Personal Use Only Not for Distribution

0 downloads 0 Views 1020KB Size Report
102 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 .... in turn may raise the high trans-endothelial electrical resis- tance, which is ...
99

Send Orders for Reprints to [email protected]

Current Pharmacogenomics and Personalized Medicine, 2015, 13, 99-116 ISSN:1875-6921 eISSN: 1875-6913

Pharmacological Benefits of Active Components of Natural Products Against Traumatic Brain Injury - A Review BENTHAM SCIENCE

Selvaraju Subash1,2, Musthafa M. Essa1,2,3*, Samir Al-Adawi2,4, Mushtaq A. Memon5, Thamilarasan Manivasagam6, Arokiasamy J. Thenmozhi6, Mohammed Akbar7, Byoung-Joon Song7 and Gilles J. Guillemin8

nl y

Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Oman; 2Ageing and Dementia Research Group, Sultan Qaboos University, Oman; 3 Food and Brain Research Foundation, India; 4College of Medicine and Health Sciences, Sultan Qaboos University, Oman; 5College of Veterinary Medicine, Washington State University, Pullman, Washington, USA; 6Department of Biochemistry and Biotechnology, Annamalai University, Tamilnadu, India; 7Section of Molecular Pharmacology and Toxicology, LMBB, NIAAA, National Institutes of Health, Rockville, Maryland, USA; 8Neuroinflammation Group, Department of Biomedical Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia

ut

rib

is t

rs on

al

U

io n

se

O

Abstract: Background: Since the last decade, the therapeutic potentials of natural phenolic antioxidants in human diseases associated with oxidative damage have received great attention. Within the last few years, a rapidly growing number of natural compounds with neuro-protective effects have been described. Many efforts have been made to explore the mechanisms for the neuro-protective properties of natural compounds. This review focuses on the beneficial effects of natural products in treating traumatic brain injury (TBI). Numerous epidemiological studies have shown consistent health benefits through the consumption of fruits, vegetables and nuts. In this review, we have summarized the protective effects of natural compounds [apocynin, (-)-epigallocatechin Gallate (EGCG), baicalein, caffeic acid, caffeic acid phenethyl ester, hydroxysaffloryellow A, osthole, oxy-resveratrol, pycnogenol, resveratrol, salvianolicacid B, triptolide and wogonin], and omega-3 fatty acid, particularly docosahexaenoic acid and its metabolites, may be used as personalized medicine against TBI and we have also discussed some of the barricades in translating these biofunctional compounds into relevant therapeutics for TBI.

rD

Conclusion: The benefits of natural products for traumatic brain injury show high inter-individual variability in their therapeutic effects and thus, this article addresses the intersection between novel therapeutics for traumatic brain injury and personalized medicine that will allow a broader range of interventions including the evidence-based natural products.

Pe

Received: October 15, 2015

INTRODUCTION

fo

Keywords: EGCG, natural products, oxidative stress, phytochemicals, resveratrol, traumatic brain injury. Revised: November 27, 2015

N ot

Current Pharmacogenomics and Personalized Medicine

1

Traumatic brain injury (TBI) is a leading cause of disability and death in healthy populations of all ages living in the industrialized world [1]. TBI has long been called a “silent epidemic” and it affects up to 10 million people globally [2, 3]. Estimates suggest that between 2.5 million and 6.5 million individuals in the U.S.A. are living with the consequences of TBI, much of which were caused by motor vehicle accidents [4, 5]. It is estimated that more than 300,000 U.S. veterans of the wars in Iraq and Afghanistan (20% of the 1.6 million) have experienced a sustained TBI [6]. Data aggregated from Europe and the U.K. suggest that 235 per 100,000 people suffer from TBI severe enough to warrant hospitalization each year, although no truly efficacious and approved specific therapies are currently available [7]. Approximately one percent population of the developed countries experience TBI each year [8]. The World Health *Address correspondence to this author at the Department of Food Science & Nutrition, P.O. 34, CAMS, Sultan Qaboos University, Al-Khoud, Muscat, P.C. 123, Sultanate of Oman; Tel: +968 2414 3604; Fax: +968 2441 3418; E-mail: [email protected]; [email protected] 1875-6913/15 $58.00+.00

Accepted: November 27, 2015

Organization, in its 2004 World Report on Road Traffic Injury Prevention, predicted that by 2020, road traffic accidents would be within the top three leading causes of the global burden of disease, ahead of HIV and tuberculosis [9]. According to the aforementioned facts, TBI is one of the most serious and prevalent public health problems worldwide [10]. The pathophysiology of TBI is complex and heterogeneous. TBI can manifest clinically from concussion to coma and death, depending on the extent of brain damage. Two major pathophysiological processes contribute to brain injury after trauma: primary injury, where damage is caused as a direct result of the mechanical impact; and secondary injury, which is initiated immediately after trauma, activates multiple pathways due to further cellular damage from the effects of primary injuries. The secondary injury can continue to develop over a period of minutes, hours, days, or weeks later following the initial traumatic assault [4, 11, 12] (Fig. 1). Neuronal damage in TBI is caused by direct mechanical forces to the brain as well as by consequent vascular, cellular and molecular alterations that result in acute and delayed neuronal death or injury [13-15]. © 2015 Bentham Science Publishers

Subash et al.

U

io n

se

O

nl y

100 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

is t

activation of a cascade of events that include activation of calcium-dependent enzymes, destruction of the cellular architectures, and ultimately the demise of various cell types in the brain [24, 25]. This cellular demise occurs via the activation of both necrotic and apoptotic cell death pathways [23].

rD

fo

N ot

Pe

rs on

The primary injury can represent any combination of skull fractures, intracranial hematomas, lacerations, contusions, and/or penetrating wounds. The primary injury results in the compression of neuronal, glial, astrocytic, and vascular tissue [16] and is associated with disruption of the cell membrane and disturbance of ionic homeostasis due to membrane leakiness [16, 17]. The initial impact to the brain was shown to stimulate multiple death pathways that include activation of cellular proteases such as calpains (papain-like calcium-dependent protease) and caspases [17, 18]. The primary injury is usually irreversible since neural cell death occurs at the moment of injury. On the contrary, the secondary injury is likely a reversible process because delayed neural cell death is a protracted event that can be regulated at many points in the death pathway. Therefore, intervention in the program of delayed neural cell death has become a therapeutic target for the successful treatment of TBI.

rib

al

ut

Fig. (1). Schematic diagram of Traumatic Brain Injury (TBI) and the time frame resulting in primary and secondary brain damage. Use of personalized medicine and natural compounds may alleviate the insults induced by primary and secondary brain damage.

The primary injury is followed by a series of secondary processes that continue to cause axonal and neuronal damage on a cellular level. Such mechanisms include amino acid excitotoxicity, lipid peroxidation, free radical damage, calcium mediated complication, neuroinflammation, hypoxia and ischemia [19-21]. The secondary injuries can proceed within minutes or days after the initial head injury and culminate in widespread cell death [18, 22]. TBI-induced cell death is accompanied by release of the excitatory amino acids, increased production of reactive oxygen species (ROS)/reactive nitrogen species (RNS), disruption of mitochondrial bioenergetics, disturbances in calcium homeostasis, and neuroinflammatory responses such as reactive gliosis [17, 23-24]. These disturbances are then accompanied by the

Neurodegeneration can continue for months after TBI, both in humans and in experimental models [14]. Importantly, the mechanisms for post-traumatic cell death can differ from those of apoptotic programmed cell death, depending on the time delay from the primary TBI. There is also evidence that genetic background, gender, age, type and severity of the insult, metabolic state of the brain and other conditions (e.g. other pre-existing diseases, medication) associated with TBI can affect the severity and/or mechanisms of neurodegeneration [26]. Morphologically, neuronal death can be divided into two major types: necrosis and apoptosis. The two categories provide some clues about the underlying mechanisms of neuronal death, even though the usefulness of morphological criteria as a starting point for understanding biochemical cascades has been recently challenged [27]. Electron microscopic morphology of necrosis consists of inflated cellular appearance with swollen mitochondria, vacuolated cytoplasm, dilated endoplasmic reticulum (ER), pycnotic nuclei and plasma membrane rupture. Necrosis can be initiated by mechanical damage that leads to neuronal membrane failure, Ca2+ influx and disruption of ionic homeostasis which triggers the release of cytotoxic glutamate [28]. Disruptions of membrane potential can also be caused by TBI-associated ischemia or hypoglycemia, leading to membrane depolarization, glutamate release and overstimulation of postsynaptic N-methyl-D-aspartate (NMDA)

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 101

preventative agents against TBI. Natural antioxidants, that can scavenge free radicals and prevent oxidative cell damage, have been evaluated for their potential neuro-protective properties [46]. Through numerous preclinical and clinical studies for potential neuro-protection, several natural compounds showed promising effects when evaluated with in vitro and in vivo models [47]. In this review, we have summarized the protective effects of a few natural compounds such as apocynin, (-)epigallocatechin gallate (EGCG), caffeic acid, resveratrol, etc., against TBI. We have also discussed some of the barricades in translating these biofunctional compounds into relevant therapeutics against TBI. APOCYNIN Apocynin (4-hydroxy-3-methoxyacetophenone) (Fig. 2) is a naturally occurring methoxy-substituted catechol, experimentally used as an inhibitor of Nicotinamide Adenine

O

and -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (vide infra). These events result in high intracellular Ca2+, increased oxidative stress, mitochondrial failure with depleted ATP, and activation of Ca2+-dependent proteases (e.g., Calpains), that cause cytoskeletal destruction of postsynaptic neurons [29]. In addition, the spillover of lysosomal cathepsins contributes to necrotic death. An inflammatory response caused by the extracellular release of cytoplasmic components and/or breakdown of the bloodbrain barrier (BBB) is another factor that exposes adjacent neurons to death signals and secondary damage (e.g., activation of caspase 1). Necrotic appearance is most common for cells dying within minutes or a few days after TBI. Neurotransmitter release, calcium over-load, free radical-mediated damage, proapoptotic gene activation, mitochondrial dysfunction, and inflammatory responses are the major pathological mechanisms of the secondary injury [30-32]. All these mechanisms can be induced via synaptic transmission and subsequent activation of postsynaptic receptors. Several proteins on the postsynaptic membrane are believed to form a special postsynaptic structure known as the postsynaptic density that serves as a multi-protein complex with a molecular network among neural cells after injury.

nl y

Pharmacological Benefits of Active Components of Natural Products

ut

rib

Fig. (2). Chemical structure of apocynin.

is t

Dinucleotide Phosphate oxidase (NADPH-oxidase). In 1971, apocynin was isolated from Apocynum cannabinum (Canadian hemp) and Picrorhiza kurroa (Scrophulariaceae) roots [48, 49]. Extracts of apocynin, a well-known traditional Indian medicine, were used to counter dropsy and heart problems [50]. The main mechanism of action of apocynin is to prevent the translocation of the cytosolic subunit, p47phox, to the NADPH-oxidase catalytic membrane domain [51]. In several different models of middle cerebral artery occlusion (MCAO), administration of apocynin has been shown to reduce infarct size in rats [52-54]. However, most of these studies used only a single dose of apocynin and those using more than a single dose noted that neuro-protection occurred within a narrow dose range when measured 24 h postMCAO [55, 56]. In addition, different outcomes of infarct size were observed with apocynin, depending on the timing of apocynin treatment, the method of MCAO, the length of reperfusion (ranging from 22 h to 3 days post-MCAO). The efficacy of apocynin in attenuating NADPH-oxidase mediated oxidative stress has been evaluated in different animal models, and it has been extensively used as an effective antioxidant and anti-inflammatory agent in oriental medicine [57].

rD

fo

N ot

Pe

rs on

al

U

io n

se

Secondary injury results from the activation of multiple pathways, leading to altered ionic balance, BBB permeability, edema, increased intracranial pressure, oxidative stress, neuronal cell death, and eventual neurologic impairment [33]. Mitochondrial dysfunction is increased in the aged TBI brain, with enhanced oxidative damage of synaptic mitochondria, contributing to significant impairments in bioenergetics function [34]. After TBI, aged animals show increased evidence of oxidative stress (e.g., elevated levels of 4hydroxynonenal and acrolein) and reduced antioxidant capacity (e.g., ascorbate), when compared with those in younger counterparts [35-37]. At the time of BBB disruption, a neuroinflammatory response is activated and this event can persist for several weeks following TBI [38, 39]. This disruption results from mechanical shearing of blood vessels at the time of injury and/or chemically mediated signaling cascades [38-40]. Infiltrating peripheral immune cells (i.e. leukocytes) activate resident astrocytes and microglia to stimulate the pro-inflammatory signaling pathways, contributing to further BBB break-down and brain edema. These events cause structural damage and functional deficits due to both primary and secondary injuries [38, 40-42]. However, it has been clearly shown that beneficial outcomes can be achieved, if neuroinflammation is properly controlled in a regulated manner and at the defined time periods [43]. Posttraumatic neuroinflammation is also significantly impacted by aging, with more pronounced and prolonged glial cell activation in the hippocampus [44] through the altered expression of CCAAT/enhancer binding protein (C/EBP) transcription factors [45]. The pathogenesis of TBI is not completely understood, because of the lack of understanding of the pathological mechanisms for the development of secondary damage. Considering the limits of existing preventive methods, intervention strategies using antioxidants and/or flavonoid-rich natural products, such as fruits, vegetables and nuts, are of paramount importance. The therapeutic potential of herbs and marine natural products can also become important as the

ROS generation and oxidative stress through NADPHoxidase have been implicated in promoting neuronal cell death and functional impairments following TBI, and neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD) [58-62]. A single intraperitoneal administration of apocynin (100 mg/kg) significantly

102 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

[85]. Remarkably, EGCG treatment significantly decreased malondialdehyde (MDA) levels in the rat model. EGCG absorbs TBI-mediated free radicals, thereby preventing apoptotic cell death in neurons and neural stem cell (NSC) populations. Consistently, other studies also showed that EGCG treatment increased the number of nestin-positive cells and NSCs around the damaged area after TBI [85-87].

nl y

Fig. (3). Chemical structure of EGCG.

BAICALEIN

Baicalein (5, 6, 7-trihydroxyflavone) (Fig. 4) is a major flavonoid extracted from the root of Scutellaria baicalensis Georgie (also called Huang Gui), an important medicinal plant widely used in China against allergic and inflammatory diseases such as hepatitis and asthma [88]. This flavonoid has been shown to exert potent anti-inflammatory effects in vitro as well as in vivo [89]. It has been shown to attenuate the focal cerebral ischemia/reperfusion injury and offer neuro-protective effects in PD while it can also inhibit amyloid-- and -synuclein-induced neuronal cell death [90-95].

O

reduced the oxidative injury in the hippocampus mediated by ischemia and hypoglycemia in rats [63]. Apocynin is a highly selective, cell-permeable inhibitor of NADPHoxidase [64] with a half-life of about 1-h [65]. The process of neuronal death occurs within 1-h after TBI. Thus, if delivered within 1-h, a single concentrated dose of apocynin can prevent TBI-induced NADPH oxidase activation. In fact, apocynin acted as a neuroprotective agent, when administered during the critical time immediately after TBI [63]. The cellular mechanisms underlying TBI-induced neuronal injury might be more complex than simple mechanical injury and involves several factors downstream of the initial injury. These factors include sustained activation of the glutamate receptors [66], poly ADP-Ribose polymerase (PARP) activation [67] and zinc translocation [68, 69]. In addition, administration of apocynin strongly attenuated the elevation of superoxide anion (O2-.), which can be converted to highly toxic free radicals such as hydroxyl ion and peroxynitrite, which can cause nitroxidative damage to neurons and other cells [70], possibly through post-translational protein modifications including nitration, as reviewed for the peripheral tissues [71, 72]. Apocynin (i.p., at 4 mg/kg) treated 20 min preTBI or 2-h post-TBI significantly reduced oxidative damage in the cortex and hippocampus in mice.

Subash et al.

io n

ut

rD

(-)-EPIGALLOCATECHIN GALLATE (EGCG)

N ot

fo

Green tea is a widely consumed beverage which contains many biologically important polyphenols. EGCG (trans-3, 4, 3’, 5’-tetrahydroxystilbene) (Fig. 3) is the major constituent, accounting for more than 10% of the extract in dry weight [74]. It exhibits a variety of biological functions and health benefits, including anti-obesity, anti-inflammatory, antiatherogenic and neuro-protective properties [75-78]. The proposed mechanisms for these beneficial effects include: inhibition of proteasome activity, inhibition of oncogene gene expression, and protection from DNA damage in normal cells [79-81]. Numerous in vitro and in vivo studies during the last decade have shown that EGCG can prevent and/or reduce the deleterious effects of oxygen-derived free radicals, which are associated with several human diseases [82]. The structural characteristics of green tea polyphenols for scavenging the superoxide anion radical have been reported [83]. In addition to ROS scavenging, green tea catechins are also able to trap reactive carbonyl species (RCS) such as glyoxal (GO), methylglyoxal (MG), and 3deoxyglucosone (3-DG) [84]. The importance of EGCG in enhancing cell resistance to oxidative stress goes beyond its simple ROS-scavenging and iron-chelating activities, and its effects are most profound in many pathological conditions where oxidative stress and iron accumulation are involved.

Pe

rib

is t

rs on

al

U

se

The results described above strongly indicate oxidative damage, BBB disruption and subsequent microglial activation as a key mechanism of neuronal death after TBI. Intraperitoneal administration of apocynin significantly decreased the levels of ROS production, oxidative stress markers of lipid peroxidation, BBB disruption, DNA oxidative damage and microglia activation and thus showed profound neuroprotective properties against TBI [63, 73].

EGCG [0.1% (w/v)] in drinking water inhibits neuronal cell death caused by TBI-related free radicals in Wistar rats

Fig. (4). Chemical structure of baicalein.

Rats, subjected to controlled cortical impact (CCI) injury, were treated with a single or 4 consecutive daily doses of baicalein (30 mg/kg/dose) or vehicle immediately after injury [96]. Post-injury administration of baicalein significantly protected the rats from long-term neurological deficits and brain damage following the CCI injury. The functional and histological improvements were accompanied with decreased levels of TNF-, IL- and IL-6 mRNA transcripts and proteins in the brain. Thus, the protective effects of baicalein are mediated, in part, through modulation of the TBIinduced proinflammatory cascades. The advantages of baicalein treatment include: 1) chronic dosing is not required; 2) baicalein has relatively low levels of toxicity; and 3) it is easy to administer in emergency situations. Thus, baicalein offers a great promise as a potential treatment for TBI. CAFFEIC ACID Caffeic acid (3,4-dihydroxycinnamic acid) (Fig. 5) is a natural phenolic compound widely distributed in medicinal plants, vegetables, honeybee propolis and beverages such as wine, coffee, tea and apple juice [97, 98]. It is known for its

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 103

anti-depressive, anti-tumor, anti-viral and anti-diabetic activities [99-102]. Furthermore, it possesses anti-oxidant properties as it scavenges a number of reactive species, including 1, 1-diphenyl-2-picryl-hydrazyl free radical (DPPH), peroxyl and hydroxyl radicals, superoxide anion and peroxynitrite [103-105]. It also inhibits 5-lipoxygenase (5-LOX) activity, protein kinase C (PKC), PKA and nuclear factor-B (NF-B) activation induced by ceramides in U937 cells [106-108].

Fig. (5). Chemical structure of caffeic acid.

Recently Kerman et al. have shown that a single dose of CAPE (10 mol/kg, i.p), given 15 min after trauma, can restore the suppressed activities of antioxidant enzymes (e.g., SOD, CAT and GPx) and thus ameliorate neuronal damage in experimental TBI model in Sprague–Dawley rats [119]. Additionally, CAPE administration inhibits lipid peroxidation in spinal cord and kidney after ischemia–reperfusion [120]. CAPE exhibits these protective effects through its high anti-oxidant capacity, but the exact mechanism of action of CAPE remains to be elucidated. Treatment with CAPE markedly reduced the immunereactivity of degenerating neurons after trauma. CAPE treatment has also been shown to inhibit apoptotic cell death by down-regulating caspase 3 in rat brain following transient focal cerebral ischemia [121], which caused shrunken cytoplasma, extensive dark pyknotic nuclei and vacuolization. These results indicate that the tissue edema were observed in the TBI neurons. However, the dark stained nuclei and the distorted nerve cells were mainly absent in the CAPE-treated traumatized rats in comparsion to the TBI animals [119].

O

Caffeic acid inhibited astrocyte proliferation (astrogliosis) and glial scar formation in the late phase of cryoinjury in rats and mice [109-111]. This could be at least associated with its anti-oxidant ability. These findings suggest that caffeic acid may represent a new prototype compound of potential neuroprotective agents in the treatment of early and late TBIs [109].

provement might be resulted from increased levels of the tight junction protein claudin-5 in brain micro-vessels. This in turn may raise the high trans-endothelial electrical resistance, which is critical for maintaining proper BBB permeability for brain homeostasis.

nl y

Pharmacological Benefits of Active Components of Natural Products

ut

HYDROXYSAFFLOR YELLOW A

fo

rD

is t

rib

The flower of the safflower plant (Carthamus tinctorius L.) is extensively used in the traditional Chinese medicine for the treatment of cardiovascular and cerebrovascular diseases and used as a natural dye. Hydroxysafflor yellow A (HSYA) (Fig. 7) is one of the most important active ingredients of the safflower plant. Chinese State Food and Drug

N ot

Pe

rs on

al

U

Caffeic acid phenethyl ester (CAPE) [3-(3, 4Dihydroxyphenyl)-2-propenoic acid 2-phenylethyl ester] (Fig. 6) is one of the most interesting bioactive compounds extracted from honeybee propolis. CAPE shows the potential therapeutic effects on many diseases and has been used for many years as a folk medicine. CAPE has been reported to decrease the inflammatory processes and brain lipid peroxidation with neuroprotective and antioxidant properties [110114]. CAPE can also modulate the Ca2+-induced release of cytochrome c in isolated brain and liver mitochondria [115, 116]. Gocer and Gulcin [117] studied the antioxidant and anti-radical activities of CAPE compared with standard antioxidant compounds. In addition, CAPE can be used as an easily accessible natural antioxidant and possibly as a food supplement. It can be also used to stabilize the food against oxidative deterioration.

io n

se

CAFFEIC ACID PHENETHYL ESTER

Fig. (6). Chemical structure of CAPE.

Zhao et al. [118] reported that post-injury administration of CAPE effectively reduces BBB permeability in both Sprague-Dawley rat and C57BL/6 mouse models of TBI. The beneficial effects of CAPE seem to be mediated through preserving claudin-5 levels. These results are associated with that activation of endogenous antioxidant proteins restores claudin-5 levels and markedly protects against TBI-induced enhanced permeability. Furthermore CAPE could be able to reduce cortical tissue loss, which is consistent with these studies [118]. However, the molecular mechanisms by which CAPE reduces BBB permeability are not known. This im-

Fig. (7). Chemical structure of HSYA.

Administration has approved HSYA (2,5-Cyclohexadien-1one,2,4-di--D-glucopyranosyl-3,4,5-trihydroxy-6-[(2E)-3(4-hydroxyphenyl)-1-oxo-2-propen-1-yl]) in March 2005 as a neuroprotective agent for the treatment of acute cerebral contusion (state drug permit document: Z20050146) [122]. Earlier data demonstrated that HSYA could provide neuroprotective effects via increasing the activity of SOD and decreasing the level of lipid peroxidation [123, 124]. It was also shown to prevent cerebral ischemia injury through its antioxidant action [125, 126]. Another study demonstrated

104 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

Fig. (8). Chemical structure of osthole.

It has been reported that osthole can reduce cerebral water content (edema), improve neurological recovery, delay neuronal death and reduce hippocampal neuron loss at 24 h after TBI in Sprague–Dawley rats. Pretreatment with osthole (40 mg/kg, i.p.) significantly decreased the levels of MDA including ROS, while it increased GSH levels and SOD activity against the progression of secondary brain injuries. In addition, osthole increased the expression of Bcl-2 and decreased the expression of Bax, thus ameliorating the decreased Bcl-2/Bax ratio induced by TBI in the cerebral cortex [141]. Wu et al. reported that osthole may reduce calcium influx through the inhibition of L-type calcium channels in neuronal cells [140]. Furthermore, the number of TUNEL-positive cells was significantly reduced by osthole pretreatment at the same sites in the brain at 24 h after TBI. Caspase-3, a key executor in apoptosis, can stimulate a DNA fragmentation factor, which activates endonucleases to cleave nucleic acids and finally leads to cell death. These results suggest that the protective effect of osthole may be attributable to the reduction in apoptotic activity of brain tissue after TBI [142].

N ot

fo

Matrix metalloproteinases (MMPs) represent a family of calcium-requiring, zinc-containing proteolytic enzymes, which degrade the proteins of the extracellular matrix. Among MMP isozymes, gelatinase A (MMP-2) and gelatinase B (MMP-9) are able to digest the components of the basement membrane. The increased MMPs are implicated in the pathogenesis of several central nervous system diseases, such as cerebral ischemia [130, 131]. When treated with HSYA at 30 min before and 6 h after the onset of TBI, the percent of MMP-9 positive cells decreased, suggesting that HYSA has beneficial effects on cerebral ischemia [124, 130, 131]. All these studies suggest that HSYA may exert potential therapeutic properties to improve the outcome following the TBI. These results may be, at least in part, resulted from improved mitochondrial activity, antioxidant capacity, and antithrombotic with fibrinolytic effects.

Pe

ut

rib

is t

rD

rs on

al

U

Tissue plasminogen activator (t-PA) is the major physiological plasminogen activator and is inhibited by Plasminogen activator inhibitor-1 (PAI-1). t-PA can cause thrombus fibrinolysis, whereas PAI-1 can lead to thrombus formation by inhibiting t-PA activity [129]. HSYA, administered at 30 min before and 6 h after the onset of TBI, could down regulate t-PA activity and upregulate PAI-1 activity in the TBI. These additional antithrombotic and fibrinolytic effects of HSYA may provide important benefits to the effective management of TBI [124].

io n

se

O

HYSA, injected via tail-vein at 4 mg/kg, significantly reduced the contusion volume in male Sprague–Dawley rats 24 h after the TBI. Morphological evidence of cell death, caused by the irreversibly impaired oxidative respiratory enzymes Ca2+-ATPase and Mg2+-ATPase after the TBI, is commonly detected by 2, 3, 5-triphenyl-tetrazolium chloride (TTC) staining, a sensitive histochemical indicator of mitochondrial respiratory enzyme function [124]. Its inhibition would result in a decline of Na+ and K + electrochemical gradients and ultimately lead to cell death as a result of osmotic damage and the accumulation of intracellular Ca2+ secondary to the decreased active Ca2+ influx by the Na+/Ca2+ exchange system [128]. When treated with HSYA at 30 min before and 6 h after the onset of TBI, the activities of Na+, K+-ATPase, Ca2+-ATPase, and Mg2+-ATPase were significantly elevated, especially at 24 and 48 h, compared with the TBI-control groups, suggesting the improvement in mitochondrial activity with reduced the contusion volume. In addition, HYSA treatment increased the SOD activity compared to that in the TBI-group after 48 h [124].

apoptotic properties in brain ischemia models and immunesuppressive actions in both in vivo and in vitro experiments were reported [135-139]. Osthole is a potent antioxidant which can eliminate oxygen free radicals, inhibit lipid peroxidation and potentially block the calcium channel in NG108-15 neuronal cells [140].

nl y

that HSYA reduced apoptosis of vascular endothelial cells under hypoxia through up-regulation of the HIF-1-VEGF pathway and the ratio of Bcl-2/Bax [127]. However, the exact mechanism is still unclear.

Subash et al.

OSTHOLE Osthole (7-methoxy-8-isopentenoxycoumarin) (Fig. 8), a natural coumarin derivative, is a component extracted from the medicinal plants Cnidium monnieri (L), Cusson and Peucedanum ostruthium, whose dried fruits have been used as a traditional folk medicine for many years. Previous studies have shown that osthole has a broad spectrum of pharmacological properties [132-134]. In addition, neuro-protective effects such as anti-inflammatory, anti-oxidative, and anti-

OXYRESVERATROL Oxyresveratrol (2, 4, 3’, 5’-tetrahydroxystilbene) (Fig. 9), isolated from mulberry wood (Morus alba L.), is a potent antioxidant and free-radical scavenger [143, 144]. Its pharmacological properties include a wide range of biological activities. However the neuroprotective effects of oxyresveratrol have been demonstrated in experimental models of ischemia/ reperfusion injury and other neurological pathologies [144146]. It also protects cultured rat cortical neurons from amyloid- and NMDA-induced neurotoxicities by reducing the number of apoptotic cells in transient cerebral ischemia and inhibiting the -secretase (BACE1) activity [147-149]. The protective effect of oxyresveratrol after middle cerebral arterial occlusion was reported to be mediated through significant reduction of the infarct volume [148]. Neuroblastoma cells exposed to 6-hydroxydopamine, used as a model of PD, were also protected by oxyresveratrol treatment [150]. In experimental models of exposures to both NMDA and 6hydroxydopamine, oxyresveratrol was more potent than resveratrol in providing neuroprotection. Oxyresveratrol differs from resveratrol due to the presence of an extra hydroxyl group, which enhances its antioxidant activity.

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 105

Fig. (9). Chemical structure of oxyresveratrol.

RESVERATROL Resveratrol (trans-3,4,5-trihydroxystilbene) (Fig. 11) is a naturally occurring non-toxic polyphenol mainly present in some plants (cranberries, grapes and peanuts) and constitutes one of the components in red wine, which shows beneficial effects on the age-related metabolic diseases, such as type-2 diabetes and obesity [161, 162]. In addition, other studies have reported that resveratrol exerts a variety of desirable biological activities, including anti-inammatory properties, cardio-protective, hypoglycemic, hypolipidemic, anti-aging, anti-cancer and neuro-protective properties [163-167]. The neuroprotective effects of this compound have been investigated in various models of neurodegenerative diseases, such as epilepsy, stroke, AD, PD, Huntington’s diseases (HD), cerebral ischemia, amyotrophic lateral sclerosis (ALS) and nerve injury [168-176]. The cellular mechanisms for resveratrol-induced neuro-protection must be better elucidated. Moreover, evidences show that resveratrol-mediated neuroprotective effects against ischemia injury are mediated by influencing the levels of certain neurotransmitters and neuromodulators released during ischemia/reperfusion. Furthermore, resveratrol has been shown to ameliorate apoptotic cell death and retinal structural damage in light-exposed mice [177]. These results showed that the neuroprotective effects of resveratrol are attributed to its antioxidant and NOscavenging properties with inhibition of excitatory synaptic transmission [178-180].

ut

is t

rD

fo

N ot

Pe

rs on

al

U

Pycnogenol® (PYC) (Fig. 10) is a patented combination of bioflavonoids extracted from the bark of the French maritime pine, Pinus maritima. Major constituents of PYC are polyphenols, specifically mono- and oligomeric units of caffeic acid, ferulic acid, catechin, epicatechin, taxifolin, and caffeic acid [107, 152]. PYC has been reported to possess a strong antioxidant property to prevent neurotoxicity and apoptotic cell death caused by various stressors such as lipid peroxides, pro-oxidants and peroxynitrite [105, 153]. Protective effect of PYC, as a potent antioxidant, on the acroleininduced cytotoxicity has been demonstrated in human neuroblastoma (SH-SY5Y) cells [154]. The cellular antioxidant network and the expression of those genes that are regulated by cell redox status have been studied in various in vitro and in vivo models [155-159].

rib

PYCNOGENOL

io n

se

O

Weber et al [151] analyzed the neuroprotective potential of oxyresveratrol in an in vitro model of stretch-induced trauma by using co-cultures of cortical neurons and glia. Oxyresveratrol (100 M) protected cortical neurons in the co-cultured cells, but not the glia as demonstrated by elevated S-100B protein release and a high proportion of cells with condensed nuclei. These results show that neuronal cells in high traumatic conditions and high glutamate treatment are differentially protected by oxyresveratrol in comparison to glia. Studies involving oxyreserveratrol in trauma models are necessary, as selective toxicity to glia could be beneficial by inhibiting reactive gliosis, which often occurs after trauma. The exact mechanisms of the observed effects are still unknown.

PYC (100 mg/kg/dose, three i.p. injections at 15 min, 3 h, and 6 h following a cortical contusion injury for 48 h), significantly reduced the levels of oxidative stress markers [e.g., TBARS, 4-hydroxynonenal (4-HNE), 3-nitrotyrosine (3-NT)] in Sprague-Dawley rats [160]. In addition, increased antioxidant levels (GSH, SOD, and catalase) and reduced levels of neuroinflammation markers (TNF- and IL-6) were observed. After 96 h, the reduction of key synaptic proteins (postsynaptic density protein 95, PSD-95) was also recognized in the cerebral cortex and hippocampus. These results support the idea that a combinational bioflavonoid can offer significant neuroprotection against acute brain trauma. However, the mechanism of action of PYC appears to be more complex than that of a single antioxidant [160].

nl y

Pharmacological Benefits of Active Components of Natural Products

Fig. (11). Chemical structure of resveratrol.

Fig. (10). Chemical structure of pycnogenol.

A single dose of resveratrol, when administered intraperitoneally at 100 mg/kg immediately after TBI, significantly reduced the levels of MDA, xanthine oxidase (XO), nitric oxide (NO), and oxidative stress in Wistar albino male rats [181]. It also increased the GSH level and decreased the area of tissue lesion in traumatic rats. These results indicated that resveratrol, treated immediately after TBI, can effectively reduce oxidative stress and the lesion volume.

106 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

TRIPTOLIDE Triptolide, a diterpene triepoxide (Fig. 13), was first isolated from the medicinal plant Tripterygium wilfordii Hook F (TWHF), which had been used for centuries in traditional Chinese medicine. Clinical and experimental studies have demonstrated that triptolide has anti-inflammatory and immunosuppressive properties. Triptolide has been reported as a strong inhibitor of NF-B, a critically important transcription factor in regulating inflammation, while it can protect donor hearts from ischemia/reperfusion-induced injury in heart transplantation [192-194].

Fig. (13). Chemical structure of triptolide.

is t

Furthermore, in vitro results showed that triptolide defends dopaminergic neurons from inflammation-mediated damage [195] and protects neural cells through the inhibition of chemokine (C-X-C motif) receptor 2 (CXCR2) activities [196]. It is also capable of promoting nerve regeneration and functional recovery after spinal cord and brain injuries [197, 198]. All these studies support the potential use of triptolide in treating peripheral and central nerve injury.

rD

fo

N ot

Pe

rs on

al

Danshen, the dried root of Salvia miltiorrhiza Bunge (labiatae), is a traditional Chinese medicine containing salvianolic acid B (Fig. 12), which is bioactive and most abundantly present [187, 188]. In animal model studies, salvianolic acid B has been shown to exhibit many beneficial properties of the danshen herb. For instance, it inhibits hydrogen peroxide-induced endothelial cell apoptosis through regulating the PI3K/Akt signaling pathway, while it protects against skin injury, heart and brain caused by ischemia-reperfusion [189-191].

rib

SALVIANOLIC ACID B

ut

U

io n

se

O

A single intraperitoneal injection of resveratrol at 100 mg/kg immediately after the trauma reduced the neuronal loss in all ipsi- and the contralateral hippocampal brain regions of Wistar Albino rats. Additionally, treatment with resveratrol significantly modified the cognitive and behavioral performances of TBI-inflicted immature rats, indicating that resveratrol has a neuroprotective effect on cognitive impairment of hippocampal neurons [186]. Improvement in cerebral function reflects neuroprotective effect of resveratrol on the hippocampal regions.

tion and microglia activation, thus preventing the cellular neuroinflammation after TBI. For instance, salvianolic acid B markedly suppressed the expression of pro-inflammatory cytokines TNF- and IL-1, while it elevated the expression of anti-inflammatory cytokines IL-10 and TGF-1 in the brain at 24 h after TBI.

nl y

Post-injury administration of resveratrol (100 mg/kg, i.p.) in the model of Sprague-Dawley rats significantly reduced the severity of cognitive and motor deficits associated with increased contusion volumes, while it preserved CA1 and CA3 hippocampal neurons with protection from overt hippocampal loss [182]. Although the underlying mechanisms by which resveratrol mediates its neuroprotection are still unclear, the study added to the growing literature of identifying resveratrol as a potential therapy for human brain injury. The two prominent brain regions affected in head trauma are the cortex and the hippocampus that play a crucial role in the processing and execution of spatial memory and learning [183, 184]. Ozdemir et al. showed that neuronal loss is not limited to ipsilateral hippocampal regions, but also in contralateral hippocampal areas at 24 h after trauma and in immature rat brains [185].

Subash et al.

Fig. (12). Chemical structure of salvianolicacid B.

Salvianolic acid B (25 mg/kg by injection), treated within 2 h after TBI in male C57BL/6J mice, significantly alleviated brain edema, reduced lesion volume, suppressed inflammatory responses, reduced motor functional deficits, and improved spatial learning and memory. Nevertheless, the molecular mechanisms for the neuroprotective properties of salvianolic acid B remain to be investigated. Reports have shown that salvianolic acid B suppressed neutrophil infiltra-

Triptolide (1 mg/kg, i.p.) treated soon after the induction of TBI showed anti-inflammatory and neuroprotective effects on the brain injury by inhibiting the levels of proinflammatory cytokines TNF-, IL-1, and IL-6, thus preventing apoptotic cell death and loss of cerebral neurons in Sprague-Dawley rats. In addition, triptolide inhibited the pro-inflammatory prostanoids (PGI2 and PGE2), that contribute to the formation of brain edema and excess cerebral cell apoptosis in several forms of brain injury. In another study, rats treated with a single dose of triptolide, displayed improvement in neurobehavioral outcomes measured. These findings suggest that triptolide confers neuroprotection against TBI and that this protection is mediated through an attenuation of neuroinflammation in the brain [199]. WOGONIN Wogonin (5, 7-dihydroxy-8-methoxyflavone) (Fig. 14), is a flavonoid found in the root of the Chinese herb Scutellaria baicalensis Georgi (also called Huang-Qin), which has been shown to exert potent anti-inflammatory effects in both in vitro and in vivo studies. Additionally, increasing evidence

Pharmacological Benefits of Active Components of Natural Products

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 107

suggests that wogonin may have neuroprotective effects on the injured brain. Wogonin also reduced early ischemic brain injury and improved acute behavioral dysfunctions caused by focal cerebral ischemia [200, 201]. Furthermore, it attenuated excitotoxic and oxidative stress-induced neuronal damage in primary cultured rat cortical cells [202] while it reduced neuronal damage caused by exposure to oxygen and glucose deprivation in cultured rat hippocampal slices [203]. In addition, wogonin attenuated the death of hippocampal neurons and inhibited microglia activation in global ischemia and excitotoxic injury models [204].

reduced (4.4-fold) prevalence of AD in India compared to that in the United States [209]. Curcumin is a potent inhibitor of NF-B [210], thus preventing neuroinflammation and neurological injury in experimental models of AD, ischemic stroke, subarachnoid hemorrhage and focal cerebral ischemia [211-215]. In addition, the curcumin treatment can protect hippocampal neurons against excitotoxic and traumatic injury [216].

Fig. (15). Chemical structure of curcumin.

N ot

fo

io n

ut

rib

rD

Wogonin treatment caused a significant reduction in both sensory-motor deficits [assessed by mNSS (modified neurological severity score)] and post-injury motor dysfunction (assessed by rotarod and beam walk tests), indicating that wogonin can be highly effective in providing long-lasting protection from the TBI. The protection afforded by wogonin was associated with a decrease in neuronal damage and apoptotic cell death with a reduced contusion volume in the cortical regions involved in motor and sensory functions [205]. Further investigations are needed to clarify the underlying mechanisms for the neuroprotective effects of wogonin on TBI.

Pe

The powerful antioxidant effects of curcumin were sufficient to reduce the action of oxidative stress on the BDNF system, synaptic plasticity, and cognitive function altered by TBI. However, more mechanistic and interventional studies are warranted to further evaluate the role of curcumin in clinical brain disorders and injuries.

is t

rs on

al

U

se

O

The post-injury treatment with wogonin (40 mg/kg, i.p.) has been shown to improve long-term functional and histological outcomes, while it reduced brain edema in a clinically relevant model of TBI in C57BL/6 mice. This could suggest that the neuroprotective effects of wogonin following TBI may be mediated, at least in part, through suppressing the TLR4/NF-kB signaling pathway and downstream factors (e.g., IL-1, IL-6, MIP-2, MMP-9 activity and COX2 expression). Thus, wogonin could be a potential therapeutic choice in the treatment of TBI [205].

nl y

Fig. (14). Chemical structure of wogonin.

The silent information regulator 2 (Sir2) has emerged as an important modulator of genomic stability and cellular homeostasis. Dietary supplementation of curcumin (500 ppm for 4 weeks) ameliorated neuronal damage partly through modulation of the expression of proteins such as AMPK, uMtCK, UCP2, COX-II and Sir2 following fluid percussion injury (FPI). These results show the potential benefits of curcumin supplementation to regulate the molecules involved in the deleterious effects on energy homeostasis, cognitive function and neuronal plasticity following TBI, as demonstrated in male Sprague–Dawley rats [217].

CURCUMIN Curcumin [1,7-bis-(4-hydroxy-3-methoxyphenyl)-1,6heptadiene-3,5-dione] (Fig. 15), a low molecular weight, lipophilic, natural yellow colored polyphenolic compound of Indian spice turmeric derived from the rhizome Curcuma longa. It has been consumed by humans for centuries as an anti-inflammatory and antioxidant in ayurvedic medicine [206]. It also reduces oxidative damage and cognitive deficits associated with aging and other pathological conditions. The curcumin is known to protect not only the brain from free radical-induced damage [207] but also for the cardiovascular, pulmonary, autoimmune and neoplastic diseases [208]. A single epidemiological study suggested that curcumin, as one of the most prevalent nutritional and medicinal compounds used by the Indian people, is responsible for the

DOCOSAHEXAENOIC ACID In addition to natural compounds that are present in various fruits, vegetables, nuts, and seeds, sea foods also contain tremendous amounts of healthy oils and fats that are equally beneficial in overall metabolic, cardiovascular and specifically neurological diseases. Dietary intake of general polyunsaturated fatty acids (PUFAs), including omega-3 fatty acids in particular, is important for the normal development and functioning of the brain. For instance, docosahexaenoic acid (DHA, 22:6, n-3) (Fig. 16), a long-chain n-3 PUFA, is highly enriched in the human central nervous system [218] and required for proper embryonic neurodevelopment. DHA along with other n-3 PUFAs have been reported to exhibit beneficial effects on the prevention and treatment of a variety of human diseases [219]. DHA offers beneficial effects in autoimmune and inflammatory disorders, cardiovascular diseases and retinopathy [220-223]. Inadequate supply of n-3 PUFAs during prenatal and postnatal development decreases the levels of DHA in neuronal tissues, leading to a variety of visual, cognitive, and/or behavioral deficits in animal models [224-226]. In addition, it has been indicated that low levels of DHA in the brain are associated with neurodegenerative diseases, such as generalized peroxisomal disorders and AD [227, 228].

108 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

Subash et al.

loss of DHA in the brain, increase in alpha spectrin II and decrease in NeuN positive cells, suggesting the importance of brain DHA status after TBI [230]. The benefit of omega-3 fatty acids including DHA, against TBI was recently reviewed [231]. CONCLUSION AND FUTURE DIRECTIONS

io n

ut

rib

is t

Resveratrol, Curcumin

Resveratrol, Salvianolicacid B, Wogonin, Docosahexaenoic acid

fo

N ot

EGCG, Baicalein, Caffeic acid, Caffeic acid phenethyl ester, Osthole, Pycnogenol, Resveratrol, Triptolide, Wogonin, Curcumin

Me mo ry an d Be ha vio r

Increased Apoptosis Astrocyte Proliferation and glial scar formation

Excitotoxicity Impaired Energy Metabolism

Inc rea sed Ap opt osi s

Apocynin, EGCG, Baicalein, Caffeic acid, Caffeic acid phenethyl ester, Hydroxysafflor yellow A, Osthole, Oxyresveratrol, Pycnogenol, Resveratrol, Salvianolicacid B, Wogonin, Curcumin, 51 |Triptolide, Page Docosahexaenoic acid

Caffeic acid, Oxyresveratrol

rD

Apocynin, Baicalein, Caffeic acid phenethyl ester, Hydroxysafflor yellow A, Osthole, Oxyresveratrol, Resveratrol, Salvianolicacid B, Triptolide, Wogonin, Curcumin

Pe

Isc he mi a, br ai n oe

rs on

al

U

se

O

Recent studies have demonstrated that DHA is beneficial in CCI-induced TBI. Mice fed with a DHA-supplemented diet for two months showed significant short- and long-term beneficial effects after induction of TBI. Supplementation of DHA indeed showed protection against hippocampal neurons and prevented loss of myelin basic protein (MBP). In addition, DHA restored the integrity of myelin sheath, resulting in the maintenance of nerve fiber conductivity in CCImodel animals [229]. In another study, in comparison to an adequate diet, the omega-3 deficient diet fed mice showed significant behavioral deficits evaluated by rotarod and beam walk tests after CCI. Mice fed the DHA-deficient diet also showed anxiety-like behavior assessed by the open field test and novel object recognition test. The dietary deficiency regime for three generations resulted in approximately 70%

nl y

Fig. (16). Chemical structure of docosahexaenoic acid.

The potential benefits of natural compounds for the prevention and treatment of TBI have been supported by numerous experimental studies. Evidences suggest that the TBI might be influenced by nutritional factors, i.e., bioactive compounds (Fig. 1 and Table 1). Naturally present bioactive compounds are good for the prevention and cure of various diseases without undesirable side effects. As multiple causative factors exist for the pathology of TBI, diverse mechanisms may be associated with the preventative effects. For instance, dietary changes provide better outcomes for patients with TBI. A ketogenic diet, which produces ketone bodies, was shown to have neuroprotective effects, since metabolism of ketone bodies decreases the amounts of ROS and exerts antioxidant, anti-inflammatory, and anti-apoptotic effects on the brain [232]. Also, magnesium, zinc, and branched-chain amino acids provide neuroprotection and increased cognitive function. Further, supplementation with DHA and other omega-3 fatty acids revealed a reduction in axonal damage and increased cognitive performance of TBIinflicted rats. On the other hand, high fat and high sucrose diets have shown to reduce spatial learning in TBI rats [232]. These results show that correcting the cerebral homeostatic dysfunction through various dietary supplements may allow for better therapeutic outcomes in patients with TBI.

TBI

Hydroxysafflor yellow A, Salvianolicacid B, Curcumin

Mitochondrial dysfunction In cr ea see d O xi da tiv ti e

EGCG, Baicalein, Caffeic acid, Caffeic acid phenethyl ester, Hydroxysafflor yellow A

Apocynin, EGCG, Caffeic acid phenethyl ester

Disruption of Blood brain barrier

Fig. (17). Graphic representation showing the targets for the TBI effects of naturally occurring compounds.

Pharmacological Benefits of Active Components of Natural Products

Shows the published reports on natural compounds against TBI

Natural Compounds

Sources

Class/mechanism/therapeutic mode

Apocynin

Apocynum cannabinum, Picrorhiza kurroa

Inhibitor of NADPH oxidase

EGCG

Green tea Camellia sinensis

Inhibits neuronal cell degeneration and death

Baicalein

Scutellaria baicalensis Georgire

Attenuated expression of TNF-, IL-1  and IL-6 mRNA and protein

Increased the reduced antioxidant enzyme activities (SOD, CAT and GPx) and reduced MDA levels

Propolis

Decreased the MMP-9 expression

Pe

• Neuroprotective effects

• Neuroprotection against brain damage • Antithrombotic and fibrinolytic effects • Reduced pathogenesis brain

rD

Restore the t-PA and PAI-1 activity

Chen et al., 2008 [96]

Zhang et al., 2007 [109]

Kerman et al., 2012 [119]

is t

Improve mitochondrial activity and reduce contusion volume

Itoh et al., 2012 [85]

• Reduced neuronal damage

ut

U

al

rs on

Carthamus tinctorius

Itoh et al., 2012 [85]

Zhao et al., 2012 [118]

• Reduced neurons distributed

Increasing the activity of SOD and decreasing the level of lipid peroxidation

References

• Reduces BBB permeability

io n

Altered Caspase-3 activity

Hydroxysafflor yellow A

• Reduce brain oedema and tissue damage

se

CAPE

• preventing free radical mediated apoptosis and cell death

• Late astrocyte proliferation and glial scar formation

Inhibition of 5-lipoxygenase (5-LOX) activity

Restores claudin-5 levels

• Reduced free radicals produced by TBI around the damaged brain region

nl y

Propolis, coffee, tea and apple

• Reduced microglial activation in the hippocampus after TBI.

O

Caffeic acid

Cimicifuga heracleifolia, Fruits, vegetables, tea,

Therapeutic effect

rib

Table 1.

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 109

Kerman et al., 2012 [119] Bie et al., 2010 [124] Zhu et al., 2003; 2005 [91, 92] Bie et al., 2010 [124] Bie et al., 2010 [124]

• Reduce cerebral water content, cerebral edema, delayed neuronal death and reduce hippocampal neuron loss

Ameliorating the Bcl-2/Bax ratio and caspase-3 activity

• Reductions in apoptotic activity of brain tissue

Ramulus mori, Morus alba, Artocarpus lakoocha

Elevated S-100B protein release and a high proportion of cells with condensed nuclei

• Inhibiting reactive gliosis

Weber et al., 2012 [151]

Pycnogenol

Pinus maritime, Pinus pinaster

Reduced levels of oxidative stress markers and neuroinflammation markers, increased antioxidants and key synaptic proteins in cortex and hippocampus

• Neuroprotection after acute brain trauma

Scheff et al., 2012 [160]

Reduced MDA, XO and NO levels, increased GSH level

• Attenuated tissue lesion area in trauma

Resveratrol

Cranberries, grapes, peanuts, red wine, mulberries and pomegranate, Veratrum grandiflorum, Polygonum cuspidatum

Oxyresveratrol

N ot

Osthole

fo

Decreased MDA, increased SOD and GSH level activity and reduce calcium influx

Cnidium monnieri, Peucedanum ostruthium, Angelica pubescens

Reduced contusion volumes, preservation of CA1 and CA3 hippocampal neurons Reduced the neuronal loss in all ipsiand the contralateral hippocampal regions

• Behavioral protection • Behavioral changes

Wu et al., 2002 [140] He et al., 2012 [141] ; Nagata, 1997 [142]

Ates et al., 2007 [181] Singleton et al., 2010 [182]

Sonmez et al., 2007 [186].

110 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

Subash et al.

Table (1) contd….

Sources

Class/mechanism/therapeutic mode

Salvianolic acid B

Danshen, Salvia miltiorrhiza

Alleviated brain edema, reduced lesion volume, suppressed inflammatory responses, reduced motor functional deficits, improved spatial learning and memory

• Recovered brain damage and inflammation

Triptolide

Tripterygium wilfordii

Ameliorated inflammatory mediators (TNF-, IL-1 and IL-6)

• Attenuation of neuroinflammation in the brain

Therapeutic effect

Modulation of the TLR4/NF-kB signaling pathway

References

Chen et al., 2011[101]

Lee et al., 2012 [198]

Chen et al., 2012 [198]

Reduction in contusion volume in the cortical regions involved in motor and sensory functions

• Decrease in neuronal damage and apoptotic cell death

Chen et al., 2012 [198]

• Regulate molecules involved in deleterious effects of energy homeostasis and cognitive function

Sharma et al., 2009 [217]

Curcumin

Curcuma longa

Decreased the expression of molecular systems such as AMPK, uMtCK, UCP2, COX-II and Sir2

Docosahexaenoic acid

Fruits, vegetables, nuts, seeds, sea foods, fat fish

Evaluated by rotarod and beam walk

O

Wogonin

nl y

• Reduced neuronal damage and behavioral impairment

Scutellaria baicalensis

ut

TBI often followed by elevated nitroxidative stress conditions. As a consequence, the administration of exogenous antioxidants could be beneficial for the proper handling of cell's oxidative stress status. In fact, epidemiological studies suggested that the regular intake of phenolic-rich vegetables and/or fruits can reduce the risk of oxidative stress-related diseases. In this regard, the important role of certain dietary components in alleviating various disorders is beginning to receive high attention, mainly due to beneficial phytochemicals contained in fruits and vegetables. All these known facts or knowledge should be utilized to find out a novel therapeutic option for the social and medical problems in human TBI survivors. The future research related to the therapeutic potential of natural phenolic antioxidants in human diseases associated with nitroxidative damage will enhance our understanding of these beneficial compounds.

is t

rD

fo

N ot

Pe

rs on

al

U

Over the past several decades, experimental research on TBI models has validated the contribution of free radicalinduced oxidative damage in the multidimensional secondary injury response following the TBI and recent reports confirm that anitoxidants and natural products could offer protection to TBI and other neurodegenerative conditions [233-253]. To date, the development of potent antioxidant compounds remains an attractive and promising strategy for the effective treatment of TBI. In the current review, we briefly described that treatment with naturally occurring compounds such as apocynin, EGCG, baicalein, caffeic acid, caffeic acid phenethyl ester, hydroxysaffloryellow A, osthole, oxyresveratrol, pycnogenol, resveratrol, salvianolicacid B, triptolide, wogonin, curcumin and omega-3 fatty acids (particularly, DHA) with antioxidant properties have been shown to reduce TBI-mediated cognitive deficits and/or functional impairments (Fig. 17 and Table 1). For the therapeutic purposes, it is ideal to focus on the inflammation process or downstream events of neuronal damage, behavioral impairment, apoptotic activity of brain tissue, BBB disruption and lipid peroxidation initiation, because these signaling events associated with the secondary injury may take several days to develop following the primary injury and thus widen the treatment window range. In these cases, the usage of natural components appears to have the significant advantage because of their relatively low toxicity, high availability with low costs and long term retention. So, a "nutritional therapy" emerges as an important strategy for preventing and/or treating TBI, contributing to the welfare of many affected individuals. However, further clinical trials and/or mechanistic studies need to be conducted to establish their efficacies.

Desai et al., 2013 [230]

io n

se

• Changed behavioral deficits

rib

Natural Compounds

Endogenous antioxidant defenses available in our body may not be proficient to block or counteract the increased production of ROS/RNS during disease conditions including

CONFLICT OF INTEREST The authors confirm that this article content has no conflict of interest. ACKNOWLEDGEMENTS This work was supported by a research grant from The Research Council of Oman (Grant # RC/AGR/FOOD/11/01 to MME), which is greatly appreciated. This work was also supported by the Intramural Program Fund of the National Institute of Alcohol Abuse and Alcoholism. The support provided by an internal grant from CAMS, SQU (IG/AGR/FOOD/14/01) is highly acknowldeged. REFERENCES [1]

Liossi C, Wood RL. Gender as a moderator of cognitive and affective outcome after traumatic brain injury. J. Neuropsychiatry Clin. Neurosci 2009; 21: 43-51.

[15] [16] [17] [18] [19]

[20]

[21] [22]

[23] [24]

[25]

[26] [27]

[34] [35]

[36] [37]

[38] [39]

io n

[14]

[33]

ut

[13]

[32]

rib

[12]

[31]

O

[11]

[30]

Povlishock JT, Katz DI. Update of neuropathology and neurological recovery after traumatic brain injury. J Head Trauma Rehabil 2005; 20: 76-94. Saatman KE, Zhang C, Bartus RT. Behavioral efficacy of posttraumatic calpain inhibition is not accompanied by reduced spectrin proteolysis, cortical lesion, or apoptosis. J Cereb Blood Flow Metab 2000; 20: 66-73. Zipfel GJ, Babcock DJ, Lee JM et al. Neuronal apoptosis after CNS injury: the roles of glutamate and calcium, J. Neurotrauma 2000; 17; 857–869. Hutchinson PJ, O'Connell MT, Rothwell NJ et al. Inflammation in human brain injury: intracerebral concentrations of IL-1alpha, IL1beta, and their endogenous inhibitor IL-1ra, J Neurotrauma 2007; 24: 1545-57. Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev 2007; 87: 99-163. Barkhoudarian G, Hovda DA, Giza CC. The molecular pathophysiology of concussive brain injury. Clin Sports Med 2011; 30: 33-48. Gilmer LK, Ansari MA, Roberts KN.et al. Age-related mitochondrial changes after traumatic brain injury. J Neurotrauma 2010; 27: 939-50. Moor E, Shohami E, Kanevsky E, et al. Impairment of the ability of the injured aged brain in elevating urate and ascorbate. Exp Gerontol 2006; 41: 303-11. Shao C, Roberts KN, Markesbery WR, et al. Oxidative stress in head trauma in aging. Free Radic Biol Med 2006; 41: 77-85. Anderson J, Sandhir R, Hamilton ES, et al. Impaired expression of neuroprotective molecules in the HIF-1alpha pathway following traumatic brain injury in aged mice. J Neurotrauma 2009; 26:15571566. Morganti-Kossmann MC, Satgunaseelan L, Bye N, et al. Modulation of immune response by head injury. Injury 2007; 38: 13921400. Shlosberg D, Benifla M, Kaufer D, et al. Blood–brain barrier breakdown as a therapeutic target in traumatic brain injury. Nat Rev Neurol 2010; 6: 393-403. Schmidt OI, Heyde CE, Ertel W, et al. Closed head injury—an inflammatory disease? Brain Res Rev 2005; 48: 388-99. Streit WJ, Mrak RE, Griffin WST. Microglia and neuroinflammation: a pathological perspective. J Neuroinflammation 2004; 1: 14. Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav Immun 2012; 26: 1191-201. Davalos D, Grutzendler J, Yang G, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci 2005; 8: 752-8. Sandhir R, Onyszchuk G, Berman NE. Exacerbated glial response in the aged mouse hippocampus following controlled cortical impact injury. Exp Neurol 2008; 213: 372-80. Sandhir R, Berman NE. Age-dependent response of CCAAT/enhancer binding proteins following traumatic brain injury in mice. Neurochem Int 2010; 56: 188-93. Slemmer J.E, Shacka JJ, Sweenev MI, et al. Antioxidants and free radical scavengers for the treatment of stroke, traumatic brain injury and aging. Curr Med Chem 2008; 15: 404-14. Jain KK. Neuroprotection in traumatic brain injury. Drug Discov Today.2008; 13: 1082-9. Connell BJ, Saleh MC, Khan BV, et al. Apocynin may limit total cell death following cerebral ischemia and reperfusion by enhancing apoptosis. Food Chemical Toxicol 2011; 49: 3063-9. Genovese T, Mazzon E, Paterniti I, et al. Modulation of NADPH oxidase activation in cerebral ischemia/reperfusion injury in rats. Brain Res 2011; 1372: 92-102. Sun AY, Wang Q, Simonyi A. Botanical phenolics and brain health,” Neuromolecular Med 2008; 10: 259-74. Stolk J, Hiltermann TJN, Dijkman JH, et al. Characteristics of the inhibition of NADPH oxidase activation in neutrophils by apocynin, a methoxy-substituted catechol. Am J Respir Cell Mol Biol 1994; 11: 95-102. Kelly KA, Li X, Tan Z, et al. NOX2 inhibition with apocynin worsens stroke outcome in aged rats. Brain Res 2009; 1292: 16572. Genovese T, Mazzon E, Pateriti I, et al. Modulation of NADPH oxidase in cerebral ischemia/reperfusion injury in rats. Brain Res 2010; 137: 92-102.

[40] [41]

is t

[10]

[29]

se

[9]

[28]

[42]

rD

[8]

U

[7]

fo

[6]

al

[5]

N ot

[4]

rs on

[3]

Coburn K. Traumatic brain injury: thesilentepidemic. AACN Clin. Issues Crit Care Nurs 1992; 3: 9-18. Hyder AA. Wunderlich CA, Puvanachandra P. et al. The impactoftraumaticbraininjuries: a global perspective. NeuroRehabilitation 2007; 22: 341-53. Maas AI, Stocchetti N, Bullock, R. Moderate and severe traumatic brain injury in adults. Lancet Neurol 2008; 8: 728-41. Faul M, Xu L, Wald M, et al. Traumatic Brain Injury in the United States: Emergency Department Visits, Hospitalizations and Deaths 2002–2006 Centers for Disease Control and Prevention National Center for Injury Prevention and Control, Atlanta, GA. 2010. Hoge CW, Goldberg, HM, Castro CA. Care of war veterans with mild traumatic brain injury – Flawed perspectives. N Engl J Med 2009; 360: 1588- 91. Tagliaferri F, Compagnone, C, Korsic M, et al. A systematic review of brain injury epidemiology in Europe. Acta Neurochir (Wien.) 2006; 148: 255-68. DeKosky ST, Blennow B, Milos D et al. Acute and chronic traumatic encephalopathies: pathogenesis and biomarkers. Nat Rev Neurol 2013; 9: 192-200. World report on traffic injury prevention, World Health Organization 2004. Mauritz W, Wilbacher I, Majdan M, et al. Epidemiology, treatment and outcome of patients after severe traumatic brain injury in European regions with different economic status. Eur J Public Health 2008; 18: 575-80. Gennarelli TA. Mechanisms of brain injury. J Emerg Med 1993; 11: 5-11. Kochanek PM, Bauman RA, Long JB. A critical problem begging for new insight and new therapies. J Neurotrauma 2009; 814: 8134. Laurer HL, McIntosh TK. Pharmacologic therapy in traumatic brain injury: update on experimental treatment strategies. Curr Pharm Des 2001; 7: 1505-16. Schwab JM, Brechtel K, Conrad S, et al. From cell death to neuronal regeneration: building a new brain after traumatic brain injury. J Neuropathol Exp Neurol 2003; 62: 801-11. Raghupathi R. Cell death mechanisms following traumatic brain injury. Brain Pathol 2004; 14: 215-22. Reilly PL. Brain injury: The pathophysiology of the first hours. “Talk and Die revisited”. J Clin Neurosci 2001; 8: 398-403. Wennersten A, Holmin S, Mathiesen T. Characterization of Bax and Bcl-2 in apoptosis after experimental traumatic brain injury in the rat. Acta Neuropathol 2003; 105: 281-8. Moppett IK. Traumatic brain injury: Assessment, resuscitation and early management. Br J Anaesth 2007; 99: 18-31. Mattson MP, Scheff SW. Endogeneous neuroprotection factors and traumatic brain injury: mechanisms of action and implications for therapy. J Neurotrauma 1994; 286: 3-33. Tymianski M, Tator CH. Normal and abnormal calcium homeostasis in neurons: A basis for the pathophysiology of traumatic and ischemic central nervous system injury. J Neurosurg 1996; 38: 1176-95. Marshall LF. Head injury: recent past, present and future. Neurosurgery 2000; 47: 546-61. Hall ED, Sullivan PG, Gibson TR, et al. Spatial and temporal characteristics of neurodegeneration after controlled cortical impact in mice: More than a focal brain injury. J Neurotrauma 2005; 22: 25265. McIntosh TK, Juhler M, Wieloch T. Novel pharmacologic strategies in the treatment of experimental traumatic brain injury. J Neurotrauma 1998; 15: 731-69. Sullivan PG, Rabchevsky AG, Waldmeier PC. Mitochondrial permeability transition in CNS trauma: Cause or effect of neuronal cell death? J Neurosci Res 2005; 79: 231-9. Rego AC, Monteiro NM, Silva AP. Mitochondrial apoptotic cell death and moderate superoxide generation upon selective activation of non-desensitizing AMPA receptors in hippocampal cultures. J Neurochem 2003; 86: 792-804. Thompson HJ, Lifshitz J, Marklund N.Lateral fluid percussion brain injury: a 15-year review and evaluation. J Neurotrauma 2005; 22: 42–75. Artal-Sanz M, Tavernarakis N. Proteolytic mechanisms in necrotic cell death and neurodegeneration. FEBS Lett 2005; 579: 3287– 3296.

Pe

[2]

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 111

nl y

Pharmacological Benefits of Active Components of Natural Products

[43]

[44] [45]

[46] [47] [48]

[49] [50] [51]

[52]

[53]

112 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

[65] [66]

[67] [68]

[69]

[70] [71] [72]

[73]

[74] [75]

[76]

[81]

[82]

O

[84]

[85]

[86]

[87]

io n

nl y

[83]

ut

[64]

[80]

rib

[63]

[79]

Wolfram S. Effects of green tea and EGCG on cardiovascular and metabolic health. J Am Coll Nutr 2007; 26: 373S-88S. Menard C, Bastianetto S, Quirion R. Neuroprotective effects of resveratrol and epigallocatechin gallate polyphenols are mediated by the activation of protein kinase C gamma. Front Cell Neurosci 2013; 7: 281. Nam S1, Smith DM, Dou QP. Ester bond-containing tea polyphenols potently inhibit proteasome activity in vitro and in vivo. J Biol Chem 2001; 276: 13322-30. Annabi B, Currie JC, Moghrabi A, et al. Inhibition of HuR and MMP-9 expression in macrophage-differentiated HL-60 myeloid leukemia cells by green tea polyphenol EGCg. Leuk Res 2007; 31: 1277-84. Fang MZ, Wang Y, Ai N, et al. Tea polyphenol (-)epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res 2003; 63: 7563-70. Weinreb O, Amit T, Mandel S, et al. Neuroprotective molecular mechanisms of (-)-epigallocatechin-3-gallate: a reflective outcome of its antioxidant, iron chelating and neuritogenic properties. Genes Nutr 2009; 4: 283-96. Sato M, Toyazaki H, Yoshioka Y, et al. Structural characteristics for superoxide anion radical scavenging and productive activities of green tea polyphenols including proanthocyanidin dimers. Chem Pharm Bull 2010; 58: 98-102. Lo CY, Li S, Tan D, et al. Trapping reactions of reactive carbonyl species with tea polyphenols in simulated physiological conditions. Mol Nutr Food Res 2006; 50: 1118-28. Itoh T, Imano M, Nishida S, et al. (2)-Epigallocatechin-3-gallate increases the number of neural stem cells around the damaged area after rat traumatic brain injury. J Neural Transm 2012; 119: 877-90. Itoh T, Satou T, Hashimoto S, et al. Immature and mature neurons coexist among glial scars after rat traumatic brain injury. Neurol Res 2007; 29: 734-42. Itoh T, Satou T, Nishida S, et al. The novel free radical scavenger, edaravone, increases neural stem cell number around the area of damage following rat traumatic brain injury. Neurotoxic Res 2009; 16: 378-89. Cheng Y, Ping J, Xu HD, et al. Synergistic effect of a novel oxymatrine-baicalin combination against hepatitis B virus replication, alpha smooth muscle actin expression and type I collagen synthesis in vitro., 2006, World J Gastroenterol 2006; 12: 5153-9. Huang WH, Lee AR, Yang CH. Antioxidative and antiinflammatory activities of polyhydroxyflavonoids of Scutellaria baicalensis Georgi. Biosci Biotechnol Biochem 2006; 70: 2371-80. Zhang ZJ, Wang Z, Zhang XY, et al. Gene expression profile induced by oral administration of baicalin and gardenin after focal brain ischemia in rats. Acta Pharmacol Sin 2005; 26: 307–314. Hamada-Hiramatsu M, Edamatsu R, Mori A. Free radical scavenging action of baicalein. Arch Biochem Biophys 1993; 306: 261-6. Van Leyen K, Kim HY, Lee SR, et al. Baicalein and 12/15lipoxygenase in the ischemic brain. Stroke 2006; 37: 3014-8. Lapchak PA, Maher P, Schubert D, et al. Baicalein, an antioxidant 12/15-lipoxygenase inhibitor improves clinical rating scores following multiple infarct embolic strokes. Neuroscience 2007; 150: 585-91. Lebeau A, Esclaire F, Rostène W, et al. Baicalein protects cortical neurons from ß-amyloid (25-35) induced toxicity. Neuroreport 2001; 12: 2199-02. Gasiorowski K, Lamer-Zarawska E, Leszek J, et al. Flavones from root of Scutellaria baicalensis Georgi: drugs of the future in neurodegeneration? CNS Neurol. Disord Drug Targets 2011; 10: 18491. Chen SF, Hsu CW, Huang WH, et al. Post-injury baicalein improves histological and functional outcomes and reduces inflammatory cytokines after experimental traumatic brain injury. Brit J Pharmacol 2008; 155: 1279–1296. Sondheimer E. On the distribution of caffeic acid and the chlorogenic acid isomers in plants. Arch Biochem Biophys 1958; 74: 131-8. Naczk M, Shahidi F. Extraction and analysis of phenolics in food. J Chromatogr A. 2004; 1054: 95-111. Takeda H, Tsuji M, Inazu M, et al. Rosmarinic acid and caffeic acid produce antidepressive-like effect in the forced swimming test in mice. Eur J Pharmacol 2002; 449 (3): 261-7.

[88]

is t

[62]

[78]

se

[61]

[77]

[89]

rD

[60]

U

[59]

fo

[58]

al

[57]

N ot

[56]

rs on

[55]

Zhao H, Mayhan WG, Arrick DM, et al. Alcohol-induced exacerbation of ischemic brain injury: role of NAD (P)H oxidase. Alcohol Clin Exp Res 2010; 14: 1-8. Jackman KA, Miller AA, DeSilva TM, et al. Reduction of cerebral infarct volume by apocynin requires pretreatment and is absent in Nox2-deficient mice. Br J Pharmacol 2009; 156: 680-8. Tang XN, Cairns B, Cairns N, et al. Apocynin improves outcome in experimental stroke with a narrow dose range. Neuroscience 2008; 154; 556-62. Wang Q, Tompkins KD, Simonyi A, et al. Apocynin protects against global cerebral ischemia-reperfusion-induced oxidative stress and injury in the gerbil hippocampus. Brain Res 2006; 1090: 182-9. Ansari MA, Roberts KN, Scheff SW. Oxidative stress and modification of synaptic proteins in hippocampus after traumatic brain injury. Free Radic Biol Med 2008; 45: 443-52. Wada K, Alonso OF, Busto R, et al. Early treatment with a novel inhibitor of lipid peroxidation (LY341122) improves histopathological outcome after moderate fluid percussion brain injury in rats. Neurosurgery 1999; 45: 601-8. Marklund N, Clausen F, McIntosh TK, et al. Free radical scavenger posttreatment improves functional and morphological outcome after fluid percussion injury in the rat. J Neurotrauma 2001; 18: 82132. Bruce-Keller A.J, Gupta S, Parrino TE, et al. NOX activity is increased in mild cognitive impairment. AntioxidRedox Signal 2010; 12: 1371-82. Wu DC, Teismann P, Tieu Ket al. NADPH oxidase mediates oxidative stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. Proc Natl Acad Sci USA 2003; 100: 6145-50. Choi BY, Jang BG, Kim JH, et al. Prevention of traumatic brain injury-induced neuronal death by inhibition of NADPH oxidase activation. Brain Res 2012; 1481: 49-58. Hart BA, Simons JM. Metabolic activation of phenols by stimulated neutrophils: a concept for as elective type of antiinflammatory drug. Biotechnol Ther 1992; 3: 119-35. Peters EA, Hiltermann JT, Stolk J. Effect of apocynin on ozoneinduced airway hyperresponsiveness to methacholine in asthmatics. Free Radic Biol Med 2001; 31: 1442-7. Carbonell WS, Grady MS. Evidence disputing the importance of excitotoxicity in hippocampal neuron death after experimental traumatic brain injury. Ann NY Acad Sci 1999; 890: 287-98. LaPlaca MC, Zhang J, Raghupathi R, et al. Pharmaco- logic inhibition of poly(ADP-ribose) polymerase is neuroprotective following traumatic brain injury in rats. J Neurotrauma 2001; 18: 369-76. Suh SW, Chen JW, Motamedi M, et al. Evidence that synapticallyreleased zinc contributes to neuronal injury after traumatic brain injury. Brain Res 2000; 852: 268-73. Suh SW, Frederickson CJ, Danscher G. Neurotoxic zinc translocation into hippocampal neurons is inhibited by hypothermia and is aggravated by hyperthermia after trau- matic brain injury in rats. J Cereb Blood Flow Metab 2006; 26: 161-9. Bergamini CM, Gambetti S, Dondi A, et al. Oxygen, reactive oxygen species and tissue damage. Curr Pharm Des 2004; 10: 1611-26. Abdelmegeed MA and Song BJ. Functional role of protein nitration in acute and chronic liver diseases. Oxid Med Cell Longev 2014; 149627. Song BJ, Akbar M, Abdelmegeed MA, et al. Mitochondrial dysfunction and tissue injury by alcohol, high fat, nonalcoholic substances and pathological conditions through post-translational protein modifications Redox Biolology 2014; 3: 109-23. Zhang QG, Laird MD, Han D, et al. Critical role of NADPH oxidase in neuronal oxidative damage and microglia activation following traumatic brain injury. PLoS One 2012; 7: 34504. Yu J, Jia Y, Guo Y, et al.Epigallocatechin-3-gallate protects motor neurons and regulates glutamate level. FEBS Lett 2010; 584: 29215. Lin JK, Lin-Shiau SY. Mechanisms of hypolipidemic and antiobesity effects of tea and tea polyphenols. Mol Nutr Food Res 2006; 50: 211-7. Calixto JB, Campos MM, Otuki MF, et al. Anti-inflammatory compounds of plant origin. Part II. Modulation of proinflammatory cytokines, chemokines and adhesion molecules. Planta Med 2004; 70: 93-103.

Pe

[54]

Subash et al.

[90] [91] [92] [93]

[94]

[95]

[96]

[97] [98] [99]

[102] [103]

[104]

[105] [106] [107]

[124] [125]

[126]

[127]

[128] [129] [130]

[131]

O

[108]

[123]

[114]

[115]

[116] [117]

[118]

[119] [120]

[121]

[122]

[133]

rib

[134]

[135]

is t

al

U

se

[132]

[136]

rD

[113]

fo

[112]

N ot

[111]

rs on

[110]

Pe

[109]

Han B, Zhao H. Effects of hydroxysafflor yellow A in the attenuation of MPTP neurotoxicity in mice. Neurochem Res 2010; 35: 107-13. Bie XD, Han J, Dai HB. Effects of hydroxysafflor yellow A on the experimental traumatic brain injury in rats. J Asian Nat Prod Res 2010; 12: 239-47. Liu YN, Zhou ZM, Chen P. Evidence that hydroxysafflor yellow A protects the heart against ischaemia–reperfusion injury by inhibiting mitochondrial permeability transition pore opening. Clin Exp Pharmacol Physiol 2008; 35(2): 211-6. Wei X, Liu H, Sun X, et al. Hydroxysafflor yellow A protects rat brains against ischemia-reperfusion injury by antioxidant action. Neurosci Lett 2005; 386 (1): 58-62. Ji DB, Zhu MC, Zhu B, et al. Hydroxysafflor yellow A enhances survival of vascular endothelial cells under hypoxia via upregulation of the HIF-1 alpha-VEGF pathway and regulation of Bcl2/Bax. J Cardiovasc Pharmacol 2008; 52: 191-202. Blaustein MP. Sodium ions, calcium ions, blood pressure regulation, and hypertension: a reassessment and a hypothesis. Am J Physiol 1977; 232: 165-73. Kohler HP, Grant PJ. Plasminogen-activator inhibitor type 1 and coronary artery disease. N Engl J Med 2000; 342: 1792-801. Marmarou A, Foda MA, van den Brink W, et al. A new model of diffuse brain injury in rats. Part I: Pathophysiology and biomechanics. J Neurosurg 1994; 80: 291-300. Borges N, Cerejo A, Santos A, et al. Changes in rat cerebral mitochondrial succinate dehydrogenase activity after brain trauma. Int J Neurosci 2004; 114: 217-27. Chou SY, Hsu CS, Wang KT et al. Antitumor effects of Osthole from Cnidium monnieri: an in vitro and in vivo study. Phytother Res 2007; 21: 226-30. Tang DZ, Hou W, Zhou Q, et al Osthole stimulates osteoblast differentiation and bone formation by activation of beta-cateninBMP signaling. J Bone Miner Res 2010; 25(6): 1234-45. Ming LG, Zhou J, Cheng GZ, et al. Osthol, a coumarin isolated from common cnidium fruit, enhances the differentiation and maturation of osteoblasts in vitro. Pharmacology 2011; 88(1–2): 33-43. Chen X, Pi R, Zou Y, et al. Attenuation of experimental autoimmune encephalomyelitis in C57 BL/6 mice by osthole, a natural coumarin. Eur. J. Pharmacol 2010; 629: 40-6. Chen T, Liu W, Chao X, et al. Neuroprotective effect of osthole against oxygen and glucose deprivation in rat cortical neurons: involvement of mitogen-activated protein kinase pathway. Neuroscience 2011; 183: 203-11. Ji HJ, Hu JF, Wang YH, et al. Osthole improves chronic cerebral hypoperfusion induced cognitive deficits and neuronal damage in hippocampus. Eur J Pharmacol 2010; 636: 96-101. Chao X, Zhou J, Chen T, et al. Neuroprotective effect of osthole against acute ischemic stroke on middle cerebral ischemia occlusion in rats. Brain Res 2010; 1363: 206-11. Liu WB, Zhou J, Qu Y, et al. Neuroprotective effect of osthole on MPP+-induced cytotoxicity in PC12 cells via inhibition of mitochondrial dysfunction and ROS production. Neurochem Int 2010; 57: 206-15. Wu SN, Lo YK, Chen CC, et al. Inhibitory effect of the plantextract osthole on L-type calcium current in NG108-15 neuronal cells. Biochem Pharmacol 2002; 63: 199-206. He Y, Qu S, Wang J, et al. Neuroprotective effects of osthole pretreatment against traumatic brain injury in rats. Brain Res 2012; 1433: 127-36. Nagata S. Apoptosis by death factor. Cell 1997; 88: 355–365. Soleas GJ, Diamandis EP, Goldberg DM. Wine as a biological fluid: history, production, and role in disease prevention. J Clin Lab Anal 1997; 11: 287-313. Dore S. Unique properties of polyphenol stilbenes in the brain: more than direct antioxidant actions; gene/protein regulatory activity. Neurosignals 2005; 14: 61-70. Sinha K, Chaudhary G, Gupta Y K. Protective effect of resveratrol against oxidative stress in middle cerebral artery occlusion model of stroke in rats. Life Sci 2002; 71: 655-65. Inoue H, Jiang XF, Katayama T, et al. Brain protection by resveratrol and fenofibrate against stroke requires peroxisome proliferatoractivated receptor alpha in mice. Neurosci Lett 2003; 352: 203-6. Ban JY, Jeon SY, Nguyen TT, et al. Neuroprotective effect of oxyresveratrol from Smilacis chinae rhizome on amyloid beta pro-

io n

[101]

Rajendra-Prasad N, Karthikeyan A, Karthikeyan S, et al. Inhibitory effect of caffeic acid on cancer cell proliferation by oxidative mechanism in human HT-1080 fibrosarcoma cell line. Mol Cell Biochem 2011; 349(1–2): 11-9. Ikeda K, Tsujimoto K, Uozaki M, et al. Inhibition of multiplication of herpes simplex virus by caffeic acid. Int J Mol Med 2011; 28(4): 595-8. Jung UJ, Lee MK, Park YB, et al. Antihyperglycemic and antioxidant properties of caffeic acid in db/db mice. J Pharmacol Exp Ther 2006; 318(2): 476-83. Castelluccio C, Paganga G, Melikian N, et al. Antioxidant potential of intermediates in phenylpropanoid metabolism in higher plants. FEBS Letters 1995; 368: 188-92. Kono Y, Kobayashi K, Tagawa S, et al. Antioxidant activity of polyphenolics in diets. Rate constants of reactions of chlorogenic acid and caffeic acid with reactive species of oxygen and nitrogen. BiochimBiophys Acta 1997; 1335: 335-42. Gulcin I. Antioxidant activity of caffeic acid (3, 4dihydroxycinnamic acid). Toxicology 2006; 217: 213-20. Koshihara Y, Neichi T, Murota S, et al. Caffeic acid is a selective inhibitor for leukotriene biosynthesis. Biochim Biophys Acta 1984; 792: 92-7. Nardini M, Scaccini C, Packer L, et al. In vitro inhibition of the activity of phosphorylase kinase, protein kinase C and protein kinase A by caffeic acid and a procyanidin-rich pine bark (Pinus marittima) extract. Biochim Biophys Acta 2000; 1474: 219-25. Nardini M, Leonardi F, Scaccini C, et al. Modulation of ceramideinduced NF-kappaB binding activity and apoptotic response by caffeic acid in U937 cells: comparison with other antioxidants. Free Radic Biol Med 2001; 30: 722-33. Zhang L, Zhang WP, Chen KD, et al. Caffeic acid attenuates neuronal damage, astrogliosis and glial scar formation in mouse brain with cryoinjury. Life Sci 2007; 80: 530-7. Hepsen IF, Bayramlar H, Gultek A, et al. Caffeic acid phenethyl ester to inhibit posterior capsule opacification in rabbits. J Cataract Refract Surg 1997; 23: 1572-6. Ilhan A, Koltuksuz U, et al. The effects of caffeic acid phenethyl ester (CAPE) on spinal cord ischemia/reperfusion injury. Eur J Cardiothorac Surg 1999; 16: 458–463. Koltuksuz U, Ozen S, Uz E, et al. Caffeic acid phenethyl ester prevents intestinal reperfusion injury in rats. J Pediatr Surg 1999; 34: 1458-62. Celik S, Erdogan S, Tuzcu M. Caffeic acid phenethyl ester (CAPE) exhibits significant potential as an antidiabetic and liver-protective agent in streptozotocin-induced diabetic rats. Pharmacol Res 2009; 60(4): 270-6. Chuu CP, Lin HP, Ciaccio MF, et al. Caffeic acid phenethyl ester suppresses the proliferation of human prostate cancer cells through inhibition of p70S6K and Akt signaling networks. Cancer Prev Res (Phila) 2012; 5(5): 788-97. Wei X, Zhao L, Ma Z, et al. Caffeic acid phenethyl ester prevents neonatal hypoxic–ischaemic brain injury. Brain 2004; 127: 262935. Noelker C, Bacher M, Gocke P, et al. The flavanoide caffeic acid phenethyl ester blocks 6-hydroxydopamineinduced neurotoxicity. Neurosci Lett 2005; 383: 39-43. Gocer H, Gulcin I. Caffeic acid phenethyl ester (CAPE): correlation of structure and antioxidant properties. Int J Food Sci Nutr 2011; 62(8): 821-5. Zhao J, Pati S, Redell JB, et al. Caffeic acid phenethyl ester protects blood-brain barrier integrity and reduces contusion volume in rodent models of traumatic brain injury. J Neurotrauma 2012; 29: 1209-18. Kerman M, Kanter M, Cokun KK, et al. Neuroprotective effects of Caffeic acid phenethyl ester on experimental traumatic brain injury in rats. J Mol Hist 2012; 43: 49-57. Ozyurt H, Irmak MK, Akyol O, et al. Caffeic acid phenethyl ester changes the indices of oxidative stress in serum of rats with renal ischemia-reperfusion injury. Cell Biochem Funct 2001; 19: 259-63. Khan M, Khan M, Elango C, et al. Caffeic acid phenethyl ester reduces neurovascular inflammation and protects rat brain following transient focal cerebral ischemia. J Neurochem 2007; 102: 36577. Tian J, Li G, Liu Z, et al. Hydroxysafflor Yellow A inhibits rat brain mitochondrial permeability transition pores by a free radical scavenging action. Pharmacology 2008; 82: 121-6.

ut

[100]

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 113

nl y

Pharmacological Benefits of Active Components of Natural Products

[137]

[138]

[139]

[140]

[141] [142] [143] [144]

[145] [146]

[147]

114 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2

[159] [160]

[161] [162]

[163]

[164] [165]

[166] [167]

[168] [169]

[170]

[176]

[177] [178]

O

[180]

[181]

[182]

[183]

io n

nl y

[179]

ut

[158]

[175]

rib

[157]

[174]

Fukui M, Choi HJ, Zhu BT. Mechanism for the protective effect of resveratrol against oxidative stress- induced neuronal death. Free Radic Biol Med 2010; 49: 800-13. Bastianetto S, Quirion R. Heme oxygenase 1: another possible target to explain the neuroprotective action of resveratrol, a multifaceted nutrient-based molecule. Exp Neurol 2010; 225: 237-9. Shetty AK. Promise of resveratrol for easing status epilepticus and epilepsy. Pharmacol Ther 2011;131: 269-86. Zhuang H, Kim YS, Koehler RC, et al. Potential mechanism by which resveratrol, a red wine constituent, protects neurons. Ann NY Acad Sci 2003; 993: 276-86. Gao D, Zhang X, Jiang X, et al. Resveratrol reduces the elevated level of MMP-9 induced by cerebral ischemia–reperfusion in mice. Life Sci 2006; 78: 2564-70. Rocha-Gonzalez HI, Ambriz-Tututi M, Granados-Soto V. Resveratrol: a natural compound with pharmacological potential in neurodegenerative diseases. CNS Neurosci Ther 2008; 14: 234-47. Kubota S, Kurihara T, Ebinuma M, et al. Resveratrol prevents light-induced retinal degeneration via suppressing activator protein1 activation. Am J Pathol 2010; 177: 1725-31. Valdecantos MP, Pérez-Matute P, Quintero P, et al. Vitamin C, resveratrol and lipoic acid actions on isolated rat liver mitochondria: all antioxidants but different. Redox Rep 2010; 15: 207-16. Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health—a comprehensive review of human clinical trials. Mol Nutr Food Res 2011; 55: 1129-41. Gao ZB, Chen XQ, Hu GY. Inhibition of excitatory synaptic transmission by trans-resveratrol in rat hippocampus. Brain Res 2006; 1111: 41-7. Ates O, Cayli S, Altinoz E, et al. Neuroprotection by resveratrol against traumatic brain injury in rats. Mol Cell Biochem 2007; 294: 137-44. Singleton RH, Yan HQ, Fellows-Mayle W, et al. Resveratrol attenuates behavioral impairments and reduces cortical and hippocampal loss in a rat controlled cortical impact model of traumatic brain injury. J Neurotrauma 2010; 27: 1091-9. Morris RGM, Garrud P, Rawlins JNP, et al. Place navigation impaired in rats with hippocampus lesions. Nature 1982; 297: 681-3. Tong W, Igarashi T, Ferriero DM, et al. Traumatic brain injury in the immature mouse brain: characterization of regional vulnerability. Exp Neurol 2002; 176: 105-16. Ozdemir D, Tugyan K, Uysal N, et al. Protective effect of melatonin against head trauma-induced hippocampal damage and spatial memory deficits in immature rats. Neurosci Lett 2005; 385: 234-9. Sonmez U, Sonmez A, Erbil G, et al. Neuroprotective effects of resveratrol against traumatic brain injury in immature rats. Neurosci Lett 2007; 420: 133-7. Ji XY, Tan BK, Zhu YZ. Salvia miltiorrhiza and ischemic diseases. Acta Pharmacol Sin 2000; 21: 1089-94. Chen J, Wang F, Lee FS, et al. Separation and identification of water-soluble salvianolicacids from Salvia miltiorrhiza Bunge by high-speed counter-current chromatography and ESI-MS analysis. Talanta 2006; 69: 172-9. Lay IS, Hsieh CC, Chiu JH, et al. Salvianolic acid B enhances in vitro angiogenesis and improves skin flap survival in Sprague– Dawleyrats. J Surg Res 2003; 115: 279-85. Tang M, Feng W, Zhang Y, et al. Salvianolic acid B improves motor function after cerebral ischemia in rats. Behav Pharmacol 2006; 17: 493-8. Liu CL, Xie LX, Li M, et al. Salvianolic acid B inhibits hydrogen peroxide-induced endothelial cell apoptosis through regulating PI3K/Akt signaling. Plos One 2007; 2: 1321. Liu H, Liu ZH, Chen ZH, et al. Triptolide: a potent inhibitor of NFkappa B in T-lymphocytes. Acta Pharmacol Sin 2000; 21: 782-6. Hu K, Liu Z, Liu D. Triptolide inhibits vascular endothelial growth factor expression and production by endothelial cells. Zhonghua Yixue Zazhi 2001; 81: 1106-9. He JK, Gu ZL, Yu SD, et al. Triptolide on inhibition of reperfusion injury and prolongation of allograft survival in rat cardiac transplants. Chin J New Drugs Clin 2001; 20: 423-6. Zhou HF, Liu XY, Niu DB, et al. Trip-tolide protects dopaminergic neurons from inflammation-mediated damageinduced by lipopolysaccharide intranigral injection. Neurobiol Dis 2005; 18: 441-9. Wang J, Shi ZQ, Xu X, et al. Triptolide inhibitsamyloid-beta production and protects neural cells by inhibiting CXCR2 activity. J Alzheim Dis 2013; 33: 217-29.

[184]

is t

[156]

[173]

se

[155]

[172]

[185]

rD

[154]

[171]

U

[153]

fo

[152]

al

[151]

N ot

[150]

rs on

[149]

Pe

[148]

tein (25–35)-induced neurotoxicity in cultured rat cortical neurons. Biol Pharm Bull 2006; 29: 2419-24. Andrabi SA, Spina MG, Lorenz P, et al. Oxyresveratrol (trans-2, 3, 4, 5-tetrahydroxystilbene) is neuroprotective and inhibits the apoptotic cell death in transient cerebral ischemia. Brain Res 2004; 1017: 98-107. Jeon SY, Kwon SH, Seong YH, et al. Beta-secretase (BACE1)inhibiting stilbenoids from smilax rhizoma. Phytomedicine 2007; 14: 403-8. Chao J, Yu MS, Ho YS, et al. Dietary oxyresveratrol prevents parkinsonian mimetic 6-hydroxydopamine neurotoxicity. Free Radic Biol Med 2008; 45: 1019-26. Weber JT, Lamont M, Chibrikova L, et al. Potential neuroprotective effects of oxyresveratrol against traumatic injury. Eur J Pharmacol 2012; 680: 55-62. Jatana M, Giri S, Ansari MA, et al. Inhibition of NF-kappaB activation by 5-lipoxygenase inhibitors protects brain against injury in a rat model of focal cerebral ischemia. J Neuroinflamm 2006; 3: 12. Peng QL, Buz Zard AR, Lau BH. Pycnogenol protects neurons from amyloid-beta peptide-induced apoptosis. Brain Res Mol Brain Res 2002; 104: 55-65. Ansari MA, Keller JN, Scheff SW. Protective effect of Pycnogenol in human neuroblastoma SH-SY5Y cells following acroleininduced cytotoxicity. Free Radic Biol Med 2008; 45: 1510-9. Rohdewald P. A review of the French maritime pine bark extract (Pycnogenol), a herbal medication with a diverse clinical pharmacology. Int J Clin Pharmacol Ther 2002; 40: 158-68. Packer L Interactions among antioxidants in health and disease: vitamin E and its redox cycle. Proc Soc Expt Biol Med 1991; 200: 271-6. Emerit I. Reactive oxygen species, chromosome mutation, and cancer: possible role of clastogenic factors in carcinogenesis. Free Radic Biol Med 1994; 16: 99-109. Nelson AB, Lau BH, Ide N. Pycnogenol inhibits macrophage oxidative burst, lipoprotein oxidation and hydroxyl radical induced DNA damage. Drug Dev Ind Med 1998; 24: 1-6. Packer L, Rimbach G, Virgili F. Antioxidant activity and biologic properties of a procyanidin-rich extract from pine (Pinus maritima) bark, pycnogenol. Free Radic Biol Med 1999; 27: 704-24. Scheff SW, Ansari MA, Roberts KN. Neuroprotective effect of Pycnogenol® following traumatic brain injury. Exp Neurol 2012; 239: 183-91. Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006; 444: 33742. Lagouge M, Argmann C, Gerhart-Hines Z, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 2006; 127: 110922. Gordon BS, Delgado Díaz DC, Kostek MC. Resveratrol decreases inflammation and increases utrophin gene expression in the mdx mouse model of duchenne muscular dystrophy. Clin Nutr 2013; 32: 104-11. Bradamante S, Barenghi L, Villa A. Cardiovascular protective effects of resveratrol. Cardiovas Drug Rev 2004; 22(3): 169-88. Su HC, Hung LM, Chen JK. Resveratrol, a red wine antioxidant, possesses an insulin-like effect in streptozotocin-induced diabetic rats. Am J Physiol Endocrinol Metab 2006; 290: 1339-46. Delmas D, Jannin B, Latruffe N. Resveratrol: preventing properties against vascular alterations and ageing. Mol Nutr Food Res 2005; 49: 377-95. Jang M, Cai L, Udeani GO, et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997; 275: 218-20. Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov 2006; 5: 493-06. Kumar A, Naidu PS, Seghal N, et al. Neuroprotective effects of resveratrol against intracerebroventricular colchicine-induced cognitive impairment and oxidative stress in rats. Pharmacology 2007; 79: 17–26. Kim D, Nguyen MD, Dobbin MM, et al. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. EMBO J 2007; 26: 3169-79.

Subash et al.

[186]

[187] [188]

[189] [190]

[191]

[192] [193]

[194] [195]

[196]

[209]

[210] [211]

[212] [213] [214]

[215] [216]

[217]

[218]

[219] [220] [221]

[227] [228]

[229]

[231]

[232] [233]

io n

[230]

ut

[208]

[226]

rib

[207]

[225]

O

[206]

[224]

Bei R, Frigiola A, Masuelli L, et al. Effects of omega-3 polyunsaturated fatty acids on cardiac myocyte protection. Front Biosci 2011; 16: 1833-43. Pawlik D, Lauterbach R, Turyk E. Fish-oil fat emulsion supplementation may reduce the risk of severe retinopathy in VLBW infants. Pediatrics 2011; 127: 23-228. Wheeler TG, Benolken RM, Anderson RE. Visual membranes: specificity of fatty acid precursors for the electrical response to illumination. Science 1975; 188: 1312-4. Moriguchi T, Greiner RS, Salem NJr. Behavioral deficits associated with dietary induction of decreased brain docosahexaenoic acid concentration. J Neurochem 2000; 75: 2563–2573. Niu SL, Mitchell DC, Lim SY, et al. Reduced G protein-coupled signaling efficiency in retinal rod outer segments in response to n-3 fatty acid deficiency. J Biol Chem 2004; 279: 31098-31104. Martinez M. Severe deficiency of docosahexaenoic acid in peroxisomal disorders: a defect of delta 4 desaturation?. Neurology 1990; 40: 1292-8. Soderberg M, Edlund C, Kristensson K, et al. Fatty acid composition of brain phospholipids in aging and in Alzheimer's disease. Lipids 1991; 26: 421-5. Pu H, Guo Y, Zhang W, et al. Omega-3 polyunsaturated fatty acid supplementation improves neurologic recovery and attenuates white matter injury after experimental traumatic brain injury. J Cereb Blood Flow Metab 2013; 33: 1474-84. Desai A, Kevala KR, Kim HY. Depletion of brain docosahexaenoic acid (DHA) deficiency impairs recovery from traumatic brain injury. PLoS ONE 2014; 9(1): e86472. Parvathy RK, Essa MM, Al-Adawi S, et al. Omega-3 fatty acids could alleviate the risks of traumatic brain injury – a mini review. J Trad Complem Med 2014; 4(2): 89-92. Greco T, Prins ML. Traumatic brain injury and diet. J Child Neurol 2013; 28(2): 983-8. Seo HB, Kang BK, Kim JH, Choi YW, Hong JW, Choi BT, Shin HK. Partially purified components of Uncaria sinensis attenuate blood brain barrier disruption after ischemic brain injury in mice. BMC Complement Altern Med 2015; 27(15): 157. Harada S, Tsujita T, Ono A, Miyagi K, Mori T, Tokuyama S. Stachys sieboldii (Labiatae, Chorogi) protects against learning and memory dysfunction associated with ischemic brain injury. J Nutr Sci Vitaminol (Tokyo). 2015; 61(2): 167-74. Zhang D, Lee B, Nutter A, Song P, Dolatabadi N, Parker J, SanzBlasco S, Newmeyer T, Ambasudhan R, McKercher SR, Masliah E, Lipton SA. Protection from cyanide-induced brain injury by the Nrf2 transcriptional activator carnosic acid. J Neurochem 2015; 133(6): 898-908. Yuan J, Zou M, Xiang X, Zhu H, Chu W, Liu W, Chen F, Lin J. Curcumin improves neural function after spinal cord injury by the joint inhibition of the intracellular and extracellular components of glial scar. J Surg Res. 2015 May 1; 195(1): 235-45. Rakhunde PB, Saher S, Ali SA. Neuroprotective effect of Feronia limonia on ischemia reperfusion induced brain injury in rats. Indian J Pharmacol. 2014 Nov-Dec; 46(6): 617-21. Yang ST, Lin JW, Chiu BY, Hsu YC, Chang CP, Chang CK. Astragaloside improves outcomes of traumatic brain injury in rats by reducing microglia activation. Am J Chin Med. 2014; 42(6): 135770. Akbar M, Essa MM, Daradkeh G, Abdelmegeed MA, Choi Y, Mahmood L, Song BJ. Mitochondrial dysfunction and cell death in neurodegenerative diseases through nitroxidative stress. Brain Res. 2016; 1637: 34-55. Nataraj J, Manivasagam T, Justin Thenmozhi A, Essa MM. Neuroprotective effect of asiatic acid on rotenone-induced mitochondrial dysfunction and oxidative stress-mediated apoptosis in differentiated SH-SYS5Y cells. Nutr Neurosci 2016 Feb 9. [Epub ahead of print] Dhanalakshmi C, Manivasagam T, Nataraj J, Justin Thenmozhi A, Essa MM. Neurosupportive role of vanillin, a natural phenolic compound, on rotenone induced neurotoxicity in SH-SY5Y neuroblastoma cells. Evid Based Complement Alternat Med. 2015; 2015: 626028. doi: 10.1155/2015/626028. Epub 2015 Nov 17. Subash S, Essa MM, Braidy N, Awlad-Thani K, Vaishnav R, AlAdawi S, Al-Asmi A, Guillemin GJ. Diet rich in date palm fruits improves memory, learning and reduces beta amyloid in transgenic mouse model of Alzheimer's disease. J Ayurveda Integr Med 2015; 6(2): 111-20.

[234]

is t

[205]

[223]

se

[204]

[222]

[235]

rD

[203]

U

[202]

fo

[201]

al

[200]

N ot

[199]

rs on

[198]

Su Z, Yuan Y, Cao L, et al. Triptolide promotes spinal cordrepair by inhibiting astrogliosis and inflammation. Glia 2010; 58: 901-15. Lee HF, Lee TS, Kou YR. Anti-inflammatory and neuroprotective effects oftriptolide on traumatic brain injury in rats. Respir Physiol Neurobiol 2012; 182: 1-8. Lee HF, Lee TS, Kou YR. Anti-inflammatory and neuroprotective effects of triptolide on traumatic brain injury in rats. Respir Physiol Neurobiol 2012; 182: 1-8. Cho J, Lee HK. Wogonin inhibits ischemic brain injury in a rat model of permanent middle cerebral artery occlusion. Biol Pharm Bull 2004; 27: 1561-4. Piao HZ, Jin S.A, Chun HS, et al. Neuroprotective effect of wogonin: potential roles of inflammatory cytokines. Arch Pharm Res 2004; 27: 930-6. Cho J, Lee HK. Wogonin inhibits excitotoxic and oxidative neuronal damage in primary cultured rat cortical cells. Eur J Pharmacol 2004; 485: 105-10. Son D, Lee P, Lee J, et al. Neuroprotective effect of wogonin in hippocampal slice culture exposed to oxygen and glucose deprivation. Eur J Pharmacol 2004; 493: 99-102. Liu XF, Liu ML, Iyanagi T, et al. Inhibition of rat liver NAD(P)H:quinone acceptor oxidoreductase (DT-diaphorase) by flavonoids isolated from the Chinese herb scutellariae radix (Huang Qin). Mol Pharmacol 1990; 37: 911-5. Chen CC, Hung TH, Wang YH, et al. Wogonin Improves Histological and Functional Outcomes, and Reduces Activation of TLR4/NF-kB Signaling after Experimental Traumatic Brain Injury. PLoS ONE 2012; 7: 30294. Garodia P, Ichikawa H, Malani N, et al. From ancient medicine to modern medicine: ayurvedic concepts of health and their role in inflammation and cancer. J Soc Integr Oncol 2007; 5: 25-37. Martin-Aragon S, Benedi JM, Villar AM. Modifications on antioxidant capacity and lipid peroxidation in mice under fraxetin treatment. J Pharm Pharmacol 1997; 49: 49-52. Shaikh J, Ankola DD, Beniwal V, et al. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur J Pharm Sci 2009; 37: 223-0. Ganguli M, Chandra V, Kamboh MI, et al. Apolipoprotein E. polymorphism and Alzheimer's disease: the Indo-US CrossNational Dementia Study. Arch Neurol 2000; 57: 824–830. Singh S, Aggarwal BB. Activation of transcription factor NF-kappa B is suppressed by curcumin (diferuloylmethane). J Biol Chem 1995; 270: 24995-5000. Lim GP, Chu T, Yang F, et al. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J Neurosci 2001; 21: 8370-7. Thiyagarajan M, Sharma SS. Neuroprotective effect of curcumin in middle cerebral artery occlusion induced focal cerebral ischemia in rats. Life Sci 2004; 74: 969-85. Cole GM, Teter B, Frautschy SA. Neuroprotective effects of curcumin. Adv Exp Med Biol 2007; 595: 197-12. Wakade C, King MD, Laird MD, et al. Curcumin attenuates vascular inflammation and cerebral vasospasm after subarachnoid hemorrhage in mice. Antioxid Redox Signal 2009; 11: 35-45. Zhao J, Zhao Y, Zheng W, et al. Neuroprotective effect of curcumin on transient focal cerebral ischemia in rats. Brain Res 2008; 1229: 224-32. Sumanont Y, Murakami Y, Tohda M, et al. Prevention of kainic acid-induced changes in nitric oxide level and neuronal cell damage in the rat hippocampus by manganese complexes of curcumin and diacetylcurcumin. Life Sci 2006; 78: 1884-91. Sharma S, Zhuang Y, Ying Z, et al. Dietary curcumin supplementation counteracts reduction in levels of molecules involved in energy homeostasis after brain trauma. Neuroscience 2009; 161: 1037-44. Salem, N Jr, Kim HY, Yergey JA. In: Simopoulos AP, Kifer RR, RE Martin, Ed. Docosahexaenoic acid: membrane function and metabolism. In: Health effects of polyunsaturated fatty acids in seafoods. Academic, New York, 1986; pp. 263–317. Calder PC, Yaqoob P Omega-3 polyunsaturated fatty acids and human health outcomes. Biofactors 2009; 35: 266–272 Mori TA, Beilin LJ. Omega-3 fatty acids and inflammation. Curr Atheroscler Rep 2004; 6: 461-7. Lavie CJ, Milani RV, Mehra MR, et al. Omega-3 polyunsaturated fatty acids and cardiovascular diseases, J Am Coll Cardiol 2009; 54: 585-94.

Pe

[197]

Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 115

nl y

Pharmacological Benefits of Active Components of Natural Products

[236]

[237]

[238]

[239]

[240]

[241]

[242]

116 Current Pharmacogenomics and Personalized Medicine, 2015, Vol. 13, No. 2 [248]

[249]

[250]

[251]

[252]

Subash S, Essa MM, Al-Adawi S, Memon MA, Manivasagam T, Akbar M. Neuroprotective effects of berry fruits on neurodegenerative diseases. Neural Regen Res. 2014 Aug 15; 9(16): 1557-66. Al-Sabahi BN, Fatope MO, Essa MM, Subash S, Al-Busafi SN, AlKusaibi FS, Manivasagam T. Pomegranate seed oil: Effect on 3nitropropionic acid-induced neurotoxicity in PC12 cells and elucidation of unsaturated fatty acids composition. Nutr Neurosci 2014 Sep 19. [Epub ahead of print] Subash S, Essa MM, Al-Asmi A, Al-Adawi S, Vaishnav R. Chronic dietary supplementation of 4% figs on the modification of oxidative stress in Alzheimer's disease transgenic mouse model. Biomed Res Int. 2014;2014:546357. doi: 10.1155/2014/546357. Epub 2014 Jun 19. Muthaiyah B, Essa MM, Lee M, Chauhan V, Kaur K, Chauhan A. Dietary supplementation of walnuts improves memory deficits and learning skills in transgenic mouse model of Alzheimer's disease. J Alzheimers Dis. 2014; 42(4): 1397-405. Subash S, Essa MM, Al-Asmi A, Al-Adawi S, Vaishnav R, Guillemin GJ. Effect of dietary supplementation of dates in Alzheimer's disease APPsw/2576 transgenic mice on oxidative stress and antioxidant status. Nutr Neurosci. 2015 Aug;18(6): 281-8. Subash S, Essa MM, Braidy N, Al-Jabri A, Vaishnav R, Al-Adawi S, Al-Asmi A, Guillemin GJ. Consumption of fig fruits grown in Oman can improve memory, anxiety, and learning skills in a transgenic mice model of Alzheimer's disease. Nutr Neurosci 2014; 18. [Epub ahead of print]

io n

se

O

nl y

[253]

rD

is t

rib

ut

U fo

[247]

al

[246]

N ot

[245]

rs on

[244]

Essa MM, Subash S, Dhanalakshmi C, Manivasagam T, Al-Adawi S, Guillemin GJ, Justin Thenmozhi A. Dietary supplementation of walnut partially reverses 1-Methyl-4-phenyl-1,2,3,6tetrahydropyridine Induced Neurodegeneration in a mouse model of Parkinson's disease. Neurochem Res 2015; 40(6): 1283-93. doi: 10.1007/s11064-015-1593-2. Essa MM, Subash S, Akbar M, Al-Adawi S, Guillemin GJ. Longterm dietary supplementation of pomegranates, figs and dates alleviate neuroinflammation in a transgenic mouse model of Alzheimer's disease. PLoS One 2015; 10(3): e0120964. doi: 10.1371/journal.pone.0120964. eCollection 2015. Nataraj J, Manivasagam T, Thenmozhi AJ, Essa MM. Lutein protects dopaminergic neurons against MPTP-induced apoptotic death and motor dysfunction by ameliorating mitochondrial disruption and oxidative stress. Nutr Neurosci. 2015 Mar 2. [Epub ahead of print] Subash S, Braidy N, Essa MM, Zayana AB, Ragini V, Al-Adawi S, Al-Asmi A, Guillemin GJ. Long-term (15 mo) dietary supplementation with pomegranates from Oman attenuates cognitive and behavioral deficits in a transgenic mice model of Alzheimer's disease. Nutrition 2015; 31(1): 223-9. doi: 10.1016/j.nut.2014.06.004. [Epub 2014 Jun 25]. Subash S, Essa MM, Al-Asmi A, Al-Adawi S, Vaishnav R, Braidy N, Manivasagam T, Guillemin GJ. Pomegranate from oman alleviates the brain oxidative damage in transgenic mouse model of Alzheimer's disease. J Tradit Complement Med. 2014; 4(4): 232-8. doi: 10.4103/2225-4110.139107.

Pe

[243]

Subash et al.