Open access peer-reviewed chapter

Neuropharmacology of Secondary Metabolites from Plants with Anxiolytic and Antidepressant Properties

Written By

Rosa Isela García-Ríos, Armando Mora-Pérez, Ana Raquel Ramos-Molina and Cesar Soria-Fregozo

Submitted: 27 May 2019 Reviewed: 20 December 2019 Published: 04 February 2020

DOI: 10.5772/intechopen.90919

From the Edited Volume

Behavioral Pharmacology - From Basic to Clinical Research

Edited by Juan Francisco Rodríguez-Landa and Jonathan Cueto-Escobedo

Chapter metrics overview

1,151 Chapter Downloads

View Full Metrics

Abstract

Depression and anxiety currently rank as the second and fifth most common causes worldwide of years lived with disability-a reality that has intensified the search for new treatments. There are many studies of herbal extracts and secondary metabolites from plants used in traditional medicine due to their antidepressant and anxiolytic properties. Clinical and preclinical studies about some of the mechanisms of action of metabolites like alkaloids, terpenes, flavonoids, and sterols, among others, have documented effects similar to those produced by clinically effective drugs. These metabolites have shown anxiolytic and antidepressant effects in various experimental models of anxiety by interacting with γ-aminobutyric acid subtype A receptors (GABAA-receptors) and by stimulating the serotonergic, noradrenergic, and dopaminergic neurotransmitter systems. These pharmacological effects can be attributed to plant metabolites that share structural similarities with monoamines, which allow them to bind to receptors. The objective of this chapter is to summarize the various mechanisms of action that have been identified in secondary metabolites with anxiolytic and antidepressant properties. Terpenes, alkaloids, flavonoids, and sterols can interact at different levels of the neurotransmission systems involved in the neurobiology of anxiety and depression, suggesting their potential for treating these mental illnesses.

Keywords

  • antidepressant
  • anxiolytic
  • active metabolites
  • plant extracts
  • herbal medicines

1. Introduction

According to the Global Burden of Disease, depression and anxiety are currently the second and fifth most common causes worldwide of years lived with disability in both sexes in the age range of 15–49 years [1]. In 2015, 4.4% (322 million people) of the world’s population suffered from depressive disorders, while 3.6% (264 million) were affected by anxiety [2]. In that year, the World Health Organization (WHO) estimated that by 2020 depression would be the second leading cause of disability; thus, its prediction has been confirmed. Depression is characterized by persistent sadness and a loss of interest in activities that an individual normally enjoys, accompanied by periods of at least 2 weeks marked by the inability to perform everyday activities [2]. Anxiety, in turn, is defined as an emotion expressed in response to stressful, dangerous, or unfamiliar situations, or some unidentified factor, that is, the feeling of unease, distress, or dread that one feels in the face of a significant event. A certain level of anxiety is necessary to keep us alert and aware, but for those who suffer from anxiety disorders, it can be totally debilitating [3]. Current pharmacological treatments for depressive disorders are mainly based on selective serotonin reuptake inhibitors (SSRIs), serotonin (5-HT) and noradrenaline (NE) reuptake inhibitors (SNRIs), and monoamine oxidase inhibitors (MAOIs), all of which act by increasing short-term levels of neurotransmitters in the brain. One consequence of treatment is the desensitization of receptors, for example, 5-HT1A, with a downregulation of autoreceptors, but no changes in the postsynaptic receptors, which leads to the recovery of neuronal activity in the long term [4]. These changes are associated with the long latency to the onset of antidepressant effects. However, up to 70% of depressed patients have residual symptoms [5], and few options exist for transitioning treatment-resistant sufferers to alternative therapies that operate through distinct mechanisms [6]. It is important to note that conventional antidepressants produce significant side effects, such as nausea and vomiting, insomnia, agitation, fatigue, sedation, sexual dysfunction, headaches, and weight gain, which contribute to poor patient compliance and, in some cases, abandonment of treatment [7]. This occurs under such anxiolytic treatments as benzodiazepine (a GABAA receptor agonist) and SSRIs [8] and is the main cause of the increasing demand for alternative medicines, such as medicinal plants, to alleviate the symptoms of these psychiatric disorders. However, reports of adverse reactions to products of this kind have increased [9], leading WHO to publish the document, “The WHO’s traditional medicine strategy: 2014-2023”, which outlines a global approach to fomenting the appropriate integration, regulation, and supervision of natural substances. This paper will be useful in countries seeking to develop a proactive policy toward this important and expanding area of health care and will contribute to the use of herbal medicines of proven quality, safety, and efficacy, providing quality medical care to all people [10]. Recent decades have witnessed efforts to gather scientific evidence that validates the efficacy of plants commonly used for their antidepressant and anxiolytic properties [11], but research has been insufficient because of the wide range of plants available worldwide. We lack solid scientific data on the neurochemical pathways and mechanisms of action of medicinal plants or their active metabolites because few clinical studies have addressed these issues. Also, reports of adverse reactions to medicinal plants [9] may reflect the broad variety of active metabolites they contain, thus highlighting the need for preclinical and clinical studies that evaluate the possible biological activity of compounds isolated from plants or standardized crude extracts, their mechanisms of action, and possible toxicity.

Fajemiroye et al. [12] proposed a hypothetical model for identifying medicinal plant extracts and phytoconstituents with anxiolytic and/or antidepressant properties that is currently used by most researchers: (i) select medicinal plants with anxiolytic and/or antidepressant potential based on local reports; (II) prepare standard crude extracts; (III) perform phytochemical studies that include sequential partitioning of crude extracts, purification and isolation of phytoconstituents, chemical elucidation or characterization of the isolates, structural modifications or syntheses of new compounds based on chemical structure of their isolates; and (IV) conduct pharmacological analyses of the anti-anxiety and antidepressant properties of the standard crude extracts, fractions, isolated compounds, or derivatives using ex vivo, in vitro, and in vivo assays (e.g., preliminary pharmacological screening, classic animal models of anxiety like the light dark box (LDB) or elevated plus-maze (EPM) tests, etc., or the forced swim test (FST) and tail suspension tests (TST), among others). These tools have allowed researchers to analyze the possible metabolites responsible for the anxiolytic or antidepressant properties of plants used by different populations, and identify how their mechanisms of action affect the functioning of the central nervous system (CNS). Their studies contribute to advancing scientific understanding of the neurobiology of depression and anxiety, and to developing new pharmacological treatments that may favorably impact public health.

Plants used in traditional medicine contain compounds in their secondary metabolism [13] such as alkaloids, phenols, sterols, carbohydrates, tannins, terpenes, and phytoalexins, all of which have important biological activities [14]. The most widely studied metabolites are terpenes, flavonoids, alkaloids, and sterols, whose mechanisms of action stimulate the serotonergic, noradrenergic, dopaminergic, or GABAergic neurotransmission systems, acting on receptors or the synthetic pathways of neurotransmitters and their transporters. However, they may also stimulate other neurotransmission systems. For example, terpenes can stimulate at the same time serotonergic, dopaminergic, and noradrenergic neurotransmission systems [12] that can produce a similar effect on mood regulation, perhaps leading to an overstimulation that may generate undesirable collateral effects.

This chapter reviews and discusses the findings from research on several metabolites of medicinal plants that have shown potential anxiolytic and antidepressant activities once screened for their biological mechanisms at various levels: receptor, transporter, synthesis, gene, protein, or metabolic. The studies analyzed were identified by a preliminary search in PubMed, Scopus and Ovid for articles on (i) the dose effects and possible mechanisms of action of metabolite(s) isolated from parts of plants with previously identified anxiolytic or antidepressant effects; and (ii) standard chemical tests performed with specific metabolites.

Advertisement

2. Terpenes with antidepressant effects

Terpenes are formed by the union of isoprene units (5 C atoms). Their classification depends on the number of units they contain: 10 C terpenes (two units) are called monoterpenes, while 15 C terpenes (three units) are called sesquiterpenes, and those with 20 C are diterpenes. Triterpenes have 30 C, tetraterpenes have 40 C, and polyterpenes are those with over 8 isoprene units. Studies have evaluated the effect of terpenes isolated from plants, including rosmanol from Rosmarinus officinalis, ursolic acid, and oleanolic acid; carnosol from Artemisa indica; and linalool and β-pinene from Litsea glaucescens. All these terpenes have proven antidepressant effects. Abdelhalim et al. [15] isolated rosmanol, an ethyl acetate diterpene, from R. officinalis. A single acute dose of 30 or 100 mg/kg i.p. of rosmanol in male Swiss mice produced an antidepressant effect on the FST and TST. The 100-mg/kg dose produced an effect similar to that of a 60-mg/kg dose of imipramine on the FST. Their study also tested the acute toxicity of administering 50, 150, and 200 mg/kg, i.p., of rosmanol. Some signs of toxic effects on grooming behavior were observed, as well as hyperactivity, sedation, respiratory arrest, convulsions, and locomotor activity. However, no cases of lethality or variations in the amount of food and water ingested were reported.

Other terpenes with antidepressant properties include phenolic diterpene, carnosol, and pentacyclic triterpenoids like betulinic, oleanolic, and ursolic acids. Carnosol generated an antidepressant-like effect at doses of 0.01 and 0.1 mg/kg [16] on TST, and 10, 30, and 100 mg/kg, i.p., on TST and FST, as did the oleanolic and ursolic acids. Meanwhile, 100 mg/kg of ursolic acid showed an effect similar to that of imipramine at 60 mg/kg [17]. Betulinic acid at 10 mg/kg was evaluated in male mice on TST [16]. None of those metabolites had effects on locomotor activity. Ursolic acid at doses of 0.01 and 0.1 mg/kg produced an effect similar to that of fluoxetine (10 mg/kg), imipramine (1 mg/kg), and bupropion (10 mg/kg) on TST [18]. Only the 10-mg/kg dose had an antidepressant effect on FST, which was similar to that of bupropion at 10 mg/kg. Studies exploring the mechanism of action of ursolic acid found the involvement of D1 and D2 receptors and a pharmacological synergism with bupropion at 1 mg/kg, p.o. (dual dopamine/noradrenaline reuptake inhibitor, DDNRI) [18], 5 mg/kg of fluoxetine, or 2 mg/kg of reboxetine (SRNI) [19]. They also observed a related increase in noradrenaline and dopamine synthesis on TST [19]. These findings are noteworthy because they suggest the possibility that various mechanisms of action may be stimulated by the same metabolite.

A study of the essential oil of Origanum majorana (OMEO) identified 14 component terpenes that represented 92.32% of yield. The five components that were more abundant in OMEO were terpinene-4-ol (32.69%), γ-terpinene (12.88%), trans-sabinene hydrate (8.47%), α-terpinene (7.98%), and sabinene (6.211%). Administered in a single acute dose of 40 or 80 mg/kg, OMEO increased swimming and climbing times in male mice on FST. These findings were interpreted as showing an antidepressant-like effect. In that study [20], pretreatment with antagonist drugs demonstrated that the terpenes act through several mechanisms: first, by activating the dopaminergic receptors D1 and D2, the noradrenergic α1- and α2-adrenoceptor, and the 5-HT1A and 5-HT2A receptors, and, second, by activating or increasing 5-HT synthesis and monoamine vesicular storage involved in reducing the immobility time produced by 80 mg/kg of OMEO [20]. A pharmacological synergism at the combined subthreshold OMEO doses of 10 mg/kg plus fluoxetine or imipramine (5 mg/kg, i.p.) was also seen. It reduced immobility time but increased swimming and climbing times, similar effects to those produced by OMEO at 80 mg/kg [20]. (A more detail view about the OMEO components can be reviewed at Tables 1 and 2 as summary). (R)-(−)-linalool produced antidepressant effects in male mice on TST at a single dose of 100 or 200 mg/kg, i.p., and on FST when administered 3 times at 100 mg/kg, i.p. [21]. That effect was produced by activation of the 5-HT1A receptor and α2-adrenoceptors [22], while (1S)-(−)-β-pinene produced an antidepressant effect after three applications at a dose of 100 mg/kg, i.p., by activating β-adrenoceptors, 5-HT1A and D1-receptors, and noradrenergic neurotransmission in the cerebral cortex [22].

Table 1.

Secondary metabolites with antidepressant properties in preclinical study.

NE = no explorated.

Table 2.

Secondary metabolites with anxiolytic properties in preclinical study.

NE = no explorated.

However, due to the terpene’s capacity to stimulate several neurotransmission systems—especially high doses of monoterpenes—possible collateral or undesirable effects as serotonergic syndrome need to be explored. The monoterpenic oxide, 1,4-cineole, for example, produced a prodespair effect at doses of 200 mg/kg, i.p., on FST and 400 mg/kg, i.p., and FST and TST, but did not induce any significant deficit in motor coordination on the rota-rod test (RRT). It did, however, have an anxiolytic effect at a dose of 400 mg/kg in male mice evaluated in EPM that was not associated with any sedative effect [23]. These findings require additional study in light of potential depressor effects on the CNS, and to elucidate the mechanisms of action involved.

Advertisement

3. Terpenes with anxiolytic effects: preclinical research

Anxiolytic properties have also been attributed to terpenes. A combination of two monoterpenoids, p-cymene + thymol, both at doses of 3 mg/kg i.p., produced anxiolytic effects on the hole-board test (HBT) and EPM [24]. Studies have also demonstrated that (−)-myrtenol, a monoterpenoid alcohol, produced an anxiolytic effect on EPM at doses of 25, 50, and 75 mg/kg, i.p., though only the 25- and 50-mg/kg doses did so on LDB. On both tests, the anxiolytic effect of 25 mg/kg of (−)-myrtenol was mediated by GABAA receptors [25]. In another study, rosmanol produced an anxiolytic effect at doses of 10, 30, and 100 mg/kg in EPM and LDB, and the 10- and 30-mg/kg doses showed an effect similar to those of diazepam at 1 mg/kg [15]. This mechanism of action acts on GABAA receptors at a distinct site from the high-affinity benzodiazepine-binding region [15]. Triterpenes as ursolic acid, oleanolic acid, and carnosol produced anxiolytic effects at doses of 10, 30, and 100 mg/kg in EPM and LDB. The effect of the 30- and 100-mg/kg doses of these three metabolites was similar to that of diazepam at 1 mg/kg. No significant effect was seen on locomotor activity. We know that this effect was produced through GABAA receptors of the α1β2γ2L conformation because 2.5 mg/kg of flumazenil blocked the anxiolytic effects of 10 mg/kg of all three in EPM [17].

Although most terpenes have a GABAergic mechanism, their action may also occur through the serotonergic system, as evidenced in the study by Costa et al. [26]. In an experiment with male rats, those authors identified that acute administration of 5 mg/kg, p.o., or 14-day repeat doses (1 mg/kg/day), of the essential oil of ripe fruits of C. aurantium (Rutaceae) (whose chemical composition includes 98.66% limonene, 0.53% β-myrcene, 0.41% β-pinene, and 0.41% unidentified compounds) produced an anxiolytic-like effect in LDB that was mediated by the serotonergic system through 5-HT1A receptors (WAY 100635 0.5 mg/kg, i.p.), not the GABAergic system (flumazenil 2 mg/kg, i.p.). That study also analyzed the antidepressant effect on FST after oral and inhaled treatment, but found that it did not modify immobility. Their results suggest that distinct mechanisms of action exist for the anxiolytic and antidepressant effects [26]. In this regard, some of the terpenes in certain essential oils produce anxiolytic effects and are often used in aromatherapy to reduce anxiety in animals and humans [27]. Inhaling emulsions of linalool oxide (a monocyclic alcohol) at 0.65, 1.25, 2.5, and 5.0% w/w during 7 min of exposure in the inhalation chamber, for example, produced anxiolytic-like effects in EPM and LDB, but no significant motor deficit on RRT [28].

Finally, songorine, a C20 diterpenoid alkaloid, produced an anxiolytic-like effect at 0.25 mg/kg v.o. for 5 days with greater efficacy than phenazepam on Vogel’s conflict test (VCT) [29]. For the terpenes described so far, we know that both the serotonergic and GABAergic systems are involved in their mechanisms of action, and these are the same systems that are activated by other groups of metabolites, such as flavonoids and sterols (see Tables 1 and 2 for summaries).

Advertisement

4. Flavonoids with antidepressant effects: preclinical research

Flavonoids are the most widely studied active metabolites (for a broad review of research, see German-Ponciano et al. [30]). Genistein is an isoflavone that can cross the blood-brain barrier in mice [31] and rats [32]. Acute oral administration of 5, 15, and 45 mg/kg genistein in male mice did not reduce immobility time on FST or TST, but chronic, dose-dependent administration for 21 days produced antidepressant-like effects on both tests, without affecting locomotor activity [33]. This effect was associated with increased NA and 5-HT concentrations in the hippocampus and frontal cortex, and of 5-HT in the hypothalamus, though it decreased the 5-HTIAA/5-HT ratio in the hippocampus and frontal cortex. These results suggest an inhibition effect of genistein on MAO-A in the hippocampus, frontal cortex, and hypothalamus and on MAO-B in the hippocampus, three brain structures involved in the neurobiology of depression and anxiety. Their results [33] also demonstrate that central depletion of 5-HT reversed the antidepressant effect of genistein, suggesting a critical role of the serotonergic system, specifically through 5-HT1A receptors. It is important to note that these results on the serotonergic metabolic ratio (5-HIAA/5-HT) may be dependent on gonadal hormones. Ovariectomized rats (OVX, surgical removal of both ovaries) showed reduced immobility times on FST after administration of genistein (10 mg/kg, p.o. [34], or by i.m.) [35] for 14 days, but the downward tendency of the serotonergic metabolic ratio caused by FST was only evident in the hippocampus [34]. Sapronov and Kasakova [35] found antidepressant-like effects on FST at the same dose of genistein (10 mg/kg) but in non-ovariectomized rats. That effect was more marked in the metestrus and diestrus phases of the estral cycle, which are characterized by low plasmatic concentrations of ovarian hormones, than during the proestrus and estrus stages with their characteristically high hormone concentrations. This suggests that these hormones play a significant role in the antidepressant effect of genistein.

A similar effect on the serotonergic system and MAO-A activity was found with quercetin 4’-O-glucoside or quercetin administered at doses of 10 and 20 mg/kg, p.o., for 7 days in Swiss albino mice of both sexes. These substances produced antidepressant-like effects in mice on FST as well as those subjected to unpredictable, chronic mild stress (CUMS, a mouse model designed to induce depression) and subsequently evaluated on FST. In that study, a 20-mg/kg dose of quercetin 4’-O-glucoside showed a similar effect to that of fluoxetine at 20 mg/kg, p.o., on FST, with or without prior exposure to CUMS [36]. A consequence of CUMS on the sucrose preference test (SPT, a test used to study anhedonia rodents, the main symptom of depression in humans) is a decrease in the consumption of a sweetened solution. In these sense, both doses of quercetin 4’-O-glucoside reverted this effect, and interpreted as being antidepressant. Other consequences of CUMS are metabolic, for example, excessive production of reactive oxygen species that was evidenced by higher brain thiobarbituric acid reactive species (TBARS). Compromising endogenous anti-oxidants, like reduced glutathione (GSH), enhances MAO-A activity in the brain and, consequently, depletes monoamine levels there, especially serotonin 5-HT. The effects observed in that study were blocked by 21 days of treatment with 10 and 20 mg/kg of quercetin 4’-O-glucoside [36], suggesting a possible mechanism of action with an antioxidant effect that impedes ROS production. Another study [37] found that 10 mg/kg of quercetin administered for 14 days reduced immobility time on TST, but not FST, while doses of 25 and 50 mg/kg produced this effect in female mice on both tests. The mechanisms of action were explored on TST, where i.c.v. administration of N-methyl-D-aspartate (NMDA at 0.1 pmol/site) and L-arginine (at 750 mg/kg, i.p., a nitric oxide inhibitor) blocked the antidepressant effect of quercetin. Hence, the antidepressant-like effect of quercetin may involve inhibiting NMDA receptors to decrease intracellular calcium that, in turn, inhibits the protein calmodulin, which then inhibits neuronal nitric oxide synthase to decrease nitric oxide levels (NO). This hypothesis is supported by the finding that administering methylene blue (a NO synthase inhibitor) at 20 mg/kg, i.p., and soluble guanylate cyclase or 7-nitroindazole (another NO synthase inhibitor) at 50 mg/kg, i.p., improved quercetin’s antidepressant-like effect on TST. This indicates that the antidepressant effect may be dependent on limiting NO synthesis, either by inhibiting the enzyme or by reducing NO production, perhaps via decreased cyclic guanosine monophosphate (cGMP), since sildenafil (a phosphodiesterase 5 selective inhibitor that increases cGMP levels) also canceled this effect [37].

A model of depression induced by olfactory bulbectomy (OB, surgical removal of the olfactory bulbs) reduced the latency to immobility and increased immobility time on FST and TST. This was accompanied by an increase in the levels of the markers of oxidative stress, for example, 116% in the case of lipid hydroperoxide content (LOOH) in the hippocampus. This effect was reverted by 52.25% by administering 25 mg/kg of genistein in the content of LOOH, as observed on the immobility on FST and TST. In sham rats only (i.e., animals subjected to the same surgical procedure but without resection of the olfactory bulbs), genistein reduced glutathione (GSH) levels, in that study by 65.94%. The authors [37] explained that “the reduction of GSH levels caused by OB and, surprisingly, quercetin, can be explained by the fact that glutathione peroxidase, in addition to reducing H2O2, decreases lipid and nonlipid hydroperoxides at the expense of GSH, causing it to become oxidized and giving rise to glutathione disulfide. Therefore, it is suggested that LOOH activated glutathione peroxidase which, in turn, oxidized GSH to normalize LOOH levels”. In this area, increased levels of the markers of oxidative stress in major depression have been associated with poor response to antidepressant treatment [38]. Therefore, a therapy that reduces the levels of markers of oxidative stress and produces antidepressant effects could be a promising form of treatment.

Additional mechanisms of the antidepressant action of flavonoids have been explored. Administering chrysin for 14 days at a dose of 20 mg/kg, for example, increased grooming time in male OB C57B/6J mice evaluated on the splash test (ST), where 200 ml of a 10% sucrose solution is squirted on the mouse’s snout to initiate grooming behavior. Here, greater grooming time is considered an antidepressant effect. Doses of 5 and 20 mg/kg impeded an increase in immobility time by these OB mice on FST, but increased 5-HT and brain-derived neurotrophic factor concentrations in the hippocampus [39]. In another study, fisetin administered at 5 mg/kg v.o. increased activation of the tropomyosin kinase B receptor (TrkB) by signaling and increasing its phosphorylation in the hippocampus. This suggests that fisetin produced pro-neurogenesis [40] related to its antidepressant effect on FST and TST after 1 or 2 weeks of treatment with a relatively short therapeutic latency compared to clinically-effective antidepressants. Fisetin also reversed depression-like behaviors induced by spatial restraint stress in mice evaluated on FST and TST [40]. Other studies have found that the chemical standard dihydromyricetin activated the ERK1/2-CREB pathway and increased glycogen synthase kinase-3 beta (GSK-3β) phosphorylation at ser-9 with upregulation of BDNF expression in the hippocampus, while inhibiting neuroinflammation. These findings may be related to the antidepressant effect seen on TST and FST after once-daily administration of 10 and 20 mg/kg, v.o. for 3 days, but not after a single acute dose [41]. Interestingly, dihydromyricetin reverted the depressogenic effect caused by CUMS in mice subjected to SPT and FST, or TST, only after administration of once daily for 7 days, but not 3 days [41]. Another flavonoid analyzed is hesperidin, which increased BDNF levels in the hippocampus after administration of once daily for 21 days (0.3 and 1 mg/kg, i.p.). These doses produced an antidepressant effect on TST similar to fluoxetine (32 mg/kg i.p.) and imipramine (15 mg/kg, i.p.). Another research has also verified that when applied acutely (1 mg/kg after 30 min) or chronically (0.1, 0.3, and 1 mg/kg for 21 days) hesperidin significantly decreased nitrate/nitrite (NOX) levels in the hippocampus of mice, suggesting a possible inhibition of the L-arginine-NO-cGMP pathway [42].

Another flavonoid that has shown effects on the CNS is baicalin, which may promote neuronal differentiation through neuronal maduration and ensure their survival via the associated Akt/FOXG1 pathway, which stimulates dendrite elongation. This is related to findings that indicated that, after 6 weeks of treatment, a 60-mg/kg dose of baicalin had an effect similar to that of fluoxetine (15 mg/kg, v.o.), because it reverted the decrease of sucrose intake on SPT and the increase in immobility on TST produced by CUMS [43]. Another flavonoid that associates antidepressant and antioxidant effects is naringin, which reduced immobility on FST at doses of 2.5, 5, and 10 mg/kg given for 7 days. The antidepressant effect of these doses correlated with enhanced cholinergic transmission due to a decrease in the activity of the enzyme acetylcholinesterase and of the antioxidant defense systems caused by higher GSH levels, as well as an increase in the activity of superoxide dismutase (SOD) and catalase (CAT) in mice brains.

Studies have demonstrated that naringin inhibits lipid peroxidation and nitrosative processes by reducing levels of ROS and nitrogen species [44]. Finally, the extract of Cirsium japonicum at doses of 200 and 400 mg/kg has shown the ability to reduce immobility time on FST in a similar manner to that of 5 mg/kg of the antidepressant imipramine. A major component of this plant is the flavonoid luteolin, which at doses of 5 and 10 mg/kg produced a similar effect to that of the complete extract, likely through a positive modulating effect on the GABAA receptor complex. This was proven in an in vitro study where extracts of both Cirsium japonicum and luteolin increased Cl influx in an effect impeded by pretreatment with bicuculline, a competitive GABAA receptor antagonist [45].

The varied mechanisms seen in flavonoids make them an important object of study, especially in the search for side effect-free treatments that can compromise their effectiveness or produce toxicity by interacting with other medications or food. This is another area of research that remains to be explored.

Advertisement

5. Flavonoids with anxiolytic effects

A particularly important fact concerning the potency of the biological activity of plants is that it depends on several factors, for instance, the part of the plant used, the region where it is gathered, the season, and harvesting time, among others [46]. For example, in male mice evaluated by HBT and EPM, a single dose of 100 mg/kg i.p. of the methanolic extract of inflorescences of Tilia americana var. mexicana collected in Morelia, Mexico, produced a more effective anxiolytic effect than those gathered in Honey, Puebla, though the leaves collected in Honey were more effective than those from Morelia or Santa María Ahuacatitlan, Mexico. These three Mexican states are located at different elevations with distinct humidity and soil types. That study quantified quercetin, rutin, and isoquercitrin in the inflorescences and leaves, determining that the concentrations of these substances differed with the part of the plant used and the collection area [46]. It also tested several standard commercial flavonoids: kaempferol (10 mg/kg), quercetin (20 mg/kg), astragalin (10 mg/kg), isoquercitrin (2 mg/kg), quercetin (10 mg/kg), and rutin (15.7 mg/kg), and a mixture of flavonoids (MIX) composed of quercetin 20 mg/kg, rutin 15.7 mg/kg, and isoquercitrin 2 mg/kg and quercetin (20 mg/kg). Results showed that a mixture of quercitin (20 mg/kg), rutin (15.70 mg/kg), and isoquercitrin (2 mg/kg) produced an anxiolytic effect in male mice tested in HBT and EPM [46] by reducing the number of head-dippings but increasing the time spent in the open arms, respectively. Finally, upon testing the anxiolytic effect of the methanolic extract of Tilia americana var. mexicana, those authors found that this produced an effect in EPM through the participation of GABA/BDZ (flumazenil 5 mg/kg) and 5HT1A serotonergic receptors (WAY 100635 0.5 mg/kg), though they were not involved in the anxiolytic effect on HBT [46].

Another flavonoid known to have anxiolytic effects is formononetin, an active metabolite of traditional Chinese medicine red clover (Trifolium pratense L.). Wang et al. [47] observed that administering 25 mg/kg of this metabolite to male mice once daily for 8 days blocked the anxiogenic effect on the open field test (OFT) produced by administering Freund’s complete adjuvant (CFA), reduced the time spent and distance traveled in the central area, and decreased the time spent in the open arms of EPM [47]. Formononetin did not modify behavior compared to the control group on either test, indicating that it demonstrated an anxiolytic effect. However, it seems that these results should be understood as a neuroprotective effect more than an anxiolytic one, at least under those study conditions. The notion of a neuroprotector effect is supported by the fact that the study found that formononetin attenuated inflammation and neuronal hyperexcitability by inhibiting NMDA receptors and the CREB signaling pathway in the basolateral amygdala (BLA) [47].

Other anxiolytic mechanisms of action seen in flavonoids are dopaminergic in nature. Theaflavins, for example, increased dopamine (DA) turnover to induce activation of the dopaminergic system in the frontal cortex in male mice in EPM and LDB [48], while chrysin at 2 and 4 mg/kg produced an anxiolytic effect in rats at 12 weeks postovariectomy on LDB by increasing the time spent in the light compartment. Those findings resembled the effect of diazepam. At doses of 1, 2, and 4 mg/kg, this flavonoid increased the frequency of entries into, and the time spent in, the open arms of EPM partially through action on GABAA receptors (pre-treatment with 1 mg/kg picrotoxin) [49]. On the other hand, neurosteroids and the serotonergic system have also been implicated in the anxiolytic effect of flavonoids, as in the case of puerarin, which increased 5-HT and allopregnanolone levels in the prefrontal cortex and hippocampus in male rats. These results have been associated with the finding that puerarin increased the time spent in the open arms and the percentage of entries into the open arms of EPM, whereas on the VCT test, it produced an increase in the number of shocks received. In both cases, the effect was similar to that of sertraline, which was used as a positive control drug to generate an anxiolytic effect on both tests [50].

In an animal model of surgically-induced menopause, genistein at 0.09 and 0.12 mg/kg, s.c., for seven consecutive days, or the same treatment regimen but with 17β-estradiol, increased the time spent in, and the percentage and frequency of entries into, the open arms of EPM. These effects were caused by activation of the β-estrogenic receptor (ERβ) since pretreatment with tamoxifen (5 mg/kg, an ERβ antagonist) blocked the anxiolytic effect. Also, genistein and 17β-estradiol increased the frequency of rearing and grooming behaviors on the locomotor activity test (LAT), associated with an anxiety-reducing effect manifested in EPM [51]. Genistein tested at doses of 0.25, 0.5, and 1 mg/kg increased the time spent in, and the frequency of exploration of, the light compartment of LDB, while doses of 0.5 and 1 mg/kg increased rearing frequency, and 1 mg/kg increased grooming time. Those studies used rats at 12 weeks postovariectomy [52]. The authors suggest that “genistein is considered a phytoestrogen that acts in a dose-dependent manner with a broader margin of safety at anxiolytic doses. However, more studies are required to take advantage of its potential therapeutic anxiolytic effects” [51].

In a distinct approach, a post-traumatic stress disorder (PTSD) model used a chamber with a grid floor connected to a system that delivered foot shocks, exposing rats to 5 shocks per day. There, an increase in the contextual freezing time on days 7, 14, and 21 indicated the induction of anxiety-like behaviors. The time spent in freezing behavior was calculated with the shock-administering system turned off. In that study, genistein at 4 and 8 mg/kg i.p. administered to male rats from day 7 reduced freezing time at 7, 14, and 21 days. Interestingly, only the 8-mg/kg dose returned freezing times to control levels on day 21 [53]. The stressed rats were also tested in EPM, where they spent less time in the open arms, indicating an anxiety-like effect that was reverted by administering 4 and 8 mg/kg of genistein. This reduced anxiety-like behavior in the stressed rats occurred in association with enhanced tryptophan hydroxylase (TPH) and 5-HT levels, but also promoted the 5-HT receptor-related CaMKII/CREB signaling pathway in the amygdala [53], likely reflecting the fact that the amygdala receives serotonergic projections from the raphe, two brain structures to which emotional valence and 5-HT synthesis, respectively, are attributed [54].

A study evaluated the pharmacokinetic profile of 6-methoxyflavanone and calculated the KP value (i.e., the tissue-to-serum partition coefficient). Molecules with KP values >0.30 are thought to be readily distributed in the brain [55]. That study determined that 30 min postadministration of a 30-mg/kg i.p. dose, the 6-methoxyflavanone had crossed the blood-brain barrier (BBB) to reach the amygdala with Kp = 0.47, and the cerebral cortex with Kp = 0.437. These two cerebral structures are known to be involved in the neurobiology of anxiety [56], so these properties were associated with the anxiolytic effect of 6-methoxyflavanone in male and female mice in EPM at doses of 10, 30, and 50 mg/kg, and on the staircase test (ScT). In this model, the lower frequency of rearings, but no reduction in the steps climbed in a 3-min period, was interpreted as indications of an anxiolytic effect [45]. Those authors verified that 6-methoxyflavanone produced its effect by activating GABAA receptors with the α2-subunit, perhaps in the amygdala and brain cortex, since pretreatment with PTZ blocked this anxiolytic effect in EPM and on ScT [56].

Another flavonoid with anxiolytic effects is rutin at doses of 300 and 562 mg/kg, i.p., or 16 nmol/site, in the basolateral amygdala of male rats tested in EPM. This involves partial GABAergic neurotransmission that was not associated with BDZ binding in the GABAA receptors [57]. Finally, viscosine administered to male mice assessed in EPM and LDB was seen to exert its action through the α1β2γ2L and α2β2γ2L modulates of the GABAA receptors at a site distinct from the one classically associated with benzodiazepine [58].

Advertisement

6. Alkaloids with antidepressant activity

Alkaloids purified from crude acid-base extracts have diverse chemical structures. They may contain one or more nitrogen atom(s) (in the heterocyclic ring) in the form of salt [59]. Pseudoalkaloids that possess nitrogen exist. They are not synthesized from amino acids, but by nitrogen transfer in the form of ammonia to a compound of terpenic origin, steroids, polyketides, monosaccharides, or fatty acids [59].

The alkaloid berberine (50 mg/kg, i.p.) decreased immobility but increased climbing behavior on FST; results are considered to reflect an antidepressant-like effect in rats after abstinence from repeated morphine administration [59]. Chronic treatment with the extract of Annona cherimola produced antidepressant-like effects in tests of mice on FST. A. cherimola contains mainly the alkaloids 1,2-dimethoxy-5,6,6a,7-tetrahydro-4H-dibenzoquinoline-3,8,9,10-tetraol, anonaine, liriodenine, and Nornuciferine, which are likely responsible for the increase in 5-HT and DA [60].

In another work, the alkaloid derivatives of the β-carbolines (harmane 5, 10, and 15 mg/kg, norharmane 2.5, 10, and 15 mg/kg, and harmine 5, 10, and 15 mg/kg, all i.p.) showed antidepressant effects in mice that were dose-dependent, suggesting that the effect occurs through an inverse agonist mechanism of the benzodiazepine receptors due to flumazenil antagonism (5 mg/kg, i.p.) [61]. In addition, anhedonia was reversed in rats subjected to the CUMS model after harmine treatment at 15 mg/kg/day for 7 days. They showed increased adrenal gland weight, ACTH levels, and BNDF protein levels produced by the CUMS [62]. Treatment for 14 days with harmine (5, 10, and 15 mg/kg) and imipramine (10, 20, and 30 mg/kg) in rats subjected to FST produced antidepressant-like effects, while harmine (10 and 15 mg/kg), but not imipramine, increased BDNF protein levels in the hippocampus of rats [62]. These results indicate that the main mechanism involved in harmine’s antidepressant effect is an increase in hippocampal BDNF, though this may be dependent on treatment time and the precise region of the hippocampus.

An infusion of harman (1-methyl-beta-carboline) into the hippocampus of rats, or through systemic administration, increased the concentration of 5-HT [63]. In addition, metabolite levels of 5-HT degradation decreased dose-dependently, probably due to inhibition of MAO-A [63]. Another study showed that harman bonds to type 5-HT2A serotonergic receptors but shows no affinity to dopaminergic or BZ receptors [64]. Injections of 2.5 and 10 mg/kg of harmane in rats under fear conditioning have been shown to increase plasma ACTH, corticosterone, 5-HT, and NA levels in limbic system structures. These results suggest that harman can modulate behavioral alterations, brain neurochemistry, and neuroendocrine functions through a mechanism that inhibits MAO-A [65].

Subchronic, oral administration for 21 days of the lyophilized extract of Rhazya stricta and alkaloid fractions (akuammidine, rhaziminine, and tetrahydrosecamine) to male rats inhibited the activity of the MAO-B enzyme, a mechanism through which the antidepressant-like effect may occur [66]. Another alkaloid in the Mitragyna speciosa plant has shown antidepressant effects. Administered to mice at doses of 10 and 30 mg/kg, i.p., mitragynine decreased immobility time on then TST and FST [67] and the release of corticosterone. Mitragynine’s effect appears to be mediated through the neuroendocrine HPA (hypothalamus-adrenal-pituitary) axis [67].

Punarnavine administered at doses of 20 and 40 mg/kg, v.o., for 14 days decreased immobility on FST, MAO-A activity, and corticosterone levels in both stressed and unstressed mice [68], while treatment with evodiamine at 10 and 20 mg/kg in rats exposed to CUMS reversed the decrease in their preference for sugared water and immobility time on FST, but increased 5-HT and NA levels and the protein expression of BNDF in the hippocampus. However, it reduced corticosterone levels, suggesting that it likely modulates monoamines and BDNF-TrkB signaling in the hippocampus [69]. Chronic administration of piperine in rats at 5, 10, and 20 mg/kg has shown antidepressant-like effects on FST, probably due to a serotonergic mechanism [70]. At a dose of 30 mg/kg, protopine produced an antidepressant effect on TST in mice, perhaps by inhibiting the 5-HT and NA transporters, since in vitro studies have reported that it produces an inhibitory effect on these elements [71].

Because alkaloids have powerful antidepressant effects, many are used in clinical practice with effective therapeutic results. Preclinical studies have clearly demonstrated the antidepressant effects of alkaloids, but evidence of their mechanisms of action is still deficient or unclear. Alkaloids isolated from plants are an option for treating depression, but more studies are needed at the preclinical level to evaluate their potency, efficacy, and safety before they can be incorporated into clinical practice.

Advertisement

7. Alkaloids with anxiolytic effect

The alkaloids gelsemine, koumine, and gelsevirine exerted anxiolytic effects in single doses of 2 and 10 mg/kg in mice in EPM and LDB [72]. Gelsemine in low doses (10−6 M and 10−14 M) administered to male rats for 7 days also showed anxiolytic effects in EPM [73]. Koumine has shown this effect on VCT in mice at doses of 0.167, 0.5, or 1.5 mg/kg [74]. Other reports indicate that the decoction of the African peach root (Nauclea latifolia) injected intraperitoneally in mice produces dose-dependent anxiolytic-like effects (16, 40, 80, and 160 mg/kg) in EPM. Its effect has been attributed to isoquinoline-type alkaloids [75], but no reports have yet substantiated this claim. One study reported that the isoquinoline alkaloid berberine hydrochloride has both antipsychotic and anxiolytic properties. In this regard, studies have shown that a dose of 100 mg/kg/day produces anxiolytic effects and can modulate the gratifying effects induced by methamphetamine in rats [76].

The aqueous extract of Eschscholzia californica Cham (200 mg/kg, p.o.) has shown anxiolytic-like effects on the LDB test in mice that have been attributed to action on GABAA receptors [77]. Administration of the aqueous extract of the stem of Uncaria rhynchophylla (200 mg/kg), which contains the alkaloid rhynchophylline, in a single dose, or for 7 days, produced an anxiolytic effect in EPM by acting on the 5-HT1A receptor [78]. Another example has been implicated to alkaloids with anxiolytic effects were the hydroethanolic extract of Davilla rugosa produced anxiolytic-like effects in EPM when administered to rats at 15 mg/kg [79]. Two other plants that contain alkaloids with anxiolytic effects (erythravine and 11a-hydroxy-eritravine) are Erythrina velutina and Erythrina mulungu. A study in mice showed that chronic administration (23–26 days) of the hydroalcoholic extract of the stem of E. velutina at 100 mg/kg produced an anxiolytic effect in EPM [80], while acute treatment with 200 mg/kg of E. mulungu showed an anxiolytic response in LDB comparable to that of diazepam [81]. Nevertheless, in this study did not identify the content or type of alkaloids in these extracts.

Turning to the species Magnolia (Magnolia spp.), we find that at least four anxiolytic components have been identified: honokiol, 4-O-methylhonokiol, magnolol, and obovatol. Administering honokiol (1 mg/kg) for 7 days had an anxiolytic-like effect on mice tested in EPM with results similar to those of diazepam [82]. That treatment increased the activity of the enzyme glutamic acid decarboxylase (GAD-subtype 65) in the hippocampus, but not the cortex of the mice brains. This, in turn, increased the release of GABA and reduced anxiety behavior. GAD65 is located on the terminal nerve and regulates the release of GABA to the synaptic cleft [83]. On this topic, there are reports that GAD65-deficient mice show higher anxiety levels [83]. Administering 4-O-methylhonokiol (0.1, 0.2, and 0.5 mg/kg) to mice in a single dose or during 7 days produced anxiolytic effects in EPM through the benzodiazepine site by binding to the GABAA receptor [84]. This is similar to observations of obovatol at doses of 0.2, 0.5, and 1 mg/kg [85]. In addition, an increase in the expression of the GABAA receptor α1 subunit [84] and of the α1 subunit in the amygdala and Cl (−) currents was observed [85]. The diterpene alkaloid songorine has shown anxiolytic effects when male mice were tested on VCT at a dose of 0.25 mg/kg [29], revealing an effect similar to that of phenazepam.

Numerous reports attribute anxiolytic activity to a broad range of plants. However, isolating and identifying the alkaloids responsible for this activity have not advanced substantially. Preclinical reports point to a common mechanism of action that modulates the GABAergic and serotonergic systems. The data described here justifies the need to conduct preclinical and clinical studies using alkaloids as alternative treatments for some anxiety disorders.

Advertisement

8. Sterols with anxiolytic and antidepressant effects

Plants synthesize a class of sterols called phytosterols, whose chemical structure is similar to that of cholesterol. Phytosterols are found in nuts, vegetable oils, cereals, fruits, vegetables, and various plants [86]. Some 40 different types have been identified, including β-sitosterol, campesterol, fucosterol, and stigmasterol. Due to their lipidic nature and glycosylated forms, they are able to cross the blood-brain barrier and impact the CNS [87]. Trevisan et al. [88] suggest that α-spinasterol has the ability to cross the blood-brain barrier and exert an antagonistic effect on the transient potential receptor V1 (TRPV1). When these receptors are expressed in various areas of the brain—prefrontal cortex, amygdala, hypothalamus, and hippocampus—their activation augments the release of glutamate and, consequently, that of GABA, DA, or other catecholamines [89]. This fact involved TRPV1 receptors in the mechanism underlying the etiology of depression and anxiety. This was corroborated by Socała and Wlaź [90] by administering (1 and 2 mg/kg, i.p.) α-spinasterol to male mice and testing them on FST. Their results suggest an antidepressant effect. Also, intracerebroventricular (i.c.v.) coadministration of 50 μg of the TRPV1 receptor antagonist capsazepine/mouse with an ineffective dose of 0.5 mg/kg, i.p., of α-spinasterol, also reduced immobility time on FST, indicating the involvement of TRPV1 in the neurobiology of depression. However, α-spinasterol itself (0.5, 1, and 2 mg/kg) was unable to produce anxiolytic-like effects in EPM or LDB. In this sense, TRPV1-knockout mice manifested less anxiety behavior on the same tests [91]. Socała and Wlaź [90] proposed that α-spinasterol may be able to activate CB1 receptors with greater affinity because those neurons coexpress these receptors in various brain structures whose activation could activate TRPV1 receptors simultaneously to block their possible anxiolytic effects. In another work, administering fucosterol (10, 20, 30, or 40 mg/kg, v.o.) to male mice produced antidepressant effects on FST and TST, with the 20 and 30 mg/kg doses achieving an effect of comparable efficacy to 20 mg/kg of fluoxetine, a standard dose in humans [92]. Those doses also exerted an acute effect that increased BDNF levels in the hippocampus, a limbic structure involved in mood regulation. Fucosterol also blocked the decrease in 5-HT, 5-HTIIA, and NA levels in mice brains generated by the stress of FST. The effect of fucosterol on that test was similar to that of the positive control drug, but it was unable to prevent the reduction of DA, another factor caused by FST. These findings suggest that the antidepressant mechanism is mediated by increasing monoamines and reducing the rate of 5-HT metabolism. Fucosterol did not modify either motor or exploratory activity and showed no neurotoxic effects [92]. Similar results were found when administering β-sitosterol at 10, 20, and 30 mg/kg for 7 days. In that case, 30 mg/kg exerted effects similar to those of 20 mg/kg of fluoxetine on FST and TST. Finally, the effects on monoamine levels in mice brains confirm that sterols modify the serotonergic and noradrenergic systems but do not impact the dopaminergic system [93].

Another case involved α- and β-amyrin (αβAMY) isolated from the resin of the stem of Protium heptaphyllum plants obtained and identified from hexane-ethyl acetate fractions analyzed by TLC. That process produced 450 mg of αβAMY made up of 67% α- and 33% β-amyrin, which were further purified and tested on FST. Administering 2.5 and 5 mg/kg of αβAMY via i.p. or p.o. decreased immobility time, but the most effective treatment was the 2.5-mg/kg dose via the p.o. route. However, the effects produced by this route were similar to those of imipramine at 30 and 10 mg/kg. Imipramine is a tricyclic antidepressant (TCA) that blocks reuptake of both serotonin and norepinephrine. In addition, a pharmacological synergism between 1 or 2.5 mg/kg of αβAMY and 10 mg/kg of imipramine was observed, but not between 2.5 mg/kg of αβAMY and 4 mg/kg of paroxetine (SSRIs). These effects were blocked by pretreatment with 2 mg/kg of reserpine, an inhibitor of the vesicular catecholamine transporter that facilitates vesicular storage. This result suggests a possible mechanism of action through activation of the noradrenergic system [94]. The base structure that cholesterol and sterols share allows the latter to exert actions at the level of the CNS, as in the case of cholesterol. Cholesterol is a vital substance for neurons because it is required for vesicle transport and neurotransmitter release and as a precursor to neurosteroids. It is also implicated in synaptic plasticity in relation to the formation of new synapses. For these reasons, studying sterols and their mechanisms of action on the CNS is extremely important because of the anxiolytic and/or antidepressant effects they exert.

Advertisement

9. Final comments and conclusion

This broad review constitutes a significant contribution to our understanding of the mechanisms of action that allow plants to produce antidepressant and anxiolytic effects (see Figure 1 for a summary). However, most of the studies reviewed were conducted with mice, due to the low yields achieved when isolating the metabolites of plant extracts [95], which limit the amount of testing that can be done. Another concern is that some infusions or extracts used as household remedies lose their antidepressant or anxiolytic effects when fractioned [96]. These findings indicate that in some cases it may be necessary to keep the metabolites together at the concentrations present in the original infusion or extract that has a proven therapeutic effect. As explained herein, some metabolites share pharmacological targets, which explains why they lose their effect when separated and emphasizes the importance of using standardized extracts with demonstrated therapeutic effects in animal and human studies [97, 98]. Unfortunately, very few clinical studies have evaluated the potential antidepressant or anxiolytic effects of isolated metabolites, so a great deal of work remains to be done.

Figure 1.

Principal mechanisms of action of flavonoids, terpenes, sterols, and alkaloids with antidepressant and anxiolytic properties. DAG: diacylglycerol; IP3: inositol triphosphate; AMPC: adenosine monophosphate 3; PKA: protein kinase A; PLC: phospholipase C; AC: adenylyl cyclase; ATP: adenosine triphosphate; GDP: guanosin trifosfato; VDC: canal dependiente de voltaje; BNDF: factor neurotrófico derivado del cerebro; TrkB: tropomyosin receptor kinase B; Ca2+: calcium ion; Cl: chloride ion; R-5HT1A: 5HT1A receptor; α2AR: alpha 2-adrenergic receptor; D1-R: dopamine receptor D1; D2-R: dopamine receptor D2; DAT: dopamine transporter; NAT: noradrenaline transporter; SERT: serotonin transporter; MAO-A: monoamine oxidase A; MAO-B: monoamine oxidase B; AD: antidepressant; AX: anxiolytic.

Several observations suggest that active metabolites share the mechanism of action of antidepressant and anxiolytic drugs like SSRIs, SNRIs, MAOIs, DDNRI, and BZDs, but we should emphasize that some metabolites—at least in preclinical studies—produced better effects than conventional drugs, even at lower doses, while others presented a pharmacological synergism between both types at suboptimal doses that improved the effects exerted separately at higher doses. A second shared characteristic is that they contain active stereoisomers and probably, some metabolites, once metabolized, could become more active. This encourages us to consider a significant number of substances with anxiolytic and/or antidepressant pharmacological profiles and invites us to take on the challenge of evaluating their pharmacokinetics, pharmacodynamics, and safety.

In conclusion, terpenes, flavonoids, alkaloids, and sterols share mechanisms of action that include activation of the critical enzyme for catecholamine synthesis (e.g., tyrosine hydroxylase) or the inhibition of their limiting enzymes, MAO-A and MAO-B, and transporters, thus stimulating the vesicular storage monoamine and the release of neurotransmitters toward the synaptic cleft. Finally, they can prevent the production of ROS and inhibit NO synthesis and, further downstream, interact with the 5-HT1A, 5-HT2A, D1, D2, GABAA receptors, and α1, α2, β-adrenoceptors that contribute to stimulating PKA. One consequence is that CREB increases BDNF levels, which foster the appearance of dendritic contacts that improve cerebral neurotransmission and modulate the emotions.

Advertisement

10. Perspective

This chapter discusses the efficacy of some plant metabolites in treating anxiety and depression disorders, as demonstrated in preclinical studies. In the future, this option for treating such disorders will allow us to reduce treatment costs and moderate the side effects produced by drugs currently in use. However, our review also points out that few clinical studies have focused on the pharmacokinetic and pharmacodynamic processes involving metabolites that would permit the safe use of these extracts. Despite this, research has shown that traditional medicine, especially forms that use medicinal plants that have been passed down through several generations, constitutes an important alternative for health care.

Acknowledgments

The third author received (Ramos-Molina Ana Raquel) fellowships from Consejo Nacional de Ciencia y Tecnología (CONACyT) for postgraduate studies in Science and Technology (Reg. 631048).

References

  1. 1. Global Burden of Disease (GBD). Available from: https://vizhub.healthdata.org/gbd-compare/. [Accessed: 05 August 2019]
  2. 2. Depression and Other Common Mental Disorders: Global Health Estimates. Geneva: World Health Organization; 2017. Licence: CC BY-NC-SA 3.0 IGO
  3. 3. Anxiety.org. [Internet] 2019. Available from: https://www.anxiety.org/what-is-anxiety [Accessed: 05 August 2019]
  4. 4. Rakofsky JJ, Holtzheimer PE, Nemeroff CB. Emerging targets for antidepressant therapies. Current Opinion in Chemical Biology. 2009;13(3):291-302
  5. 5. Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: Implications for clinical practice. The American Journal of Psychiatry. 2006;163(1):28-40
  6. 6. Rush AJ, Trivedi MH, Wisniewski SR, Stewart JW, Nierenberg AA, Thase ME, et al. Bupropion-SR, sertraline, or venlafaxine-XR after failure of SSRIs for depression. The New England Journal of Medicine. 2006;354(12):1231-1242
  7. 7. Papakostas GI. Tolerability of modern antidepressants. The Journal of Clinical Psychiatry. 2008;69(E1):8-13
  8. 8. Griebel G, Holmes A. 50 years of hurdles and hope in anxiolytic drug discovery. Nature Reviews. Drug Discovery. 2013;12:667-687
  9. 9. New WHO guidelines to promote proper use of alternative medicines [Internet] 2004. Available from: https://www.who.int/mediacentre/news/releases/2004/pr44/en/. WHO Guidelines: Developing Information on Proper Use of Traditional, Complementary and Alternative Medicine [pdf 3.1Mb] [Accessed: 01 July 2019]
  10. 10. WHO Traditional Medicine Strategy: 2014–2023. Hong Kong SAR, China: Organización Mundial de la Salud; 2013. ISBN: 978 92 4 350609 8 (Clasificación NLM: WB 55)
  11. 11. López-Rubalcava C, Estrada-Camarena E. Mexican medicinal plants with anxiolytic or antidepressant activity: Focus on preclinical research. Journal of Ethnopharmacology. 2016;186:377-391
  12. 12. Fajemiroye JO, da Silva DM, de Oliveira DR, Costa EA. Treatment of anxiety and depression: Medicinal plants in retrospect. Fundamental & Clinical Pharmacology. 2016;30(3):198-215
  13. 13. Lundstrom K, Pham HT, Dinh LD. Interaction of plant extracts with central nervous system receptors. Medicines (Basel). 2017;4(1):1-13
  14. 14. Arnason JT, Catling PM, Small E, Dang PT, Lambert JDH. Biodiversity & health: Focusing research to policy. In: Proceeding of the International Symposium, NRC Research Press, Canada. 2005
  15. 15. Abdelhalim A, Karim N, Chebib M, Aburjai T, Khan I, Johnston GA, et al. Antidepressant, anxiolytic and antinociceptive activities of constituents from Rosmarinus officinalis. Journal of Pharmacy & Pharmaceutical Sciences. 2015;18(4):448-459
  16. 16. Machado DG, Cunha MP, Neis VB, Balen GO, Colla A, Bettio LE, et al. Antidepressant-like effects of fractions, essential oil, carnosol and betulinic acid isolated from Rosmarinus officinalis L. Food Chemistry. 2013;136(2):999-1005
  17. 17. Khan I, Karim N, Ahmad W, Abdelhalim A, Chebib M. GABA-A receptor modulation and anticonvulsant, anxiolytic, and antidepressant activities of constituents from Artemisia indica Linn. Evidence-based Complementary and Alternative Medicine. 2016;1215393
  18. 18. Machado DG, Neis VB, Balen GO, Colla A, Cunha MP, Dalmarco JB, et al. Antidepressant-like effect of ursolic acid isolated from Rosmarinus officinalis L. in mice: Evidence for the involvement of the dopaminergic system. Pharmacology, Biochemistry, and Behavior. 2012;103(2):204-211
  19. 19. Colla AR, Oliveira A, Pazini FL, Rosa JM, Manosso LM, Cunha MP, et al. Serotonergic and noradrenergic systems are implicated in the antidepressant-like effect of ursolic acid in mice. Pharmacology, Biochemistry, and Behavior. 2014;124:108-116
  20. 20. Saeid A-M, Zohre K, Rahim T-F. The antidepressant-like effects of Origanum majorana essential oil on mice through monoaminergic modulation using the forced swimming test. Journal of Traditional and Complementary Medicine. DOI: 10.1016/j.jtcme.2019.01.003. [Accessed: 14 January 2019] in press
  21. 21. Coelho V, Mazzardo-Martins L, Martins DF, Santos AR, da Silva Brum LF, Picada JN, et al. Neurobehavioral and genotoxic evaluation of (−)-linalool in mice. Journal of Natural Medicines. 2013;67(4):876-880
  22. 22. Guzmán-Gutiérrez SL, Bonilla-Jaime H, Gómez-Cansino R, Reyes-Chilpa R. Linalool and β-pinene exert their antidepressant-like activity through the monoaminergic pathway. Life Sciences. 2015;128:24-29
  23. 23. Gomes PB, Feitosa ML, Silva MI, Noronha EC, Moura BA, Venâncio ET, et al. Anxiolytic-like effect of the monoterpene 1,4-cineole in mice. Pharmacology, Biochemistry, and Behavior. 2010;96(3):287-293
  24. 24. González-Trujano ME, Hernández-Sánchez LY, Muñoz Ocotero V, Dorazco-González A, Guevara Fefer P, Aguirre-Hernández E. Pharmacological evaluation of the anxiolytic-like effects of Lippia graveolens and bioactive compounds. Pharmaceutical Biology. 2017;55(1):1569-1576
  25. 25. Moreira MR, Salvadori MG, de Almeida AA, de Sousa DP, Jordán J, Satyal P, et al. Anxiolytic-like effects and mechanism of (−)-myrtenol: A monoterpene alcohol. Neuroscience Letters. 2014;579:119-124
  26. 26. 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 and Alternative Medicine. 2013;13:42
  27. 27. Zhang N, Yao L. Anxiolytic effect of essential oils and their constituents: A review. Journal of Agricultural and Food Chemistry. 2019. DOI: 10.1021/acs.jafc.9b00433 in press
  28. 28. Souto-Maior FN, de Carvalho FL, de Morais LC, Netto SM, de Sousa DP, de Almeida RN. Anxiolytic-like effects of inhaled linalool oxide in experimental mouse anxiety models. Pharmacology, Biochemistry, and Behavior. 2011;100(2):259-263
  29. 29. Nesterova YV, Povet’eva TN, Suslov NI, Shults EE, Ziuz’kov GN, Aksinenko SG, et al. Anxiolytic activity of diterpene alkaloid songorine. Bulletin of Experimental Biology and Medicine. 2015;159(5):620-622
  30. 30. German-Ponciano LJ, Rosas-Sánchez GU, Rivadeneyra-Domínguez E, Rodríguez-Landa JF. Advances in the preclinical study of some flavonoids as potential antidepressant agents. Scientifica. 2018;1:2963565
  31. 31. Liu LX, Chen WF, Xie JX, Wong MS. Neuroprotective effects of genistein on dopaminergic neurons in the mice model of Parkinson’s disease. Neuroscience Research. 2008;60:156-161
  32. 32. Tsai TH. Concurrent measurement of unbound genistein in the blood, brain and bile of anesthetized rats using microdialysis and its pharmacokinetic application. Journal of Chromatography. A. 2005;1073(1–2):317-322
  33. 33. Hu P, Ma L, Wang YG, Ye F, Wang C, Zhou WH, et al. Genistein, a dietary soy isoflavone, exerts antidepressant-like effects in mice: Involvement of serotonergic system. Neurochemistry International. 2017;108:426-435
  34. 34. Kageyama A, Sakakibara H, Zhou W, Yoshioka M, Ohsumi M, Shimoi K, et al. Genistein regulated serotonergic activity in the hippocampus of ovariectomized rats under forced swimming stress. Bioscience, Biotechnology, and Biochemistry. 2010;74(10):2005-2010
  35. 35. Sapronov NS, Kasakova SB. Effects of synthetic and plant-derived selective modulators of estrogen receptors on depression-like behavior of female rats. Bulletin of Experimental Biology and Medicine. 2008;146(1):73-76
  36. 36. Singh V, Chauhan G, Shri R. Anti-depressant like effects of quercetin 4'-O-glucoside from Allium cepa via regulation of brain oxidative stress and monoamine levels in mice subjected to unpredictable chronic mild stress. Nutritional Neuroscience. 2019;1:1-10
  37. 37. Holzmann I, da Silva LM, Corrêa da Silva JA, Steimbach VM, de Souza MM. Antidepressant-like effect of quercetin in bulbectomized mice and involvement of the antioxidant defenses, and the glutamatergic and oxidonitrergic pathways. Pharmacology Biochemistry and Behavior. 2015;136:55-63
  38. 38. Lindqvist D, Dhabhar FS, James SJ, Hough CM, Jain FA, Bersani FS, et al. Oxidative stress, inflammation and treatment response in major depression. Psychoneuroendocrinology. 2017;76:197-205
  39. 39. Filho CB, Jesse CR, Donato F, Del Fabbro L, Gomes de Gomes M, Rossito Goes AT, et al. Chrysin promotes attenuation of depressive-like behavior and hippocampal dysfunction resulting from olfactory bulbectomy in mice. Chemico-Biological Interactions. 2016;260:154-162
  40. 40. Wang Y, Wang B, Lu J, Shi H, Gong S, Wang Y, et al. Fisetin provides antidepressant effects by activating the tropomyosin receptor kinase B signal pathway in mice. Journal of Neurochemistry. 2017;143(5):561-568
  41. 41. Ren Z, Yan P, Zhu L, Yang H, Zhao Y, Kirby BP, et al. Dihydromyricetin exerts a rapid antidepressant-like effect in association with enhancement of BDNF expression and inhibition of neuroinflammation. Psychopharmacology. 2018;235(1):233-244
  42. 42. Donato F, de Gomes MG, Goes AT, Filho CB, Del Fabbro L, Antunes MS, et al. Hesperidin exerts antidepressant-like effects in acute and chronic treatments in mice: Possible role of l-arginine-NO-cGMP pathway and BDNF levels. Brain Research Bulletin. 2014;104:19-26
  43. 43. Zhang R, Ma Z, Liu K, Li Y, Liu D, Xu L, et al. Baicalin exerts antidepressant effects through Akt/FOXG1 pathway promoting neuronal differentiation and survival. Life Sciences. 2019;15(221):241-248
  44. 44. Ben-Azu B, Nwoke EE, Aderibigbe AO, Omogbiya IA, Ajayi AM, Olonode ET, et al. Possible neuroprotective mechanisms of action involved in the neurobehavioral property of naringin in mice. Biomedicine & Pharmacotherapy. 2019;109:536-546
  45. 45. de la Peña JB, Kim CA, Lee HL, Yoon SY, Kim HJ, Hong EY, et al. Luteolin mediates the antidepressant-like effects of Cirsium japonicum in mice, possibly through modulation of the GABAA receptor. Archives of Pharmacal Research. 2014;37(2):263-269
  46. 46. Aguirre-Hernández E, González-Trujano ME, Terrazas T, Herrera Santoyo J, Guevara-Fefer P. Anxiolytic and sedative-like effects of flavonoids from Tilia americana var. mexicana: GABAergic and serotonergic participation. Salud Mental. 2001;39(1):37-46
  47. 47. Wang XS, Guan SY, Liu A, Yue J, Hu LN, Zhang K, et al. Anxiolytic effects of formononetin in an inflammatory pain mouse model. Molecular Brain. 2019;12(1):36
  48. 48. Kita M, Uchida S, Yamada K, Ano Y. Anxiolytic effects of theaflavins via dopaminergic activation in the frontal cortex. Bioscience, Biotechnology, and Biochemistry. 2019;83(6):1157-1162
  49. 49. Rodríguez-Landa JF, Hernández-López F, Cueto-Escobedo J, Herrera-Huerta EV, Rivadeneyra-Domínguez E, Bernal-Morales B, et al. Chrysin (5,7-dihydroxyflavone) exerts anxiolytic-like effects through GABAA receptors in a surgical menopause model in rats. Biomedicine & Pharmacotherapy. 2019;109:2387-2395
  50. 50. Qiu ZK, Zhang GH, Zhong DS, He JL, Liu X, Chen JS, et al. Puerarin ameliorated the behavioral deficits induced by chronic stress in rats. Scientific Reports. 2017;7(1):6266
  51. 51. Rodríguez-Landa JF, Cueto-Escobedo J, Puga-Olguín A, Rivadeneyra-Domínguez E, Bernal-Morales B, Herrera-Huerta EV, et al. The phytoestrogen genistein produces similar effects as 17β-estradiol on anxiety-like behavior in rats at 12 weeks after ovariectomy. BioMed Research International. 2017;2017:9073816
  52. 52. Rodríguez-Landa JF, Hernández-Figueroa JD, Hernández-Calderón BC, Saavedra M. Anxiolytic-like effect of phytoestrogen genistein in rats with long-term absence of ovarian hormones in the black and white model. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2009;33(2):367-372
  53. 53. Wu ZM, Ni GL, Shao AM, Cui R. Genistein alleviates anxiety-like behaviors in post-traumatic stress disorder model through enhancing serotonergic transmission in the amygdala. Psychiatry Research. 2017;255:287-291
  54. 54. Kelmendi B, Adams TG, Yarnell S, Southwick S, Abdallah CG, Krystal JH. PTSD: From neurobiology to pharmacological treatments. European Journal of Psychotraumatology. 2016;8(7):31858
  55. 55. Di L, Kerns EH, Carter GT. Strategies to assess blood-brain barrier penetration. Expert Opinion on Drug Discovery. 2008;3(6):677-687
  56. 56. Akbar S, Subhan F, Karim N, Aman U, Ullah S, Shahid M, et al. Characterization of 6-methoxyflavanone as a novel anxiolytic agent: A behavioral and pharmacokinetic approach. European Journal of Pharmacology. 2017;801:19-27
  57. 57. Hernandez-Leon A, González-Trujano ME, Fernández-Guasti A. The anxiolytic-like effect of rutin in rats involves GABAA receptors in the basolateral amygdala. Behavioural Pharmacology. 2017;28(4):303-312
  58. 58. Karim N, Irshad S, Khan I, Mohammad A, Anis I, Shah MR, et al. GABA(A) receptor modulation and neuropharmacological activities of viscosine isolated from Dodonaea viscosa (Linn). Pharmacology, Biochemistry, and Behavior. 2015;136:64-72
  59. 59. Lee B, Sur B, Yeom M, Shim I, Lee H, Hahm DH. Effect of berberine on depression- and anxiety-like behaviors and activation of the noradrenergic system induced by development of morphine dependence in rats. The Korean Journal of Physiology & Pharmacology. 2012;16(6):379-386
  60. 60. Martínez-Vázquez M, Estrada-Reyes R, Araujo Escalona AG, Velázquez IL, Martínez-Mota L, Morenod J, et al. Antidepressant-like effects of an alkaloid extract of the aerial parts of Annona cherimolia in mice. Journal of Ethnopharmacology. 2012;139:164-170
  61. 61. Farzin D, Mansouri N. Antidepressant-like effect of harmane and other β-carbolines in the mouse forced swim test. European Neuropsychopharmacology. 2006;16:324-328
  62. 62. Fortunato JJ, Réus GZ, Kirsch TR, Stringari RB, Fries GR, Kapczinski F, et al. Effects of beta-carboline harmine on behavioral and physiological parameters observed in the chronic mild stress model: Further evidence of antidepressant properties. Brain Research Bulletin. 2010;81(4–5):491-496
  63. 63. Adell A, Biggs TA, Myersb RD. Action of Harman (l-methyl-P-carboline) on the brain: Body temperature and in viva efflux of 5-HT from hippocampus of the rat. Neuropharmacology. 1996;35:1101-1107
  64. 64. Glennon RA, Dukat M, Grella B, Hong S, Costantino L, Teitler M, et al. Binding of b-carbolines and related agents at serotonin (5-HT2 and 5-HT1A), dopamine (D2) and benzodiazepine receptors. Drug and Alcohol Dependence. 2000;60:121-132
  65. 65. Smith KL, Ford GK, Jessop DS, Finn DP. Behavioural, neurochemical and neuroendocrine effects of the endogenous β-carboline harmane in fear-conditioned rats. Journal of Psychopharmacology. 2013;2:162-170
  66. 66. Ali BH, Bashir AK, Tanira MOM, Medvedev AE, Jarrett N, Sandler M, et al. Effect of extract of Rhazya stricta, a traditional medicinal plant, on rat brain tribulin. Pharmacology, Biochemistry, and Behavior. 1998;59:671-675
  67. 67. Idayu NF, Hidayata MT, Moklasa MAM, Sharidaa F, Raudzaha ARN, Shamimaa AR, et al. Antidepressant-like effect of mitragynine isolated from Mitragyna speciosa Korth in mice model of depression. Phytomedicine. 2011;18:402-407
  68. 68. Dhingra D, Valecha R. Punarivine, an alkaloid isolated from ethanolic extract of Boerhaavia diffusa Linn, reverses depression-like behaviour in mice subject to chronic unpredictable mild stress. Indian Journal of Experimental Biology. 2014;52:799-807
  69. 69. Jiang ML, Zhang ZX, Li YZ, Wang XH, Yan W, Gong GQ. Antidepressant-like effect of evodiamine on chronic unpredictable mild stress rats. Neuroscience Letters. 2015;588:154-158
  70. 70. Wattanathorn J, Chonpathompikunlert P, Muchimapura S, Priprem A, Tankamnerdthai O. Piperine, the potential functional food for mood and cognitive disorders. Food and Chemical Toxicology. 2008;46:3106-3110
  71. 71. Xu LF, Chu WJ, Qing XY, Li S, Wang XS, Qing GW, et al. Protopine inhibits serotonin transporter and noradrenaline transporter and has the antidepressant-like effect in mice models. Neuropharmacology. 2006;50:934-940
  72. 72. Liu M, Huang HH, Yang J, Su YP, Lin HW, Lin LQ, et al. The active alkaloids of Gelsemium elegans Benth., are potent anxiolytics. Psychopharmacology. 2013;225(4):839-851
  73. 73. Meyer L, Boujedaini N, Patte-Mensah C, Mensah-Nyagan AG. Pharmacological effect of gelsemine on anxiety-like behavior in rat. Behavioural Brain Research. 2013;253:90-94
  74. 74. Chen CJ, Zhong ZF, Xin ZM, Hong LH, Su YP, Yu CX. Koumine exhibits anxiolytic properties without inducing adverse neurological effects on functional observation battery, open-field and Vogel conflict tests in rodents. Journal of Natural Medicines. 2017;71(2):397-408
  75. 75. Ngo Bum E, Taiwe GS, Moto FC, et al. Anticonvulsant, anxiolytic, and sedative properties of the roots of Nauclea latifolia Smith in mice. Epilepsy & Behavior. 2009;15(4):434-440
  76. 76. Alavijeh MM, Vaezi G, Khaksari M, Hojati V. Berberine hydrochloride attenuates voluntary methamphetamine consumption and anxiety-like behaviors via modulation of oxytocin receptors in methamphetamine addicted rats. Physiology & Behavior. 2019;206:157-165
  77. 77. Rolland A, Fleurentin J, Lanhers MC, et al. Neurophysiological effects of an extract of Eschscholzia californica Cham. (Papaveraceae). Phytotherapy Research. 2001;15(5):377-381
  78. 78. Jung JW, Ahn NY, Oh HR, Lee BK, Lee KJ, Kim SY, et al. Anxiolytic effects of the aqueous extract of Uncaria rhynchophylla. Journal of Ethnopharmacology. 2006;108(2):193-197
  79. 79. Guaraldo L, Chagas DA, Konno AC, et al. Hydroalcoholic extract and fractions of Davilla rugosa Poiret: Effects on spontaneous motor activity and elevated plus-maze behavior. Journal of Ethnopharmacology. 2000;72(1–2):61-67
  80. 80. Raupp IM, Sereniki A, Virtuoso S, et al. Anxiolytic-like effect of chronic treatment with Erythrina velutina extract in the elevated plus-maze test. Journal of Ethnopharmacology. 2008;118(2):295-299
  81. 81. Onusic GM, Nogueira RL, Pereira AM, et al. Effect of acute treatment with a water-alcohol extract of Erythrina mulungu on anxiety-related responses in rats. Brazilian Journal of Medical and Biological Research. 2002;35(4):473-477
  82. 82. Ku TH, Lee YJ, Wang SJ, et al. Effect of honokiol on activity of GAD(65) and GAD(67) in the cortex and hippocampus of mice. Phytomedicine. 2011;18(13):1126-1129
  83. 83. Kash SF, Tecott LH, Hodge C, Baekkeskov S. Increased anxiety and altered responses to anxiolytics in mice deficient in the 65-kDa isoform of glutamic acid decarboxylase. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(4):1698-1703
  84. 84. Han H, Jung JK, Han SB, et al. Anxiolytic-like effects of 4-Omethylhonokiol isolated from Magnolia officinalis through enhancement of GABAergic transmission and chloride influx. Journal of Medicinal Food. 2011;14(7–8):724-731
  85. 85. Seo JJ, Lee SH, Lee YS, et al. Anxiolytic-like effects of obovatol isolated from Magnolia obovata: Involvement of GABA/benzodiazepine receptors complex. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2007;31(7):1363-1369
  86. 86. Klingberg S, Andersson H, Mulligan A, Bhaniani A, Welch A, Bingham S. Food sources of plant sterols in the EPIC Norfolk population. European Journal of Clinical Nutrition. 2008;62:695-703
  87. 87. Tabata RC, Wilson JM, Zwiegers P, Kwok D, Van Kampen JM. Chronic exposure to dietary sterol glucosides is neurotoxic to motor neuronsand induces an ALS-PDC phenotype. NeuroMolecular Medicine. 2008;10:24-39
  88. 88. Trevisan G, Rossato MF, Walker CI, Klafke JZ, Rosa F, Oliveira SM. Identification of the plant steroid alpha-spinasterol as a novel transient receptor potential vanilloid 1 antagonist with antinociceptive properties. The Journal of Pharmacology and Experimental Therapeutics. 2012;343(2):258-269
  89. 89. Martins D, Tavares I, Morgado C. Hotheaded: The role of TRPV1 in brain functions. Neuropharmacology. 2014;85:151-157
  90. 90. Socała K, Wlaź P. Evaluation of the antidepressant- and anxiolytic-like activity of α-spinasterol, a plant derivative with TRPV1 antagonistic effects, in mice. Behavioural Brain Research. 2016;5(303):19-25
  91. 91. Marsch R, Foeller E, Rammes G, Bunck M, Kössl M, Holsboer F, et al. Reduced anxiety, conditioned fear, and hippocampal long-term potentiation in transient receptor potential vanilloid type 1 receptor-deficient mice. The Journal of Neuroscience. 2007;27(4):832-839
  92. 92. Zhen XH, Quan YC, Jiang HY, Wen ZS, Qu YL, Guan LP. Fucosterol, a sterol extracted from Sargassum fusiforme, shows antidepressant and anticonvulsant effects. European Journal of Pharmacology. 2015;768:131-138
  93. 93. Zhao D, Zheng L, Qi L, Wang S, Guan L, Xia Y, et al. Structural features and potent antidepressant effects of total sterols and β-sitosterol extracted from Sargassum horneri. Marine Drugs. 2016;14:123
  94. 94. Aragão GF, Carneiro LM, Junior APF, Vieira LC, Bandeira PN, Lemos TLG, et al. A possible mechanism for anxiolytic and antidepressant effects of alpha- and beta-amyrin from Protium heptaphyllum (Aubl.) March. Pharmacology, Biochemistry and Behavior. 2007;85(4):827-834
  95. 95. Cassani J, Dorantes-Barrón AM, Novales LM, Real GA, Estrada-Reyes R. Anti-depressant-like effect of kaempferitrin isolated from Justicia spicigera Schltdl (Acanthaceae) in two behavior models in mice: Evidence for the involvement of the serotonergic system. Molecules. 2014;19(12):21442-21461
  96. 96. Nogueira E, Rosa GJ, Haraguchi M, Vassilieff VS. Anxiolytic effect of Rubus brasilensis in rats and mice. Journal of Ethnopharmacology. 1998;61(2):111-117
  97. 97. Liu R, Ahmed F, Cayer C, Mullally M, Carballo AF, Rojas MO, et al. New botanical anxiolytics for use in companion animals and humans. The AAPS Journal. 2017;19(6):1626-1631
  98. 98. Lima EBC, de Sousa CNS, Meneses LN, E Silva Pereira YF, Matos NCB, de Freitas RB, et al. Involvement of monoaminergic systems in anxiolytic and antidepressive activities of the standardized extract of Cocos nucifera L. Journal of Natural Medicines. 2017;71(1):227-237

Written By

Rosa Isela García-Ríos, Armando Mora-Pérez, Ana Raquel Ramos-Molina and Cesar Soria-Fregozo

Submitted: 27 May 2019 Reviewed: 20 December 2019 Published: 04 February 2020