenoside Rb(1), that upregulates Bcl-x(L) expression. J. Cereb. Blood Flow Metab. 2006, 26, 708–721. [Google Scholar] [CrossRef]
  33. Pendurthi, U.R.; Williams, J.T.; Rao, L.V. Inhibition of tissue factor gene activation in cultured endothelial cells by curcumin. Suppression of activation of transcription factors Egr-1, AP-1, and NF-kappa B. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 3406–3413. [Google Scholar] [CrossRef]
  34. Weber, W.M.; Hunsaker, L.A.; Gonzales, A.M.; Heynekamp, J.J.; Orlando, R.A.; Deck, L.M.; Vander Jagt, D.L. TPA-induced up-regulation of activator protein-1 can be inhibited or enhanced by analogs of the natural product curcumin. Biochem. Pharmacol. 2006, 72, 928–940. [Google Scholar] [CrossRef]
  35. Hanai, H.; Iida, T.; Takeuchi, K.; Watanabe, F.; Maruyama, Y.; Andoh, A.; Tsujikawa, T.; Fujiyama, Y.; Mitsuyama, K.; Sata, M.; Yamada, M.; Iwaoka, Y.; Kanke, K.; Hiraishi, H.; Hirayama, K.; Arai, H.; Yoshii, S.; Uchijima, M.; Nagata, T.; Koide, Y. Curcumin maintenance therapy for ulcerative colitis: randomized, multicenter, double-blind, placebo-controlled trial. Clin. Gastroenterol. Hepatol. 2006, 4, 1502–1506. [Google Scholar] [CrossRef]
  36. He, L.F.; Chen, H.J.; Qian, L.H.; Chen, G.Y.; Buzby, J.S. Curcumin protects pre-oligodendrocytes from activated microglia in vitro and in vivo. Brain Res. 2010, 1339, 60–69. [Google Scholar] [CrossRef]
  37. Lim, G.P.; Chu, T.; Yang, F.; Beech, W.; Frautschy, S.A.; Cole, G.M. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J. Neurosci. 2001, 21, 8370–8377. [Google Scholar]
  38. Kim, H.J.; Moon, K.D.; Lee, D.S.; Lee, S.H. Ethyl ether fraction of Gastrodia elata Blume protects amyloid beta peptide-induced cell death. J. Ethnopharmacol. 2003, 84, 95–98. [Google Scholar] [CrossRef]
  39. Wang, H.M.; Zhao, Y.X.; Zhang, S.; Liu, G.D.; Kang, W.Y.; Tang, H.D.; Ding, J.Q.; Chen, S.D. PPARgamma agonist curcumin reduces the amyloid-beta-stimulated inflammatory responses in primary astrocytes. J. Alzheimers Dis. 2010, 20, 1189–1199. [Google Scholar]
  40. Cole, G.M.; Teter, B.; Frautschy, S.A. Neuroprotective effects of curcumin. Adv. Exp. Med. Biol. 2007, 595, 197–212. [Google Scholar] [CrossRef]
  41. Frautschy, S.A.; Hu, W.; Kim, P.; Miller, S.A.; Chu, T.; Harris-White, M.E.; Cole, G.M. Phenolic anti-inflammatory antioxidant reversal of Abeta-induced cognitive deficits and neuropathology. Neurobiol. Aging 2001, 22, 993–1005. [Google Scholar] [CrossRef]
  42. Garcia-Alloz, M.; Dodwell, L.; Borelli, A.; Raju, S.; Backskai, B. In vivo reduction of plaque size in APPswe/PS1D9 mice treated with curcumin. Alzheimer's Dementia 2006, 2, S617. [Google Scholar]
  43. Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; Cole, G.M. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar]
  44. Neyestani, T.R.; Gharavi, A.; Kalayi, A. Selective effects of tea extract and its phenolic compounds on human peripheral blood mononuclear cell cytokine secretions. Int. J. Food Sci. Nutr. 2009, 60 (Suppl. 1), 79–88. [Google Scholar]
  45. Mandel, S.A.; Avramovich-Tirosh, Y.; Reznichenko, L.; Zheng, H.; Weinreb, O.; Amit, T.; Youdim, M.B. Multifunctional activities of green tea catechins in neuroprotection. Modulation of cell survival genes, iron-dependent oxidative stress and PKC signaling pathway. Neurosignals 2005, 14, 46–60. [Google Scholar] [CrossRef]
  46. Bazinet, L.; Labb, D.; Tremblay, A. Production of green tea EGC- and EGCG-enriched fractions by a two-step extraction procedure. Separation Purification Technol. 2007, 56, 53–56. [Google Scholar] [CrossRef]
  47. Hu, B.; Wang, L.; Zhou, B.; Zhang, X.; Sun, Y.; Ye, H.; Zhao, L.; Hu, Q.; Wang, G.; Zeng, X. Efficient procedure for isolating methylated catechins from green tea and effective simultaneous analysis of ten catechins, three purine alkaloids, and gallic acid in tea by high-performance liquid chromatography with diode array detection. J. Chromatogr. A 2009, 1216, 3223–3231. [Google Scholar] [CrossRef]
  48. Perva-Uzunalića, A.; Škergeta, M.; Kneza, Ž.; Weinreichb, B.; Ottob, F.; Grünerb, S. Extraction of active ingredients from green tea (Camellia sinensis): Extraction efficiency of major catechins and caffeine. Food Chem. 2006, 96, 597–605. [Google Scholar] [CrossRef]
  49. Chen, C.; Yu, R.; Owuor, E.D.; Kong, A.N. Activation of antioxidant-response element (ARE), mitogen-activated protein kinases (MAPKs) and caspases by major green tea polyphenol components during cell survival and death. Arch. Pharm. Res. 2000, 23, 605–612. [Google Scholar] [CrossRef]
  50. Hou, R.R.; Chen, J.Z.; Chen, H.; Kang, X.G.; Li, M.G.; Wang, B.R. Neuroprotective effects of (-)-epigallocatechin-3-gallate (EGCG) on paraquat-induced apoptosis in PC12 cells. Cell Biol. Int. 2008, 32, 22–30. [Google Scholar] [CrossRef]
  51. Rezai-Zadeh, K.; Shytle, D.; Sun, N.; Mori, T.; Hou, H.; Jeanniton, D.; Ehrhart, J.; Townsend, K.; Zeng, J.; Morgan, D.; Hardy, J.; Town, T.; Tan, J. Green tea epigallocatechin-3-gallate (EGCG) modulates amyloid precursor protein cleavage and reduces cerebral amyloidosis in Alzheimer transgenic mice. J. Neurosci. 2005, 25, 8807–8814. [Google Scholar]
  52. Okello, E.J.; Savelev, S.U.; Perry, E.K. In vitro anti-beta-secretase and dual anti-cholinesterase activities of Camellia sinensis L. (tea) relevant to treatment of dementia. Phytother. Res. 2004, 18, 624–627. [Google Scholar] [CrossRef]
  53. Li, R.; Huang, Y.G.; Fang, D.; Le, W.D. (-)-Epigallocatechin gallate inhibits lipopolysaccharide-induced microglial activation and protects against inflammation-mediated dopaminergic neuronal injury. J. Neurosci. Res. 2004, 78, 723–731. [Google Scholar] [CrossRef]
  54. Salah, N.; Miller, N.J.; Paganga, G.; Tijburg, L.; Bolwell, G.P.; Rice-Evans, C. Polyphenolic flavanols as scavengers of aqueous phase radicals and as chain-breaking antioxidants. Arch. Biochem. Biophys. 1995, 322, 339–346. [Google Scholar] [CrossRef]
  55. LI, Y.; Ouyang, C.; Shu, S. Protective Effect of EGCG on Focal Cerebral Ischemia-Reperfusion Injury in Rats. Journal of Xianning College(Medical Sciences) 2006, 20, 373–376. [Google Scholar]
  56. Yang, S.; Zhu, G.; Xie, D. Epigallocatechin Gallate Ameliorates Accumulated β-amyloid Protein Caused by Oxidative Free Radicals in Cortex Neuron in vitro. J. Clinical Res. 2007, 24, 1065–1067. [Google Scholar]
  57. Mandel, S.A.; Amit, T.; Kalfon, L.; Reznichenko, L.; Weinreb, O.; Youdim, M.B. Cell signaling pathways and iron chelation in the neurorestorative activity of green tea polyphenols: special reference to epigallocatechin gallate (EGCG). J. Alzheimers Dis. 2008, 15, 211–222. [Google Scholar]
  58. Aktas, O.; Prozorovski, T.; Smorodchenko, A.; Savaskan, N.E.; Lauster, R.; Kloetzel, P.M.; Infante-Duarte, C.; Brocke, S.; Zipp, F. Green tea epigallocatechin-3-gallate mediates T cellular NF-kappa B inhibition and exerts neuroprotection in autoimmune encephalomyelitis. J. Immunol. 2004, 173, 5794–5800. [Google Scholar]
  59. Rietveld, A.; Wiseman, S. Antioxidant effects of tea: evidence from human clinical trials. J. Nutr. 2003, 133, 3285S–3292S. [Google Scholar]
  60. Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: the in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar]
  61. Bradamante, S.; Barenghi, L.; Villa, A. Cardiovascular protective effects of resveratrol. Cardiovasc. Drug Rev. 2004, 22, 169–188. [Google Scholar]
  62. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.; Fong, H.H.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; Moon, R.C.; Pezzuto, J.M. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997, 275, 218–220. [Google Scholar]
  63. Das, S.; Das, D.K. Anti-inflammatory responses of resveratrol. Inflamm. Allergy Drug Targets 2007, 6, 168–173. [Google Scholar] [CrossRef]
  64. Bi, X.L.; Yang, J.Y.; Dong, Y.X.; Wang, J.M.; Cui, Y.H.; Ikeshima, T.; Zhao, Y.Q.; Wu, C.F. Resveratrol inhibits nitric oxide and TNF-alpha production by lipopolysaccharide-activated microglia. Int. Immunopharmacol. 2005, 5, 185–193. [Google Scholar] [CrossRef]
  65. Meng, X.L.; Yang, J.Y.; Chen, G.L.; Wang, L.H.; Zhang, L.J.; Wang, S.; Li, J.; Wu, C.F. Effects of resveratrol and its derivatives on lipopolysaccharide-induced microglial activation and their structure-activity relationships. Chem. Biol. Interact. 2008, 174, 51–59. [Google Scholar] [CrossRef]
  66. Candelario-Jalil, E.; de Oliveira, A.C.; Graf, S.; Bhatia, H.S.; Hull, M.; Munoz, E.; Fiebich, B.L. Resveratrol potently reduces prostaglandin E2 production and free radical formation in lipopolysaccharide-activated primary rat microglia. J. Neuroinflammation 2007, 4, 25. [Google Scholar] [CrossRef]
  67. Lorenz, P.; Roychowdhury, S.; Engelmann, M.; Wolf, G.; Horn, T.F. Oxyresveratrol and resveratrol are potent antioxidants and free radical scavengers: effect on nitrosative and oxidative stress derived from microglial cells. Nitric Oxide 2003, 9, 64–76. [Google Scholar] [CrossRef]
  68. Wang, Q.; Xu, J.; Rottinghaus, G.E.; Simonyi, A.; Lubahn, D.; Sun, G.Y.; Sun, A.Y. Resveratrol protects against global cerebral ischemic injury in gerbils. Brain Res. 2002, 958, 439–447. [Google Scholar] [CrossRef]
  69. Okawara, M.; Katsuki, H.; Kurimoto, E.; Shibata, H.; Kume, T.; Akaike, A. Resveratrol protects dopaminergic neurons in midbrain slice culture from multiple insults. Biochem. Pharmacol. 2007, 73, 550–560. [Google Scholar] [Green Version]
  70. Anekonda, T.S. Resveratrol--a boon for treating Alzheimer's disease? Brain Res. Rev. 2006, 52, 316–326. [Google Scholar] [CrossRef]
  71. Chen, J.; Zhou, Y.; Mueller-Steiner, S.; Chen, L.F.; Kwon, H.; Yi, S.; Mucke, L.; Gan, L. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J. Biol. Chem. 2005, 280, 40364–40374. [Google Scholar]
  72. Parker, J.A.; Arango, M.; Abderrahmane, S.; Lambert, E.; Tourette, C.; Catoire, H.; Neri, C. Resveratrol rescues mutant polyglutamine cytotoxicity in nematode and mammalian neurons. Nat. Genet. 2005, 37, 349–350. [Google Scholar]
  73. Bournival, J.; Quessy, P.; Martinoli, M.G. Protective effects of resveratrol and quercetin against MPP+ -induced oxidative stress act by modulating markers of apoptotic death in dopaminergic neurons. Cell. Mol. Neurobiol. 2009, 29, 1169–1180. [Google Scholar] [CrossRef]
  74. Chen, C.Y.; Jang, J.H.; Li, M.H.; Surh, Y.J. Resveratrol upregulates heme oxygenase-1 expression via activation of NF-E2-related factor 2 in PC12 cells. Biochem. Biophys. Res. Commun. 2005, 331, 993–1000. [Google Scholar] [CrossRef]
  75. Lu, X.; Ji, C.; Xu, H.; Li, X.; Ding, H.; Ye, M.; Zhu, Z.; Ding, D.; Jiang, X.; Ding, X.; Guo, X. Resveratrol-loaded polymeric micelles protect cells from Abeta-induced oxidative stress. Int. J. Pharm. 2009, 375, 89–96. [Google Scholar] [CrossRef]
  76. Lee, M.K.; Kang, S.J.; Poncz, M.; Song, K.J.; Park, K.S. Resveratrol protects SH-SY5Y neuroblastoma cells from apoptosis induced by dopamine. Exp. Mol. Med. 2007, 39, 376–384. [Google Scholar]
  77. Tang, W. Gastrodia elata Bl. In Chinese Drugs of Plant Origin: Chemistry, Pharmacology, and Use in Traditional and Modern Medicine; Springer-Verlag: Berlin, Heidelberg, 1992; pp. 545–548. [Google Scholar]
  78. Zeng, X.; Zhang, S.; Zhang, L.; Zhang, K.; Zheng, X. A study of the neuroprotective effect of the phenolic glucoside gastrodin during cerebral ischemia in vivo and in vitro. Planta Med. 2006, 72, 1359–1365. [Google Scholar] [CrossRef]
  79. An, H.; Kim, I.S.; Koppula, S.; Kim, B.W.; Park, P.J.; Lim, B.O.; Choi, W.S.; Lee, K.H.; Choi, D.K. Protective effects of Gastrodia elata Blume on MPP+-induced cytotoxicity in human dopaminergic SH-SY5Y cells. J. Ethnopharmacol. 2010, 130, 290–298. [Google Scholar] [CrossRef]
  80. Huang, N.K.; Chern, Y.; Fang, J.M.; Lin, C.I.; Chen, W.P.; Lin, Y.L. Neuroprotective principles from Gastrodia elata. J. Nat. Prod. 2007, 70, 571–574. [Google Scholar] [CrossRef]
  81. Kim, S.T.; Kim, J.D.; Lyu, Y.S.; Lee, M.Y.; Kang, H.W. Neuroprotective effect of some plant extracts in cultured CT105-induced PC12 cells. Biol. Pharm. Bull. 2006, 29, 2021–2024. [Google Scholar] [CrossRef]
  82. Jung, J.W.; Yoon, B.H.; Oh, H.R.; Ahn, J.H.; Kim, S.Y.; Park, S.Y.; Ryu, J.H. Anxiolytic-like effects of Gastrodia elata and its phenolic constituents in mice. Biol. Pharm. Bull. 2006, 29, 261–265. [Google Scholar] [CrossRef]
  83. Ahn, E.K.; Jeon, H.J.; Lim, E.J.; Jung, H.J.; Park, E.H. Anti-inflammatory and anti-angiogenic activities of Gastrodia elata Blume. J. Ethnopharmacol. 2007, 110, 476–482. [Google Scholar] [CrossRef]
  84. Hsieh, C.L.; Chen, C.L.; Tang, N.Y.; Chuang, C.M.; Hsieh, C.T.; Chiang, S.Y.; Lin, J.G.; Hsu, S.F. Gastrodia elata BL mediates the suppression of nNOS and microglia activation to protect against neuronal damage in kainic acid-treated rats. Am. J. Chin. Med. 2005, 33, 599–611. [Google Scholar] [CrossRef]
  85. Huang, N.K.; Lin, Y.L.; Cheng, J.J.; Lai, W.L. Gastrodia elata prevents rat pheochromocytoma cells from serum-deprived apoptosis: the role of the MAPK family. Life Sci. 2004, 75, 1649–1657. [Google Scholar] [CrossRef]
  86. Baek, N.I.; Choi, S.Y.; Park, J.K.; Cho, S.W.; Ahn, E.M.; Jeon, S.G.; Lee, B.R.; Bahn, J.H.; Kim, Y.K.; Shon, I.H. Isolation and identification of succinic semialdehyde dehydrogenase inhibitory compound from the rhizome of Gastrodia elata Blume. Arch. Pharm. Res. 1999, 22, 219–224. [Google Scholar] [CrossRef]
  87. Wu, C.R.; Hsieh, M.T.; Huang, S.C.; Peng, W.H.; Chang, Y.S.; Chen, C.F. Effects of Gastrodia elata and its active constituents on scopolamine-induced amnesia in rats. Planta Med. 1996, 62, 317–321. [Google Scholar] [CrossRef]
  88. Wu, H.Q.; Xie, L.; Jin, X.N.; Ge, Q.; Jin, H.; Liu, G.Q. The effect of vanillin on the fully amygdala-kindled seizures in the rat. Yao Xue Xue Bao 1989, 24, 482–486. [Google Scholar]
  89. Hwang, S.M.; Lee, Y.J.; Kang, D.G.; Lee, H.S. Anti-inflammatory effect of Gastrodia elata rhizome in human umbilical vein endothelial cells. Am. J. Chin. Med. 2009, 37, 395–406. [Google Scholar] [CrossRef]
  90. Lee, Y.J.; Hwang, S.M.; Kang, D.G.; Kim, J.S.; Lee, H.S. Effect of Gastrodia elata on tumor necrosis factor-alpha-induced matrix metalloproteinase activity in endothelial cells. J. Nat. Med. 2009, 63, 463–467. [Google Scholar] [CrossRef]
  91. Heo, J.C.; Woo, S.U.; Son, M.; Park, J.Y.; Choi, W.S.; Chang, K.T.; Kim, S.U.; Yoon, E.K.; Kim, Y.H.; Shin, H.M.; Lee, S.H. Anti-tumor activity of Gastrodia elata Blume is closely associated with a GTP-Ras-dependent pathway. Oncol. Rep. 2007, 18, 849–853. [Google Scholar]
  92. Hsieh, M.T.; Wu, C.R.; Chen, C.F. Gastrodin and p-hydroxybenzyl alcohol facilitate memory consolidation and retrieval, but not acquisition, on the passive avoidance task in rats. J. Ethnopharmacol. 1997, 56, 45–54. [Google Scholar] [CrossRef]
  93. Jung, T.Y.; Suh, S.I.; Lee, H.; Kim, I.S.; Kim, H.J.; Yoo, H.S.; Lee, S.R. Protective effects of several components of Gastrodia elata on lipid peroxidation in gerbil brain homogenates. Phytother. Res. 2007, 21, 960–964. [Google Scholar] [CrossRef]
  94. Kim, H.J.; Lee, S.R.; Moon, K.D. Ether fraction of methanol extracts of Gastrodia elata, medicinal herb protects against neuronal cell damage after transient global ischemia in gerbils. Phytother. Res. 2003, 17, 909–912. [Google Scholar] [CrossRef]
  95. Liu, J.; Mori, A. Antioxidant and free radical scavenging activities of Gastrodia elata Bl. and Uncaria rhynchophylla (Miq.) Jacks. Neuropharmacology 1992, 31, 1287–1298. [Google Scholar] [CrossRef]
  96. Sun, X.F.; Wang, W.; Wang, D.Q.; Du, G.Y. Research progress of neuroprotective mechanisms of Gastrodia elata and its preparation. Zhongguo Zhong Yao Za Zhi 2004, 29, 292–295. [Google Scholar]
  97. Ojemann, L.M.; Nelson, W.L.; Shin, D.S.; Rowe, A.O.; Buchanan, R.A. Tian ma, an ancient Chinese herb, offers new options for the treatment of epilepsy and other conditions. Epilepsy Behav. 2006, 8, 376–383. [Google Scholar] [CrossRef]
  98. Afzal, M.; Al-Hadidi, D.; Menon, M.; Pesek, J.; Dhami, M.S. Ginger: an ethnomedical, chemical and pharmacological review. Drug Metabol. Drug Interact. 2001, 18, 159–190. [Google Scholar]
  99. Awang, D. Ginger. Can. Pharmaceut. J. 1992, 125, 309–311. [Google Scholar]
  100. Leung, A. Encyclopedia of Common Natural Ingredients Used in Food, Drugs and Cosmetics. In Encyclopedia of Common Natural Ingredients Used in Food, Drugs and Cosmetics; John Wiley and Sons: New York, 1980. [Google Scholar]
  101. Tripathi, S.; Bruch, D.; Kittur, D.S. Ginger extract inhibits LPS induced macrophage activation and function. BMC Complement. Altern. Med. 2008, 8, 1. [Google Scholar] [CrossRef]
  102. Grzanna, R.; Phan, P.; Polotsky, A.; Lindmark, L.; Frondoza, C.G. Ginger extract inhibits beta-amyloid peptide-induced cytokine and chemokine expression in cultured THP-1 monocytes. J. Altern. Complement. Med. 2004, 10, 1009–1013. [Google Scholar] [CrossRef]
  103. Jung, H.W.; Yoon, C.H.; Park, K.M.; Han, H.S.; Park, Y.K. Hexane fraction of Zingiberis Rhizoma Crudus extract inhibits the production of nitric oxide and proinflammatory cytokines in LPS-stimulated BV2 microglial cells via the NF-kappaB pathway. Food Chem. Toxicol. 2009, 47, 1190–1197. [Google Scholar] [CrossRef]
  104. Kim, S.O.; Kundu, J.K.; Shin, Y.K.; Park, J.H.; Cho, M.H.; Kim, T.Y.; Surh, Y.J. [6]-Gingerol inhibits COX-2 expression by blocking the activation of p38 MAP kinase and NF-kappaB in phorbol ester-stimulated mouse skin. Oncogene 2005, 24, 2558–2567. [Google Scholar] [CrossRef]
  105. Tripathi, S.; Maier, K.G.; Bruch, D.; Kittur, D.S. Effect of 6-gingerol on pro-inflammatory cytokine production and costimulatory molecule expression in murine peritoneal macrophages. J. Surg. Res. 2007, 138, 209–213. [Google Scholar] [CrossRef]
  106. Lee, S.K.; Kim, H.N.; Kang, Y.R.; Lee, C.W.; Kim, H.M.; Han, D.C.; Shin, J.; Bae, K.; Kwon, B.M. Obovatol inhibits colorectal cancer growth by inhibiting tumor cell proliferation and inducing apoptosis. Bioorg. Med. Chem. 2008, 16, 8397–8402. [Google Scholar]
  107. Pyo, M.K.; Lee, Y.; Yun-Choi, H.S. Anti-platelet effect of the constituents isolated from the barks and fruits of Magnolia obovata. Arch. Pharm. Res. 2002, 25, 325–328. [Google Scholar] [CrossRef]
  108. Ito, K.; Iida, T.; Ichino, K.; Tsunezuka, M.; Hattori, M.; Namba, T. Obovatol and obovatal, novel biphenyl ether lignans from the leaves of Magnolia obovata Thunb. Chem. Pharm. Bull. (Tokyo) 1982, 30, 3347–3353. [Google Scholar] [CrossRef]
  109. Choi, M.S.; Lee, S.H.; Cho, H.S.; Kim, Y.; Yun, Y.P.; Jung, H.Y.; Jung, J.K.; Lee, B.C.; Pyo, H.B.; Hong, J.T. Inhibitory effect of obovatol on nitric oxide production and activation of NF-kappaB/MAP kinases in lipopolysaccharide-treated RAW 264.7cells. Eur. J. Pharmacol. 2007, 556, 181–189. [Google Scholar] [CrossRef]
  110. Seo, J.J.; Lee, S.H.; Lee, Y.S.; Kwon, B.M.; Ma, Y.; Hwang, B.Y.; Hong, J.T.; Oh, K.W. Anxiolytic-like effects of obovatol isolated from Magnolia obovata: involvement of GABA/benzodiazepine receptors complex. Prog. Neuropsychopharmacol. Biol. Psychiatry 2007, 31, 1363–1369. [Google Scholar] [CrossRef]
  111. Ock, J.; Han, H.S.; Hong, S.H.; Lee, S.Y.; Han, Y.M.; Kwon, B.M.; Suk, K. Obovatol attenuates microglia-mediated neuroinflammation by modulating redox regulation. Br. J. Pharmacol. 2010, 159, 1646–1662. [Google Scholar] [CrossRef]
  112. Jung, B.; Shin, M. Encyclopedia of illustrated Korea Natural Drugs. In Encyclopedia of illustrated Korea Natural Drugs; Yong Lim Sa: Seoul, 1990; pp. 845–846. [Google Scholar]
  113. Lee, C. Illustrated Flora of Korea. In Illustrated Flora of Korea; Hyang Moon Sa: Seoul, 1989; p. 661. [Google Scholar]
  114. Hong, S.S.; Lee, S.A.; Han, X.H.; Jin, H.Z.; Lee, J.H.; Lee, D.; Lee, J.J.; Hong, J.T.; Kim, Y.; Ro, J.S.; Hwang, B.Y. Kaurane diterpenoids from Isodon excisus inhibit LPS-induced NF-kappaB activation and NO production in macrophage RAW264.7 cells. J. Nat. Prod. 2007, 70, 632–636. [Google Scholar] [CrossRef]
  115. Ko, H.M.; Koppula, S.; Kim, B.W.; Kim, I.S.; Hwang, B.Y.; Suk, K.; Park, E.J.; Choi, D.K. Inflexin attenuates proinflammatory responses and nuclear factor-kappaB activation in LPS-treated microglia. Eur. J. Pharmacol. 2010, 633, 98–106. [Google Scholar] [CrossRef]
  116. Kim, H.S.; Lim, J.Y.; Sul, D.; Hwang, B.Y.; Won, T.J.; Hwang, K.W.; Park, S.Y. Neuroprotective effects of the new diterpene, CBNU06 against beta-amyloid-induced toxicity through the inhibition of NF-kappaB signaling pathway in PC12 cells. Eur. J. Pharmacol. 2009, 622, 25–31. [Google Scholar] [CrossRef]
  117. Zhang, R.; Xu, S.; Cai, Y.; Zhou, M.; Zuo, X.; Chan, P. Ganoderma lucidum Protects Dopaminergic Neuron Degeneration Through Inhibition of Microglial Activation. Evid. Based Complement. Alternat. Med. 2009.
  118. Neto, C.C. Cranberry and blueberry: evidence for protective effects against cancer and vascular diseases. Mol. Nutr. Food Res. 2007, 51, 652–664. [Google Scholar] [CrossRef]
  119. Seeram, N.P.; Adams, L.S.; Zhang, Y.; Lee, R.; Sand, D.; Scheuller, H.S.; Heber, D. Blackberry, black raspberry, blueberry, cranberry, red raspberry, and strawberry extracts inhibit growth and stimulate apoptosis of human cancer cells in vitro. J. Agric. Food Chem. 2006, 54, 9329–9339. [Google Scholar] [CrossRef]
  120. Zhu, Y.; Bickford, P.C.; Sanberg, P.; Giunta, B.; Tan, J. Blueberry opposes beta-amyloid peptide-induced microglial activation via inhibition of p44/42 mitogen-activation protein kinase. Rejuvenation Res. 2008, 11, 891–901. [Google Scholar] [CrossRef]
  121. Lu, D.Y.; Tang, C.H.; Chen, Y.H.; Wei, I.H. Berberine suppresses neuroinflammatory responses through AMP-activated protein kinase activation in BV-2 microglia. J. Cell. Biochem. 2010, 110, 697–705. [Google Scholar] [CrossRef]
  122. Wang, Y.K.; Huang, Z.Q. Protective effects of icariin on human umbilical vein endothelial cell injury induced by H2O2 in vitro. Pharmacol. Res. 2005, 52, 174–182. [Google Scholar] [CrossRef]
  123. Liang, H.R.; Vuorela, P.; Vuorela, H.; Hiltunen, R. Isolation and immunomodulatory effect of flavonol glycosides from Epimedium hunanense. Planta Med. 1997, 63, 316–319. [Google Scholar] [CrossRef]
  124. Chung, B.H.; Kim, J.D.; Kim, C.K.; Kim, J.H.; Won, M.H.; Lee, H.S.; Dong, M.S.; Ha, K.S.; Kwon, Y.G.; Kim, Y.M. Icariin stimulates angiogenesis by activating the MEK/ERK- and PI3K/Akt/eNOS-dependent signal pathways in human endothelial cells. Biochem. Biophys. Res. Commun. 2008, 376, 404–408. [Google Scholar] [CrossRef]
  125. Ding, L.; Liang, X.G.; Hu, Y.; Zhu, D.Y.; Lou, Y.J. Involvement of p38MAPK and reactive oxygen species in icariin-induced cardiomyocyte differentiation of murine embryonic stem cells in vitro. Stem Cells Dev. 2008, 17, 751–760. [Google Scholar] [CrossRef]
  126. Wu, J.F.; Dong, J.C.; Xu, C.Q. [Effects of icariin on inflammation model stimulated by lipopolysaccharide in vitro and in vivo]. Zhongguo Zhong Xi Yi Jie He Za Zhi 2009, 29, 330–334. [Google Scholar]
  127. Liu, M.H.; Sun, J.S.; Tsai, S.W.; Sheu, S.Y.; Chen, M.H. Icariin protects murine chondrocytes from lipopolysaccharide-induced inflammatory responses and extracellular matrix degradation. Nutr. Res. 2010, 30, 57–65. [Google Scholar] [CrossRef]
  128. Zeng, K.W.; Fu, H.; Liu, G.X.; Wang, X.M. Icariin attenuates lipopolysaccharide-induced microglial activation and resultant death of neurons by inhibiting TAK1/IKK/NF-kappaB and JNK/p38 MAPK pathways. Int. Immunopharmacol. 2010, 10, 668–678. [Google Scholar] [CrossRef]
  129. Li, B.; Tian, X. Enmein type diterpenoids from Isodon japonica. Phytochemistry 2001, 58, 543–546. [Google Scholar] [CrossRef]
  130. Hong, S.S.; Lee, S.A.; Han, X.H.; Hwang, J.S.; Lee, C.; Lee, D.; Hong, J.T.; Kim, Y.; Lee, H.; Hwang, B.Y. ent-Kaurane diterpenoids from Isodon japonicus. J. Nat. Prod. 2008, 71, 1055–1058. [Google Scholar] [CrossRef]
  131. Lim, J.Y.; Won, T.J.; Hwang, B.Y.; Kim, H.R.; Hwang, K.W.; Sul, D.; Park, S.Y. The new diterpene isodojaponin D inhibited LPS-induced microglial activation through NF-kappaB and MAPK signaling pathways. Eur. J. Pharmacol. 2010, 642, 10–18. [Google Scholar] [CrossRef]
  132. Tak, P.P.; Firestein, G.S. NF-kappaB: a key role in inflammatory diseases. J. Clin. Invest. 2001, 107, 7–11. [Google Scholar] [CrossRef]
  133. Teh, B.S.; Seow, W.K.; Li, S.Y.; Thong, Y.H. Inhibition of prostaglandin and leukotriene generation by the plant alkaloids tetrandrine and berbamine. Int. J. Immunopharmacol. 1990, 12, 321–326. [Google Scholar] [CrossRef]
  134. Xue, Y.; Wang, Y.; Feng, D.C.; Xiao, B.G.; Xu, L.Y. Tetrandrine suppresses lipopolysaccharide-induced microglial activation by inhibiting NF-kappaB pathway. Acta Pharmacol. Sin. 2008, 29, 245–251. [Google Scholar] [CrossRef]
  135. Klingelhoefer, S.; Obertreis, B.; Quast, S.; Behnke, B. Antirheumatic effect of IDS 23, a stinging nettle leaf extract, on in vitro expression of T helper cytokines. J. Rheumatol. 1999, 26, 2517–2522. [Google Scholar]
  136. Konrad, A.; Mahler, M.; Arni, S.; Flogerzi, B.; Klingelhofer, S.; Seibold, F. Ameliorative effect of IDS 30, a stinging nettle leaf extract, on chronic colitis. Int. J. Colorectal Dis. 2005, 20, 9–17. [Google Scholar] [CrossRef]
  137. Teucher, T.; Obertreis, B.; Ruttkowski, T.; Schmitz, H. Cytokine secretion in whole blood of healthy subjects following oral administration of Urtica dioica L. plant extract. Arzneimittelforschung 1996, 46, 906–910. [Google Scholar]
  138. Cheon, B.S.; Kim, Y.H.; Son, K.S.; Chang, H.W.; Kang, S.S.; Kim, H.P. Effects of prenylated flavonoids and biflavonoids on lipopolysaccharide-induced nitric oxide production from the mouse macrophage cell line RAW 264.7. Planta Med. 2000, 66, 596–600. [Google Scholar] [CrossRef]
  139. Chi, Y.S.; Jong, H.G.; Son, K.H.; Chang, H.W.; Kang, S.S.; Kim, H.P. Effects of naturally occurring prenylated flavonoids on enzymes metabolizing arachidonic acid: cyclooxygenases and lipoxygenases. Biochem. Pharmacol. 2001, 62, 1185–1191. [Google Scholar] [CrossRef]
  140. Kim, H.K.; Cheon, B.S.; Kim, Y.H.; Kim, S.Y.; Kim, H.P. Effects of naturally occurring flavonoids on nitric oxide production in the macrophage cell line RAW 264.7 and their structure-activity relationships. Biochem. Pharmacol. 1999, 58, 759–765. [Google Scholar]
  141. Krol, W.; Czuba, Z.P.; Threadgill, M.D.; Cunningham, B.D.; Pietsz, G. Inhibition of nitric oxide (NO.) production in murine macrophages by flavones. Biochem. Pharmacol. 1995, 50, 1031–1035. [Google Scholar] [CrossRef]
  142. Rezai-Zadeh, K.; Ehrhart, J.; Bai, Y.; Sanberg, P.R.; Bickford, P.; Tan, J.; Shytle, R.D. Apigenin and luteolin modulate microglial activation via inhibition of STAT1-induced CD40 expression. J. Neuroinflammation 2008, 5, 41. [Google Scholar] [CrossRef]
  143. Sung, B.; Pandey, M.K.; Aggarwal, B.B. Fisetin, an inhibitor of cyclin-dependent kinase 6, down-regulates nuclear factor-kappaB-regulated cell proliferation, antiapoptotic and metastatic gene products through the suppression of TAK-1 and receptor-interacting protein-regulated IkappaBalpha kinase activation. Mol. Pharmacol. 2007, 71, 1703–1714. [Google Scholar] [CrossRef]
  144. Maher, P.; Akaishi, T.; Abe, K. Flavonoid fisetin promotes ERK-dependent long-term potentiation and enhances memory. Proc. Natl. Acad. Sci. U S A 2006, 103, 16568–16573. [Google Scholar]
  145. Hanneken, A.; Lin, F.F.; Johnson, J.; Maher, P. Flavonoids protect human retinal pigment epithelial cells from oxidative-stress-induced death. Invest. Ophthalmol. Vis. Sci. 2006, 47, 3164–3177. [Google Scholar] [CrossRef]
  146. Park, H.H.; Lee, S.; Oh, J.M.; Lee, M.S.; Yoon, K.H.; Park, B.H.; Kim, J.W.; Song, H.; Kim, S.H. Anti-inflammatory activity of fisetin in human mast cells (HMC-1). Pharmacol. Res. 2007, 55, 31–37. [Google Scholar]
  147. Zheng, L.T.; Ock, J.; Kwon, B.M.; Suk, K. Suppressive effects of flavonoid fisetin on lipopolysaccharide-induced microglial activation and neurotoxicity. Int. Immunopharmacol. 2008, 8, 484–494. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Choi, D.K.; Koppula, S.; Suk, K. Inhibitors of Microglial Neurotoxicity: Focus on Natural Products. Molecules 2011, 16, 1021-1043. https://doi.org/10.3390/molecules16021021

AMA Style

Choi DK, Koppula S, Suk K. Inhibitors of Microglial Neurotoxicity: Focus on Natural Products. Molecules. 2011; 16(2):1021-1043. https://doi.org/10.3390/molecules16021021

Chicago/Turabian Style

Choi, Dong Kug, Sushruta Koppula, and Kyoungho Suk. 2011. "Inhibitors of Microglial Neurotoxicity: Focus on Natural Products" Molecules 16, no. 2: 1021-1043. https://doi.org/10.3390/molecules16021021

Article Metrics

Back to TopTop