Open access peer-reviewed chapter

Extracts and Essential Oils from Medicinal Plants and Their Neuroprotective Effect

Written By

Ianara Mendonça da Costa, Elaine Cristina Gurgel Andrade Pedrosa, Ana Paula de Carvalho Bezerra, Luciana Cristina Borges Fernandes, José Rodolfo Lopes de Paiva Cavalcanti, Marco Aurélio Moura Freire, Dayane Pessoa de Araújo, Amália Cinthia Meneses do Rego, Irami Araujo Filho, Francisco Irochima Pinheiro and Fausto Pierdoná Guzen

Submitted: 24 June 2019 Reviewed: 19 December 2019 Published: 28 February 2020

DOI: 10.5772/intechopen.90903

From the Edited Volume

Neuroprotection - New Approaches and Prospects

Edited by Matilde Otero-Losada, Francisco Capani and Santiago Perez Lloret

Chapter metrics overview

1,068 Chapter Downloads

View Full Metrics

Abstract

Current therapies for neurodegenerative diseases offer only limited benefits to their clinical symptoms and do not prevent the degeneration of neuronal cells. Neurological diseases affect millions of people around the world, and the economic impact of treatment is high, given that health care resources are scarce. Thus, many therapeutic strategies to delay or prevent neurodegeneration have been the subject of research for treatment. One strategy for this is the use of herbal and essential oils of different species of medicinal plants because they have several bioactive compounds and phytochemicals with neuroprotective capacity. In addition, they respond positively to neurological disorders, such as dementia, oxidative stress, anxiety, cerebral ischemia, and oxidative toxicity, suggesting their use as complementary treatment agents in the treatment of neurological disorders.

Keywords

  • neuroprotection
  • herbal medicines
  • neurological disorders
  • oxidative stress

1. Introduction

A number of complementary treatment are currently being investigated to provide neuroprotection or to treat neurodegenerative diseases. Some therapies are known to provide limited benefits because, despite their treating the clinical symptoms, they are not effective in preventing neuronal cell degeneration.

The economic impact of treating neurodegenerative disorders is also high with disproportionately scarce neurological services and resources that patient survival may depend on. Studies have shown that over 80% of natural deaths in low- and middle-income countries may be attributed to stroke [1]. In the United States alone, the combined annual costs of neurological diseases total nearly $ 800 billion, expected to increase in the coming years due to an aging population, resulting in a severe economic burden to the health system [2].

Recent advances in understanding the pathophysiological mechanisms of neurological disorders have led to new strategies in drug development. Animal models have contributed considerably to these advances, as they play an important role in evaluating potential drugs that can alleviate these conditions and also delay their processes [3].

Interest in natural products has increased significantly, resulting in the increasing use of herbal medicines [4]. In a recent review, Izzo et al. report a 6.8% increase in US herbal and food supplement sales in 2014, with an estimated over $ 6.4 billion in total sales [5].

The clinical and social repercussions of neuropathologies reveal an important theme of study and commitment to structure strategies that can contribute to the quality of life of society. Scientific research has explored which stimuli and substances can contribute to neural cell plasticity, resulting in improved quality of life for people with depression, Alzheimer’s Disease (AD), Parkinson’s Disease (PD), among other nervous system-related disorders [6].

Increased neurogenesis and the facilitating effects of plasticity can be produced by a variety of treatments, including enriched environment, physical activity or drug action [7]. A complementary treatment proposed is the use of herbal medicines, which have scientific relevance in the treatment of neurological diseases because they contain multiple compounds and phytochemicals that can have neuroprotective effect, with a consequent beneficial action for health in different neuropsychiatric and neurodegenerative disorders [8].

Advertisement

2. Neuroprotective effect of extracts

Studies have investigated therapies that can alleviate the symptoms of neurodegenerative disorders and also avoid the multiple pathogenic factors involved in these diseases. One promising approach is the use of herbal extracts and their isolated bioactive compounds for the treatment of conditions such as Parkinson’s, Alzheimer’s, cerebral ischemia. Behavioral analysis has shown them to have neurochemical activity and symptom reduction [9].

Recent advances in understanding the pathophysiological mechanisms related to neurodegenerative diseases point to new strategies in drug development [10]. Animal models have contributed considerably to these advances and play an even greater role in evaluating possible drugs with therapeutic potential, not only to alleviate these pathologies, but also to modify the disease process [3]. Rodents are suitable models for these studies because of their well-characterized brain organization and the magnitude of information focused on altered states of the nervous system [11, 12].

Phytotherapics have scientific relevance in the treatment of neurological diseases, as they contain multiple compounds and phytochemicals that can have neuroprotective effects, with consequent beneficial health action between different neuropsychiatric and neurodegenerative disorders [8, 9, 10]. Several extracts that have shown beneficial action in these disorders as will be addressed in this paper.

2.1 Alzheimer’s disease

Alzheimer’s disease (AD) is a neurodegenerative pathology that results in progressive loss of cell function, structure and number, leading to widespread brain atrophy and profound cognitive and behavioral deficit [13]. Histopathologically, it is characterized by accumulation of beta-amyloid peptide (ßA), which can initiate a cascade of oxidative events and chronic inflammation leading to neuronal death [14].

Several studies have investigated the action of Piper methysticum in experimental models of neurodegenerative diseases, specifically in AD, demonstrating the neuroprotective effect of this herbal medicine [15, 16, 17].

Piper methysticum is popularly known as Kava or Kava-kava, a perennial shrub belonging to the Pacific Ocean pepper family (Piperaceae) with historical and cultural significance is described in the literature as a compound that has neuroprotective action and anxiolytic effects and is used in sedatives, and analgesics, being anti-inflammatory, anticonvulsant and anti-ischemic. Most of these pharmacological effects have been attributed to six kavalactones isolated from kava extracts, including yangonin, kawain and methysticin, dihydromethysticin, dihydrokavain and desmethoxyyangonin [18].

Recent studies, such as Fragoulis et al. have shown that one of the possible explanations for the action of piper mechanism in AD is associated with the activation of the erythroid2-related nuclear factor (NrF2) [15].

Nrf2 is the major regulator of phase II detoxifying/antioxidant enzymes, including heme oxygenase 1 (HO-1). Transcription factor Nrf2 binds to ARE (antioxidant response element), transcribing a battery of genes involved in redox status, anti-inflammatory response and detoxification [19]. A study by Lobota et al. reports that Nrf2 activation and HO-1 induction are involved in the regulation of inflammation [20].

Another study developed to find agents that activate the Nrf2 factor was performed and three analytically pure kavalactones - Methysticin, Yangonin and Kavain - were researched. The effects of kavalactones on the protection of neural cells against beta-amyloid peptide (ßA)-induced neurotoxicity were evaluated using the ARE-luciferase and Western blot assay. The results indicated that kavalactones Methysticin, Yangonin and Kavain activate time and dose-dependent Nrf2/ARE in astroglial PC-12 and C6 neural C6 cells and thus up-regulate cytoprotective genes. At the same time, viability and cytotoxicity assays have shown that Nrf2 activation is able to protect neuronal cells from neurotoxicity by attenuating neuronal cell death caused by β amyloid [14].

Taken together, it is understood that the Nrf2/ARE signaling pathway is an attractive therapeutic target for neurodegenerative diseases and that chemically modified kavalactones as well as naturally occurring kavalactones can attenuate neurological damage by reducing oxidative stress and neuroinflammation.

Some herbal medicines have shown neuroprotective effects, such as curcumin, which is the main polyphenol found in turmeric (Curcuma longa), belonging to the Zingiberaceae family, native to South Asia and cultivated in the tropics [21]. It has been reported that this compound has properties that can prevent or ameliorate pathological processes related to neurodegenerative diseases such as cognitive decline, dementia and mood disorders [22]. In addition, curcumin has been investigated for experimental models of treatment for Parkinson’s disease and has shown hopeful results [9].

Saffron compounds have been linked to beneficial biological properties such as anti-inflammatory, pro-apoptotic, antiproliferative, anti-amyloidogenic, antioxidant, antiviral, and antidiabetic [23, 24]. Saffron’s most bioactive constituents are curcuminoids, including curcumin and its derivatives such as demethoxycurcumin and bisdemetoxycurcumin [25, 26].

The features attributed to curcumin, such as inhibition of amyloid pathology, protection against inflammation and oxidative stress, inhibition of beta amyloid plaque aggregation and tau protein hyperphosphorylation, suggest that this compound may prevent or improve pathological processes related to cognitive decline and dementia, as occur in the symptomatology of AD patients [27, 28].

A systematic review study showed that curcumin has a positive action on AD symptoms, both when assessing biochemical and behavioral symptoms. The proposed mechanisms of its action in AD show that it is able to act by preventing the formation and aggregation of β-amyloid protein and tau protein hispanphosphorylation [10], in addition curcumin has also been shown to prevent neural damage, mitochondrial disorders, cellular stress and glial hyperactivation, as shown in Figure 1.

Figure 1.

Active curcumin mechanisms after experimental treatment in AD models. Curcumin acts by preventing the formation and aggregation of β-amyloid protein and hyperphosphorylation of tau protein, stabilizing microtubules and preventing the formation of neurofibrillary tangles that occur due to deposition of this protein. It has also been shown to prevent mitochondrial damage favoring the increase of cellular ATP and the healthy maintenance of mitochondria, avoiding excessive Ca2+ intake. Curcumin is also able to counteract cellular stress and glial overactivation.

Another compound that represents a promising approach is astragaloside IV (AS-IV), a triterpenoid saponin present in the root of Astragalus membranaceus (Fisch.) Bge. It is part of Chinese traditional culture [29], first described in the Chinese book Shen Nong Ben Cao Jing in 200 AD with a number of beneficial effects and no toxicity.

The biological and pharmacological properties of AS-IV include its protective effect on pathologies due to its wide range of beneficial actions, such as antioxidant, antibacterial, antiviral [30, 31], anti-inflammatory, anti-asthmatic, antidiabetic, antifibrotic, immunoregulatory and antimicrobial, and cardioprotective effects, preventing myocardial insufficiency in rats [29, 30, 31, 32], able to improve the immune system, digestion and promote wound healing [33].

Astragalus action can be understood based on the regulation of the release of caspases and cytochrome c (both being inducers of apoptosis), since cytochrome binds to Apaf-1 and Procaspase-9c when released into cytosol, forming a functional apoptosome and subsequently triggering the sequential activation of caspase-3 and 9 [34]. Several stimuli that induce apoptosis, leading to the release of mitochondrial cytochrome c which plays a key role in a common pathway of caspase activation [34, 35]. In addition, caspase-3 activation has been shown to be a fundamental step in the apoptosis process and its inhibition may block cellular apoptosis.

In addition, Chang et al. evaluated the action of AS-IV on the cerebral cortex after Aβ infusion, showing that i.p. Administration of 40 mg/kg/day of the herbal compound once daily for 14 days reduced the levels of mitochondrial dysfunction apoptosis in cortical cells blocked by inhibition of phosphoinositol 3-kinase (PI3K) protein kinase, known as AKT [36].

The beneficial effects of AS-IV administration in experimental models of neurodegenerative diseases proved to be effective in both in vivo and in vitro models, such as PD and AD, cerebral ischemia and encephalomyelitis by characterizing the antioxidant, antiapoptotic and anti-inflammatory action of this bioactive compound on the various neurochemical substances and behavioral mechanisms. This suggests that the mechanisms presented by AS-IV offer a possible future complementary treatment for the potential treatment of these pathologies [10].

2.2 Parkinson’s disease

Parkinson’s disease (PD) is a condition that causes progressive neurodegeneration of dopaminergic neurons with the consequent reduction of dopamine content in the substantia nigra. The 6-hydroxydopamine neurotoxin (6-OHDA) is widely used to mimic the neuropathology of PD [37].

There are reports in the literature analyzing the effect of supplementation, including Chinese herbs and herbal extracts that have shown clinical potential to attenuate the progression of PD in humans. In addition, plant extracts act on the neurochemical or motor profile in isolation [38]. It is known, however, that this pathology involves symptomatology related to both characteristics.

A recently published systematic review study discussed studies showing neuroprotective properties of medicinal plants and their bioactive compounds. These included Amburana cearenses (Amburoside A), Camellia sinensis (Catechins and Polyphenols), Gynostemma pentaphyllum (Saponin Extract), Pueraria lobata (Puerarin), Alpinia oxyphylla (Protocatechuic Acid), Cistanches salsa (Glycosides or Phenylethanoids), Spirulina platensis (Polysaccharide), and Astragalus Membranaceus - AS IV Tetracillic Saponin Triterpenoid [9].

As previously mentioned, Astragaloside showed a neuroprotective effect on several AD models. In addition, studies have shown the positive action of AS-IV in PD models. One of the studies induced Parkinson by the action of 6-OHDA, where AS IV attenuated the loss of dopaminergic neurons and the treated group presented intact germination, neurite growth and increased immunoreactive TH and NOS. In addition, when the pathology was induced in SH-SY5Y cell culture by MPP + (DP inducing drug) action, it also significantly reversed cell loss, nuclear condensation, intracellular generation of reactive oxygen species and pathway inhibition as mediated by Bax; these effects, however, were related only to neurochemical analysis. Behavioral findings were not reported [39].

One legume that has become the target of scientific research for its neuroprotective properties is Mucuna pruriens. Behavioral analysis studies have been carried out with Mucuna pruriens (Alkaloids, coumarins, flavonoids, triterpenes, saponins, carotenoids) and Baicalein (Flavonoids) for PD, but no neurochemical evaluation has been performed. There are also publications demonstrating in vivo behavioral effects and in vitro neurochemical analyses, such as a recent publication showing the effect of Ligusticum officinale (Makino) on MPTP (1-methyl-4-phenyl-1,2,3,6)-induced with an animal model and tetrahydropyridine, a neurotoxin capable of permanently causing symptoms of Parkinson’s disease by destroying the dopaminergic neurons of the substantia nigra. This drug has been used to study the disease in experiments with animals; the treatment restored behavior when compared to the control group. In this study, Withania somnifera (Ashwagandha) extract also showed improvement in all these physiological anomalies [9, 40, 41, 42, 43].

Another study investigated the ability of guanosine to protect neuronal PC12 cells from toxicity induced by 1-methyl-4-phenylpyridinium (MPP), the active metabolite of 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP), which mediates selective damage to dopaminergic neurons and causes irreversible Parkinson-like symptoms in humans and primates. The results demonstrated that MPP-induced apoptosis of PC12 cells (cell line derived from a rat adrenal membrane pheochromocytoma) was significantly prevented by guanosine pretreatment for 3 h. In addition, guanosine attenuated the collapse of the MPP-induced mitochondrial transmembrane potential and prevented subsequent activation of caspase-3, thus protecting dopaminergic neurons against mitochondrial stress-induced damage [44].

Other studies have shown plants with neuroprotective properties capable of protecting from PD damage. These include plants such as Amburana Cearenses (Amburoside A) [5], Myracrodruon urundeuva (tannins and chalcones) [45], Camellia sinensis (catechins and polyphenols) [46], Gynostemma pentaphyllum (saponin extract) [47], Pueraria lobata (Puerarin) [48], Alpinia oxyphylla (protocatecholic citric acid) [49], parsley Cistanches salsa (phenylethane glycosides) [50, 51], Spirulina platensis (polysaccharide) [22] and Astragalus membranaceus (triterpenoid saponin), as mentioned in a review study [9, 39].

Current PD medications treat symptoms; none prevent or retard the degeneration of dopaminergic neurons. It is understood that the above-mentioned herbal medicines have neuroprotective properties.

2.3 Neurological disorders/cerebrovascular diseases/brain dysfunctions

Stroke is the second leading cause of death in industrialized countries and the leading medical cause of acquired adult disability [52].

Piper methysticum is cited as a multi-potent phytopharmaceutical due to its numerous pharmacological effects including anxiolytic, sedative, anticonvulsant, anti-ischemic, local anesthetic, anti-inflammatory and analgesic activities. The use of Kava in brain dysfunctions has clinical and financial advantages, acting as an adjunct or complementary treatment to existing medications [53].

Chang et al. [9] have shown that the use of combined glucose and oxygen administration of guanosine (100 μM) significantly reduced the proportion of apoptosis. To determine whether guanosine was also neuroprotective in vivo, middle cerebral artery occlusion (CoA) was performed in male Wistar rats and guanosine (8 mg/kg) intraperitoneally or saline (control vehicle) was administered daily for 7 days. Guanosine prolongs survival and decreased neurological deficits and tissue damage resulting from CoA. These data are the first to demonstrate that guanosine is neuroprotective in stroke.

Through an experimental study developed by Backhauss and Krieglstein [55] that induced focal cerebral ischemia in rodents, through left middle cerebral artery (MCA) microbipolar coagulation, with the objective of evaluating whether kava extract and its constituents, kawain, dihydrokawain, Methysticin, dihydromethysticin and yangonin, are capable of reducing the size of a heart attack zone in rats and mice, providing protection against ischemic brain damage. Compounds were administered ip, except kava extract, which was administered orally. The results demonstrated that Kava extract decreased the infarct area in mouse brains and the infarct volume in rat brains. Methysticin and dihydromethysticin significantly reduced the infarct area in mouse brain, thus evidencing neuroprotective activity of the mice. Kava extract works by the action of its constituent’s methysticin and dihydromethysticin. The other Kavapyronas could not produce a beneficial effect on the infarct area.

The study by Deng et al. examined whether late administration of GUO (guanosine) improved long-term functional recovery after stroke. Late administration of GUO improved functional recovery from day 14 after stroke when compared with the vehicle group [56].

Gerbatin et al. evaluated the effect of guanosine on TBI-induced neurological damage. The findings showed that a single dose of guanosine (7.5 mg/kg), intraperitoneally (i.p) injected 40 min after fluid percussion injury (IPF) in rats protected them from locomotor and exploratory impairment, observed 8 h after injury, guanosine protected against neuronal death and activation of caspase 3 (protein responsible for cleaving genetic material.) This study suggests that guanosine plays a neuroprotective role in TBI and can be explored as a new pharmacological strategy [57].

Experimental models of ischemic stroke help our understanding of the events that occur in the ischemic and reperfused brain. One of the main developments in the treatment of acute ischemic stroke is neuroprotection.

2.4 Psychological disorders/anxiety/depression

Depression and stress-related disorders affect approximately 17% of the population, resulting in enormous personal suffering as well as social and economic burdens [58].

Guanosine is a nucleoside that has a neuroprotective effect. Current studies have analyzed the action of guanosine as an antidepressant. One study investigated the effects of guanosine on the tail suspension test (TT), open field test and adult hippocampal neurogenesis. The results suggest that the antidepressant effect of chronic guanosine use causes an increase in neuronal differentiation, suggesting that this nucleoside may be an endogenous mood modulator [59].

The ability of this nucleoside to nullify acute stress-induced behavioral and biochemical changes has not been evaluated in female mice, given that depression has a greater impact on women. A study aimed at investigating the protective effect of this nucleoside against oxidative damage and stress response evaluated this using the FST (forced swimming test). The Acute Containment Stress Protocol (ARS) has been proposed as a model that triggers biochemical changes in the rat brain that may be detrimental to CNS (central nervous system) function, implicated in several psychiatric disorders, including major depression [60].

Considering that the hippocampus plays a key role in mood regulation, numerous studies have evaluated whether adult hippocampal neurogenesis is altered in psychiatric disorders. Stress is a risk factor for depression that can manifest itself years after the stressful event [54].

Behavioral studies have shown that guanosine produces anxiolytic substances and amnesic effects. Other analyses have shown that reductions induced by hippocampal stress, cell proliferation and/or neuronal differentiation cause depressive symptoms. Deng et al. explain that hippocampal neurogenesis in humans is affected by various neurological disorders, including depression [56].

According to Duman et al. chronic administration of an antidepressant regulates neurogenesis in the hippocampus of adult rodents. Overregulation of neurogenesis could block or reverse the effects of stress on hippocampal neurons, which include downregulation of neurogenesis as well as atrophy. The possibility that the cAMP signal transduction cascade contributes to antidepressant regulation of neurogenesis is supported by previous studies and recent work [61].

Disturbances in hippocampal neurogenesis may be involved in the pathophysiology of depression. It has been argued that an increase in the generation of new hippocampal nerve cells is involved in the mechanism of action of antidepressants. This study, using adult Wistar rats given fluoxetine, showed that a significant behavioral effect occurred. It also pointed out that chronic antidepressant treatment increases cell proliferation as well as neurogenesis in the dentate gyrus. Neurogenesis may serve as an important parameter for examining the efficacy and mechanism of action of new drugs [61].

Anxiety is a diffuse mental condition manifested through unpleasant feelings of fear and apprehension without specific cause [62].

Currently, the psychotherapeutic complementary treatment chosen to treat patients is through antidepressant drugs such as selective serotonin and serotonin-norepinephrine reuptake inhibitors (SSRIs), tricyclic antidepressants and benzodiazepines [63]. Due to the undesirable and destructive side effects of these drugs, including drowsiness, cognitive impairment, and symptoms of dependence and withdrawal, many patients prefer herbal remedies. Several plants with anxiolytic activity have been studied in clinical trials, and Kava (Piper methysticum) has been shown to be effective and is mentioned as a nonadditive, nonhypnotic anxiolytic with phytotherapeutic potential to act as an adjuvant or complementary treatment to anxiolytic drugs [64, 65].

A meta-analysis review by Pittler et al. evaluated the efficacy and safety of kava extract versus placebo for treating anxiety. Seven randomized controlled trials using Piper methysticum indicated that kava extract is superior to placebo and relatively safe as a treatment option for anxiety [66].

Another recent meta-analysis, conducted by Ooi et al., revealed similar results, mentioning that there is promising evidence from well-designed clinical studies suggesting Kava, particularly aqueous extracts, as an effective treatment for generalized anxiety disorder (GAD) [67]. The authors add that the effect of Kava is comparable to commonly prescribed pharmacological drugs (buspirone and opipramol), but with fewer adverse consequences.

It is suggested that the progression of new treatments for psychological disorders is described in the identification of neural substrates and mechanisms underlying their etiology and pathophysiology. Adult hippocampal neurogenesis is a candidate mechanism for the etiology of depression and may be used as a substrate for antidepressant action, as it may also be important for some of the behavioral effects of antidepressants [68].

The therapeutic properties of kava are supported by the six major kavalactones (dihydromethysticin, kavain, dihydrokavain, methysticin, yangonin and desmethoxyyangonin), of which kawain and dihydrokawain have more intense anxiolytic activity [69].

Advertisement

3. Neuroprotective effect of essential oils

In recent years, growing interest in research on medicinal plants and the effects of essential oils (EOs), especially for the treatment of neuropathologies, has emerged [70]. EOs (also called volatile or ethereal oils) are odorous and volatile compounds found only in 10% of the plant kingdom [71, 72, 73, 74, 75, 76].

Secondary metabolites present in SOs have been widely used as antibacterial, antifungal and insecticidal agents. Their chemical and biological properties, especially antioxidants, have been considered important tools for the management of various neurological disorders [76].

Natural antioxidants derived from herbaceous plants have demonstrated in vitro cytoprotective properties and have a long history of providing benefits to human health [70]. Evidence of oxidative stress in neuronal damage and the benefits of antioxidant therapy have elucidated the importance of eliminating free radicals as a fundamental principle for the prevention and treatment of neurological disorders [77]. In addition, OEs derived antioxidants have been considered as a complementary treatment against neuronal loss as they have the ability to counteract the activity of free radicals responsible for neurodegeneration [78], protecting against cellular stress, as outlined in Figure 2.

Neural cells suffer functional or sensory loss due to neurological disorders and, in addition to other environmental or genetic factors that contribute to this loss, oxidative stress is a major contributor to neurodegeneration. Therefore, excess reactive oxygen species and an unbalanced metabolism lead to a number of neurological disorders, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD) [78].

Figure 2.

Main identified mechanisms for the action of medicinal plant essential oils in experimental models of neurological disorders. In step A: In an experimental model of cerebral ischemia after occlusion of the common carotid artery followed by reperfusion (BCCAO/R), it was observed that occlusion in the frontal cortex caused a decrease in docosahexaenoic acid (DHA). with Pistacia lentiscus L. showing positive plasma levels in the proportion of DHA-for its precursor, eicosapentaenoic acid (EPA) and levels of palmitoylethanolamide (PEA) and oleoylethanolamine (OAS), reversing its reduction, consequently decreasing the susceptibility to oxidation. In step B: essential oils from different medicinal plants demonstrated positive effect on the cellular antioxidant system. Salvia lavandulifolia Vahl, Lavandula angustifolia SSP. mill and Lavandula hybrida acted by increasing the multiple enzyme system (Cat, SOD, GPX, GSH and GSSG), Salvia l. (GR) and propolis (SOD) after induction of oxidative stress induced by different oxidants. Salvia essential oil reduced the expression of malondialdehyde (MDA), a marker of lipid peroxidation, thus inhibiting effector caspase-3 by preventing cellular apoptosis and preventing mitochondrial damage. The essential oil of Lavandula angustifolia ssp. angustifolia Mill also potentiated the described antioxidant enzyme system, reversing the scopolamine-induced damage (simulating a dementia model) as well as decreasing MDA levels. Aloysia citrodora acted upon damage in an experimental model of H2O2 and B-amyloid induced AD and its ability to act on the antioxidant system was observed due to the ability of its compounds to act as free radical scavengers or hydrogen donors. Increasing the antioxidant defense system through the action of these essential oils assists in reducing hydrogen peroxide (H2O2) to H20 and O2. Essential oils from other medicinal plants also regulated MDA (Lavandula hybrida, Aloysia citrodora and Propolis) levels. In step C: Lavandula A. Mill. and Lavandula hybrida demonstrated effects on DNA fragmentation (cleavage patterns were absent in the treated groups suggesting their antiapoptotic activity), similar effects were also observed in Rosa damascena mill treatment. Which also slowed the oxidative degradation of DNA, lipids and proteins due to the presence of phenols in their composition. In step D: Aloysia citrodora essential oils acted on the cell membrane, helping to increase iron chelation in vitro through Fe3 + to Fe2 + hydroximation, an important mechanism because the transition metal ions contribute to the oxidative damage in Neurodegenerative disorders, thus the chelation of transition metals, prevent catalysis of hydrogen peroxide decomposition via Fenton-type reaction. L. angustifolia Mill, acting on MDA levels and in the formation of reactive oxygen species (ROS), which are oxidative markers, consequently also prevents lipid peroxidation (determined by MDA level) in rat temporal lobe homogenates. Pistacia lenticus also acts by attenuating the levels of the enzyme Cox-2 cyclooxygenase 2, consequently decreasing the inflammation and oxidative stress observed in untreated groups. In step E: The essential oils of S. lavandulifolia are capable of activating the transcription factor Nrf2 - Nuclear factor (erythroid-derived 2) -like 2, a regulator of antioxidant genes, since protein expression and enzyme activity CAT, SOD, GR, GPx and HO-1 is markedly reduced, correlating with a decrease in nuclear Nrf2 protein. After treatment with S. lavandulifolia, regulation of Nrf2 was identified with a concomitant increase in antioxidant enzymes and HO-1, avoiding the formation of ROS, oxidation and decreased cell viability.

3.1 Alzheimer’s disease

Alzheimer’s disease (AD) is an age related neurodegenerative disease of the brain and this disease is characterized by a progressive deterioration of cognitive functions [79]. Oxidative stress, a detrimental factor during aging and pathologies, is involved in various neurodegenerative disorders [80, 81].

Studies suggest that caspase activation and apoptosis play important roles in AD neuropathogenesis [82]. Thus, SOs have been considered multi-potent agents against neurological disorders, being able to improve cognitive performance [83].

Lavender EO has several protective properties for the nervous system, as evidenced by its effectiveness in controlling depression, anxiety, stress, and cerebral ischemia [84, 85]. Some experimental models of AD have confirmed the neuroprotective effect and cognitive improvement of lavender OS, whose properties have been attributed to its antioxidant activity [86, 87].

EO (100 mg/kg) presented significant protection in the cognitive deficits evaluated, where the mechanism involved seems to be by a protection against decrease in the cellular antioxidant defense system, thus avoiding the reduction of the activity of superoxide dismutase, glutathione peroxidase and protection of the increase acetylcholinesterase malondialdehyde activity. The authors also demonstrated that lavender EO and its active component linalool protect against oxidative stress, cholinergic function and Nrf2/HO-1 pathway protein expression and synaptic plasticity. Therefore, it is suggested that linalool extracted from lavender OS may be a potential agent for improving cognitive impairment, especially in AD [88].

3.2 Oxidative stress

Oxidative stress occurs when the balance between antioxidants and reactive oxygen species (ROS) occurs negatively due to the depletion of antioxidants or the accumulation of ROS (Reactive Oxygen Species) [89]. Hydrogen peroxide (H2O2) is a major ROS and is involved in most cellular oxidative stresses [90, 91]. Several plants are considered a rich source of antioxidants because they inhibit or retard ROS-induced oxidative degradation [92, 93, 94].

Porres-Martínez et al. demonstrated that Salvia lavandulifolia E.O has neuroprotective activity against H2O2-induced oxidative stress in PC12 cells [93]. These effects appear to be related to Salvia lavandulifolia EO’s ability to activate the transcription factor Nrf2. Therefore, pretreatments with S. lavandulifolia EO resulted in decreased lipid peroxidation, ROS levels and caspase-3 activity, showing cell viability and morphological recovery.

Natural antioxidants present in some herbal plants are responsible for inhibiting or preventing oxidative stress, one of the agents that acts in AD, due to their ability to eliminate free radicals. The neuroprotective effect of A. citrodora has been attributed to its chelating activity. As described in the literature, an important mechanism of antioxidant effect is the chelation of transition metals, thus avoiding the catalysis of hydrogen peroxide decomposition via the Fenton type reaction [95]. The main proposed mechanisms regarding the action of A. citrodora and Salvia lavandulifolia OE in vitro models of neurological disorders.

A pioneering study by Abuhamdah et al. showed that Aloysia citrodora EO provides complete and partial protection from oxidative stress in an experimental model with H2O2-induced Alzheimer’s disease and β-amyloid-induced neurotoxicity using neuroblastoma cells. This study showed that 250 μm H2O2 could not trigger its neurotoxic effect when in the presence of 0.01 and 0.001 mg/mL O. citrodora EO, exhibiting neuroprotective activity at both concentrations [70].

Chelating agents have been reported to be effective as secondary antioxidants because they reduce the redox potential of transition metals, thereby stabilizing the oxidized form of the metal ion [96]. This seems to be one of the mechanisms involved in the antioxidant activity of some essential oils, as some of them were able to effectively chelate iron (II).

3.3 Brain ischemia

Cerebral ischemia consists in decreased blood flow in specific areas of the brain, causing hypoxia, which leads to an insufficient supply of glucose and oxygen, the magnitude of which disturbs the development of normal brain functions [97].

Currently, treatments that minimize neuronal damage after cerebral ischemia are limited, thus leading to the search for new complementary treatment therapies [98]. Terpenoids present in some essential oils constitute the largest group of secondary metabolites with neurological properties, including sedative, antidepressant and antinociceptive activities [99].

Another metabolite with neuroprotective function is linalool, a monoterpene present in volatile lavender oil, responsible for important therapeutic properties [100]; its activity on nerve disorders is well documented [101, 102]. Vakili et al. demonstrated that Lavandula angustifolia had a protective effect on focal cerebral ischemia in Wistar rats, especially when the treatment was performed with Lavandula angustifolia EO at 200 and 400 mg/kg. The results of the administration of this EO were an avoidance of a total antioxidant defense, reduced cerebral edema, and reduced infarct size. In addition, it improved functional performance after cerebral ischemia [103].

Advertisement

4. Concluding remarks

Neurodegenerative and neuropsychiatric diseases have multiple etiology. Multiple studies have been developed to clarify which approaches might be promising in prevention and treatment. We have targeted studies that present a neuroprotective perspective, herbal medicines and essential oils from different species of medicinal plants. These have various bioactive and phytochemical compounds with neuroprotective capacity, and also have given positive responses in studies on neurological disorders such as dementia, oxidative stress, anxiety, cerebral ischemia and oxidative toxicity. We suggest that these present a potential as agents in the treatment of neurological disorders.

Advertisement

Acknowledgments

The English text of this paper has been revised by Sidney Pratt, Canadian, MAT (The Johns Hopkins University), RSAdip - TESL (Cambridge University).

References

  1. 1. World Health Organization. Neurological Disorders: Public Health Challenges. Geneva: WHO; 2006. Available from: http://www.who.int/mental_health/neurology/neurodiso/en/. [Accessed: 18 May 2018]
  2. 2. Shaw G. The economic burden of neurologic disease—$800 billion annually in the US. Neurology Today. 2017;17(12):1-14. DOI: 10.1097/01.NT.0000521169.52982.7f
  3. 3. Van Dam D, De Deyn PP. Drug discovery in dementia: The role of rodent models. Nature Reviews. Drug Discovery. 2006;5:956-970. DOI: 10.1038/nrd2075
  4. 4. Sachan A, Singh S, Singh H, et al. An experimental study to evaluate the effect of Mucuna pruriens on learning and memory in mice. Journal of Innovation Sciences and Research. 2015;4(4):144-148
  5. 5. Izzo AA, Hoon-Kim S, Radhakrishnan R, et al. A critical approach to evaluating clinical efficacy, adverse events and drug interactions of herbal remedies. Phytotherapy Research. 2016;30:691-700. DOI: 10.1002/ptr.5591
  6. 6. Faillace MP, Zwiller J, Bernabeu RO. Effects of combined nicotine and fluoxetine treatment on adult hippocampal neurogenesis and conditioned place preference. Neuroscience. 2015;300:104-115. DOI: 10.1016/j.neuroscience.2015.05.017
  7. 7. Zhang QJ, Li LB, Niu XL, et al. The pyramidal neurons in the medial prefrontal cortex show decreased response to 5-hydroxytryptamine-3 receptor stimulation in a rodent model of Parkinson’s disease. Brain Research. 2011;1384:69-79. DOI: 10.1016/j.brainres.2011.01.086
  8. 8. Kumar GP, Khanum F. Neuroprotective potential of phytochemicals. Pharmacognosy Reviews. 2012;6:81. DOI: 10.4103/0973-7847.99898
  9. 9. Costa IM, Cavalcanti JRLP, Queiroz DB, et al. Supplementation with herbal extracts to promote behavioral and neuroprotective effects in experimental models of Parkinson’s disease: A systematic review. Phytotherapy Research. 2017;31:959-970. DOI: 10.1002/ptr.5813
  10. 10. Costa IM, Freire MAM, Cavalcanti JRLP, et al. Supplementation with Curcuma longa reverses neurotoxic and behavioral damage in models of Alzheimer’s disease: A systematic review. Current Neuropharmacology. 2019;17(5):406-421. DOI: 10.2174/0929867325666180117112610
  11. 11. Hellewell SC, Ziebell JM, Lifshitz J, et al. Impact acceleration model of diffuse traumatic brain injury. Methods in Molecular Biology. 2016;1462:253-266. DOI: 10.1007/978-1-4939-3816-2_15
  12. 12. Santiago LF, Rocha EG, Freire MA, et al. The organizational variability of the rodent somatosensory cortex. Reviews in the Neurosciences. 2007;18:283-294. DOI: 10.1515/REVNEURO.2007.18.3-4.283
  13. 13. Rodríguez JJ, Verkhratsky A. Neuroglial roots of neurodegenerative diseases? Molecular Neurobiology. 2011;43(2):87-96. DOI: 10.1007/s12035-010-8157-x. Available from: https://www.ncbi.nlm.nih.gov/pubmed/21161612. [Accessed: 18 May 2018]
  14. 14. Wruck CJ, Götz ME, Herdegen T, et al. Kavalactones protect neural cells against amyloid beta peptide-induced neurotoxicity via extracellular signal-regulated kinase 1/2-dependent nuclear factor erythroid 2-related factor 2 activation. Molecular Pharmacology. 2008;73(6):1785-1795. DOI: 10.1124/mol.107.042499. Available from: https://www.ncbi.nlm.nih.gov/pubmed/18334601. [Accessed: 18 May 2018]
  15. 15. Fragoulis A, Siegl S, Fendt M, et al. Oral administration of methysticin improves cognitive deficits in a mouse model of Alzheimer’s disease. Redox Biology. 2017;12:843-853. DOI: 10.1016/j.redox.2017.04.024. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5406548/. [Accessed: 18 May 2018]
  16. 16. Tanaka A, Hamada N, Fujita Y, et al. A novel kavalactone derivative protects against H2O2-induced PC12 cell death via Nrf2/ARE activation. Bioorganic and Medicinal Chemistry. 2010;18:3133-3139. DOI: 10.1016/j.bmc.2010.03.034. Available from: https://www.sciencedirect.com/science/article/pii/S0968089610002385?via%3Dihub. [Accessed: 18 May 2018]
  17. 17. Garrett KM, Basmadjian G, Khan IA, et al. Extracts of kava (Piper methysticum) induce acute anxiolytic-like behavioral changes in mice. Psychopharmacology. 2003;170:33-41. DOI: 10.1007/s00213-003-1520-0. Available from: https://link.springer.com/article/10.1007/s00213-003-1520-0. [Accessed: 18 May 2018]
  18. 18. Terazawa R, Akimoto N, Kato T, et al. A kavalactone derivative inhibits lipopolysaccharide-stimulated iNOS induction and NO production through activation of Nrf2 signaling in BV2 microglial cells. Pharmacological Research. 2013;71:34-43. DOI: 10.1016/j.phrs.2013.02.002 [Accessed: May 2018]
  19. 19. Joshi G, Johnson JA. The Nrf2-ARE pathway: A valuable therapeutic target for the treatment of neurodegenerative diseases. Recent Patents on CNS Drug Discovery. 2012;7(3):218-229. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3625035/. [Accessed: 18 May 2018]
  20. 20. Lobota A, Damulewicz M, Pyza E, et al. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cellular and Molecular Life Sciences. 2016;73:3221-3247. DOI: 10.1007/s00018-016-2223-0. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4967105/. [Accessed: 18 May 2018]
  21. 21. Lorenzi H, Matos FJ, Francisco JM. Plantas medicinais no Brasil: nativas e exóticas. 2nd ed. Nova Odessa: Plantarum; 2002
  22. 22. Zhang L, Fang Y, Xu Y, et al. Curcumin improves amyloid β-peptide (1-42) induced spatial memory deficits through BDNF-ERK signaling pathway. PLoS One. 2015;10:e0131525. DOI: 10.1371/journal.pone.0131525
  23. 23. Darvesh AS, Carroll RT, Bishayee A, et al. Curcumin and neurodegenerative diseases: A perspective. Expert Opinion on Investigational Drugs. 2012;21:1123-1140. DOI: 10.1517/13543784.2012.693479
  24. 24. Strimpakos AS, Sharma RA. Curcumin: Preventive and therapeutic properties in laboratory studies and clinical trials. Antioxidants and Redox Signaling. 2008;10:511-546. DOI: 10.1089/ars.2007
  25. 25. Ahmed T, Gilani AH. Therapeutic potential of turmeric in Alzheimer’s disease: Curcumin or curcuminoids? 2014;28(4):517-525. DOI: 10.1002/ptr.5030
  26. 26. Wright L, Frye JB, Gorti B, et al. Bioactivity of turmeric-derived curcuminoids and related metabolites in breast cancer. Current Pharmaceutical Design. 2013;19:6218-6225
  27. 27. Garcia-Alloza M, Borrelli LA, Rozkalne A, et al. Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model. Journal of Neurochemistry. 2007;102:1095-1104. DOI: 10.1111/j.1471-4159.2007.04613.x
  28. 28. Lim GP, Chu T, Yang F, et al. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. The Journal of Neuroscience. 2001;21:8370-8377
  29. 29. Li M, Li H, Fang F, et al. Astragaloside IV attenuates cognitive impairments induced by transient cerebral ischemia and reperfusion in mice via anti-inflammatory mechanisms. Neuroscience Letters. 2017;639:114-119
  30. 30. Wagner H, Bauer R, Xiao PG, et al. Chinese Drug Monographs and Analysis: Radix Astragali (Huangqi). Verlag Wald Germany; 1997. pp. 1-17
  31. 31. Zheng XY. Pharmacopoeia of the People’s Republic of China. Vol. 1. Beijing: Chemical Industry Press; 2005
  32. 32. Li ZP, Cao Q . Effects of astragaloside IV on myocardial calcium transport and cardiac function in ischemic rats. Acta Pharmacologica Sinica. 2002;23(10):898-904
  33. 33. Yang J, Wang HX, Zhang YJ, et al. Astragaloside IV attenuates inflammatory cytokines by inhibiting TLR4/NF-кB signaling pathway in isoproterenol-induced myocardial hypertrophy. Journal of Ethnopharmacology. 2013;150:1062-1070
  34. 34. Mancini M, Nicholson DW, Roy S, et al. The caspase-3 precursor has a cytosolic and mitochondrial distribution: Implications for apoptotic signaling. The Journal of Cell Biology. 1998;140:1485-1495. DOI: 10.1083/jcb.140.6.1485
  35. 35. Mulugeta S, Maguire JA, Newitt JL, et al. Misfolded BRICHOS SP-C mutant proteins induce apoptosis via caspase-4- and cytochrome c-related mechanisms. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2007;293:L720-L729. DOI: 10.1152/ajplung.00025.2007
  36. 36. Chang CP, Liu YF, Lin HJ, et al. Beneficial effect of astragaloside on Alzheimer’s disease condition using cultured primary cortical cells under beta-amyloid exposure. Molecular Neurobiology. 2016;53(10):7329-7340. DOI: 10.1007/s12035-015-9623-2
  37. 37. Su C, Elfeki N, Ballerini P, et al. Guanosine improves motor behavior, reduces apoptosis, and stimulates neurogenesis in rats with parkinsonism. Journal of Neuroscience Research. 2009;87(3):617-625. DOI: 10.1002/jnr.21883
  38. 38. Pérez-Hernández ZM, Villanueva-Porras D, Veja-Avila E, et al. A potential alternative against neurodegenerative diseases: Phytodrugs. Oxidative Medicine and Cellular Longevity. 2016;2016:8378613. DOI: 10.1155/2016/8378613
  39. 39. Chan WS, Durairajan SS, Lu JH, et al. Neuroprotective effects of astragaloside IV in 6-hydroxydopamine-treated primary nigral cell culture. Neurochemistry International. 2009;55(6):414-442. DOI: 10.1016/j.neuint.2009.04.012
  40. 40. Arulkumar S, Sabesan M. The behavioral performance tests of Mucuna pruriens gold nanoparticles in the 1-methyl4- phenyl-1,2,3,6-tetrahydropiridina treated mouse model of parkinsonism. Asian Pacific Journal of Tropical Disease. 2012;2:499-502. DOI: 10.1016/S2222-1808(12)60210-2
  41. 41. Yu X, He GR, Sun L, et al. Assessment of the treatment effect of baicalein on a model Parkinsonian tremor and elucidation of the mechanism. Life Sciences. 2012;91(1-2):5-13. DOI: 10.1016/j.lfs.2012.05.005
  42. 42. Kim BW, Koppula S, Park SY, et al. Attenuation of neurinflamatory responses and behavioral déficits by Ligusticum officinale (Makino) kitag in stimulated microglia and MPTP-induced mouse model of Parkinson’s disease. Journal of Ethnopharmacology. 2015;164:388-397. DOI: 10.1016/j.jep.2014.11.004
  43. 43. Sankar RS, Manivasagam T, Surendran S. Ashwagandha leaf extract: A potential agent in treating oxidative damage and physiological abnormalities seen in a mouse model of Parkinson’s disease. Neuroscience Letters. 2009;454(1):11-15. DOI: 10.1016/j.neulet.2009.02.044
  44. 44. Jiang S, Bendjelloul F, Ballerini P, et al. Guanosine reduces apoptosis and inflammation associated with restoration of function in rats with acute spinal cord injury. Purinergic Signalling. 2007;3(4):411-442. Available from: https://link.springer.com/article/10.1007/s11302-007-9079-6. [Accessed: 31 July 2018]
  45. 45. Calou I, Bandeira MA, Galvão WA, et al. Neuroprotective properties of a standardized extract from Myracrodruon urundeuva Fr. All. (Aroeira-do-Sertão), as evaluated by a Parkinson’s disease model in rats. Parkinson’s Disease. 2014;2014:1-11. DOI: 10.1155/2014/519615
  46. 46. Guo S, Yan J, Yang T, et al. Protective effect of green tea polyphenols in the 6-OHDA rat model of Parkinson’s disease through inhibition of ROS–NO pathway. Biological Psychiatry. 2007;62(12):1353-1362. DOI: 10.1016/j.biopsych.2007.04.020
  47. 47. Choi HS, Park MS, Km SH, et al. Neuroprotective effects of herbal ethanol extracts from Gynostemma pentaphyllum in the 6-hydroxydopamine lesioned rat model of Parkinson’s disease. Molecules. 2010;15(4):2814-2824. DOI: 10.3390/molecules15042814
  48. 48. Zhu G, Wang X, Chen Y, et al. Puerarin protects dopaminergic neurons against 6-hydroxydopamine neurotoxicity via inhibiting apoptosis and upregulating glial cell line derived neurotrophic fator in a rat model of Parkinson’s disease. Planta Medica. 2010;76(16):1820-1826. DOI: 10.1055/s-0030-1249976
  49. 49. Zhang HN, An CN, Zhang HN, et al. Protocatechuic acid inhibits neurotoxicity induced by MPTP in vivo. Neuroscience Letters. 2010;474(2):99-103. DOI: 10.1016/j.neulet.2010.03.016
  50. 50. Chen H, Jing FC, Li CH, et al. Echinacoside prevents the striatal extracellular levels of monoamine neurotransmitters from diminution in 6- hydroxydopamine lesion rats. Journal of Ethnopharmacology. 2007;114(3):285-289. DOI: 10.1016/j.jep.2007.07.035
  51. 51. Zhao L, Pux XP. Neuroprotective effect of acteoside against MPTP-induced mouse model of Parkinson’s disease. Chinese Pharmacological Bulletin. 2007;23(1):42-46
  52. 52. Molz S, Dal-Cim T, Budni J, et al. Neuroprotective effect of guanosine against glutamate-induced cell death in rat hippocampal slices is mediated by the phosphatidylinositol-3 kinase/Akt/glycogen synthase kinase 3β pathway activation and inducible nitric oxide synthase inhibition. Journal of Neuroscience Research. 2011;89(9):1400-1408. DOI: 10.1002/jnr.22681. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/jnr.22681?scrollTo=references. [Accessed: July 31, 2018]
  53. 53. Singh YN. Kava: An overview. Journal of Ethnopharmacology. 1992;37(1):13-45. DOI: 10.1016/0378-8741(92)90003-a. Available from: https://www.sciencedirect.com/science/article/pii/037887419290003A. [Accessed: 18 May 2018]
  54. 54. Chang R, Algird A, Bau C, et al. Neuroprotective effects of guanosine on stroke models in vitro and in vivo. Neuroscience Letters. 2008;431(2):101-105. DOI: 10.1016/j.neulet.2007.11.072. Available from: https://www.ncbi.nlm.nih.gov/pubmed/18191898. [Accessed: 31 July 2018]
  55. 55. Backhauss C, Krieglstein J. Extract of kava (Piper methysticum) and its methysticin constituents protect brain tissue against ischemic damage in rodents. European Journal of Pharmacology. 1992;215(2-3):265-269. DOI: 10.1016/0014-2999(92)90037-5. Available from: https://www.ncbi.nlm.nih.gov/pubmed/1396990. [Accessed: 18 May 2018]
  56. 56. Deng W, Aimone JB, Gage FH. New neurons and new memories: How does adult hippocampal neurogenesis affect learning and memory? Nature Reviews Neuroscience. 2010;11:339-350. Available from: https://www.ncbi.nlm.nih.gov/pubmed/20354534. [Accessed: 18 May 2018]
  57. 57. Gerbatin RR, Cassol G, Dobrachinski F, et al. Guanosine protects against traumatic brain injury-induced functional impairments and neuronal loss by modulating excitotoxicity, mitochondrial dysfunction, and inflammation. Molecular Neurobiology. 2017;54(10):7585-7596. DOI: 10.1007/s12035-016-0238-z. Available from: https://www.ncbi.nlm.nih.gov/pubmed/27830534. [Accessed: 18 May 2018]
  58. 58. Schmidt AP, Lara DR, Souza DO. Proposal of a guanine-based purinergic system in the mammalian central nervous system. Pharmacology and Therapeutics. 2007;116(3):401-416. DOI: 10.1016/j.pharmthera.2007.07.004. Available from: https://www.sciencedirect.com/science/article/pii/S0163725807001568. [Accessed: 31 July 2018]
  59. 59. Bettio LEB, Freitas AE, Neis VB, et al. Guanosine prevents behavioral alterations in the forced swimming test and hippocampal oxidative damage induced by acute restraint stress. Pharmacology Biochemistry and Behavior. 2014;127:7-14. DOI: 10.1016/j.pbb.2014.10.002. Available from: https://www.sciencedirect.com/science/article/pii/S0091305714002767. [Accessed:31 July 2018]
  60. 60. Bettio LEB, Neis VB, Pazini FL, et al. The antidepressant-like effect of chronic guanosine treatment is associated with increased hippocampal neuronal differentiation. European Journal of Neuroscience. 2016;43(8):1006-1015. DOI: 10.1111/ejn.13172. Available from: https://onlinelibrary.wiley.com/doi/abs/10.1111/ejn.13172. [Accessed: 31 July 2018]
  61. 61. Duman RS, Nakagawa S, Malberg J. Regulation of adult neurogenesis by antidepressant treatment. Neuropsychopharmacology. 2001;25(6):836-844. DOI: 10.1016/S0893-133X(01)00358-X. Available from: https://www.ncbi.nlm.nih.gov/pubmed/11750177. [Accessed: 31 July 2018]
  62. 62. Singh YN, Singh NN. Therapeutic potential of kava in the treatment of anxiety disorders. CNS Drugs. 2002;16(11):731-743. DOI: 10.2165/00023210-200216110-00002
  63. 63. Bandelow B, Boerner JR, Kasper S, et al. The diagnosis and treatment of generalized anxiety disorder. Deutsches Ärzteblatt International. 2013;110(17):300-309. DOI: 10.3238/arztebl.2013.0300. Available from: https://www.ncbi.nlm.nih.gov/pubmed/23671484. [Accessed: 18 May 2018]
  64. 64. Saki K, Bahmani M, Rafieian-Kopaei M. The effect of most important medicinal plants on two importnt psychiatric disorders (anxiety and depression)—A review. Asian Pacific Journal of Tropical Medicine. 2014;7(1):S34-S42. DOI: 10.1016/S1995-7645(14)60201-7. Available from: https://www.sciencedirect.com/science/article/pii/S1995764514602017. [Accessed: 18 May 2018]
  65. 65. Savage KM, Stough CK, Byrne GJ, et al. Kava for the treatment of generalised anxiety disorder (K-GAD): Study protocol for a randomised controlled trial. Trials. 2015;16(493):1-13. DOI: 10.1186/s13063-015-0986-5
  66. 66. Pittler MH, Ernst E. Kava Extract Versus Placebo for Treating Anxiety (Review). New Jersey: John Wiley & Sons; 2010. Available from: https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD003383/epdf/abstract. [Accessed: 18 May 2018]
  67. 67. Ooi SL, Henderson P, Pak SC. Kava for generalized anxiety disorder: A review of current evidence. Journal of Alternative and Complementary Medicine. 2018;24(8):770-780. DOI: 10.1089/acm.2018.0001
  68. 68. Sahay A, Hen R. Adult hippocampal neurogenesis in depression. Nature Neuroscience. 2007;10(9):1110-1115. DOI: 10.1038/nn1969
  69. 69. Wu D, Yu L, Nair MG, et al. Cyclooxygenase enzyme inhibitory compounds with antioxidant activities from Piper methysticum (kava kava) roots. Phytomedicine. 2002;9(1):41-47. DOI: 10.1078/0944-7113-00068
  70. 70. Abuhamdah S, Abuhamdah R, Howes MJ, et al. Pharmacological and neuroprotective profile of an essential oil derived from leaves of Aloysia citrodora Palau. The Journal of Pharmacy and Pharmacology. 2015;67(9):1306-1315. DOI: 10.1111/jphp.12424
  71. 71. Ahmadi L, Mirza M, Shahmir F. The volatile constituents of Artemisia marschaliana Sprengel and its secretory elements. Flavour and Fragrance Journal. 2002;17:141-143. DOI: 10.1002/ffj.1055
  72. 72. Bezić N, Šamanić I, Dunkić V, et al. Essential oil composition and internal transcribed spacer (ITS) sequence variability of four south-Croatian Satureja species (Lamiaceae). Molecules. 2009;14:925-938. DOI: 10.3390/molecules14030925
  73. 73. Ciccarelli D, Garbari F, Pagni AM. The flower of Myrtus communis (Myrtaceae): Secretory structures, unicellular papillae, and their ecological role. Flora. 2008;203(15):85-93. DOI: 10.1016/j.flora.2007.01.002
  74. 74. Gershenzon J. Metabolic costs of terpenoid accumulation in higher plants. Journal of Chemical Ecology. 1994;20(6):1281-1328. DOI: 10.1007/BF02059810
  75. 75. Liolios CC, Graikou K, Skaltsa E, et al. Dittany of Crete: A botanical and ethnopharmacological review. Journal of Ethnopharmacology. 2010;131:229-241. DOI: 10.1016/j.jep.2010.06.005
  76. 76. Misharina TA, Polshkov AN. Antioxidant properties of essential oils: Autoxidation of essential oils from laurel and fennel and effects of mixing with essential oil from coriander. Prikladnaia Biokhimiia i Mikrobiologiia. 2005;41:693-702
  77. 77. Santos JR, Gois AM, Mendonca DM, et al. Nutritional status, oxidative stress and dementia: The role of selenium in Alzheimer’s disease. Frontiers in Aging Neuroscience. 2014;6:206. DOI: 10.3389/fnagi.2014.00206
  78. 78. Uttara B, Singh AV, Zamboni P, et al. Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Current Neuropharmacology. 2009;7(1):65-74. DOI: 10.2174/157015909787602823
  79. 79. Dunne TE. Alzheimer’s Disease: Overview. 2nd ed. Oxford, UK: Academic Press; 2016. pp. 58-63
  80. 80. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000;408:239-247. DOI: 10.1038/35041687
  81. 81. Freire MAM. Pathophysiology of neurodegeneration following traumatic brain injury. The West Indian Medical Journal. 2012;61:751-755
  82. 82. Mattson MP. Contributions of mitochondrial alterations, resulting from bad genes and a hostile environment, to the pathogenesis of Alzheimer’s disease. International Review of Neurobiology. 2002;53:387-409
  83. 83. Ayaz M, Sadiq A, Junaid M, et al. Neuroprotective and anti-aging potentials of essential oils from aromatic and medicinal plants. Frontiers in Aging Neuroscience. 2017;9:168. DOI: 10.3389/fnagi.2017.00168
  84. 84. Koulivand PH, Khaleghi Ghadiri M, Gorji A. Lavender and the nervous system. Evidence-based Complementary and Alternative Medicine. 2013;2013:681304. DOI: 10.1155/2013/681304
  85. 85. Takahashi M, Satou T, Ohashi M, et al. Interspecies comparison of chemical composition and anxiolytic-like effects of lavender oils upon inhalation. Natural Product Communications. 2011;6(11):1769-1774
  86. 86. Hancianu M, Cioanca O, Mihasan M, et al. Neuroprotective effects of inhaled lavender oil on scopolamine-induced dementia via anti-oxidative activities in rats. Phytomedicine. 2013;20(5):446-452. DOI: 10.1016/j.phymed.2012.12.005
  87. 87. Hritcu L, Cioanca O, Hancianu M. Effects of lavender oil inhalation on improving scopolamine-induced spatial memory impairment in laboratory rats. Phytomedicine. 2012;19(6):529-534. DOI: 10.1016/j.phymed.2012.02.002
  88. 88. Xu P, Wang K, Lu C, et al. The protective effect of lavender essential oil and its main component linalool against the cognitive deficits induced by D-Galactose and aluminum trichloride in mice. Evidence-based Complementary and Alternative Medicine. 2017;2017:7426538. DOI: 10.1155/2017/7426538
  89. 89. Emerit J, Edeas M, Bricaire F. Neurodegenerative diseases and oxidative stress. Biomedicine and Pharmacotherapy. 2004;58:39-46. DOI: 10.1038/nrd1330
  90. 90. Dasuri K, Zhang L, Keller JN. Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis. Free Radical Biology and Medicine. 2013;62:170-185. DOI: 10.1016/j.freeradbiomed.2012.09.016
  91. 91. Halliwell B. Free radicals and antioxidants: Updating a personal view. Nutrition Reviews. 2012;70(5):257-265. DOI: 10.1111/j.1753-4887.2012.00476.x
  92. 92. Gonzalez-Burgos E, Gomez-Serranillos MP. Terpene compounds in nature: A review of their potential antioxidant activity. Current Medicinal Chemistry. 2012;19(31):5319-5341. DOI: 10.2174/092986712803833335
  93. 93. Guerra-Araiza C, Alvarez-Mejia AL, Sanchez-Torres S, et al. Effect of natural exogenous antioxidants on aging and on neurodegenerative diseases. Free Radical Research. 2013;47:451-462. DOI: 10.3109/10715762.2013.795649
  94. 94. Porres-Martinez M, Gonzalez-Burgos E, Carretero ME, et al. Protective properties of Salvia lavandulifolia Vahl. essential oil against oxidative stress-induced neuronal injury. Food and Chemical Toxicology. 2015;80:154-162. DOI: 10.1016/j.fct.2015.03.002
  95. 95. Gil A, Van Baren CM, Di Leo Lira PM, et al. Identification of the genotype from the content and composition of the essential oil of lemon verbena (Aloysia citriodora Palau). Journal of Agricultural and Food Chemistry. 2007;55(21):8664-8669. DOI: 10.1021/jf0708387
  96. 96. Bush AI. The metallobiology of Alzheimer’s disease. Trends in Neurosciences. 2003;26(4):207-214. DOI: 10.1016/S0166-2236(03)00067-5
  97. 97. Small DL, Buchan AM. Mechanisms of cerebral ischemia: Intracellular cascades and therapeutic interventions. Journal of Cardiothoracic and Vascular Anesthesia. 1996;10:139-146. DOI: 10.1016/s1053-0770(96)80189-3
  98. 98. Jivad N, Rabiei Z. Review on herbal medicine on brain ischemia and reperfusion. Asian Pacific Journal of Tropical Biomedicine. 2015;5(10):789-795. DOI: 10.1016/j.apjtb.2015.07.015
  99. 99. Perazzo FF, Lima LM, Maistro EL, et al. Effect of Artemisia annua L. leaves essential oil and ethanol extract on behavioral assays. Revista Brasileira de Farmacognosia. 2008;18:686-689. DOI: 10.1590/S0102-695X2008000500008
  100. 100. Peana AT, De Montis MG, Nieddu E, et al. Profile of spinal and supra-spinal antinociception of (−)-linalool. European Journal of Pharmacology. 2004;485:165-174. DOI: 10.1016/j.ejphar.2003.11.066
  101. 101. Devi SL, Kannappan S, Anuradha CV. Evaluation of in vitro antioxidant activity of Indian bay leaf, Cinnamomum tamala (Buch. -ham.) T. Nees and Eberm using rat brain synaptosomes as model system. Indian Journal of Experimental Biology. 2007;45(9):778-784
  102. 102. Batish DR, Singh HP, Setia N, et al. Chemical composition and inhibitory activity of essential oil from decaying leaves of Eucalyptus citriodora. Zeitschrift für Naturforschung. Section C. 2006;61:52-56. DOI: 10.1515/znc-2006-1-210
  103. 103. Vakili A, Sharifat S, Akhavan MM, et al. Effect of lavender oil (Lavandula angustifolia) on cerebral edema and its possible mechanisms in an experimental model of stroke. Brain Research. 2014;1548:56-62. DOI: 10.1016/j.brainres.2013.12.019

Written By

Ianara Mendonça da Costa, Elaine Cristina Gurgel Andrade Pedrosa, Ana Paula de Carvalho Bezerra, Luciana Cristina Borges Fernandes, José Rodolfo Lopes de Paiva Cavalcanti, Marco Aurélio Moura Freire, Dayane Pessoa de Araújo, Amália Cinthia Meneses do Rego, Irami Araujo Filho, Francisco Irochima Pinheiro and Fausto Pierdoná Guzen

Submitted: 24 June 2019 Reviewed: 19 December 2019 Published: 28 February 2020