Open access peer-reviewed chapter

Physiological and Cellular Targets of Neurotrophic Anxiolytic Phytochemicals in Food and Dietary Supplements

Written By

Benjamin S. Weeks, Samuel D. Weeks, Amanda Kim, Landon Kessler and Pedro P. Perez

Submitted: 27 October 2020 Reviewed: 02 April 2021 Published: 26 April 2021

DOI: 10.5772/intechopen.97565

From the Edited Volume

Functional Foods - Phytochemicals and Health Promoting Potential

Edited by Muhammad Sajid Arshad and Muhammad Haseeb Ahmad

Chapter metrics overview

549 Chapter Downloads

View Full Metrics

Abstract

Diet impacts anxiety in two main ways. First anxiety can be caused by deficiencies in antioxidants, neurotransmitter precursors, amino acids, cations and vitamins and other cofactors. Second, anxiety can be reduced by anxiolytic nutraceuticals which are food molecules that bind to molecular targets of the amygdala and the hypothalamus-pituitary–adrenal axis (HPA-axis). Anxiety is a feeling of fear that arises from a perceived threat and can be a beneficial coping mechanism to threats and stressors. However excessive anxiety is a disorder that interferes with healthy responses to stressors. The amygdala is responsible for assigning value to a threat or stressor and triggering the HPA-axis to support the body wide system responses to the threat. The amygdala also communicates with the neuroplastic learning and memory centers of the hippocampus to fix or set a learned value to the threat. Interestingly, many anxiolytic nutraceuticals that show benefits in human clinical trials have neurotrophic activity and increase neuronal plasticity. Moreover, anxiolytic nutraceuticals either act like the neurotrophins, nerve growth factor (NGF), brain derived neurotrophic factor (BDNF and neurotrophin-3 (NT3) by either directly binding to or potentiating the tyrosine receptor kinase (TRK) family of receptors (TRKA, TRKB and TRKC) and activating the ERK1/2 signal transduction pathway associated with neurite outgrowth and neural plasticity. This chapter will explore the neuritogenic activity of clinically proven plant-based anxiolytic nutraceuticals and examine the commonality of TRKA-C receptors and the ERK1/2 signaling pathway in the pharmacological and nutraceutical treatment of anxiety disorders.

Keywords

  • Anxiety
  • Anxiolytic
  • Nutraceutical
  • Cannabidiol
  • Neurotrophin
  • Neurite Outgrowth
  • NGF
  • BDNF
  • NT3
  • TRKA
  • TRKB
  • TRKC
  • ERK1/2

1. Introduction

In humans, the appropriate and measured behavioral responses to environmental cues are under control of the limbic nervous system which is composed primarily of the amygdala, hippocampus, thalamus, and hypothalamus [1]. In order for sensory inputs to the cerebral cortex to result in the appropriate responses in the body, sensory inputs relay from the cerebrum, to the limbic system and then from the limbic system to the body either through the brainstem or through the pituitary gland. It is when relaying sensory inputs from the cerebral cortex to the body that the limbic system also assigns emotional value to sensory input and sets or fixes that value by learning and remembering the rewards and punishments associated with specific environmental cues. The amygdala is known for assigning a scaled value to negative threats and stressors which the amygdala then communicates to learning and memory centers in the hippocampus so that human behavioral responses to negative cues can be consistent and appropriate. The amygdala also stimulates the hypothalamus to secrete corticotrophin-releasing hormone (CRH) which in turn stimulates the pituitary to release adrenocorticotropin hormone (ACTH), which in turn stimulates the adrenal cortex to secrete glucocorticoids including primarily, cortisol in what is known as the HPA-axis [2, 3, 4]. The hypothalamus can also send signals through the brainstem and activate the adrenal medulla to secrete epinephrine and norepinephrine. Cortisol, epinephrine and norepinephrine are hormones that can signal body wide changes in metabolic rates, breathing, heart rate, blood pressure and a variety of other appropriate body responses to the presence of an environmental threat or stressor [2, 3]. Anxiety is the feeling of fear or worry that arises from the neurochemistry of the amygdala in response to negative environmental cues and the activation of the HPA-axis and the overall preparation of the body to meet the challenges of a threat or stressor and while anxiety is a negative feeling, when it is in proportion to the actual threat a stressor presents, anxiety can be a normal and even healthy part of an adequate response to the stressor [5, 6, 7]. However, excessive and prolonged anxiety that is unwarranted by the environmental cue and exaggerated in proportion to the actual threat level leads to inappropriate and prolonged activation of the HPA-axis and cortisol release which is associated with inflammatory damage and other pathophysiologies that further stresses the human body system [2, 3, 4]. In these cases anxiety interferes with normal and health everyday life and is considered an anxiety disorder [8, 9].

People suffer from five different types of anxiety disorders; generalized anxiety disorder (GAD, obsessive compulsive disorder (OCD), panic disorder (PD), social anxiety disorder (SAD), and posttraumatic stress disorder (PTSD) [8, 9]. Each of these anxiety disorders can be described by the level of synaptic neurotransmitters and cell surface neurotransmitter receptors in the amygdala [1, 8, 9]. For example, GAD is associated with decreased activity of the inhibitory neurotransmitter, GABA. GABA acts on GABAA receptors on neurons within the amygdala to inhibit signals and help to assign lower threat values to certain stressor. Down regulation of the GABAA receptor and the subsequent reduction of GABA signaling in the amygdala leads GAD through elevated valuation of threats [10]. Similarly, PD is also associated with decreased GABAnergic transmission and subsequent over stimulation of neural pathways, however in PD the decrease GABAnergic signaling may be due to reduced level of the GABA neurotransmitter itself and not due to decreased GABAA receptors as seen in GAD [9, 11, 12]. While GABAnergic pathways in the amygdala are inhibitory and stress reducing, glutamate, the major stimulatory neurotransmitter, when over active in the amygdala enhances stress and can lead to OCD. Pharmacological enhancement of glutaminergic signals in the frontolimbic regions of the brain enhance anxiety and imaging studies have shown increased glutaminergic activity in various structures of the limbic system in the brain [13, 14, 15]. PTSD and SAD also appear involve increased glutaminergic activity in the amygdala [9, 16]. GABA and glutamate influence the feeling of anxiety be reducing and enhancing the perceived threats, while the neurotransmitters, serotonin and dopamine are associated with the reward and pleasure pathways of the limbic system and can influence the overall perception of environmental stressors generally reducing anxiety. For example, SAD is associated with both decreased activity at serotonin receptors and also decreased dopamine levels in limbic neurocircuitry [9, 16, 17]. Taken together, anxiety disorders involve irregularities in the levels of neurotransmitters and neurotransmitter receptors in the neurocircuitry of the limbic system. The inappropriate levels of neurotransmitters and their receptors can lead to hyper activity in regions of the limbic system such as the amygdale and lead to incorrect and unhealthy assessment of the risks and threats associated with stressors or lack of stressors and lead to anxiety and fear potentially even in the absence of threat. Activation of the HPA-axis can contribute to both the clinical signs and symptoms of anxiety and also lead to chronic glucocorticoid induced pathologies which serve and further internal stressors and add to anxiety. Treatments for anxiety disorders have therefore focused on developing drugs that correct and manage the levels of neurotransmitters and neurotransmitters receptors and signaling in the limbic system pathways and particularly in the amygdala.

GABAnergic benzodiazepines are the favored class of anxiolytic medications [10, 11, 18]. The diazepine ring is a seven membered ring structure containing two nitrogens and this diazepine ring and when fuses with a benzene ring forms a benzodiazepine that can bind to GABAA receptors on neurons in the brain [18]. Benzodiazepines are favored due to their lesser side-effects compared to other anxiolytic drugs, although side effects are still concerns [18]. The mechanism of benzodiazepine signaling is binding to either GABAA or GABAB receptors and allowing either chlorine ions into the cell at the synapse or stimulating the release of potassium from the cell into the synapse respectively [10, 11, 18]. In the cells of the amygdala, the chlorine influx inhibits the signaling of the pathway and diminishes the level of potential threat assigned to a sensory input or any external or internal stressor. People with GAD and PD express low levels of GABAA and produce less GABA respectively thereby limiting the patient’s ability diminish the signals from stressors is associate with a heightened sense of fear and worry. By being GABAnergic the benzodiazepines help to restore or boost the GABAnergic pathway and the therefore the reduction of anxiety. Alternatively to drugs that act in a GABAnergic fashion, serotonin and dopamine uptake inhibitors, often used for depression, reduce anxiety and fear by increasing levels of these “feel good” neurotransmitters in the limbic neurocircuitry. Low synaptic serotonin and dopamine in the amygdale and nucleus accumbens is associated SAD. Serotonin uptake inhibitors (SSRIs) and noradrenalin and dopamine reuptake inhibitors (NDRIs) increased the level of serotonin and dopamine in the synapse and have been used to treat depression and also provide relief from anxiety and anxiety disorders. [18, 19, 20, 21, 22].

In addition to the development of new drugs that interact with the amygdala and HPA-axis, anxiety can also be addressed by diet. The diet can be associated with anxiety in two main ways. First, if a diet is deficient in nutrients such as selenium, lysine, magnesium and inositol, changes in food consumptions or dietary supplementation can replace the deficient nutrient, balance the diet and alleviate anxiety [23]. Further, dietary deficiencies in antioxidants can lead to the buildup of reactive oxygen species (ROSs) that form as a part of normal metabolism and are reactive chemicals that can bind to DNA, lipids and proteins leading to DNA and membrane damage and cellular toxicity. This cellular damage serves as a stress signal and is associated with anxiety [24, 25]. Therefore, increasing dietary antioxidant intake can help with anxiety. Second, food nutrients can directly affect the neurochemistry of the limbic system by either directly boosting GABA or Serotonin levels or by binding to neurotransmitter receptors. For example, GABA is an amino acid is available directly in the diet. Further the amino acid, 5-hydroxytryptophan is a serotonin precursor and is a popular dietary supplement taken to easy feelings of anxiety and stress. While it is not clear if increasing oral consumption of GABA and 5-hydroxytrptophan can increase brain GABA and serotonin levels, clinical studies have shown and relaxing effect of GABA and 5-HTP supplementation [23]. The neurochemistry of the brain can also be altered by food chemicals eaten from bacteria, fungi and plants that have nutraceutical effects by acting in a drug-like fashion as cell signaling molecules and alerting cellular behavior. In this chapter we focus on food nutraceuticals that are anxiolytic in humans and alter the neurochemistry and the amygdala and other limbic structures in the brain. Of particular interest are anxiolytic phytochemicals that in addition to changing the brain neurotransmitter physiology also stimulate neuronal plasticity through the activation and or potentiating of neurotrophin receptors and signal transduction pathways.

Recent studies have revealed that numerous anxiolytic substances, including endogenous neurotransmitters, anxiolytic drugs, and nutraceuticals, are also neurotrophic in that they also activate the brain derived neurotrophic factor (BDNF) pathway, the neurotrophin-3 (NT-3) pathways and the nerve growth factor (NGF) pathway by binding to or potentiating the TRKA – C neurotrophin receptors and directly activating the ERK1/2 signaling pathway leading to neuroplasticity [26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37]. This is important because neurotrophins can regulate neuroplasticity not only during development but also during learning and the establishment of memories [35, 36, 37]. Neurotrophins are small soluble signaling molecules that can diffuse between cells to play a role in cell–cell communication [35, 36, 37]. These neurotrophic factors include BDNF, NGF and NT3 bind to cell surface molecules on neuronal cells known as the tropomyosin receptor kinases (TRK) A – C respectively [35, 36, 37]. Neurotrophin signaling is associated with neuritogenesis or new neurite formation in neuronal cells. The changes in cell shape associated with the establishment of new neurites and therefore potentially new connections is known as neuroplasticity [35, 36, 37]. Recent attention has been brought to the idea that in so far as anxiety is related to the memories of trauma and the establishment of a learned threat level in the perception of stressors through neuroplasticity, perhaps anxiolytic phytochemicals with neurotrophic activity can be used to reduce anxiety not only through changes neurotransmitter activity, but also by providing the plasticity required to relearn and reduce the emotional value ascribed to a stressor thereby also facilitating the reduction in anxiety [26, 27, 28, 29, 30, 31, 32, 33, 34]. Therefore anxiolytic phytochemical neurotrophins are important because they offer a new area of research into not simply adjusting neurotransmitter activity, but to the development of natural treatments and drugs that can actually reverse the neurocircuitry associated with anxiety through neuroplasticity and relearning. It is important to note however, not all anxiolytic phytochemicals are capable of stimulating neuroplasticity. The following section of this chapter will present all nutraceutical phytochemicals that are anxiolytic in human clinical trials that also show potential for stimulating neuroplasticity either by directly stimulating neuritogenesis or neurite outgrowth neuronal cells or by binding to the TRKA-C neurotrophin receptors and or by the activation of the neurotrophin ERK1/2 signal transduction pathway and others associated with neurite formation.

Advertisement

2. Clinically relevant anxiolytic phytochemicals with neurotrophic activity

In this chapter we present only plants and plant extracts that contain phytochemicals that are both shown to be anxiolytic in human clinical trials and also possess neuroplastic properties (Table 1). The specific anxiolytic nutraceutical or phytochemical in the plant is in most cases not known, in part because neuroactive plants usually contain many nervine agents. Often however there is a suspected phytochemical or group of phytochemicals thought to be responsible for the anxiolytic activity. In some cases the anxiolytic nutraceutical in the plant extract is the same phytochemical that has the neurotrophic activity, while in other cases it may be a different phytochemical in the plant extract. Anxiolytic drugs adjust neurotransmitter and neurotransmitter receptors levels which leads to increased drug insensitivity, extreme withdrawal effects and a return to imbalance neurotransmitter and neurotransmitter receptor levels when and if the drug is removed. In addition to altering neurotransmitter and receptor levels neuroplastic anxiolytics also stimulate the new neurite connections associated with learning and remembering appropriate responses to stressors. If a new response to a threat is learned, then treatment of the anxiety disorder may not require dosage increases and the newly learned healthy perceptions of threats could remain with the patient even if the drug or treatment is removed or reduced. This would represent a tremendous advancement in the treatment of anxiety disorders. Table 1 is a list of the fourteen clinically supported anxiolytic plants that also have neuroplastic properties.

AnxiolyticNeurotrophic Activities.
Plant/NutraceuticalNeurotrophin PathwaysNeurite OutgrowthReferences
Green Tea/theanineIncreases brain BDNF levelshippocampal cells[45, 46]
Increases brain NGF synthesisNeural stem cells
Potentiates NGF at TRKA
Chamomile/apigeninIncreases hippocampal BDNFN2a cells[55, 56, 57, 58]
ERK1/2 kinase activation
Lavender/N.D.Increases brain NGFRneuronal cells[67, 68]
Ashwagandha/N.D.Increases brain GDNFhippocampal cells[75]
Passion Flower/apigeninPotentiates NGFPC12 cells[80]
Cannabis/CBDBinds TRKAPC12 cells[88]
Activates ERK1/2
Valerian/SesquiterpenesIncreases BDNF secretionPC12 cells[102, 103]
NGF potentiation
Citrus/limoleneERK1/2 activationPC12 cells[110, 111, 112, 113]
Saffron/N.D.Increases BDNF and GDNFN.D.[116, 117]
Bacopa Monieri/saponinsIncreases brain NGF andN.D.[120, 121]
BDNF levels
Skullcap/baicalinIncreases brain BDNFN.D.[126, 127]
ERK1/2 activation
Rhodiola rosea/salidrosideIncreases NT-3, BDNFstem cells[130, 131]
and NGF; ERK1/2 activation
Hops/prenylflavinoidsTRKA signalingPC12 cells[135, 136]
dorsal root ganglia
Nigella sativa/thymoquinoneN.D.hippocampal cells[141, 142]
dorsal root ganglia

Table 1.

The neurotrophic activities of anxiolytic plant extracts and phytochemical nutraceuticals.

All plants listed above have been shown to be anxiolytic in human clinical trials. In some cases the anxiolytic molecules and neurotrophic activities have not been determined (N.D.). Neurotrophic activities are those associated with activating neurotrophin signaling pathways by increasing levels of neurotrophin (NGF, BDNF, NT3, GDBF) synthesis, or by directly binding to neurotrophin receptors (TRKA, TRKB, TRKC and NGFR) or by activating the ERK1/2 signaling pathway. Another neurotrophic activity is the induction of neurite outgrowth or neuritogenesis in neuronal cell cultures and in. In these cases the names of the cells or tissues showing a neuroplastic response is provided.

2.1 Theanine

Theanine is an amino acid that when taken as a green tea extract or in a purified form is able to reduce anxiety in clinical trials [38, 39, 40]. When administered in a double blind placebo controlled study, theanine was shown to reduce stress-induced salivary cortisol levels [39]. However in other studies, while theanine did improve the sleep in people with GAD, theanine did not reduced anxiety scores on the HAMA scale [41]. Both animal and in vitro studies have suggested that theanine supplementation increases brain serotonin, dopamine and GABA levels and that the cellular target for theanine includes glutamate receptors to which theanine binds and antagonizes the stimulating action of glutamate on neurons [42, 43, 44]. With regard to neuroplasticity, theanine facilitates neuritogenesis in the developing rat hippocampus and enhances object learning memory [45]. Further, dietary theanine increases nerve growth factor (NGF) levels in the developing rat brain [46]. Theanine is not the only green tea molecule that can affect neurotrophin activity. The catechins from green tea have been show to potentiate BDNF binding to TRKB receptors in PC12 cells and enhance neurite outgrowth [47], and potentiate NGF signaling through TRKA receptors and enhance neurite outgrowth also in PC12 cells [48]. Further, the green tea catechin, green tea polyphenol (−)-epigallocatechin-3-gallate (EGCG) also stimulates neurite outgrowth in cultured PC12 neurons [49].

2.2 Chamomile

In clinical trials, chamomile has been shown to decrease the symptoms of general anxiety disorder [50, 51], in part by exerting an effect on diurnal cortisol changes [52]. While not yet known, apigenin is a plant flavone component of chamomile which is thought to contribute to the anxiolytic effects of chamomile [53]. Interestingly, apigenin increases neurite formation in murine N2a cells [54] and reverses PTZ induced behavioral impairments in mice by increasing hippocampal levels of brain derived neurotrophic factor (BDNF) [55]. Apigenin also has been shown to increase hippocampal BDNF levels in a chronic corticosteroid treatment model of depression in mice [56]. Apigenin also activates the ERK1/2 pathway in PC12 cells and while not sufficient to stimulate differentiation in PC12 cells [57], apigenin does increases neurite outgrowth in estrogen receptor expressing PC12 cells [58] again linking dietary phytochemicals that are anxiolytic to neural plasticity.

2.3 Lavender

Lavender oil also has anxiolytic effects in clinical trials in which it can both reduce anxiety associated with stressful event such as surgeries and recovery and also reducing anxiety in anxiety disorders [49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63]. Targets for lavender oil include the 5-HT1A serotonin receptor, the NMDA receptor and the serotonin transporter (SERT) [64, 65]. Linalool, a lavender oil terpene in specific can bind to SERT [64]. In a clinical trial where subjects were subjected to stress, linalool helped to reduce stress as measured by salivary cortisol levels, blood pressure and heart rate [66]. While linalool may be responsible for much of the anxiolytic effects of lavender oil, linalool has not been shown to have neurotrophic activity, however, lavender oil has be shown to increase neurite outgrowth and synapse formation in neuronal cell cultures [67] and increase both BDNF and nerve growth factor receptor (NGFR) levels in mouse brain [68]. Activation of NGFR is associated with enhanced TRKA receptor activity in neurons which triggers neurite outgrowth in response to NGF signaling [69, 70].

2.4 Ashwagandha

Ashwagandha is a plant used in Ayeurvic medicine from which the roots and berries have been used as adaptagens and also to relieve stress. In double blind placebo controlled clinical trials Ashwagandha supplementation has been shown to reduce anxiety based reducing both scores on the Hamiltion-Anxiety (HAMA) scale and morning salivary cortisol [71] and reduce anxiety in a variety of other contexts including schizophrenia and sleep disorders [71, 72, 73]. With regard to brain neurochemistry, Ashwagandha does not appear to affect sertotonergic, GABAnergic, or glutaminergic pathways but instead increases cholinergic signaling in the cortical and basal forebrain [74]. While the specific bioactive molecule(s) in Ashwagandha that are anxiolytic have not been specifically identified, sominone, an aglycone derivative of Withanoside IV when injected into mice stimulated neurite outgrowth in the hippocampus and increased production of the neurotrophin, Glial Derived neurotrophic Factor (GDNF) [75]. Further injection of sominone into mice enhances spatial memory, again suggesting that anxioloytic phytochemicals that are neurotrophic may ease anxiety by providing signals to enhance neural plasticity and learning [75].

2.5 Passion flower

Passion flower also shows anxiolytic properties in clinical trials that are as effective as midazolam and oxezepam [76, 77] and can reduce anxiety associated with ambulatory surgery and dental extraction [77, 78, 79]. The anxiolytic molecule from passion flower has not been identified and the effects of passion flower on brain neurochemistry is not well studied. It is interesting to note however that C-dideoxyhexosyl flavones from passion flower have been shown to enhance NGF-induced neurite outgrowth in PC12 cells [80].

2.6 Cannabidiol

Cannabidiol (CBD) is anxiolytic and has been shown in clinical trials to reduce social stress [81] and reduces anxiety in social phobia patients [82]. CBD also reduces anxiety associated with drug-craving during recovery from heroin addiction [83]. With regard to brain neurochemistry in clinical trials, CBD reduction in SAD was associated with increased blood flow in the limbic and paralimbic brain areas [84]. CBD is anxiolytic through direct binding of the GABAA receptor and activating the GABAnergic pathway [85, 86, 87]. CBD also bind to the NGF receptor, TRKA which signals the ERK1/2 signal transduction pathway and stimulates neurite outgrowth in PC12 cells [88]. Indeed the mechanism of action of CBD is recognized to help with the neuronal plasticity through autophagy and neuritogenesis and may help not only with anxiety, but also with other psychiatric disorders [88, 89]. Due to the lipophilicity of CBD, there is interest in developing emulsification techniques to increase CBD bioavailability when taken in the diet. For example, nanoemulsification [90] and lipid extractions [91] and lipid-vehicles [92] and piperine nanoliposhperes of CBD [93, 94] have been investigated for better oral absorption and better bioavailability for cellular targeting. Nanoemulsified, versus lipid emulsified CBD were tested for their ability to stimulate neurite outgrowth in PC12 cells (Figure 1). Continuous lipid extracted CBD shows greater bioavailability and activity compared to nanoemulsification and piperine nanoliposheperes (Figure 1).

Figure 1.

The effects of CBD on PC12 cell neurite outgrowth. PC12 cells were seeded on tissue culture plastic in a serum free defined medium and the percentage of cells that formed neurites were counted by visual inspection over a five day period. Cells were either untreated (blank) or treated with 100 ng/ml nerve growth factor (NGF) or with 10 uM of five different CBD formulations (CBD1-CBD5). CBD designations are as follows: CBD isolated by continuous lipid extraction, (CBD1), CBD1 + vitamin C (CBD2), nano-emulsion CBD (CBD3), liposomal-emulsion CBD (CBD 4) and piperine nanoliposhpere preparation CBD (CBD5). CBD1 and CBD2 were statistically significantly more neuritogenic at 95% confidence (*) on days three and five (p < 0.05, t-test) compared to any of the other CBD formulations.

2.7 Valerian root

The anxiolytic activity seen in patients supplementing with Valerian root extract [95], is known to be due to the sesquiterpene, valerenic acid [96]. While there is evidence to suggest that valerenic acid activates the GABAnergic pathway [97, 98], growing evidence suggests that valerenic acid mediates anxiolytic effects also by both antagonizing glutaminegric pathways [99, 100] and agonizing the serotonin receptor [101]. Valerenic acid also activates secretion of BDNF in cultured SH-SY5Y neurons [102]. Interestingly, germacrane, another sesquiterpene extracted from Valerian root, while not associated with an anxiolytic activity, has been shown to potentiate NGF and TRKA signaling and neurite outgrowth in PC12 cells [103]. An aqueous extract of valerian root enhances NGF-mediated neurite outgrowth and neuroplasticity but unlike CBD, the valerian extract is not neurotrophic in PC12 cells in the absence of NGF stimulation (Figure 2).

Figure 2.

The effect of valerian root extract and valerenic acid on neurite outgrowth in PC12 cell cultures. PC12 cells were seeded in serum free defined medium and treated as indicated with 10 ng/ml NGF, 100 μM valerenic acid and 50 mg/ml of 4:1 aqueous valerian root extract. The percentage of cells that formed neurites was counted by visual inspection over a five day period. More neurites were seen in PC12 cells treated with the valerian root extract and NGF when compared to NGF alone and these differences were statistically significantly at 95% confidence (*) on days one, three and five (p < 0.05, t-test). These data suggest valerian root extract phytochemical can potentiate NGF activity.

2.8 Citrus

Citrus plant extracts, including those from lemon, bitter orange, and bergamot relieve anxiety in clinical trials. For example, lemon inhalation reduced anxiety in myocardial infarction patients [104] and bergamot aromatherapy reduced preoperative anxiety [105] and bitter orange aroma therapy relieves anxiety in patients with acute coronary syndrome [106] and chronic myeloid leukemia [107] and preoperative anxiety [108]. Bitter orange extract contains primarily limolene and b-myrcrene, appear to act on the 5-HT serotonergic pathway [109]. When tested in PC12 cells, citrus phytochemicals such as nobilitin, gardenin A and auraptene all stimulate neurite outgrowth [110, 111, 112] and 5-Hydroxy-3,6,7,8,3′,4′-hexamethoxyflavone from sweet orange peel stimulates neurite outgrowth in and NGF-like fashion activating the ERK1/2 signaling pathway suggesting binding to TRKA [113].

2.9 Saffron

Saffron has been shown to be anxiolytic in two double blind placebo controlled clinical trials. [114, 115] and while the active anxiolytic molecule in Saffron has not been identified, crocin, a carotinoid in Saffron, has been shown to increase BDNF and GDNF expression in neuronal stem cells [116] and also increase hippocampal BDNF and protect the murine brain from methamphetamine toxicity [117].

2.10 Bacopa monnieri

In double blind placebo controlled trials, Bacopa monnieri, an adaptagen of Ayruvic medical tradition, has been show to enhance cognition and reduce anxiety [118, 119]. This anxiolytic adaptagen has been shown to increase nerve NGF expression in rats [120]. A saponin isolated from Bacopa monnieri, Bacopacide I, has been shown to have antidepressant activity in mice by modulating the HPA axis and enhancing BDNF mRNA expression in the hippocampus and prefrontal cortex of mice [121].

2.11 Skullcap

Skuttleria is a genus of plants known as the skullcaps that include scutellaria Radix and Scutellaria lateriflora (American skullcap). American skullcap has been shown to be anxiolytic in humans as shown by a reduction in anxiety in healthy volunteers using the Beck Anxiety Inventory (BAI) [122, 123]. The Skullcap flavone, baicalin which is found in american sckullcap and other members of the skuttleria genus has been shown to be anxiolytic by binding to GABAA receptors in mice [124, 125]. Interestingly biacalin increases hippocampal BDNF expression and in doing so protects the hippocampus from corticosterone induced depression in mice [126]. In addition biacalin stimulates neurite outgrowth in C17.2 neuronal stem cells by signaling through the ERK1/2 pathways, the known signal transduction triggered by NGF binding to TRKA [127].

2.12 Rhodeola Rosea

Rhodeola Rosea has been shown to reduce GAD in small pilot and self reporting clinical trials [128, 129]. While the effects of Rhodeola on brain neurochemistry has not been well studied, salidroside, a glycoside from Rhodiola has been shown to increase stem cells expression of neurotrophin-3 (NT-3), BDNF, NGF mRNA and induce differentiation into neurons [130] and also activates the ERK1/2 pathway in NGF treated PC12 cells [131].

2.13 Hops

One study shows Hops to be anxiolytic in clinical trials [132]. Prenylflavinoids from extracted from Hops can both bind to the benzodiazepine binding site on GABAA receptors [133, 134] and stimulate neurite outgrowth through TRKA a signaling in PC12 cells and cultures of dorsal root ganglia neurons [135, 136].

2.14 Nigella sativa

One clinical study shows that Nigella sativa seeds are anxiolytic in clinical trials [137]. Oral administration of Nigella extracts increase brain serotonin level in rats [138, 139] and thymoquinone, a terpine from Nigella is anxiolytic through a GABAnergic pathway when orally administered to mice [140]. Thymoquinone promotes neurite outgrowth in rat hippocampal neurons and dorsal root ganglion neurons [141, 142].

Advertisement

3. Conclusion

Phytochemical nutrients that are used to reduce anxiety may have this affect in part by stimulating neuroplasticity and altering the brain neurocircuitry associated with learned responses to stressors and threats. Fourteen of the roughly forty-five plant and plant extracts proven to reduce anxiety in humans in clinical trials are also able to act like neurotrophins; endogenous molecules that stimulate neuroplasticity in the human brain. Anxiolytic drugs are harsh and symptoms return when the drug is removed because the neurotransmitter chemistry returns to an imbalance. Neuroplasticity offers an opportunity to use food phytochemicals along with drugs or in their place to learn to establish more appropriate responses to perceived threats by reworking neural connections. These new neural connections may not be lost even when the anxiolytic treatment is removed. Using neuroplastic drugs and foods to not only alter brain chemistry, but also the circuitry, would be a tremendous advancement in the treatment of anxiety.

Advertisement

Acknowledgments

The funding to support this publication were provided by a grant from One Innovation Labs, 35 San Lorenzo Ave, Suite 850, Coral Gables, FL 33146 to Benjamin S. Weeks at Adelphi University, Garden City, NY 11530.

Advertisement

Acronyms and abbreviations

BDNF

Brain derive neurotrophic factor

NGF

Nerve growth factor

NT3

Neurotrophin-3

GDNF

Glial derived neurotrophic factor

TRK

Tropomyosin receptor kinase

GABA

Gama miniobutyric acid

CBD

Cannabidiol

GAD

Generalized anxiety disorder

PTSD

Post traumatic stress disorder

OCD

Obsessive compulsive disorder

PD

Panic disorder

SAD

Social anxiety disorder

References

  1. 1. Rajmohan V, Mohandas E. The limbic system. Indian J Psychiatry. 2007;49(2):132-139. doi:10.4103/0019-5545.33264
  2. 2. Smith SM, Vale WW. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin Neurosci. 2006;8(4):383-395. doi:10.31887/DCNS.2006.8.4/ssmith
  3. 3. Jankord R, Herman JP. Limbic regulation of hypothalamo-pituitary-adrenocortical function during acute and chronic stress. Ann N Y Acad Sci. 2008;1148:64-73. doi:10.1196/annals.1410.012
  4. 4. Romero LM, Gormally BMG. How Truly Conserved Is the "Well-Conserved" Vertebrate Stress Response?. Integr Comp Biol. 2019;59(2):273-281. doi:10.1093/icb/icz011
  5. 5. Chand SP, Marwaha R. Anxiety. In: StatPearls. Treasure Island (FL): StatPearls Publishing; November 29, 2020
  6. 6. Munir S, Takov V. Generalized Anxiety Disorder. In: StatPearls. Treasure Island (FL): StatPearls Publishing; November 19, 2020
  7. 7. Crocq MA. A history of anxiety: from Hippocrates to DSM. Dialogues Clin Neurosci. 2015;17(3):319-325. doi:10.31887/DCNS.2015.17.3/macrocq
  8. 8. Bao AM, Swaab DF. The human hypothalamus in mood disorders: The HPA axis in the center. IBRO Rep. 2018;6:45-53. Published 2018 Dec 14. doi:10.1016/j.ibror.2018.11.008
  9. 9. Martin EI, Ressler KJ, Binder E, Nemeroff CB. The neurobiology of anxiety disorders: brain imaging, genetics, and psychoneuroendocrinology. Psychiatr Clin North Am. 2009;32(3):549-575. doi:10.1016/j.psc.2009.05.004
  10. 10. Nemeroff CB. The role of GABA in the pathophysiology and treatment of anxiety disorders. Psychopharmacol Bull. 2003;37(4):133-146
  11. 11. Goddard AW, Mason GF, Appel M, et al. Impaired GABA neuronal response to acute benzodiazepine administration in panic disorder. Am J Psychiatry. 2004;161(12):2186-2193. doi:10.1176/appi.ajp.161.12.2186
  12. 12. Ham BJ, Sung Y, Kim N, et al. Decreased GABA levels in anterior cingulate and basal ganglia in medicated subjects with panic disorder: a proton magnetic resonance spectroscopy (1H-MRS) study. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31(2):403-411. doi:10.1016/j.pnpbp.2006.10.011
  13. 13. Karthik S, Sharma LP, Narayanaswamy JC. Investigating the Role of Glutamate in Obsessive-Compulsive Disorder: Current Perspectives. Neuropsychiatr Dis Treat. 2020;16:1003-1013. Published 2020 Apr 17. doi:10.2147/NDT.S211703
  14. 14. Rotge JY, Guehl D, Dilharreguy B, et al. Meta-analysis of brain volume changes in obsessive-compulsive disorder. Biol Psychiatry. 2009;65(1):75-83. doi:10.1016/j.biopsych.2008.06.019
  15. 15. Simpson HB, Shungu DC, Bender J Jr, et al. Investigation of cortical glutamate-glutamine and γ-aminobutyric acid in obsessive-compulsive disorder by proton magnetic resonance spectroscopy. Neuropsychopharmacology. 2012;37(12):2684-2692. doi:10.1038/npp.2012.132
  16. 16. Cortese BM, Phan KL. The role of glutamate in anxiety and related disorders. CNS Spectr. 2005;10(10):820-830. doi:10.1017/s1092852900010427
  17. 17. Engel K, Bandelow B, Gruber O, Wedekind D. Neuroimaging in anxiety disorders. J Neural Transm (Vienna). 2009;116(6):703-716. doi:10.1007/s00702-008-0077-9
  18. 18. Garakani A, Murrough JW, Freire RC, et al. Pharmacotherapy of Anxiety Disorders: Current and Emerging Treatment Options. Front Psychiatry. 2020;11:595584. Published 2020 Dec 23. doi:10.3389/fpsyt.2020.595584
  19. 19. Hjorth OR, Frick A, Gingnell M, et al. Expression and co-expression of serotonin and dopamine transporters in social anxiety disorder: a multitracer positron emission tomography study [published online ahead of print, 2019 Dec 10]. Mol Psychiatry. 2019;10.1038/s41380-019-0618-7. doi:10.1038/s41380-019-0618-7
  20. 20. Taylor DP, Riblet LA, Stanton HC, Eison AS, Eison MS, Temple DL Jr. Dopamine and antianxiety activity. Pharmacol Biochem Behav. 1982;17 Suppl 1:25-35. doi:10.1016/0091-3057(82)90507-x
  21. 21. Den Boer JA, Bosker FJ, Slaap BR. Serotonergic drugs in the treatment of depressive and anxiety disorders. Hum Psychopharmacol. 2000;15(5):315-336. doi:10.1002/1099-1077(200007)15:5<315::AID-HUP204>3.0.CO;2-Y
  22. 22. Zarrindast MR, Khakpai F. The Modulatory Role of Dopamine in Anxiety-like Behavior. Arch Iran Med. 2015;18(9):591-603
  23. 23. Alramadhan E, Hanna MS, Hanna MS, Goldstein TA, Avila SM, Weeks BS. Dietary and botanical anxiolytics. Med Sci Monit. 2012;18(4):RA40-RA48. doi:10.12659/msm.882608
  24. 24. Fedoce ADG, Ferreira F, Bota RG, Bonet-Costa V, Sun PY, Davies KJA. The role of oxidative stress in anxiety disorder: cause or consequence?. Free Radic Res. 2018;52(7):737-750. doi:10.1080/10715762.2018.1475733
  25. 25. Bouayed J, Rammal H, Soulimani R. Oxidative stress and anxiety: relationship and cellular pathways. Oxid Med Cell Longev. 2009;2(2):63-67. doi:10.4161/oxim.2.2.7944
  26. 26. Borodinsky LN, O'Leary D, Neale JH, Vicini S, Coso OA, Fiszman ML. GABA-induced neurite outgrowth of cerebellar granule cells is mediated by GABA(A) receptor activation, calcium influx and CaMKII and erk1/2 pathways. J Neurochem. 2003;84(6):1411-1420. doi:10.1046/j.1471-4159.2003.01638.x
  27. 27. Busch R, Baldus M, Vogt MA, et al. Effects of p75NTR deficiency on cholinergic innervation of the amygdala and anxiety-like behavior. J Neurochem. 2017;141(3):461-471. doi:10.1111/jnc.14006
  28. 28. Ribeiro DE, Casarotto PC, Spiacci AJ, et al. Activation of the TRKB receptor mediates the panicolytic-like effect of the NOS inhibitor aminoguanidine. Prog Neuropsychopharmacol Biol Psychiatry. 2019;93:232-239. doi:10.1016/j.pnpbp.2019.04.007
  29. 29. de Miranda AS, de Barros JLVM, Teixeira AL. Is neurotrophin-3 (NT-3): a potential therapeutic target for depression and anxiety?. Expert Opin Ther Targets. 2020;24(12):1225-1238. doi:10.1080/14728222.2020.1846720
  30. 30. Shah AP, Carreno FR, Wu H, Chung YA, Frazer A. Role of TrkB in the anxiolytic-like and antidepressant-like effects of vagal nerve stimulation: Comparison with desipramine. Neuroscience. 2016;322:273-286. doi:10.1016/j.neuroscience.2016.02.024
  31. 31. Règue-Guyon M, Lanfumey L, Mongeau R. Neuroepigenetics of Neurotrophin Signaling: Neurobiology of Anxiety and Affective Disorders. Prog Mol Biol Transl Sci. 2018;158:159-193. doi:10.1016/bs.pmbts.2018.03.002
  32. 32. Martinowich K, Manji H, Lu B. New insights into BDNF function in depression and anxiety. Nat Neurosci. 2007;10(9):1089-1093. doi:10.1038/nn1971
  33. 33. Zhou X, He X, He B, et al. Etifoxine promotes glial-derived neurotrophic factor-induced neurite outgrowth in PC12 cells. Mol Med Rep. 2013;8(1):75-80. doi:10.3892/mmr.2013.1474
  34. 34. Fox AS, Souaiaia T, Oler JA, et al. Dorsal Amygdala Neurotrophin-3 Decreases Anxious Temperament in Primates. Biol Psychiatry. 2019;86(12):881-889. doi:10.1016/j.biopsych.2019.06.022
  35. 35. Reichardt LF. Neurotrophin-regulated signalling pathways. Philos Trans R Soc Lond B Biol Sci. 2006;361(1473):1545-1564. doi:10.1098/rstb.2006.1894
  36. 36. Huang EJ, Reichardt LF. Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem. 2003;72:609-642. doi:10.1146/annurev.biochem.72.121801.161629
  37. 37. Skaper SD. Neurotrophic Factors: An Overview. Methods Mol Biol. 2018;1727:1-17. doi:10.1007/978-1-4939-7571-6_1
  38. 38. Unno K, Furushima D, Hamamoto S, et al. Stress-Reducing Function of Matcha Green Tea in Animal Experiments and Clinical Trials. Nutrients. 2018;10(10):1468. Published 2018 Oct 10. doi:10.3390/nu10101468
  39. 39. Unno K, Yamada H, Iguchi K, et al. Anti-stress Effect of Green Tea with Lowered Caffeine on Humans: A Pilot Study. Biol Pharm Bull. 2017;40(6):902-909. doi:10.1248/bpb.b17-00141
  40. 40. White DJ, de Klerk S, Woods W, Gondalia S, Noonan C, Scholey AB. Anti-Stress, Behavioural and Magnetoencephalography Effects of an L-Theanine-Based Nutrient Drink: A Randomised, Double-Blind, Placebo-Controlled, Crossover Trial. Nutrients. 2016;8(1):53. Published 2016 Jan 19. doi:10.3390/nu8010053
  41. 41. Sarris J, Byrne GJ, Cribb L, et al. L-theanine in the adjunctive treatment of generalized anxiety disorder: A double-blind, randomised, placebo-controlled trial. J Psychiatr Res. 2019;110:31-37. doi:10.1016/j.jpsychires.2018.12.014
  42. 42. Nathan PJ, Lu K, Gray M, Oliver C. The neuropharmacology of L-theanine(N-ethyl-L-glutamine): a possible neuroprotective and cognitive enhancing agent. J Herb Pharmacother. 2006;6(2):21-30
  43. 43. Cho HS, Kim S, Lee SY, Park JA, Kim SJ, Chun HS. Protective effect of the green tea component, L-theanine on environmental toxins-induced neuronal cell death. Neurotoxicology. 2008;29(4):656-662. doi:10.1016/j.neuro.2008.03.004
  44. 44. Yokogoshi H, Kobayashi M, Mochizuki M, Terashima T. Effect of theanine, r-glutamylethylamide, on brain monoamines and striatal dopamine release in conscious rats. Neurochem Res. 1998;23(5):667-673. doi:10.1023/a:1022490806093
  45. 45. Takeda A, Sakamoto K, Tamano H, et al. Facilitated neurogenesis in the developing hippocampus after intake of theanine, an amino acid in tea leaves, and object recognition memory. Cell Mol Neurobiol. 2011;31(7):1079-1088. doi:10.1007/s10571-011-9707-0
  46. 46. Yamada T, Terashima T, Wada K, et al. Theanine, r-glutamylethylamide, increases neurotransmission concentrations and neurotrophin mRNA levels in the brain during lactation. Life Sci. 2007;81(16):1247-1255. doi:10.1016/j.lfs.2007.08.023
  47. 47. Gundimeda U, McNeill TH, Fan TK, et al. Green tea catechins potentiate the neuritogenic action of brain-derived neurotrophic factor: role of 67-kDa laminin receptor and hydrogen peroxide. Biochem Biophys Res Commun. 2014;445(1):218-224. doi:10.1016/j.bbrc.2014.01.166
  48. 48. Gundimeda U, McNeill TH, Schiffman JE, Hinton DR, Gopalakrishna R. Green tea polyphenols potentiate the action of nerve growth factor to induce neuritogenesis: possible role of reactive oxygen species. J Neurosci Res. 2010;88(16):3644-3655. doi:10.1002/jnr.22519
  49. 49. Reznichenko L, Amit T, Youdim MB, Mandel S. Green tea polyphenol (−)-epigallocatechin-3-gallate induces neurorescue of long-term serum-deprived PC12 cells and promotes neurite outgrowth. J Neurochem. 2005;93(5):1157-1167. doi:10.1111/j.1471-4159.2005.03085.x
  50. 50. Mao JJ, Xie SX, Keefe JR, Soeller I, Li QS, Amsterdam JD. Long-term chamomile (Matricaria chamomilla L.) treatment for generalized anxiety disorder: A randomized clinical trial. Phytomedicine. 2016;23(14):1735-1742. doi:10.1016/j.phymed.2016.10.012
  51. 51. Amsterdam JD, Li Y, Soeller I, Rockwell K, Mao JJ, Shults J. A randomized, double-blind, placebo-controlled trial of oral Matricaria recutita (chamomile) extract therapy for generalized anxiety disorder. J Clin Psychopharmacol. 2009;29(4):378-382. doi:10.1097/JCP.0b013e3181ac935c
  52. 52. Keefe JR, Guo W, Li QS, Amsterdam JD, Mao JJ. An exploratory study of salivary cortisol changes during chamomile extract therapy of moderate to severe generalized anxiety disorder. J Psychiatr Res. 2018;96:189-195. doi:10.1016/j.jpsychires.2017.10.011
  53. 53. Schäfer AM, Gilgen PM, Spirgi C, Potterat O, Meyer Zu Schwabedissen HE. Constituents of Passiflora incarnata, but Not of Valeriana officinalis, Interact with the Organic Anion Transporting Polypeptides (OATP)2B1 and OATP1A2 [published online ahead of print, 2021 Jan 28]. Planta Med. 2021;10.1055/a-1305-3936. doi:10.1055/a-1305-3936
  54. 54. Namsi A, Nury T, Hamdouni H, Yammine A, Vejux A, Vervandier-Fasseur D, Latruffe N, Masmoudi-Kouki O, Lizard G. Induction of Neuronal Differentiation of Murine N2a Cells by Two Polyphenols Present in the Mediterranean Diet Mimicking Neurotrophins Activities: Resveratrol and Apigenin. Diseases. 2018 Jul 22;6(3):67. doi: 10.3390/diseases6030067. PMID: 30037152; PMCID: PMC6165409
  55. 55. Sharma P, Sharma S, Singh D. Apigenin reverses behavioural impairments and cognitive decline in kindled mice via CREB-BDNF upregulation in the hippocampus. Nutr Neurosci. 2020 Feb;23(2):118-127. doi: 10.1080/1028415X.2018.1478653. Epub 2018 May 30. PMID: 29847220
  56. 56. Weng L, Guo X, Li Y, Yang X, Han Y. Apigenin reverses depression-like behavior induced by chronic corticosterone treatment in mice. Eur J Pharmacol. 2016;774:50-54. doi:10.1016/j.ejphar.2016.01.015
  57. 57. Llorens F, Garcia L, Itarte E, Gómez N. Apigenin and LY294002 prolong EGF-stimulated ERK1/2 activation in PC12 cells but are unable to induce full differentiation. FEBS Lett. 2002;510(3):149-153. doi:10.1016/s0014-5793(01)03252-5
  58. 58. Lecomte S, Lelong M, Bourgine G, Efstathiou T, Saligaut C, Pakdel F. Assessment of the potential activity of major dietary compounds as selective estrogen receptor modulators in two distinct cell models for proliferation and differentiation. Toxicol Appl Pharmacol. 2017;325:61-70. doi:10.1016/j.taap.2017.04.005
  59. 59. Kasper S, Möller HJ, Volz HP, Schläfke S, Dienel A. Silexan in generalized anxiety disorder: investigation of the therapeutic dosage range in a pooled data set. Int Clin Psychopharmacol. 2017;32(4):195-204. doi:10.1097/YIC.0000000000000176
  60. 60. Seifritz E, Schläfke S, Holsboer-Trachsler E. Beneficial effects of Silexan on sleep are mediated by its anxiolytic effect. J Psychiatr Res. 2019;115:69-74. doi:10.1016/j.jpsychires.2019.04.013
  61. 61. Karan NB. Influence of lavender oil inhalation on vital signs and anxiety: A randomized clinical trial. Physiol Behav. 2019;211:112676. doi:10.1016/j.physbeh.2019.112676
  62. 62. Woelk H, Schläfke S. A multi-center, double-blind, randomised study of the Lavender oil preparation Silexan in comparison to Lorazepam for generalized anxiety disorder. Phytomedicine. 2010;17(2):94-99. doi:10.1016/j.phymed.2009.10.006
  63. 63. Bozkurt P, Vural Ç. Effect of Lavender Oil Inhalation on Reducing Presurgical Anxiety in Orthognathic Surgery Patients. J Oral Maxillofac Surg. 2019;77(12):2466.e1-2466.e7. doi:10.1016/j.joms.2019.08.022
  64. 64. López V, Nielsen B, Solas M, Ramírez MJ, Jäger AK. Exploring Pharmacological Mechanisms of Lavender (Lavandula angustifolia) Essential Oil on Central Nervous System Targets. Front Pharmacol. 2017;8:280. Published 2017 May 19. doi:10.3389/fphar.2017.00280
  65. 65. Chioca LR, Ferro MM, Baretta IP, et al. Anxiolytic-like effect of lavender essential oil inhalation in mice: participation of serotonergic but not GABAA/benzodiazepine neurotransmission. J Ethnopharmacol. 2013;147(2):412-418. doi:10.1016/j.jep.2013.03.028
  66. 66. Höferl M, Krist S, Buchbauer G. Chirality influences the effects of linalool on physiological parameters of stress. Planta Med. 2006;72(13):1188-1192. doi:10.1055/s-2006-947202
  67. 67. Friedland K, Silani G, Schuwald A, et al. Neurotrophic Properties of Silexan, an Essential Oil from the Flowers of Lavender-Preclinical Evidence for Antidepressant-Like Properties. Pharmacopsychiatry. 2021;54(1):37-46. doi:10.1055/a-1293-8585
  68. 68. Takahashi M, Yoshino A, Yamanaka A, et al. Effects of inhaled lavender essential oil on stress-loaded animals: changes in anxiety-related behavior and expression levels of selected mRNAs and proteins. Nat Prod Commun. 2012;7(11):1539-1544
  69. 69. Soligo M, Albini M, Bertoli FL, et al. Different responses of PC12 cells to different pro-nerve growth factor protein variants. Neurochem Int. 2019;129:104498. doi:10.1016/j.neuint.2019.104498
  70. 70. Bradshaw RA, Pundavela J, Biarc J, Chalkley RJ, Burlingame AL, Hondermarck H. NGF and ProNGF: Regulation of neuronal and neoplastic responses through receptor signaling. Adv Biol Regul. 2015;58:16-27. doi:10.1016/j.jbior.2014.11.003
  71. 71. Lopresti AL, Smith SJ, Malvi H, Kodgule R. An investigation into the stress-relieving and pharmacological actions of an ashwagandha (Withania somnifera) extract: A randomized, double-blind, placebo-controlled study. Medicine (Baltimore). 2019;98(37):e17186. doi:10.1097/MD.0000000000017186
  72. 72. Gannon JM, Brar J, Rai A, Chengappa KNR. Effects of a standardized extract of Withania somnifera (Ashwagandha) on depression and anxiety symptoms in persons with schizophrenia participating in a randomized, placebo-controlled clinical trial. Ann Clin Psychiatry. 2019;31(2):123-129
  73. 73. Deshpande A, Irani N, Balakrishnan R. Study protocol and rationale for a prospective, randomized, double-blind, placebo-controlled study to evaluate the effects of Ashwagandha (Withania somnifera) extract on nonrestorative sleep. Medicine (Baltimore). 2018;97(26):e11299. doi:10.1097/MD.0000000000011299
  74. 74. Schliebs R, Liebmann A, Bhattacharya SK, Kumar A, Ghosal S, Bigl V. Systemic administration of defined extracts from Withania somnifera (Indian Ginseng) and Shilajit differentially affects cholinergic but not glutamatergic and GABAergic markers in rat brain. Neurochem Int. 1997;30(2):181-190. doi:10.1016/s0197-0186(96)00025-3
  75. 75. Tohda C, Joyashiki E. Sominone enhances neurite outgrowth and spatial memory mediated by the neurotrophic factor receptor, RET. Br J Pharmacol. 2009;157(8):1427-1440. doi:10.1111/j.1476-5381.2009.00313.x
  76. 76. Akhondzadeh S, Naghavi HR, Vazirian M, Shayeganpour A, Rashidi H, Khani M. Passionflower in the treatment of generalized anxiety: a pilot double-blind randomized controlled trial with oxazepam. J Clin Pharm Ther. 2001;26(5):363-367. doi:10.1046/j.1365-2710.2001.00367.x
  77. 77. Dantas LP, de Oliveira-Ribeiro A, de Almeida-Souza LM, Groppo FC. Effects of Passiflora incarnata and midazolam for control of anxiety in patients undergoing dental extraction. Med Oral Patol Oral Cir Bucal. 2017;22(1):e95-e101. Published 2017 Jan 1. doi:10.4317/medoral.21140
  78. 78. Movafegh A, Alizadeh R, Hajimohamadi F, Esfehani F, Nejatfar M. Preoperative oral Passiflora incarnata reduces anxiety in ambulatory surgery patients: a double-blind, placebo-controlled study. Anesth Analg. 2008;106(6):1728-1732. doi:10.1213/ane.0b013e318172c3f9
  79. 79. da Cunha RS, Amorim KS, Gercina AC, et al. Herbal medicines as anxiolytics prior to third molar surgical extraction. A randomized controlled clinical trial. Clin Oral Investig. 2021;25(3):1579-1586. doi:10.1007/s00784-020-03468-1
  80. 80. Xu F, Wang C, Yang L, et al. C-dideoxyhexosyl flavones from the stems and leaves of Passiflora edulis Sims. Food Chem. 2013;136(1):94-99. doi:10.1016/j.foodchem.2012.07.101
  81. 81. Appiah-Kusi E, Petros N, Wilson R, et al. Effects of short-term cannabidiol treatment on response to social stress in subjects at clinical high risk of developing psychosis. Psychopharmacology (Berl). 2020;237(4):1121-1130. doi:10.1007/s00213-019-05442-6
  82. 82. Bergamaschi MM, Queiroz RH, Chagas MH, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naïve social phobia patients. Neuropsychopharmacology. 2011;36(6):1219-1226. doi:10.1038/npp.2011.6
  83. 83. Hurd YL, Spriggs S, Alishayev J, et al. Cannabidiol for the Reduction of Cue-Induced Craving and Anxiety in Drug-Abstinent Individuals With Heroin Use Disorder: A Double-Blind Randomized Placebo-Controlled Trial [published correction appears in Am J Psychiatry. 2020 Jul 1;177(7):641]. Am J Psychiatry. 2019;176(11):911-922. doi:10.1176/appi.ajp.2019.18101191
  84. 84. Crippa JA, Derenusson GN, Ferrari TB, et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J Psychopharmacol. 2011;25(1):121-130. doi:10.1177/0269881110379283
  85. 85. Bakas T, van Nieuwenhuijzen PS, Devenish SO, McGregor IS, Arnold JC, Chebib M. The direct actions of cannabidiol and 2-arachidonoyl glycerol at GABAA receptors. Pharmacol Res. 2017;119:358-370. doi:10.1016/j.phrs.2017.02.022
  86. 86. Pretzsch CM, Freyberg J, Voinescu B, et al. Effects of cannabidiol on brain excitation and inhibition systems; a randomised placebo-controlled single dose trial during magnetic resonance spectroscopy in adults with and without autism spectrum disorder. Neuropsychopharmacology. 2019;44(8):1398-1405. doi:10.1038/s41386-019-0333-8
  87. 87. Cifelli P, Ruffolo G, De Felice E, et al. Phytocannabinoids in Neurological Diseases: Could They Restore a Physiological GABAergic Transmission?. Int J Mol Sci. 2020;21(3):723. Published 2020 Jan 22. doi:10.3390/ijms21030723
  88. 88. Santos NA, Martins NM, Sisti FM, et al. The neuroprotection of cannabidiol against MPP+-induced toxicity in PC12 cells involves trkA receptors, upregulation of axonal and synaptic proteins, neuritogenesis, and might be relevant to Parkinson's disease. Toxicol In Vitro. 2015;30(1 Pt B):231-240. doi:10.1016/j.tiv.2015.11.004
  89. 89. Campos AC, Fogaça MV, Scarante FF, et al. Plastic and Neuroprotective Mechanisms Involved in the Therapeutic Effects of Cannabidiol in Psychiatric Disorders. Front Pharmacol. 2017;8:269. Published 2017 May 23. doi:10.3389/fphar.2017.00269
  90. 90. Izgelov D, Shmoeli E, Domb AJ, Hoffman A. The effect of medium chain and long chain triglycerides incorporated in self-nano emulsifying drug delivery systems on oral absorption of cannabinoids in rats. Int J Pharm. 2020;580:119201. doi:10.1016/j.ijpharm.2020.119201
  91. 91. Knaub K, Sartorius T, Dharsono T, Wacker R, Wilhelm M, Schön C. A Novel Self-Emulsifying Drug Delivery System (SEDDS) Based on VESIsorb® Formulation Technology Improving the Oral Bioavailability of Cannabidiol in Healthy Subjects. Molecules. 2019;24(16):2967. Published 2019 Aug 16. doi:10.3390/molecules24162967
  92. 92. Izgelov D, Regev A, Domb AJ, Hoffman A. Using the Absorption Cocktail Approach to Assess Differential Absorption Kinetics of Cannabidiol Administered in Lipid-Based Vehicles in Rats. Mol Pharm. 2020;17(6):1979-1986. doi:10.1021/acs.molpharmaceut.0c00141
  93. 93. Cherniakov I, Izgelov D, Barasch D, Davidson E, Domb AJ, Hoffman A. Piperine-pro-nanolipospheres as a novel oral delivery system of cannabinoids: Pharmacokinetic evaluation in healthy volunteers in comparison to buccal spray administration. J Control Release. 2017;266:1-7. doi:10.1016/j.jconrel.2017.09.011
  94. 94. Izgelov D, Domb AJ, Hoffman A. The effect of piperine on oral absorption of cannabidiol following acute vs. chronic administration. Eur J Pharm Sci. 2020;148:105313. doi:10.1016/j.ejps.2020.105313
  95. 95. Pakseresht S, Boostani H, Sayyah M. Extract of valerian root (Valeriana officinalis L.) vs. placebo in treatment of obsessive-compulsive disorder: a randomized double-blind study. J Complement Integr Med. 2011;8:/j/jcim.2011.8.issue-1/1553-3840.1465/1553-3840.1465.xml. Published 2011 Oct 11. doi:10.2202/1553-3840.1465
  96. 96. Becker A, Felgentreff F, Schröder H, Meier B, Brattström A. The anxiolytic effects of a Valerian extract is based on valerenic acid. BMC Complement Altern Med. 2014;14:267. Published 2014 Jul 28. doi:10.1186/1472-6882-14-267
  97. 97. Orhan IE. A Review Focused on Molecular Mechanisms of Anxiolytic Effect of Valerina officinalis L. in Connection with Its Phytochemistry through in vitro/in vivo Studies [published online ahead of print, 2021 Jan 18]. Curr Pharm Des. 2021;10.2174/1381612827666210119105254. doi:10.2174/1381612827666210119105254
  98. 98. Thomas K, Canedo J, Perry PJ, et al. Effects of valerian on subjective sedation, field sobriety testing and driving simulator performance. Accid Anal Prev. 2016;92:240-244. doi:10.1016/j.aap.2016.01.019
  99. 99. Del Valle-Mojica LM, Ayala-Marín YM, Ortiz-Sanchez CM, Torres-Hernández BA, Abdalla-Mukhaimer S, Ortiz JG. Selective Interactions of Valeriana officinalis Extracts and Valerenic Acid with [H]Glutamate Binding to Rat Synaptic Membranes. Evid Based Complement Alternat Med. 2011;2011:403591. doi:10.1155/2011/403591
  100. 100. Del Valle-Mojica LM, Ortíz JG. Anxiolytic properties of Valeriana officinalis in the zebrafish: a possible role for metabotropic glutamate receptors. Planta Med. 2012;78(16):1719-1724. doi:10.1055/s-0032-1315240
  101. 101. Dietz BM, Mahady GB, Pauli GF, Farnsworth NR. Valerian extract and valerenic acid are partial agonists of the 5-HT5a receptor in vitro. Brain Res Mol Brain Res. 2005;138(2):191-197. doi:10.1016/j.molbrainres.2005.04.009
  102. 102. Gonulalan EM, Bayazeid O, Yalcin FN, Demirezer LO. The roles of valerenic acid on BDNF expression in the SH-SY5Y cell. Saudi Pharm J. 2018;26(7):960-964. doi:10.1016/j.jsps.2018.05.005
  103. 103. Chen HW, Chen L, Li B, et al. Three new germacrane-type sesquiterpenes with NGF-potentiating activity from Valeriana officinalis var. latiofolia. Molecules. 2013;18(11):14138-14147. Published 2013 Nov 14. doi:10.3390/molecules181114138
  104. 104. Rambod M, Rakhshan M, Tohidinik S, Nikoo MH. The effect of lemon inhalation aromatherapy on blood pressure, electrocardiogram changes, and anxiety in acute myocardial infarction patients: A clinical, multi-centered, assessor-blinded trial design. Complement Ther Clin Pract. 2020;39:101155. doi:10.1016/j.ctcp.2020.101155
  105. 105. Pasyar N, Rambod M, Araghi F. The effect of bergamot orange essence on anxiety, salivary cortisol, and alpha amylase in patients prior to laparoscopic cholecystectomy: A controlled trial study. Complement Ther Clin Pract. 2020;39:101153. doi:10.1016/j.ctcp.2020.101153
  106. 106. Moslemi F, Alijaniha F, Naseri M, Kazemnejad A, Charkhkar M, Heidari MR. Citrus aurantium Aroma for Anxiety in Patients with Acute Coronary Syndrome: A Double-Blind Placebo-Controlled Trial. J Altern Complement Med. 2019;25(8):833-839. doi:10.1089/acm.2019.0061
  107. 107. Pimenta FC, Alves MF, Pimenta MB, et al. Anxiolytic Effect of Citrus aurantium L. on Patients with Chronic Myeloid Leukemia. Phytother Res. 2016;30(4):613-617. doi:10.1002/ptr.5566
  108. 108. Akhlaghi M, Shabanian G, Rafieian-Kopaei M, Parvin N, Saadat M, Akhlaghi M. Citrus aurantium blossom and preoperative anxiety. Rev Bras Anestesiol. 2011;61(6):702-712. doi:10.1016/S0034-7094(11)70079-4
  109. 109. Costa CA, Cury TC, Cassettari BO, Takahira RK, Flório JC, Costa M. citrus aurantium L. essential oil exhibits anxiolytic-like activity mediated by 5-HT(1A)-receptors and reduces cholesterol after repeated oral treatment. BMC Complement Altern Med. 2013;13:42. Published 2013 Feb 23. doi:10.1186/1472-6882-13-42
  110. 110. Chiu SP, Wu MJ, Chen PY, et al. Neurotrophic action of 5-hydroxylated polymethoxyflavones: 5-demethylnobiletin and gardenin A stimulate neuritogenesis in PC12 cells. J Agric Food Chem. 2013;61(39):9453-9463. doi:10.1021/jf4024678
  111. 111. Nagase H, Omae N, Omori A, et al. Nobiletin and its related flavonoids with CRE-dependent transcription-stimulating and neuritegenic activities. Biochem Biophys Res Commun. 2005;337(4):1330-1336. doi:10.1016/j.bbrc.2005.10.001
  112. 112. Furukawa Y, Watanabe S, Okuyama S, Nakajima M. Neurotrophic effect of citrus auraptene: neuritogenic activity in PC12 cells. Int J Mol Sci. 2012;13(5):5338-5347. doi:10.3390/ijms13055338
  113. 113. Lai HC, Wu MJ, Chen PY, et al. Neurotrophic effect of citrus 5-hydroxy-3,6,7,8,3′,4′-hexamethoxyflavone: promotion of neurite outgrowth via cAMP/PKA/CREB pathway in PC12 cells. PLoS One. 2011;6(11):e28280. doi:10.1371/journal.pone.0028280
  114. 114. Lopresti AL, Drummond PD, Inarejos-García AM, Prodanov M. affron®, a standardised extract from saffron (Crocus sativus L.) for the treatment of youth anxiety and depressive symptoms: A randomised, double-blind, placebo-controlled study. J Affect Disord. 2018;232:349-357. doi:10.1016/j.jad.2018.02.070
  115. 115. Ghajar A, Neishabouri SM, Velayati N, et al. Crocus sativus L. versus Citalopram in the Treatment of Major Depressive Disorder with Anxious Distress: A Double-Blind, Controlled Clinical Trial. Pharmacopsychiatry. 2017;50(4):152-160. doi:10.1055/s-0042-116159
  116. 116. Baharvand Z, Nabiuni M, Tahmaseb M, Amini E, Pandamooz S. Investigating the synergic effects of valproic acid and crocin on BDNF and GDNF expression in epidermal neural crest stem cells. Acta Neurobiol Exp (Wars). 2020;80(1):38-46
  117. 117. Mozaffari S, Ramezany Yasuj S, Motaghinejad M, Motevalian M, Kheiri R. Crocin Acting as a Neuroprotective Agent against Methamphetamine-induced Neurodegeneration via CREB-BDNF Signaling Pathway. Iran J Pharm Res. 2019;18(2):745-758. doi:10.22037/ijpr.2019.2393
  118. 118. Benson S, Downey LA, Stough C, Wetherell M, Zangara A, Scholey A. An acute, double-blind, placebo-controlled cross-over study of 320 mg and 640 mg doses of Bacopa monnieri (CDRI 08) on multitasking stress reactivity and mood. Phytother Res. 2014;28(4):551-559. doi:10.1002/ptr.5029
  119. 119. Downey LA, Kean J, Nemeh F, et al. An acute, double-blind, placebo-controlled crossover study of 320 mg and 640 mg doses of a special extract of Bacopa monnieri (CDRI 08) on sustained cognitive performance. Phytother Res. 2013;27(9):1407-1413. doi:10.1002/ptr.4864
  120. 120. Priyanka HP, Bala P, Ankisettipalle S, ThyagaRajan S. Bacopa monnieri and L-deprenyl differentially enhance the activities of antioxidant enzymes and the expression of tyrosine hydroxylase and nerve growth factor via ERK 1/2 and NF-κB pathways in the spleen of female wistar rats. Neurochem Res. 2013;38(1):141-152. doi:10.1007/s11064-012-0902-2
  121. 121. Zu X, Zhang M, Li W, et al. Antidepressant-like Effect of Bacopaside I in Mice Exposed to Chronic Unpredictable Mild Stress by Modulating the Hypothalamic-Pituitary-Adrenal Axis Function and Activating BDNF Signaling Pathway. Neurochem Res. 2017;42(11):3233-3244. doi:10.1007/s11064-017-2360-3
  122. 122. Brock C, Whitehouse J, Tewfik I, Towell T. American Skullcap (Scutellaria lateriflora): a randomised, double-blind placebo-controlled crossover study of its effects on mood in healthy volunteers. Phytother Res. 2014;28(5):692-698. doi:10.1002/ptr.5044
  123. 123. Wolfson P, Hoffmann DL. An investigation into the efficacy of Scutellaria lateriflora in healthy volunteers. Altern Ther Health Med. 2003;9(2):74-78
  124. 124. Liao JF, Hung WY, Chen CF. Anxiolytic-like effects of baicalein and baicalin in the Vogel conflict test in mice. Eur J Pharmacol. 2003;464(2-3):141-146. doi:10.1016/s0014-2999(03)01422-5
  125. 125. Wang F, Xu Z, Ren L, Tsang SY, Xue H. GABA A receptor subtype selectivity underlying selective anxiolytic effect of baicalin. Neuropharmacology. 2008;55(7):1231-1237. doi:10.1016/j.neuropharm.2008.07.040
  126. 126. Li YC, Wang LL, Pei YY, et al. Baicalin decreases SGK1 expression in the hippocampus and reverses depressive-like behaviors induced by corticosterone. Neuroscience. 2015;311:130-137. doi:10.1016/j.neuroscience.2015.10.023
  127. 127. Li M, Tsang KS, Choi ST, Li K, Shaw PC, Lau KF. Neuronal differentiation of C17.2 neural stem cells induced by a natural flavonoid, baicalin. Chembiochem. 2011;12(3):449-456. doi:10.1002/cbic.201000570
  128. 128. Bystritsky A, Kerwin L, Feusner JD. A pilot study of Rhodiola rosea (Rhodax) for generalized anxiety disorder (GAD). J Altern Complement Med. 2008;14(2):175-180. doi:10.1089/acm.2007.7117
  129. 129. Cropley M, Banks AP, Boyle J. The Effects of Rhodiola rosea L. Extract on Anxiety, Stress, Cognition and Other Mood Symptoms. Phytother Res. 2015;29(12):1934-1939. doi:10.1002/ptr.5486
  130. 130. Zhao HB, Ma H, Ha XQ , et al. Salidroside induces rat mesenchymal stem cells to differentiate into dopaminergic neurons. Cell Biol Int. 2014;38(4):462-471. doi:10.1002/cbin.10217
  131. 131. Yu S, Shen Y, Liu J, Ding F. Involvement of ERK1/2 pathway in neuroprotection by salidroside against hydrogen peroxide-induced apoptotic cell death. J Mol Neurosci. 2010;40(3):321-331. doi:10.1007/s12031-009-9292-6
  132. 132. Kyrou I, Christou A, Panagiotakos D, et al. Effects of a hops (Humulus lupulus L.) dry extract supplement on self-reported depression, anxiety and stress levels in apparently healthy young adults: a randomized, placebo-controlled, double-blind, crossover pilot study. Hormones (Athens). 2017;16(2):171-180. doi:10.14310/horm.2002.1738
  133. 133. Benkherouf AY, Soini SL, Stompor M, Uusi-Oukari M. Positive allosteric modulation of native and recombinant GABAA receptors by hops prenylflavonoids. Eur J Pharmacol. 2019;852:34-41. doi:10.1016/j.ejphar.2019.02.034
  134. 134. Benkherouf AY, Logrén N, Somborac T, et al. Hops compounds modulatory effects and 6-prenylnaringenin dual mode of action on GABAA receptors. Eur J Pharmacol. 2020;873:172962. doi:10.1016/j.ejphar.2020.172962
  135. 135. Bieler L, Vogl M, Kirchinger M, et al. The Prenylflavonoid ENDF1 Overrules Central Nervous System Growth Inhibitors and Facilitates Regeneration of DRG Neurons. Front Cell Neurosci. 2019;13:332. Published 2019 Jul 24. doi:10.3389/fncel.2019.00332
  136. 136. Oberbauer E, Urmann C, Steffenhagen C, et al. Chroman-like cyclic prenylflavonoids promote neuronal differentiation and neurite outgrowth and are neuroprotective. J Nutr Biochem. 2013;24(11):1953-1962. doi:10.1016/j.jnutbio.2013.06.005
  137. 137. Bin Sayeed MS, Shams T, Fahim Hossain S, et al. Nigella sativa L. seeds modulate mood, anxiety and cognition in healthy adolescent males. J Ethnopharmacol. 2014;152(1):156-162. doi:10.1016/j.jep.2013.12.050
  138. 138. Perveen T, Haider S, Zuberi NA, Saleem S, Sadaf S, Batool Z. Increased 5-HT Levels Following Repeated Administration of Nigella sativa L. (Black Seed) Oil Produce Antidepressant Effects in Rats. Sci Pharm. 2013;82(1):161-170. Published 2013 Nov 5. doi:10.3797/scipharm.1304-19
  139. 139. Perveen T, Haider S, Kanwal S, Haleem DJ. Repeated administration of Nigella sativa decreases 5-HT turnover and produces anxiolytic effects in rats. Pak J Pharm Sci. 2009;22(2):139-144
  140. 140. Gilhotra N, Dhingra D. Thymoquinone produced antianxiety-like effects in mice through modulation of GABA and NO levels. Pharmacol Rep. 2011;63(3):660-669. doi:10.1016/s1734-1140(11)70577-1
  141. 141. Beker M, Dallı T, Elibol B. Thymoquinone Can Improve Neuronal Survival and Promote Neurogenesis in Rat Hippocampal Neurons. Mol Nutr Food Res. 2018;62(5):10.1002/mnfr.201700768. doi:10.1002/mnfr.201700768
  142. 142. Üstün R, Oğuz EK, Şeker A, Korkaya H. Thymoquinone prevents cisplatin neurotoxicity in primary DRG neurons. Neurotoxicology. 2018;69:68-76. doi:10.1016/j.neuro.2018.09.001

Written By

Benjamin S. Weeks, Samuel D. Weeks, Amanda Kim, Landon Kessler and Pedro P. Perez

Submitted: 27 October 2020 Reviewed: 02 April 2021 Published: 26 April 2021