Back to Journals » Neuropsychiatric Disease and Treatment » Volume 21

Anxiolytic, Antidepressant and Healthy Sleep-Promoting Potential of Rosmarinic Acid: Mechanisms and Molecular Targets

Authors Priya V, Srinivasan D , Priyadarsini S, Dabaghzadeh F, Rana SS, Chengaiyan JG, Sudesh R, Ahmad F 

Received 18 October 2024

Accepted for publication 20 January 2025

Published 20 March 2025 Volume 2025:21 Pages 641—661

DOI https://doi.org/10.2147/NDT.S501597

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Yu-Ping Ning



Vijayan Priya,1 Dhiyanesh Srinivasan,1 Swagatika Priyadarsini,2 Fatemeh Dabaghzadeh,3 Sandeep Singh Rana,4 Jeevitha Gada Chengaiyan,4 Ravi Sudesh,5 Faraz Ahmad1

1Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India, 632014; 2ICAR-National Research Centre on Camel (NRCC), Bikaner, India, 334001; 3Department of Clinical Pharmacy, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran; 4Department of Biosciences, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014 India; 5Department of Biomedical Science, School of Bio Sciences and Technology (SBST) Vellore Institute of Technology (VIT), Vellore, 632014, India

Correspondence: Faraz Ahmad, Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, India, Email [email protected] Ravi Sudesh, Department of Biomedical Science, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, India, Email [email protected]

Abstract: The etiology of psychiatric disorders is complex and results from intricate interactions among multiple neurobiological, psychological, environmental, and genetic factors. Furthermore, the roles of gut microbiome dyshomeostasis in their pathogeneses are just beginning to be uncovered, adding to another level of complexity. In recent years, significant efforts have been directed toward discovering multimodal yet safe therapeutics to counteract psychological deficits. Rosmarinic acid (RA), a polyphenol found in several medicinal herbs, has received considerable attention as a potential multifaceted therapeutic agent, particularly for neuropsychiatric conditions. In order to critically evaluate this aspect, data was compiled and consolidated after extensive searches on scholarly databases like PubMed, Google Scholar, and Web of Science. Peer-reviewed publications which focused on RA as a therapeutic agent for psychiatric disorders were included regardless of the year of publication and country of origin. Based on pre-clinical and clinical evidence, this review delves into the various mechanistic aspects of the antidepressant, anxiolytic, and sleep-promoting functions of RA. The beneficial effects of RA on the gut-microbiome-brain (GMB) axis and their implications for the regulation of neuroprotective pathways are also discussed, with a particular focus on exploiting them to ameliorate neuropsychiatric conditions. Our assessment indicated that RA is a multimodal neuroprotectant against psychiatric conditions and beneficially influences a plethora of targets related to redox, inflammatory, synaptic, cell death, neurotrophic, and cell signaling pathways. As a dietary agent, RA may also be relevant in favorably altering the GMB axis, indicating its prospects as a potential multimodal adjuvant therapeutic agent in regulating the pathogenic mechanisms underlying neuropsychiatric conditions. However, more extensive clinical studies are required to ascertain the neuromodulatory actions of RA in neuronal pathophysiologies, including psychiatric ailments.

Keywords: psychiatric disorders, circadian rhythm, gut-microbiome-brain axis, phytotherapy, traditional medicine

Introduction

Psychological conditions, such as stress, anxiety, and depression, have a tremendous impact on disability rates and overall quality of life. While the pathogenic mechanisms of these disorders rely on an intricately complex interaction between multiple neurobiological, genetic, and environmental factors, resident gut microbial species are also increasingly being perceived as a critical underlying element.1 This emphasizes the necessity for thorough evaluations of multimodal treatment plans to address mental health disorders. Furthermore, the regimens should be safe with minimal or no adverse effects. In this regard, plant-based therapeutic agents isolated from traditional medicinal herbs are coming to the forefront.2 Indeed, medicinal plants and their bioactive components have been shown to have promising therapeutic potential in neurological and psychiatric conditions.3

Rosmarinic acid (RA) is a naturally occurring polyphenolic compound derived from hydroxycinnamic acid, which is found in many plants, predominantly in the Lamiaceae and Boraginaceae families.4 It is a major bioactive compound found in medicinal herbs, such as rosemary (Salvia rosmarinus), lemon balm (Melissa officinalis), and spearmint (Mentha spicata), which are also used as dietary supplements.5 RA extracted from these natural sources has been found to have immense potential applications for human health.6 It offers a variety of medicinal uses, including anti-inflammatory, anti-cancer, antioxidant, anti-apoptotic, anti-aging, and neuroprotective applications.7–9 With regard to central nervous system (CNS) disorders, RA supplementation is regarded as a potent multifaceted therapeutic intervention for ameliorating several pathological dysfunctions, lowering the risk of psychiatric conditions, and improving memory and cognition.9,10 RA is also perceived as a promising therapeutic for sleep dysfunction,11 which is often associated with psychiatric conditions, such as depression and anxiety.12

While the neuromodulatory and neuroprotective potential of RA is well known, only recently have research studies associated with RA supplementation shown beneficial alterations in the gut-microbiome-brain (GMB) axis. While previously published reviews on the neuroprotective activities of RA primarily cover its antioxidant, anti-inflammatory, and neurogenesis-promoting activities in neuropathologies in general,9,10,13,14 the objective of the current review is to specifically delineate the underlying molecular and cellular mechanisms of RA-mediated antidepressant, anxiolytic, and healthy sleep-promoting actions. Furthermore, evidence for its utility, primarily as a prominent component of various plant-based extracts, in attenuating psychological dysfunction in humans is discussed in detail. Clinical evidences discussed in this review may bridge the gap between preclinical findings and their possible practical therapeutic applications. Indeed, such comprehensive and parallel assessment of the mechanisms and therapeutic potential of RA coupled with clinical insights may provide avenues for RA-based future clinical interventional approaches and for the development of personalized regimens for the treatment of psychological conditions. Additionally, RA’s potential ability to positively modulate the GMB axis is currently under exploration, and the data suggests promising results, emphasizing its dietary relevance. Hence, based on recent reports, we implicate RA as a potent regulator of resident microflora, with a particular focus on the mental and psychological well-being of the host. Finally, we outline the limitations that have hindered the therapeutic use of RA to its full potential and suggest possible strategies to overcome these challenges.

Materials and Methods

The selection of pertinent primary research and review articles for compilation of this narrative review was performed using scholarly databases, such as PubMed (Medline), Google Scholar, and Web of Science. Multiple keyword combinations were employed for the literature search; {(“rosmarinic acid”) AND (“depression” OR “anxiolytic” OR “neuropsychiatric” OR “anxiety” OR “sleep” OR “gut microbiota” etc.)}. Few more search keywords such as “neuroprotection”, “clinical trials”, “gut-microbiome-brain”, etc. were used. The retrieved articles were included in the study only after individualistic assessment of the titles and abstracts as well as a detailed inspection of the full texts. Furthermore, cited references in the retrieved articles and articles citing them were screened for pertinence and included if relevant. Articles were retrieved solely based on relevance, regardless of the publication timeframe. Articles in languages other than English were excluded, primarily because of the inability of the authors to comprehend other languages (and the ineffectiveness of language translation platforms in scholarly literature). The literature retrieved in this manner allowed for the collection of different articles presenting pre-clinical and clinical evidence for RA-mediated amelioration of psychiatric conditions.

RA and Mental Health

Several independent research groups have implicated RA as a potent anxiolytic, antidepressant, and normal sleep-promoting therapeutic.9,15,16 It also elicits beneficial effects against psycho-behavioral anxiety- and depression-like comorbidities in neuropathologies, such as neuropathic pain,17,18 epilepsy,19,20 and Alzheimer’s disease.21,22 The underlying mechanisms of RA-mediated therapeutic actions for neuropsychiatric conditions of anxiety and depression appear to be multimodal and encompass the regulation of a plethora of endocrine, inflammatory, redox, and synaptic targets (Figure 1).

Figure 1 Molecular targets and mechanisms underlying the multimodal anxiolytic and antidepressant actions of RA. The figure illustrates the interactions of RA with different receptors, including TNF-α, 5-HT1, and GABA and glutamate receptors, resulting in beneficial modulation of second messenger intracellular pathways involving cAMP, IP3, and DAG. Downstream activation of cell signaling cascades involving protein kinases PKA, and PKC, as well as nuclear factors, CREB and GSK-3β mediate multifaceted neuroprotective effects. These include regulation of pro-inflammatory cytokine release, signaling thorough neurotrophic factors like BDNF, and anti-inflammatory mediators such as IL-10 and TGF-β, resulting in reduced inflammation, enhanced neuroprotection, and mood stabilization.

Abbreviations: TNF-α, tumor necrosis factor alpha; 5-HT1 receptor, 5-hydroxytryptamine (serotonin) receptor 1; GABA, gamma-aminobutyric acid; cAMP, cyclic adenosine monophosphate; IP3, inositol 1,4,5-trisphosphate; DAG, diacylglycerol; PKA, protein kinase A; PKC, protein kinase C; CREB, cAMP response element-binding protein; GSK-3β, glycogen synthase kinase-3 beta; BDNF, brain-derived neurotrophic factor; IL-10, interleukin-10; TGF-β, transforming growth factor beta.

RA is the most relevant phytochemical constituent underlying the suppression of depression- and anxiety-like behavior of several traditional medicinal plants, such as Perilla frutescens,23 Micromeria myrtifolia,24 Satureja montana,25 and Origanum majorana.26 In this section, we focus on the anxiolytic and antidepressant actions of RA, detailing the cellular and molecular mechanisms underlying the inflammatory, oxidative, and synaptic regulation elicited by RA in neuropsychiatric conditions (Table 1). Implications of oral supplementation of RA as a therapeutic strategy against psychological and sleep disturbances in humans are discussed in a subsequent section. Furthermore, given the critical roles of the gut microbiome in the pathophysiology of the CNS,27 we examined the implications of RA as an exogenous agent that regulates the GMB axis as a key facet of its therapeutic actions against neuropsychiatric ailments.

Table 1 Cellular and Molecular Mechanisms of RA-Mediated Anxiolytic and Antidepressant Actions

Redox Regulation

The brain is particularly sensitive to oxidative stress and neuronal injury, and redox dysregulation has been implicated as a primary mechanism of several neuropathologies, including neuropsychiatric ailments.47 Amelioration of dysregulated redox pathways is a critical facet of the potent anxiolytic and antidepressant functions of RA. Thus, RA was shown to ameliorate behavioral deficits in open field, elevated plus maze, tail suspension and forced swim paradigms in mice challenged with restraint stress, in part by rescuing dysfunction of the neuronal antioxidant pathways, particularly those centered on superoxide dismutase (SOD) and glutathione peroxidase (GPx) enzymes.28 An Iranian research group identified RA as the major bioactive accounting for the antidepressant action of medicinal herbs, Nepeta menthoides and Melissa officinalis.40 Further experimental analyses indicated that the antidepressant potential of these RA-containing herbal extracts was due to the ability of RA to stimulate brain catalase activity. Verma et al recently showed that RA rescues depression-like behavior in chronic unpredictable stress (CUS)-exposed,30 and in chronic maternal separation-challenged rats, probably via mitigation of deficits in the antioxidant, inflammatory, and serotonergic pathways. The authors proposed restoration of the defects in corticosterone, brain-derived neurotrophic factor (BDNF), creatine kinase, oxidative (glutathione and SOD), inflammatory (interleukin 10; IL-10), and cell death pathways as the major mechanisms underlying RA-mediated antidepressant effects.31 These results indicate that the antioxidant system of SOD is a primary target of RA-mediated antidepressant actions in CUS, in agreement with those obtained by Gavzan et al.48 Depressive comorbidity linked to lipopolysaccharide (LPS)-induced hippocampal neuroinflammation has been shown to be repressed by RA via activation of nuclear factor erythroid 2-related factor 2 (NRF-2) signaling, resulting in significant induction of antioxidant enzymes, heme oxygenase 1 (HO-1), NAD(P)H-quinone oxidoreductase (NQO1), and glutamate–cysteine ligase catalytic subunit (GCLC).29

Interestingly, gender appears to be a significant mediator of the therapeutic actions of RA, including neuroprotection. For instance, supplementation of RA has been shown to elicit sex-dependent modulation of hippocampal functions, including oxidative, immune-related and sex hormone signaling.49 While RA supplementation is associated with memory improvements in both aged male and female rodents, it leads to altered lipid metabolism in young adults. The underlying mechanisms of these alterations appear to be gender-specific; in males, glycemic signaling is impaired, while in females, BDNF signaling may be the more prominent target.50 Hence, it is plausible that peripheral metabolic and endocrinological parameters influence RA’s actions. Indeed, previous literature suggests that RA, along with other polyphenols, is involved in regulating sex hormone pathways. Thus, a study done on screening natural compounds and potential drugs against breast cancer indicated that RA binds strongly to the active site and inhibits aromatase, an enzyme responsible for estrogen biosynthesis, supporting its potential in regulating sex hormone pathways.51 Along similar lines, Zych et al52 have suggested that RA can modulate certain metabolic and oxidative parameters under conditions of estrogen deprivation, such as those experienced by ovariectomized rats. These findings indicate that RA, in addition to its antioxidant effects, could modulate hormone levels and control metabolic parameters, possibly in a gender-specific manner. However, further studies are warranted to clearly establish gender as a moderator of the actions of RA on pathophysiological processes, particularly those related to emotionality and psychological functions.

Neuroinflammation

Dyshomeostasis of neuroinflammatory signaling is another relevant therapeutic target for psychological conditions such as depression and anxiety. Given the potent anti-inflammatory actions of RA,53 it is hardly surprising that studies have implicated neuroinflammatory pathways as potential underlying mediators of RA’s beneficial effects. Hence, RA was found to prevent depressive and anxiety-like behavior in rodents subjected to the cecal ligation and puncture (CLP) paradigm of sepsis.44 In the CLP-induced sepsis in male mice, RA had shown to prevent neuronal damage and induce synaptogenesis, likely due to normalization of neuroinflammatory responses, including attenuation of microglial M1 polarization, as assessed by the levels of ionized calcium-binding adapter molecule 1 (IBA-1), inducible nitric oxide synthase (iNOS), and tumor necrosis factor alpha (TNF-α). The authors further implicated the receptor for activated C kinase/hypoxia-inducible factor 1 alpha (RACK/HIF-1α) signaling as a key target of RA-mediated antidepressant and anxiolytic actions in sepsis. In a recently published study, RA was found to attenuate the effects of maternal separation-induced psychological stress in mice by prompting appreciable reductions in the levels of pro-inflammatory mediators, TNF-α, IL-1β, toll-like receptor 4 (TLR4), and iNOS.42 Mitigation of neuroinflammatory responses as a module of antidepressant activities of RA has also been proposed by Yu et al, who showed that RA markedly reduced the expression of pro-inflammatory mediators, iNOS, CD44, TNF-α, and IL-1β, by rescuing the deficits in BDNF/Nrf2 signaling.29

Synaptic Signaling

Monoaminergic signaling has been known as a therapeutic target for RA-mediated antidepressant and anxiolytic actions. Indeed, pharmacological assessments in animal models of stress and depression have established serotonergic, noradrenergic and dopaminergic systems as chief underlying players.54 Lin et al have also implicated serotonin and signaling through its postsynaptic receptors as mediators of antidepressant actions of RA.41 Kondo et al evaluated the underlying mechanisms of RA’s antidepressant in rodents challenged with tail suspension stress, and found that it causes significant upregulation of tyrosine hydroxylase (TH) and pyruvate carboxylase, enzymes which are known regulators of dopaminergic, serotoninergic and GABAergic signaling.34,35 Further, stimulated hippocampal catecholaminergic signaling in their study was found to be associated with repressed mitogen-activated protein kinase phosphatase-1 (MKP-1) signaling, as well as with enhanced BDNF signaling.34 Cell culture experiments using RA in corticosterone-challenged PC12 cells confirmed its cytoprotective actions and positive influences on dopaminergic, serotonergic, and GABAergic pathways. Increased production of the neurotransmitters dopamine, serotonin, norepinephrine, and acetylcholine was proposed to be dependent on the cell kinase systems of mitogen-activated protein kinase (MAPK) and ERK-1/2.35

While previous studies have suggested that the antidepressant activities of RA may not depend on monoamine oxidase (MAO) or synaptic uptake of monoaminergic neurotransmitters,55 recent studies indicate that RA can influence the monoaminergic system by altering the expression and activation status of monoamine oxidase isoforms A (MAO-A)56 and B (MAO-B).57 Indeed, IC50 of RA for MAO-A and MAO-B have been found to be 6.5 and 5.3 mg/mL.24 Acetylcholinesterase (AChE) hydrolyzes acetylcholine in the synaptic cleft, and thereby influences signaling at the cholinergic synapses. AChE appears to be a key synaptic target for the RA-mediated antidepressant and anxiolytic actions. Cellular models have confirmed the ability of RA to significantly alter AChE activity.58 In fact, direct high-affinity binding of RA to AChE has been confirmed using in silico analyses.59

GABAergic signaling is another target of RA in the psychological conditions of anxiety and depression. For instance, Ibarra et al found that RA, as a principal bioactive in Melissa officinalis extract, repressed anxiety-like behavior of rodents in open field and elevated plus maze tasks by repressing GABA transaminase, elevating brain GABA levels.60 These findings are in concurrence with a recent study, wherein the authors proposed that the anxiolytic efficacy of RA-containing Melissa officinalis extract is due to its ability to inhibit GABA transaminase and MAO-A enzymes, and stimulate the cellular endogenous antioxidant potentials.61 Further, anxiolytic and hypnotic actions of Persian medicinal herb Salvia limbata in pentobarbital-challenged mice have been proclaimed to be dependent principally on RA as the major constituent; pharmacological assessment indicated the involvement of α1 subunit-containing GABA-A receptors in RA-mediated anxiolysis and sedation.37 Similarly, RA was found to potentiate hypnotic and anxiolytic actions of pentobarbital by stimulating the expressions of GABA producing enzyme, glutamic acid decarboxylase (GAD) and GABA-A receptor subunits, α3, α4, α5, β2 and γ3.62 Recently, based upon ex vivo synaptosomal and in silico experiments, a direct binding and activation of GABA-A receptors has been proposed for RA.63 Interestingly, Kim et al have also proposed involvement of adenosine receptors, A1-R and A2A-R, in normal sleep-promoting functions of RA, which probably also involved altered neuronal activity of cholinergic and GABAergic signaling in two key sleep-regulating brain regions, lateral hypothalamus and ventrolateral preoptic nucleus, respectively.64

Anti-Apoptotic Signaling

RA, a significant bioactive compound in Melissa officinalis hydroalcoholic extract, has been shown to restore anti-apoptotic/pro-apoptotic balance in the prefrontal cortical and hippocampal brain regions of mice challenged with chronic restraint stress.28 Specifically, the authors found that repression of depression-like behavior, as assessed by elevated plus maze, forced swimming, and tail suspension paradigms, was associated with increased expression of anti-apoptotic marker, Bcl-2; as well as depressed levels of pro-apoptotic markers Bax and cleaved caspase 3/pro-caspase 3 ratio. In addition to relieving hyperalgesia in neuropathic pain, RA mitigated comorbid symptoms of anxiety, depression, and sleep disturbances in mice. RA-induced neuroprotective effects in a spared nerve injury (SNI) model were possibly due to suppression of neuroinflammatory responses, which in turn caused attenuation of cellular senescence, as assessed by the diminished levels of marker protein, β-galactosidase.17,18 Furthermore, Shahrestani et al have also proposed apoptotic mediator, caspase 3 as a critical mediator of neuroprotective and anxiolytic functions of RA.65 Mitigation of genotoxic brain DNA damage as assayed by the comet assay in rats may be another mechanism underlying RA-mediated anxiolysis.43 More recently, results from pentylenetetrazole (PTZ)- and pilocarpine-challenged mice indicate that sleep promoting activities of RA may stem from its ability to limit redox alterations and prevent DNA damage in the hippocampal and cortical brain regions.20

Neurotrophic and Neurogenic Pathways

Neurotrophic signaling involving BDNF and its TrkB receptor has been implicated as a major factor in the regulation of RA-mediated anxiolytic and antidepressant functions. Therefore, Kondo et al reported robust antidepressant effect of RA when administered orally at doses 5 and 10 mg/kg for 7 consecutive days in depressive male mice, likely via stimulatory actions on BDNF signaling.34 Probably, the best example of involvement of BDNF signaling in mediating the beneficial actions of RA in depressive states comes from animal studies, which show that RA can efficiently reduce depressive symptoms associated with brain ageing. For example, RA supplementation in aged mice with weak psychological stress-induced depression appears to result in appreciable antidepressant actions.33 These have been shown to alleviate stress-induced alterations in the SUV39H1 histone methyltransferase-related epigenetic pathways controlling the expression of BDNF and mitogen-activated protein kinase phosphatase-1 (MKP-1). Furthermore, RA-mediated mitigation of depression-like psycho-behavioral abnormalities in mice subjected to chronic restraint stress may also be dependent on its ability to alter epigenetic players, such as histone deacetylase 2 (HDAC2) and SUV39H1, in a CREB and peroxisome proliferator-activated receptor-gamma (PPARγ) signaling-dependent manner.32 Subsequently, the activation of HDAC2 and SUV39H1 caused by RA ultimately weakened MKP-1 signaling, concomitantly with stimulation of the extracellular signal-regulated kinase 1/2 (ERK1/2), p38, and c-Jun N-terminal kinase (JNK) cascades. Anxiogenic effects associated with exposure to HIV protein tat and repetitive restrain stress were observed to be significantly attenuated by RA, possibly via normalization of HPA axis signaling, and stimulation of BDNF signaling.45 Similarly, the ability of RA to normalize BDNF signaling in rodent models of depression associated with LPS-induced neuroinflammation has been implicated in attenuation of neuronal damage, normalization of hyperactivated inflammatory responses, activation of NRF-2 antioxidant signaling, stimulation of mitochondrial bioenergetic functions, and correction of autophagic imbalances.29 Hippocampal BDNF and ERK1/2 signaling cascades have also been evidenced to the chief targets of RA-mediated repression of depressive behavior in CUS-exposed Sprague-Dawley rats.39

In a chemically induced rodent model of AD, RA was shown to harbor anxiolytic and memory-enhancing activities, possibly because of its ability to stimulate hippocampal neurogenic and synaptogenic pathways, as assessed by the expressions of marker of proliferation Kiel 67 (Ki67), neuronal nuclear protein (NeuN) and doublecortin (DCX); and synaptophysin, postsynaptic density protein-95 (PSD-95), and synapsin I–III, respectively.22 Similarly, the antidepressant actions of RA in a rodent model of forced swim-induced depression were proposed to be reliant on hippocampal dentate gyrus neurogenesis, as assessed by significant increases in BrdU-positive cells.38 More recently, stimulation of BNDF/TrkB/CREB- and PI3K/Akt/mTOR-mediated hippocampal neurogenesis and synaptogenesis has been implicated as critical aspect of RA-induced antidepressant actions.36 This study also identified glucocorticoid receptor (GR) signaling, glucocorticoid-inducible kinase 1 (SGK1), heat shock protein 90 (HSP90), and FK506-binding protein 51 (FKBP51) as key targets of RA in preventing neuronal loss in corticosterone-challenged mice. Lataliza et al have additionally implicated PPARγ and cannabinoid receptors CB-1/2 receptor signaling as a target of RA-mediated antidepressant functions in a chemically induced rodent model of neuroinflammation.66 Lastly, using a combinatorial paradigm of multiple sequential stressful events (physical restraint, forced swimming, loss of consciousness induced by exposure to anesthetic diethyl ether, and mild electric foot shock), Nie et al confirmed the potent anxiolytic actions of RA in rats.46 Further, they found that RA robustly stimulated hippocampal neurogenesis in an ERK-1/2 signaling cascade-dependent manner.

RA and Human Psychiatric Health: Evidences from Clinical Studies

RA, either alone or as the major bioactive component of herbal extracts, has been tested for its anxiolytic, antidepressant, and sleep-promoting potential in several studies (Table 2). Lemon balm, which is enriched in RA, is the most widely used plant source for the formulation of oral herbal extracts in human studies. Indeed, a meta-analysis of multiple trials conducted in diverse groups of human subjects has confirmed that supplementation of its extract is associated with robust improvements in depression and anxiety scores.67 Further, dried leaf extract from lemon balm has been shown to improve mood and cognition attributes of young healthy adults.68,69 An extract formulated from lemon balm dried leaves, both as an aqueous beverage and as an added ingredient in yogurt-based drink, has been evidenced to uplift mood and have anxiolytic and cognition-enhancing effects in healthy young adults.70 Supplementation of RA-rich lemon balm leaf extract in emotionally distressed subjects with sleep dysregulation was also found to result in significantly improved psychological, emotional and sleep functions.71 Similar results of elevated mood and sleep attributes in diabetic patients orally supplemented with lemon balm hydroalcoholic extract have been reported by Safari et al.72 Lemon balm extract has also been evaluated for its beneficial effects on psychological and sleep attributes in patients suffering from chronic angina.73 Questionnaire (depression anxiety stress scale; DASS-21 and Pittsburgh sleep quality index; PQSI)-based results indicated robust declines in the depression, anxiety, stress and sleep disturbance scores in subjects supplemented with lemon balm formulation, over the placebo-controls. More recently, lemon balm, as part of a combinatorial nutraceutical formulation, has been tested for its beneficial effects on neuropsychological parameters in Colombian subjects with sleep impairments. Interestingly, no beneficial effects on sleep parameters were observed in subjects supplemented with the formulation when compared to the placebo-controls.74 It is possible that complementation of lemon balm with other ingredients of the nutraceutical concoction results in the loss of sleep-promoting properties. In a recent study of subjects diagnosed with Alzheimer’s disease and mild dementia, Noguchi-Shinohara et al observed improvements in neuropsychiatric scores upon oral supplementation with RA-enriched lemon balm extract; however, no effects on performance in cognitive tests were observed.75 In a subsequent study, the same authors showed that supplementation of the extract prevented cognitive decline in aged adults without hypertension.76

Table 2 Summary of Recent Studies Evaluating the Antidepressant, Anxiolytic and Hypnotic Activities of RA-Containing Herbal Formulations in Human Clinical Trials

Other medicinal plant extracts rich in RA have also been tested in human trials. For instance, supplementation of spearmint extract in aged subjects with memory impairments has been observed to result in attenuation of mood and sleep disturbances, in addition to the beneficial effects on working and spatial memory domains of cognition.79 On the other hand, this extract did not alter the mood, sleep and cognitive (except the attention module) attributes of normal healthy young adults and middle-aged subjects, over the placebo-controls,80 indicating that the extract may not result in any additional changes in subjects with normal sleep and psychological functions . Interestingly, Dabagzadeh et al used RA-containing rosemary extract in university students and provided evidence for its potent beneficial actions on psychological (depressive and anxious behavior), sleep, and cognitive parameters.77 Subsequently, they extended their findings of anti-anxiety, antidepressant, and cognition-stimulating potencies of rosemary extract in subjects diagnosed with major depressive disorder (MDD).78 Lastly, an herbal formulation comprising RA, among other polyphenols, was shown to attenuate sub-clinical sleep disturbances and promote healthy sleep patterns.81

RA, GMB Axis and Psychiatric Health

The resident microbial population is instrumental for the intricate network of communication between the gut and the brain, controlling multiple aspects of CNS functions.27,82 Recent research has confirmed tremendous involvement of the GMB axis in controlling the pathophysiology of psychiatric conditions,83,84 as well as sleep disorders.85,86 Indeed, dysbiosis of the resident microbial populations, and hence alterations in the levels of metabolites derived from them, may predispose individuals to mental health abnormalities such as depression, schizophrenia, and autism spectrum disorder.87 On the other hand, psychological stress-induced variations in HPA axis activity may affect microbial composition.88 Genetic factors add another level of complexity to this bidirectional interaction connecting the GMB axis and the pathogeneses of psychological conditions.89 Recent research has uncovered the involvement of a plethora of cellular mechanisms linked to neurotransmitter, endocrine, metabolic, redox and immune signaling in these intricate connections. Given the multifaceted interactions between the GMB and etiology of psychological conditions, it is not surprising that the GMB axis is increasingly being regarded as a critical therapeutic target for mental health conditions of depression and anxiety.90,91 In this regard, medicinal plants-derived polyphenols, such as RA, have the potential to serve as safe and effective ameliorative agents.92,93 Interestingly, gut microflora may produce catabolic enzymes capable of converting polyphenols such as RA to more active, better-absorbed metabolites, further increasing their therapeutic potential as antidepressants.94 Indeed, gut microbe-mediated metabolism of RA into metabolites such as caffeic acid may also direct enhanced production of short chain fatty acids (SCFAs), regulating pathways of host inflammation and oxidative stress, and eventually enhancing its therapeutic efficiency.95 Several recent reports support the hypothesis that RA influences the GMB in neuropsychiatric conditions (Figure 2). The key aspects of these findings are discussed below.

Figure 2 RA as a therapeutic agent to ameliorate gut microflora dysbiosis. A number of pathways including redox, inflammatory, metabolic and cell survival are targets of RA-gut microbiota interactions.

Abbreviations: PUFA, polyunsaturated fatty acids; ROS, reactive oxygen species; SCFA, short-chain fatty acids; SOD, superoxide dismutase.

RA-rich Rosmarinus officinalis extract mitigates depressive phenotype in a mouse model of chronic restrain stress-induced behavioral changes, as assessed by time spent in the central region of open field maze, and immobility time in tail suspension and forced swim tests.96 Upon metabolomic analyses, it was found that the Rosmarinus officinalis extract significantly enhanced proportions of the species of the Lactobacillus and Firmicutes phyla, while reducing the Bacteroidetes and Proteobacteria content in the gut of the treated stressed mice. The changes in the resident intestinal microflora induced by RA-containing herbal concoction were associated with attenuation of the activities of neuroinflammatory mediators, IL-1β, TNF-α, NF-κB, p65 and IBA-1, as well as activation of neurogenic (BDNF) and cell survival (Akt) pathways in hippocampi.96 Ou et al observed significant attenuation of dysbiosis of the resident microflora upon supplementing diabetic rats with RA at a dose of 30 mg/kg for 8 weeks.97 Specifically, RA increased the quantities of bacterial species from the Actinobacteria, Proteobacteria, Verrucomicrobia and Fusobacteria phyla. Prominent bacteria whose intestinal levels were upregulated and restored by RA are Anaerostipes, Coprococcus and Sutterella, all of which have been shown to be negatively associated with depressive phenotypes in humans.98,99 On the other hand, RA repressed the elevated levels of unfavorable bacteria, Desulfovibrio and Flavonifractor, which are positively associated with depression.100,101 Similarly, a two-week Melissa officinalis extract (containing 2.76 ± 0.05 mg RA/100 mg of dried extract) oral supplementation paradigm in obese mice abolished dysbiosis of gut microflora, specifically resulting in increased expression of Porphyromonadaceae species.102 Of note, intestinal numbers of Porphyromonadaceae have been previously shown to be reduced in association with depressive/anxious behavior induced by “emotional-single prolonged stress”,103 and elevated in response to treatment with antidepressants.104 Another study conducted in high-fat diet-induced obese mice confirmed the therapeutic actions of RA, as a principal component of Thymus serpyllum extract, in mitigating oxidative and inflammatory damage and promoting metabolic and cell survival pathways.105 These physiological alterations occurred concomitantly with significant restoration of resident microbial ecological diversity (as measured by the Bacteroidetes/ Firmicutes ratio) and upregulation of SCFA-producing bacteria. Furthermore, intestinal abundances of Faecalibaculum, Mucispirillum and Rikenella were reduced upon supplementation with Thymus serpyllum extract in these obese mice. Notably, Faecalibaculum spp. have been shown to induce depressive behavior in mice.106 Similarly, Mucispirillum107 and Rikenella108 are thought to positively regulate the development of depressive behavior.

Chemically induced rodent models of colitis have been particularly useful for studying the effects of RA-enriched natural medicinal formulations on the intestinal microflora. For instance, oral supplementation of monofloral honey from Prunella vulgaris, containing RA as the major phenolic compound, was observed to restore gut microbial homeostasis in favor of Firmicutes/Bacteroidetes ratio, particularly increasing the abundances of Lactobacillus bacteria in colitic rats.109 Similarly, rape bee pollen extract has been shown to increase the abundances of Lactobacillus, Prevotella and Akkermansia and Adlercreutzia species, and reduce the amounts of Paraprevotella and Allobaculum in the gastrointestinal tract of colitic mice.110 While probiotic bacteria of Lactobacillus111 and Adlercreutzia112 genera are known to repress depression-like behavior, high relative abundances of Paraprevotella are thought to show positive associations with neuropsychological dysfunctions.113 Similarly, Akkermansia, one of the bacterial species favorably affected by RA-containing rape bee pollen-derived extract supplementation, may positively regulate certain pathophysiology aspects of psychological disorders.114 Further, Prevotella elicits relatively lowered expression in depressive individual, compared to healthy controls. Likewise, fecal expression of Allobaculum is probably associated with attenuation of the depressive phenotype.115

The favorable effects of RA-containing honey on resident microbial populations are supported by results from studies that have assessed the supplementation of RA-enriched plant-based extracts as a therapeutic strategy against colitis. Indeed, the traditional Chinese herbal concoction Mosla chinensis Maxim. cv. Jiangxiangru (JXR), which harbors RA as the major bioactive, was shown to stimulate the expression of gut bacteria belonging to the Bifidobacteriales and Melainabacteria phyla, while repressing the amounts of Bacteroidaceae species.116 Interestingly, a recent study has proposed that reductions in copy numbers of gut Melainabacteria may contribute to the development of depression-like phenotype.107 Zhou et al successfully used RA-containing Thymus vulgaris extract to reduce colitic and inflammatory damage to dextran sulfate sodium (DSS)-challenged mice and reported enhanced proportions of SCFA-producing bacteria belonging to the Blautia, Bacteroides, Romboutsia, and Faecalibaculum phyla.117 On the other hand, the extract repressed gut expression of harmful microflora, Escherichia, Shigella and Muribaculum, which are implicated as pro-inflammatory mediators in the pathophysiology of depression.118,119 Repression of host pro-inflammatory signaling, via enhanced intestinal abundances of Lactobacillus and reduced amounts of Romboutsia, has also been suggested as the therapeutic basis for the anti-colitis effects of RA-containing Dracocephalum moldavica extract in rodents.120 Notably, intestinal expression of Romboutsia is thought to be positively associated with resilience to depression and anxiety.121,122

RA-encapsulated nanoformulations (corresponding to daily administration of 1 and 10 mg/kg RA for 4 days) have been shown to repress the intestinal copy numbers of Firmicutes, Bacteroidetes, Clostridium leptum, and Bacteroides in rats, resulting in increased fecal levels of SCFAs, such as butyrate and reduced levels of poly-unsaturated fatty acids (PUFAs), including omega-3 and 6 fatty acids.123 Interestingly, PUFAs are positively associated with the pathogeneses of depressive symptoms;124 while bacterial production of SCFA butyrate may be antidepressive.125 As part of its remedial actions on oxidative, inflammatory and apoptotic injuries, RA as the major bioactive in the ethanolic extract derived from the leaves of Trichodesma khasianum, has also been shown to attenuate ethanol-induced dysbiosis of the resident microflora and stimulate the gut microbial production of SCFA by increasing the amounts of Muribaculaceae and Ruminococcaceae bacteria in mice.126 Results from a subsequent study from the same lab further provided evidence for the involvement of gut microbial species in the ameliorative effects of Trichodesma khasianum extract in high-fat diet fed obese mice, wherein it stimulated Bacteroidetes/Firmicutes proportions, specifically resulting in increased copy numbers of Lactobacillus, Muribaculaceae and Ruminococcaceae bacteria.127 Interestingly, Ruminococcaceae family of bacteria has been previously shown to be significantly repressed in the intestines of depressive individuals.128

In contrast to the gut microbial-modifying actions of phyto-concoctions, RA alone has been shown to significantly and beneficially alter gut microflora. Hence, in a mouse model of ovalbumin-induced intestinal allergy, supplementation with RA was shown to repress pro-inflammatory signaling, in part by beneficially altering the Firmicutes/Bacteroidetes ratio, specifically changing the abundance of Muribaculaceae, Lactobacillus and Prevotella,129 all of which have been implicated in the pathogenesis of depression.113,130 RA as an individual therapeutic has also been evaluated for its effectiveness in ameliorating DSS-induced inflammatory and colitic injuries in mouse intestinal tissues. The results indicated significant mediation of the gastrointestinal microflora on the remedial actions of RA, involving an increased abundance of probiotics of the Bacteroidaceae, Alistipes and Muribaculaceae phyla. Furthermore, RA supplementation was associated with significant upregulation of gut microflora-mediated SCFA production.131 Of note, Alistipes and Muribaculaceae species have been implicated as possible regulators of host butyrate-producing capacity in the pathophysiology of depression.130,132 Finally, Li et al proposed that the anti-inflammatory, antioxidant, and anti-apoptotic actions of RA in a murine model of inflammatory bowel disease (IBD) may rely on its ability to favorably alter gut microbiota. Specifically, they showed that oral RA supplementation reduced the intestinal amounts of Bifidobacterium pseudolongum, Escherichia coli, and Romboutsia ilealis and increased the quantity of Lactobacillus johnsonii.133 Interestingly, studies indicate that Lactobacillus johnsonii may be associated with improvements in depressive symptoms.134

In conclusion, recent studies suggest that RA, both alone and as a prominent component of natural medicinal preparations, ameliorates resident gut microbial dyshomeostasis and reverses pathophysiological changes in multiple disease states (Figure 3). The mechanisms underlying the therapeutic connections between RA supplementation and microflora involve the regulation of inflammatory, metabolic, oxidative, and cell survival signaling cascades. These pathways form the basis for the therapeutic potential of RA in depression and anxiety. Hence, although few studies have explicitly evaluated the relevance of the intestinal microbiota in RA-mediated antidepressant and anxiolytic functions, it is likely that host-bacterial interactions form a key aspect of RA-based therapeutic regimens. However, this needs to be experimentally tested in future pre-clinical and clinical studies.

Figure 3 Proposed mechanisms linking therapeutic actions of RA and gut microbial dysbiosis in psychological disorders. RA-mediated beneficial effects on the host–microbe interactions in psychological conditions may involve regulation of inter-connected target pathways of redox, immune, endocrine and synaptic signaling.

Abbreviations: PUFAs, polyunsaturated fatty acids; SCFAs, short-chain fatty acids.

Discussion

Two important aspects must be conidered in order to fully establish RA a therapeutic agent; its safety and its bioavailability. RA and plant-based extracts containing RA have been widely recognized for safe administration at physiologically relevant doses. Studies pointed out the negligible toxicity effects of RA-containing plant extracts in rats and mice.135,136 Further, one of the significant studies has reported no toxicity effect upon administration of 500 mg of RA extract from lemon balm (Melissa officinalis) in healthy human subjects. The results from hematological assessment were favorable, indicating no adverse effects on blood and liver as well as kidney functions.137 Similar findings have been reported upon oral administration of lemon balm extract in patients with mild cognitive impairment76 and human subjects suffering with Alzheimer’s disease.75 In concurrence, a placebo-controlled randomized clinical study using RA extract of aqueous spearmint on human subjects with aging-associated memory impairments found no significant changes in different biochemical and hematological toxicity parameters.138 Studies have also confirmed the insignificant deleterious effects of different nanoformulations loaded with RA on multiple parameters indicative of gut microbiome dysbiosis, liver and kidney dysfunction, and hematological, biochemical, genotoxic and cytotoxic, and oxidative damage.123,139 In contrast, minor levels of toxic effects on reproductive and organ functioning, as well as genetic integrity, have been reported for exceedingly high and chronic doses of RA-containing extracts.140 This indicates that the necessity of an in-depth research on the safety and drug interaction aspects of high doses of rosemary and other RA-rich plant extracts.

Given their potent therapeutic potential, several studies have been conducted to identify ways to increase the yield of RA. Novel biotechnological modifications have resulted in high RA yield from hairy root cultures, suspension cultures, undifferentiated cell cultures, and plant metabolic engineering production.5 In this regard, it should be noted that pharmacokinetics and metabolism of RA following its oral ingestion has been described somewhat in greater detail,9,141 compared to most phytochemicals. The majority of absorbed RA is gradually eliminated in the urine after oral consumption.142 In addition, RA may be excreted in the form of its sulfate-conjugated derivatives in urine, as observed in rodents.143 The low bioavailability of orally supplemented RA is a major obstacle,144 however, multiple groups have proposed measures to increase the bioavailability. These include the creation of RA-phospholipid complexes,145 chitosan-based polymer conjugates,146,147 esterification using alkyl chains of varying lengths,148 and co-employment of piperidine to prevent RA glucuronidation.149 More recently, hydrophobic ion pair (HIP) complexes and HIP-loaded lipid nanocapsules have been proposed as effective nanofabrication methods to address the low bioavailability of RA.150 Moreover, solid lipid nanoformulations of RA have been shown not to induce damages associated with renotoxicity, hepatotoxicity, hematotoxicity, genotoxicity, cytotoxicity, and oxidative and metabolic stress.123 With regard to CNS disorders, nanotechnological procedures may represent a suitable strategy for enhancing the brain bioavailability of RA, particularly through the nasal route (reviewed by Fachel et al).151 Nevertheless, further studies are warranted to improve the permeability and absorption of orally supplemented RA and enhance its sustained and efficient delivery to maximally exploit RA’s therapeutic potential in neuropsychiatric conditions.

Conclusion

In conclusion, this review tracks the multifaceted effects of RA associated with psychological disorders with a focus on the GMB axis, supported by evidence from clinical and non-clinical data. To convert these discoveries into practical applications, additional extensive research is necessary to optimize the doses of RA in different medical conditions, address long-term safety concerns, and discern the precise mechanisms of action. Further research is required to determine and regulate the long-term effects of RA on the gut microbiota and brain inflammation to clarify and evaluate the safety and efficacy of RA at different doses. Furthermore, specific biological and neurological mechanisms underlying the beneficial actions of RA on gut microbiota, brain functions, and mental health, in association with other antidepressant treatments and dietary practices, and their potential synergistic effect, should also be discovered to draw appropriate therapeutic possibilities. We hope that the present review will serve as a relevant research platform for further investigation into these undiscerned aspects of the influence of RA on the pathophysiology of psychological disorders.

Abbreviations

AChE, acetylcholinesterase; BDNF, brain-derived neurotrophic factor; CB-1/2, cannabinoid receptors 1/2; CNS, central nervous system; CUS, chronic unpredictable stress; DASS-21, depression anxiety stress scale; DCX, doublecortin; DSS, dextran sulfate sodium; ERK1/2, extracellular signal-regulated kinase 1/2; FKBP51, FK506-binding protein 51; GAD, glutamic acid decarboxylase; GCLC, glutamate–cysteine ligase catalytic subunit; GMB, gut-microbiome-brain; GPx, glutathione peroxidase; GR, glucocorticoid receptor; HDAC2, histone deacetylase 2; HIP, hydrophobic ion pair; HO-1, heme oxygenase 1; HSP90, heat shock protein 90; IBA-1, ionized calcium-binding adapter molecule 1; IBD, inflammatory bowel disease; IL-10, interleukin 10; iNOS, inducible nitric oxide synthase; JNK, c-Jun N-terminal kinase; Ki67, Kiel 67; LPS, lipopolysaccharide; MAO, monoamine oxidase; MAO-A, monoamine oxidase isoform A; MAO-B, monoamine oxidase isoform B; MDD, major depressive disorder; MKP-1, mitogen-activated protein kinase phosphatase-1; NeuN, neuronal nuclear protein; NQO1, NAD(P)H-quinone oxidoreductase; NRF-2, nuclear factor erythroid 2-related factor 2; PPARγ, peroxisome proliferator-activated receptor-gamma; PQSI, Pittsburgh sleep quality index; PSD-95, postsynaptic density protein-95; PTZ, pentylenetetrazole; PUFA, poly-unsaturated fatty acid; RA, rosmarinic acid; RACK/HIF-1α, receptor for activated C kinase/hypoxia-inducible factor 1 alfa; SCFA, short-chain fatty acid; SGK1, glucocorticoid-inducible kinase 1; SOD, superoxide dismutase; TH, tyrosine hydroxylase; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor alfa.

Acknowledgments

We thank the Science and Engineering Board (SERB), Department of Science and Technology (DST), India, for providing the funds required (grant number SUR/2022/005429) for the completion of this work.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Shin C, Kim YK. Microbiota-gut-brain axis: pathophysiological mechanism in neuropsychiatric disorders. Adv Exp Med Biol. 2023;1411:17–37. doi:10.1007/978-981-19-7376-5_2

2. Din I, Anwar M, Rayees S, Malik F. Traditional plant compounds for the treatment of neuropsychiatric disorders. In: Plant-Derived Bioactives. Springer Singapore; 2020:435–459. doi:10.1007/978-981-15-1761-7_18

3. Grosso C, Santos M, Barroso MF. From plants to psycho-neurology: unravelling the therapeutic benefits of bioactive compounds in brain disorders. Antioxidants. 2023;12(8):1603. doi:10.3390/antiox12081603

4. Mekinic IG, Crmaric M, Skroza D, Burcul F, Blazevic I, Katalinic V. Rosmarinic acid - biologically active compound of Lamiaceae plants. In: Proceedings of the 24th International Scientific-Expert-Conference of Agriculture and Food Industry, Sarajevo, Bosnia and Herzegovina, 2013:255–259.

5. Gonçalves S, Romano A. Rosmarinic acid: sources, properties, applications and biotechnological production. In: Medicinal Plants. CRC Press; 2019:161–171. doi:10.1201/9780429259968-12

6. Ijaz S, Iqbal J, Abbasi BA, et al. Rosmarinic acid and its derivatives: current insights on anticancer potential and other biomedical applications. Biomed Pharmacother. 2023;162:114687. doi:10.1016/j.biopha.2023.114687

7. Noor S, Mohammad T, Rub MA, et al. Biomedical features and therapeutic potential of rosmarinic acid. Arch Pharm Res. 2022;45(4):205–228. doi:10.1007/s12272-022-01378-2

8. Nadeem M, Imran M, Aslam Gondal T, et al. Therapeutic potential of rosmarinic acid: a comprehensive review. Appl Sci. 2019;9(15):3139. doi:10.3390/app9153139

9. Ravaria P, Saxena P, Laksmi BSS, et al. Molecular mechanisms of neuroprotective offerings by rosmarinic acid against neurodegenerative and other CNS pathologies. Phyther Res. 2023;37(5):2119–2143. doi:10.1002/ptr.7825

10. Rahbardar GM, Hosseinzadeh H. Effects of rosmarinic acid on nervous system disorders: an updated review. Naunyn Schmiedebergs Arch Pharmacol. 2020;393(10):1779–1795. doi:10.1007/s00210-020-01935-w

11. Morde A, Acharya M, Tahilani P, Banweer J. Rosmarinic acid - A potential therapeutic agent for sleep disorders. Int J Pharm Sci. 2023;3(1):1–6. doi:10.51626/ijps.2023.03.00010

12. Pogach MS. Impact of psychiatric disorders. Encyclopedia Sleep Circadian Rhythms Elsevier. 2023;529–539. doi:10.1016/B978-0-12-822963-7.00278-4

13. Habtemariam S. Molecular pharmacology of rosmarinic and salvianolic acids: potential seeds for Alzheimer’s and vascular dementia drugs. Int J mol Sci. 2018;19(2):458. doi:10.3390/ijms19020458

14. Yamada M, Ono K, Hamaguchi T, Noguchi-Shinohara M. Natural phenolic compounds as therapeutic and preventive agents for cerebral amyloidosis. Natural Compounds Therapeutic Agents Amyloidogenic Dis. 2015;79–94. doi:10.1007/978-3-319-18365-7_4

15. Dahchour A. Anxiolytic and antidepressive potentials of rosmarinic acid: a review with a focus on antioxidant and anti-inflammatory effects. Pharmacol Res. 2022;184:106421. doi:10.1016/j.phrs.2022.106421

16. Verma H, Shivavedi N, Kumar M, Nayak PK. Pharmacology of rosmarinic acid against psychological disorders. Trad Med Neuronal Health BENTHAM SCIENCE PUBLISHERS. 2023;191–212. doi:10.2174/9789815040197123010013

17. Borgonetti V, Galeotti N. Rosmarinic acid reduces microglia senescence: a novel therapeutic approach for the management of neuropathic pain symptoms. Biomedicines. 2022;10(7):1468. doi:10.3390/biomedicines10071468

18. Borgonetti V, Galeotti N. Microglia senescence is related to neuropathic pain–associated comorbidities in the spared nerve injury model. Pain. 2023;164(5):1106–1117. doi:10.1097/j.pain.0000000000002807

19. Grigoletto J, de OCV, Grauncke ACB, et al. Rosmarinic acid is anticonvulsant against seizures induced by pentylenetetrazol and pilocarpine in mice. Epilepsy Behav. 2016;62:27–34. doi:10.1016/j.yebeh.2016.06.037

20. Coelho VR, Vieira CG, de Souza LP, et al. Behavioral and genotoxic evaluation of rosmarinic and caffeic acid in acute seizure models induced by pentylenetetrazole and pilocarpine in mice. Naunyn Schmiedebergs Arch Pharmacol. 2016;389(11):1195–1203. doi:10.1007/s00210-016-1281-z

21. Malik N, Amber S, Zahid S. Rosmarinus officinalis and methylphenidate exposure improves cognition and depression and regulates anxiety-like behavior in AlCl3-induced mouse model of Alzheimer’s disease. Front Pharmacol. 2022;13:943163. doi:10.3389/fphar.2022.943163

22. Mirza FJ, Amber S, Hassan D, Ahmed T, Zahid S. Rosmarinic acid and ursolic acid alleviate deficits in cognition, synaptic regulation and adult hippocampal neurogenesis in an Aβ1-42-induced mouse model of Alzheimer’s disease. Phytomedicine. 2021;83:153490. doi:10.1016/j.phymed.2021.153490

23. Takeda H, Tsuji M, Matsumiya T, Kubo M. Identification of rosmarinic acid as a novel antidepressive substance in the leaves of Perilla frutescens Britton var. acuta Kudo (Perillae Herba). Nihon Shinkei Seishin Yakurigaku Zasshi. 2002;22(1):15–22.

24. Akkol KE, Gürağaç Dereli FT, Ilhan M. Assessment of antidepressant effect of the aerial parts of Micromeria myrtifolia Boiss. & Hohen on mice. Molecules. 2019;24(10):1869. doi:10.3390/molecules24101869

25. Vilmosh N, Delev D, Kostadinov I, et al. Anxiolytic effect of Satureja Montana dry extract and its active compounds rosmarinic acid and carvacrol in acute stress experimental model. J Integr Neurosci. 2022;21(5):124. doi:10.31083/j.jin2105124

26. Amaghnouje A, Mechchate H, Es-safi I, et al. Subacute assessment of the toxicity and antidepressant-like effects of Origanum Majorana L. polyphenols in Swiss albino mice. Molecules. 2020;25(23):5653. doi:10.3390/molecules25235653

27. Vijay A, Ahmad F, Ravi S, Balakrishnan A. The gut microbiome and the central nervous system (CNS). In: Gut Microbiome and Brain Ageing. Springer Nature Singapore; 2024:43–70. doi:10.1007/978-981-99-8803-7_3

28. Ghazizadeh J, Hamedeyazdan S, Torbati M, et al. Melissa officinalis L. hydro‐alcoholic extract inhibits anxiety and depression through prevention of central oxidative stress and apoptosis. Exp Physiol. 2020;105(4):707–720. doi:10.1113/EP088254

29. Yu Y, Li Y, Qi K, Xu W, Wei Y. Rosmarinic acid relieves LPS-induced sickness and depressive-like behaviors in mice by activating the BDNF/Nrf2 signaling and autophagy pathway. Behav Brain Res. 2022;433:114006. doi:10.1016/j.bbr.2022.114006

30. Verma H, Shivavedi N, GNVC T, Kumar M, Nayak PK. Prophylactic administration of rosmarinic acid ameliorates depression‐associated cardiac abnormalities in Wistar rats: evidence of serotonergic, oxidative, and inflammatory pathways. J Biochem mol Toxicol. 2022;36(10):e23160. doi:10.1002/jbt.23160

31. Verma H, Bhattacharjee A, Shivavedi N, Nayak PK. Evaluation of rosmarinic acid against myocardial infarction in maternally separated rats. Naunyn Schmiedebergs Arch Pharmacol. 2022;395(10):1189–1207. doi:10.1007/s00210-022-02273-9

32. Lee JE, Kwon HJ, Choi J, Han PL. Stress-induced epigenetic changes in hippocampal Mkp-1 promote persistent depressive behaviors. mol Neurobiol. 2019;56(12):8537–8556. doi:10.1007/s12035-019-01689-4

33. Lee JE, Park SY, Han PL. Aging-dependent downregulation of SUV39H1 histone methyltransferase increases susceptibility to stress-induced depressive behavior. mol Neurobiol. 2021;58(12):6427–6442. doi:10.1007/s12035-021-02529-0

34. Kondo S, El Omri A, Han J, Isoda H. Antidepressant-like effects of rosmarinic acid through mitogen-activated protein kinase phosphatase-1 and brain-derived neurotrophic factor modulation. J Funct Foods. 2015;14:758–766. doi:10.1016/j.jff.2015.03.001

35. Sasaki K, El Omri A, Kondo S, Han J, Isoda H. Rosmarinus officinalis polyphenols produce anti-depressant like effect through monoaminergic and cholinergic functions modulation. Behav Brain Res. 2013;238:86–94. doi:10.1016/j.bbr.2012.10.010

36. Zeng J, Xie Z, Chen L, et al. Rosmarinic acid alleviate CORT-induced depressive-like behavior by promoting neurogenesis and regulating BDNF/TrkB/PI3K signaling axis. Biomed Pharmacother. 2024;170:115994. doi:10.1016/j.biopha.2023.115994

37. Jahani R, Behzad S, Saffariha M, Toufan Tabrizi N, Faizi M. Sedative-hypnotic, anxiolytic and possible side effects of Salvia limbata C. A. Mey. Extracts and the effects of phenological stage and altitude on the rosmarinic acid content. J Ethnopharmacol. 2022;282:114630. doi:10.1016/j.jep.2021.114630

38. Ito N, Yabe T, Gamo Y, et al. Rosmarinic acid from Perillae herba produces an antidepressant-like effect in mice through cell proliferation in the hippocampus. Biol Pharm Bull. 2008;31(7):1376–1380. doi:10.1248/bpb.31.1376

39. Jin X, Liu P, Yang F, hong ZY, Miao D. Rosmarinic acid ameliorates depressive-like behaviors in a rat model of CUS and up-regulates BDNF Levels in the hippocampus and hippocampal-derived astrocytes. Neurochem Res. 2013;38(9):1828–1837. doi:10.1007/s11064-013-1088-y

40. Talebi S, Rahmati B, Jorjani M, Emadi F, Ghaffari F, Naseri M. Synergistic effects of Nepeta menthoides and Melissa officinalis aqueous extracts on reserpine‐induced depressive‐like behaviors in mice. Phyther Res. 2022;36(6):2481–2494. doi:10.1002/ptr.7457

41. Lin SH, Chou ML, Chen WC, et al. A medicinal herb, Melissa officinalis L. ameliorates depressive-like behavior of rats in the forced swimming test via regulating the serotonergic neurotransmitter. J Ethnopharmacol. 2015;175:266–272. doi:10.1016/j.jep.2015.09.018

42. Mahmoudian M, Lorigooini Z, Rahimi-Madiseh M, Shabani S, Amini-Khoei H. Protective effects of rosmarinic acid against autistic-like behaviors in a mouse model of maternal separation stress: behavioral and molecular amendments. Naunyn Schmiedebergs Arch Pharmacol. 2024; 397(10):7819–7828. doi:10.1007/s00210-024-03143-2

43. Pereira P, Tysca D, Oliveira P, Dasilvabrum L, Picada J, Ardenghi P. Neurobehavioral and genotoxic aspects of rosmarinic acid. Pharmacol Res. 2005;52(3):199–203. doi:10.1016/j.phrs.2005.03.003

44. yang LD, Wu Y, qi FZ, et al. Rosmarinic acid against cognitive impairment via RACK1/HIF-1α regulated microglial polarization in sepsis-surviving mice. Chem Biol Interact. 2024;388:110830. doi:10.1016/j.cbi.2023.110830

45. Makhathini KB, Mabandla MV, Daniels WMU. Rosmarinic acid reverses the deleterious effects of repetitive stress and tat protein. Behav Brain Res. 2018;353:203–209. doi:10.1016/j.bbr.2018.07.010

46. Nie H, Peng Z, Lao N, et al. Rosmarinic acid ameliorates PTSD-like symptoms in a rat model and promotes cell proliferation in the hippocampus. Prog Neuro Psychopharmacol Biol Psychiatry. 2014;51:16–22. doi:10.1016/j.pnpbp.2014.01.002

47. Cecerska-Heryć E, Polikowska A, Serwin N, et al. Importance of oxidative stress in the pathogenesis, diagnosis, and monitoring of patients with neuropsychiatric disorders, a review. Neurochem Int. 2022;153:105269. doi:10.1016/j.neuint.2021.105269

48. Gavzan H, Araghi A, Marzban Abbasabadi B, Talebpour N, Golshahi H. Antidepressant effects of a Persian herbal formula on mice with chronic unpredictable mild stress. Avicenna J Phytomed. 2023;13(5):562–574. doi:10.22038/AJP.2023.22191

49. Giona L, Musillo C, De Cristofaro G, et al. Western diet-induced cognitive and metabolic dysfunctions in aged mice are prevented by rosmarinic acid in a sex-dependent fashion. Clin Nutr. 2024;43(10):2236–2248. doi:10.1016/j.clnu.2024.08.012

50. Musillo C, Giona L, Ristow M, et al. Rosmarinic acid improves cognitive abilities and glucose metabolism in aged C57Bl/6N mice while disrupting lipid profile in young adults in a sex-dependent fashion. Nutrients. 2023;15(15):3366. doi:10.3390/nu15153366

51. Rampogu S, Park C, Ravinder D, et al. Pharmacotherapeutics and molecular mechanism of phytochemicals in alleviating hormone-responsive breast cancer. Oxid Med Cell Longev. 2019;2019:1–14. doi:10.1155/2019/5189490

52. Zych M, Kaczmarczyk-Sedlak I, Wojnar W, Folwarczna J. Effect of rosmarinic acid on the serum parameters of glucose and lipid metabolism and oxidative stress in estrogen-deficient rats. Nutrients. 2019;11(2):267. doi:10.3390/nu11020267

53. Colica C, Di Renzo L, Aiello V, De Lorenzo A, Abenavoli L. Rosmarinic acid as potential anti-inflammatory agent. Rev Recent Clin Trials. 2018;13(4):240–242. doi:10.2174/157488711304180911095818

54. Machado DG, Bettio LEB, Cunha MP, et al. Antidepressant-like effect of the extract of Rosmarinus officinalis in mice: involvement of the monoaminergic system. Prog Neuro Psychopharmacol Biol Psychiatry. 2009;33(4):642–650. doi:10.1016/j.pnpbp.2009.03.004

55. Takeda H, Tsuji M, Inazu M, Egashira T, Matsumiya T. Rosmarinic acid and caffeic acid produce antidepressive-like effect in the forced swimming test in mice. Eur J Pharmacol. 2002;449(3):261–267. doi:10.1016/S0014-2999(02)02037-X

56. Presti-Silva SM, Herlinger AL, Martins-Silva C, Pires RGW. Biochemical and behavioral effects of rosmarinic acid treatment in an animal model of Parkinson’s disease induced by MPTP. Behav Brain Res. 2023;440:114257. doi:10.1016/j.bbr.2022.114257

57. Hase T, Shishido S, Yamamoto S, et al. Rosmarinic acid suppresses Alzheimer’s disease development by reducing amyloid β aggregation by increasing monoamine secretion. Sci Rep. 2019;9(1):8711. doi:10.1038/s41598-019-45168-1

58. El Omri A, Han J, Yamada P, Kawada K, Abdrabbah MB, Isoda H. Rosmarinus officinalis polyphenols activate cholinergic activities in PC12 cells through phosphorylation of ERK1/2. J Ethnopharmacol. 2010;131(2):451–458. doi:10.1016/j.jep.2010.07.006

59. Ferlemi AV, Katsikoudi A, Kontogianni VG, et al. Rosemary tea consumption results to anxiolytic- and anti-depressant-like behavior of adult male mice and inhibits all cerebral area and liver cholinesterase activity; phytochemical investigation and in silico studies. Chem Biol Interact. 2015;237:47–57. doi:10.1016/j.cbi.2015.04.013

60. Ibarra A, Feuillere N, Roller M, Lesburgere E, Beracochea D. Effects of chronic administration of Melissa officinalis L. extract on anxiety-like reactivity and on circadian and exploratory activities in mice. Phytomedicine. 2010;17(6):397–403. doi:10.1016/j.phymed.2010.01.012

61. Kara M, Sahin S, Rabbani F, et al. An in vitro analysis of an innovative standardized phospholipid carrier-based Melissa officinalis L. extract as a potential neuromodulator for emotional distress and related conditions. Front Mol Biosci. 2024;11:1359177. doi:10.3389/fmolb.2024.1359177

62. Kwon YO, Hong JT, Oh KW. Rosmarinic acid potentiates pentobarbital-induced sleep behaviors and non-rapid eye movement (NREM) sleep through the activation of GABAA-ergic systems. Biomol Ther. 2017;25(2):105–111. doi:10.4062/biomolther.2016.035

63. Wang CC, Hsieh PW, Kuo JR, Wang SJ. Rosmarinic acid, a bioactive phenolic compound, inhibits glutamate release from rat cerebrocortical synaptosomes through GABAA receptor activation. Biomolecules. 2021;11(7):1029. doi:10.3390/biom11071029

64. Kim TH, Bormate KJ, Custodio RJP, et al. Involvement of the adenosine A1 receptor in the hypnotic effect of rosmarinic acid. Biomed Pharmacother. 2022;146:112483. doi:10.1016/j.biopha.2021.112483

65. Shahrestani NV, Haddadi M, Samzadeh Kermani AR. Behavioral and molecular analysis of antioxidative potential of rosmarinic acid against methamphetamine-induced augmentation of Casp3a mRNA in the zebrafish brain. Basic Clin Neurosci J. 2021;12(2):243–254. doi:10.32598/bcn.12.2.1777.1

66. Lataliza AAB, de Assis PM, da Rocha Laurindo L, Gonçalves ECD, Raposo NRB, Dutra RC. Antidepressant‐like effect of rosmarinic acid during LPS-induced neuroinflammatory model: the potential role of cannabinoid receptors/ PPAR‐γ signaling pathway. Phyther Res. 2021;35(12):6974–6989. doi:10.1002/ptr.7318

67. Ghazizadeh J, Sadigh‐Eteghad S, Marx W, et al. The effects of lemon balm (Melissa officinalis L.) on depression and anxiety in clinical trials: a systematic review and meta‐analysis. Phyther Res. 2021;35(12):6690–6705. doi:10.1002/ptr.7252

68. Kennedy DO, Wake G, Savelev S, et al. Modulation of mood and cognitive performance following acute administration of single doses of Melissa officinalis (Lemon Balm) with human CNS nicotinic and muscarinic receptor-binding properties. Neuropsychopharmacology. 2003;28(10):1871–1881. doi:10.1038/sj.npp.1300230

69. Kennedy DO, Scholey AB, Tildesley NTJ, Perry EK, Wesnes KA. Modulation of mood and cognitive performance following acute administration of Melissa officinalis (lemon balm). Pharmacol Biochem Behav. 2002;72(4):953–964. doi:10.1016/s0091-3057(02)00777-3

70. Scholey A, Gibbs A, Neale C, et al. Anti-stress effects of lemon balm-containing foods. Nutrients. 2014;6(11):4805–4821. doi:10.3390/nu6114805

71. Bano A, Hepsomali P, Rabbani F, et al. The possible “calming effect” of subchronic supplementation of a standardised phospholipid carrier-based Melissa officinalis L. extract in healthy adults with emotional distress and poor sleep conditions: results from a prospective, randomised, double-blinded, placebo-controlled clinical trial. Front Pharmacol. 2023;14:1250560. doi:10.3389/fphar.2023.1250560

72. Safari M, Asadi A, Aryaeian N, et al. The effects of Melissa officinalis on depression and anxiety in type 2 diabetes patients with depression: a randomized double-blinded placebo-controlled clinical trial. BMC Complement Med Ther. 2023;23(1):140. doi:10.1186/s12906-023-03978-x

73. Haybar H, Javid AZ, Haghighizadeh MH, Valizadeh E, Mohaghegh SM, Mohammadzadeh A. The effects of Melissa officinalis supplementation on depression, anxiety, stress, and sleep disorder in patients with chronic stable angina. Clin Nutr ESPEN. 2018;26:47–52. doi:10.1016/j.clnesp.2018.04.015

74. Gutiérrez-Romero SA, Torres-Narváez ES, Zamora-Gómez AC, et al. Effect of a nutraceutical combination on sleep quality among people with impaired sleep: a randomised, placebo-controlled trial. Sci Rep. 2024;14(1):8062. doi:10.1038/s41598-024-58661-z

75. Noguchi-Shinohara M, Ono K, Hamaguchi T, et al. Safety and efficacy of Melissa officinalis extract containing rosmarinic acid in the prevention of Alzheimer’s disease progression. Sci Rep. 2020;10(1):18627. doi:10.1038/s41598-020-73729-2

76. Noguchi-Shinohara M, Hamaguchi T, Sakai K, et al. Effects of Melissa officinalis extract containing rosmarinic acid on cognition in older adults without dementia: a randomized controlled trial. J Alzheimer’s Dis. 2023;91(2):805–814. doi:10.3233/JAD-220953

77. Nematolahi P, Mehrabani M, Karami-Mohajeri S, Dabaghzadeh F. Effects of Rosmarinus officinalis L. on memory performance, anxiety, depression, and sleep quality in university students: a randomized clinical trial. Complement Ther Clin Pract. 2018;30:24–28. doi:10.1016/j.ctcp.2017.11.004

78. Azizi S, Mohamadi N, Sharififar F, Dehghannoudeh G, Jahanbakhsh F, Dabaghzadeh F. Rosemary as an adjunctive treatment in patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial. Complement Ther Clin Pract. 2022;49:101685. doi:10.1016/j.ctcp.2022.101685

79. Herrlinger KA, Nieman KM, Sanoshy KD, et al. Spearmint extract improves working memory in men and women with age-associated memory impairment. J Altern Complement Med. 2018;24(1):37–47. doi:10.1089/acm.2016.0379

80. Falcone PH, Nieman KM, Tribby AC, et al. The attention-enhancing effects of spearmint extract supplementation in healthy men and women: a randomized, double-blind, placebo-controlled, parallel trial. Nutr Res. 2019;64:24–38. doi:10.1016/j.nutres.2018.11.012

81. Tubbs AS, Kennedy KER, Alfonso-Miller P, Wills CCA, Grandner MA. A randomized, double-blind, placebo-controlled trial of a polyphenol botanical blend on sleep and daytime functioning. Int J Environ Res Public Health. 2021;18(6):3044. doi:10.3390/ijerph18063044

82. Mitra S, Dash R, Al NA, Habiba SU, Moon IS. Brain modulation by the gut microbiota: from disease to therapy. J Adv Res. 2023;53:153–173. doi:10.1016/j.jare.2022.12.001

83. Afroz KF, Manchia M. Gut microbiome and psychiatric disorders. BMC Psychiatry. 2023;23(1):488. doi:10.1186/s12888-023-05003-4

84. Socała K, Doboszewska U, Szopa A, et al. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. 2021;172:105840. doi:10.1016/j.phrs.2021.105840

85. Wang Z, Wang Z, Lu T, et al. The microbiota-gut-brain axis in sleep disorders. Sleep Med Rev. 2022;65:101691. doi:10.1016/j.smrv.2022.101691

86. Han M, Yuan S, Zhang J. The interplay between sleep and gut microbiota. Brain Res Bull. 2022;180:131–146. doi:10.1016/j.brainresbull.2021.12.016

87. Mohan A, Godugu S, Joshi SS, et al. Gut-brain axis: altered microbiome and depression - review. Ann Med Surg. 2023;85(5):1784–1789. doi:10.1097/MS9.0000000000000573

88. de RS, Garcia Pádua A O, Gonçalves Lima CM, Rodrigues R de O, Bacelar de Assis I, Pagnossa JP. Gut-brain axis as a key aspect of clinical conditions. Int J Fam Community Med. 2022;6(5):285–289. doi:10.15406/ijfcm.2022.06.00291

89. Gong W, Guo P, Li Y, et al. Role of the gut-brain axis in the shared genetic etiology between gastrointestinal tract diseases and psychiatric disorders. JAMA Psychiatry. 2023;80(4):360. doi:10.1001/jamapsychiatry.2022.4974

90. Pferschy-Wenzig EM, Pausan MR, Ardjomand-Woelkart K, et al. Medicinal plants and their impact on the gut microbiome in mental health: a systematic review. Nutrients. 2022;14(10):2111. doi:10.3390/nu14102111

91. Kargbo RB. Microbiome–gut–brain axis modulation: new approaches in treatment of neuropsychological and gastrointestinal functional disorders. ACS Med Chem Lett. 2023;14(6):692–695. doi:10.1021/acsmedchemlett.3c00168

92. Álvarez SA, Rocha-Guzmán NE, González-Laredo RF, Gallegos-Infante JA, Moreno-Jiménez MR, Bravo-Muñoz M. Ancestral food sources rich in polyphenols, their metabolism, and the potential influence of gut microbiota in the management of depression and anxiety. J Agric Food Chem. 2022;70(4):944–956. doi:10.1021/acs.jafc.1c06151

93. Korczak M, Pilecki M, Granica S, Gorczynska A, Pawłowska KA, Piwowarski JP. Phytotherapy of mood disorders in the light of microbiota-gut-brain axis. Phytomedicine. 2023;111:154642. doi:10.1016/j.phymed.2023.154642

94. Zhou N, Gu X, Zhuang T, Xu Y, Yang L, Zhou M. Gut microbiota: a pivotal hub for polyphenols as antidepressants. J Agric Food Chem. 2020;68(22):6007–6020. doi:10.1021/acs.jafc.0c01461

95. Thumann TA, Pferschy-Wenzig EM, Moissl-Eichinger C, Bauer R. The role of gut microbiota for the activity of medicinal plants traditionally used in the European Union for gastrointestinal disorders. J Ethnopharmacol. 2019;245:112153. doi:10.1016/j.jep.2019.112153

96. Guo Y, Xie J, Li X, et al. Antidepressant effects of rosemary extracts associate with anti-inflammatory effect and rebalance of gut microbiota. Front Pharmacol. 2018:9. doi:10.3389/fphar.2018.01126.

97. Ou J, Huang J, Zhao D, Du B, Wang M. Protective effect of rosmarinic acid and carnosic acid against streptozotocin-induced oxidation, glycation, inflammation and microbiota imbalance in diabetic rats. Food Funct. 2018;9(2):851–860. doi:10.1039/C7FO01508A

98. Taylor AM, Thompson SV, Edwards CG, Musaad SMA, Khan NA, Holscher HD. Associations among diet, the gastrointestinal microbiota, and negative emotional states in adults. Nutr Neurosci. 2020;23(12):983–992. doi:10.1080/1028415X.2019.1582578

99. Simpson CA, Diaz-Arteche C, Eliby D, Schwartz OS, Simmons JG, Cowan CSM. The gut microbiota in anxiety and depression – a systematic review. Clin Psychol Rev. 2021;83:101943. doi:10.1016/j.cpr.2020.101943

100. Coello K, Hansen TH, Sørensen N, et al. Affective disorders impact prevalence of Flavonifractor and abundance of Christensenellaceae in gut microbiota. Prog Neuro Psychopharmacol Biol Psychiatry. 2021;110:110300. doi:10.1016/j.pnpbp.2021.110300

101. Sanada K, Nakajima S, Kurokawa S, et al. Gut microbiota and major depressive disorder: a systematic review and meta-analysis. J Affect Disord. 2020;266:1–13. doi:10.1016/j.jad.2020.01.102

102. Brochot A, Azalbert V, Landrier J, Tourniaire F, Serino M. A two-week treatment with plant extracts changes gut microbiota, caecum metabolome, and markers of lipid metabolism in ob/ob mice. mol Nutr Food Res. 2019;63(17):1900403. doi:10.1002/mnfr.201900403

103. Marques LS, Jung JT, Zborowski VA, Pinheiro RC, Nogueira CW, Zeni G. Emotional-single prolonged stress: a promising model to illustrate the gut-brain interaction. Physiol Behav. 2023;260:114070. doi:10.1016/j.physbeh.2022.114070

104. Zhang W, Qu W, Wang H, Yan H. Antidepressants fluoxetine and amitriptyline induce alterations in intestinal microbiota and gut microbiome function in rats exposed to chronic unpredictable mild stress. Transl Psychiatry. 2021;11(1):131. doi:10.1038/s41398-021-01254-5

105. Ruiz-Malagón AJ, Rodríguez-Sojo MJ, Hidalgo-García L, et al. The antioxidant activity of Thymus serpyllum extract protects against the inflammatory state and modulates gut dysbiosis in diet-induced obesity in mice. Antioxidants. 2022;11(6):1073. doi:10.3390/antiox11061073

106. Wang S, Ishima T, Qu Y, et al. Ingestion of Faecalibaculum rodentium causes depression-like phenotypes in resilient Ephx2 knock-out mice: a role of brain–gut–microbiota axis via the subdiaphragmatic vagus nerve. J Affect Disord. 2021;292:565–573. doi:10.1016/j.jad.2021.06.006

107. Sun Y, Pei J, Chen X, et al. The role of the gut microbiota in depressive-like behavior induced by chlorpyrifos in mice. Ecotoxicol Environ Saf. 2023;250:114470. doi:10.1016/j.ecoenv.2022.114470

108. Bai S, Wang W, Wang T, et al. CD36 deficiency affects depressive-like behaviors possibly by modifying gut microbiota and the inflammasome pathway in mice. Transl Psychiatry. 2021;11(1):16. doi:10.1038/s41398-020-01130-8

109. Wang K, Wan Z, Ou A, et al. Monofloral honey from a medical plant, Prunella vulgaris, protected against dextran sulfate sodium-induced ulcerative colitis via modulating gut microbial populations in rats. Food Funct. 2019;10(7):3828–3838. doi:10.1039/C9FO00460B

110. Chen S, Zhao H, Cheng N, Cao W. Rape bee pollen alleviates dextran sulfate sodium (DSS)-induced colitis by neutralizing IL-1β and regulating the gut microbiota in mice. Food Res Int. 2019;122:241–251. doi:10.1016/j.foodres.2019.04.022

111. Barrio C, Arias-Sánchez S, Martín-Monzón I. The gut microbiota-brain axis, psychobiotics and its influence on brain and behaviour: a systematic review. Psychoneuroendocrinology. 2022;137:105640. doi:10.1016/j.psyneuen.2021.105640

112. Zhang X, Chen S, Zhang M, et al. Effects of fermented milk containing Lacticaseibacillus paracasei strain shirota on constipation in patients with depression: a randomized, double-blind, placebo-controlled trial. Nutrients. 2021;13(7):2238. doi:10.3390/nu13072238

113. Barandouzi ZA, Starkweather AR, Henderson WA, Gyamfi A, Cong XS. Altered composition of gut microbiota in depression: a systematic review. Front Psychiatry. 2020;11:541. doi:10.3389/fpsyt.2020.00541

114. Lei W, Cheng Y, Gao J, et al. Akkermansia muciniphila in neuropsychiatric disorders: friend or foe? Front Cell Infect Microbiol. 2023;13:1224155. doi:10.3389/fcimb.2023.1224155

115. Watanabe C, Oyanagi E, Aoki T, et al. Antidepressant properties of voluntary exercise mediated by gut microbiota. Biosci Biotechnol Biochem. 2023;87(11):1407–1419. doi:10.1093/bbb/zbad115

116. Wang X, Cheng K, Liu Z, et al. Bioactive constituents of Mosla chinensis-cv. Jiangxiangru ameliorate inflammation through MAPK signaling pathways and modify intestinal microbiota in DSS-induced colitis mice. Phytomedicine. 2021;93:153804. doi:10.1016/j.phymed.2021.153804

117. Zhou Z, He W, Tian H, Zhan P, Liu J. Thyme (Thymus vulgaris L.) polyphenols ameliorate DSS-induced ulcerative colitis of mice by mitigating intestinal barrier damage, regulating gut microbiota, and suppressing TLR4/NF-κB-NLRP3 inflammasome pathways. Food Funct. 2023;14(2):1113–1132. doi:10.1039/D2FO02523J

118. Liu L, Wang H, Zhang H, et al. Toward a deeper understanding of gut microbiome in depression: the promise of clinical applicability. Adv Sci. 2022;9(35):e2203707. doi:10.1002/advs.202203707

119. Shen CL, Wang R, Santos JM, et al. Ginger alleviates mechanical hypersensitivity and anxio-depressive behavior in rats with diabetic neuropathy through beneficial actions on gut microbiome composition, mitochondria, and neuroimmune cells of colon and spinal cord. Nutr Res. 2024;124:73–84. doi:10.1016/j.nutres.2024.01.014

120. Gang S, Bai W, Yu H, G A, Wang Z. Dracocephalum moldavica L. extract alleviates experimental colitis in rats by modulating gut microbiome and inflammatory pathways. Mol Med Rep. 2023;28(6):228. doi:10.3892/mmr.2023.13115

121. He H, Zhao Z, Xiao C, et al. Gut microbiome promotes mice recovery from stress-induced depression by rescuing hippocampal neurogenesis. Neurobiol Dis. 2024;191:106396. doi:10.1016/j.nbd.2023.106396

122. Zheng S, Zhu Y, Wu W, et al. A correlation study of intestinal microflora and first‐episode depression in Chinese patients and healthy volunteers. Brain Behav. 2021;11(8):e02036. doi:10.1002/brb3.2036

123. Madureira AR, Nunes S, Campos DA, et al. Safety profile of solid lipid nanoparticles loaded with rosmarinic acid for oral use: in vitro and animal approaches. Int J Nanomed. 2016;11:3621–3640. doi:10.2147/IJN.S104623

124. Davyson E, Shen X, Gadd DA, et al. Metabolomic investigation of major depressive disorder identifies a potentially causal association with polyunsaturated fatty acids. Biol Psychiatry. 2023;94(8):630–639. doi:10.1016/j.biopsych.2023.01.027

125. Suda K, Matsuda K. How microbes affect depression: underlying mechanisms via the gut–brain axis and the modulating role of probiotics. Int J mol Sci. 2022;23(3):1172. doi:10.3390/ijms23031172

126. Wang GY, Chen SY, Chen YY, Hong CJ, Hsu YH, Yen GC. Protective effect of rosmarinic acid-rich Trichodesma khasianum Clarke leaves against ethanol-induced gastric mucosal injury in vitro and in vivo. Phytomedicine. 2021;80:153382. doi:10.1016/j.phymed.2020.153382

127. Lai KM, Chen SY, Wang GY, Shahidi F, Yen GC. Protective effect of rosmarinic acid-rich extract from Trichodesma khasianum Clarke against microbiota dysbiosis in high-fat diet-fed obese mice. Food Res Int. 2023;164:112344. doi:10.1016/j.foodres.2022.112344

128. Knuesel T, Mohajeri MH. The role of the gut microbiota in the development and progression of major depressive and bipolar disorder. Nutrients. 2021;14(1):37. doi:10.3390/nu14010037

129. Yang Q, Jia B, Shang J, et al. Effects of rosmarinic acid on immune response and intestinal microbiota in ovalbumin‐induced intestinal allergy mice. J Sci Food Agric. 2024;104(5):3002–3012. doi:10.1002/jsfa.13192

130. Liu X, Li X, Teng T, et al. Comparative analysis of gut microbiota and fecal metabolome features among multiple depressive animal models. J Affect Disord. 2022;314:103–111. doi:10.1016/j.jad.2022.06.088

131. Wang Q, Xu K, Cai X, Wang C, Cao Y, Xiao J. Rosmarinic acid restores colonic mucus secretion in colitis mice by regulating gut microbiota-derived metabolites and the activation of inflammasomes. J Agric Food Chem. 2023;71(11):4571–4585. doi:10.1021/acs.jafc.2c08444

132. Yang Y, Mori M, Wai KM, et al. The association between gut microbiota and depression in the Japanese population. Microorganisms. 2023;11(9):2286. doi:10.3390/microorganisms11092286

133. Li K, Wu J, Xu S, Li X, Zhang Y, Jiao GX. Rosmarinic acid alleviates intestinal inflammatory damage and inhibits endoplasmic reticulum stress and smooth muscle contraction abnormalities in intestinal tissues by regulating gut microbiota. Microbiol Spectr. 2023;11(5):e0191423. doi:10.1128/spectrum.01914-23

134. Jia L, Xiao L, Fu Y, et al. Neuroprotective effects of probiotics on anxiety- and depression-like disorders in stressed mice by modulating tryptophan metabolism and the gut microbiota. Food Funct. 2024;15(6):2895–2905. doi:10.1039/D3FO03897A

135. Lasrado JA, Trinker D, Ceddia MA, Herrlinger KA. The safety of a dry spearmint extract in vitro and in vivo. Regul Toxicol Pharmacol. 2015;71(2):213–224. doi:10.1016/j.yrtph.2014.12.007

136. Calil Brondani J, Reginato FZ, da Silva Brum E, et al. Evaluation of acute and subacute toxicity of hydroethanolic extract of Dolichandra unguis-cati L. leaves in rats. J Ethnopharmacol. 2017;202:147–153. doi:10.1016/j.jep.2017.03.011

137. Noguchi-Shinohara M, Ono K, Hamaguchi T, et al. Pharmacokinetics, safety and tolerability of Melissa officinalis extract which contained rosmarinic acid in healthy individuals: a randomized controlled trial. PLoS One. 2015;10(5):e0126422. doi:10.1371/journal.pone.0126422

138. Nieman KM, Sanoshy KD, Bresciani L, et al. Tolerance, bioavailability, and potential cognitive health implications of a distinct aqueous spearmint extract. Funct Foods Heal Dis. 2015;5(5):165. doi:10.31989/ffhd.v5i5.181

139. Vieira LC, de S MCP, Castro BFM, et al. Rosmarinic acid intravitreal implants: a new therapeutic approach for ocular neovascularization. Planta Med. 2020;86(17):1286–1297. doi:10.1055/a-1223-2525

140. Ghasemzadeh Rahbardar M, Hosseinzadeh H. Toxicity and safety of rosemary (Rosmarinus officinalis): a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol. 2024;2024:1–5. doi:10.1007/s00210-024-03336-9

141. Hitl M, Kladar N, Gavarić N, Božin B. Rosmarinic acid–human pharmacokinetics and health benefits. Planta Med. 2021;87(04):273–282. doi:10.1055/a-1301-8648

142. Baba S, Osakabe N, Natsume M, Terao J. Orally administered rosmarinic acid is present as the conjugated and/or methylated forms in plasma, and is degraded and metabolized to conjugated forms of caffeic acid, ferulic acid and m-coumaric acid. Life Sci. 2004;75(2):165–178. doi:10.1016/j.lfs.2003.11.028

143. Nakazawa T, Ohsawa K. Metabolism of rosmarinic acid in rats. J Nat Prod. 1998;61(8):993–996. doi:10.1021/np980072s

144. Wang J, Li G, Rui T, et al. Pharmacokinetics of rosmarinic acid in rats by LC-MS/MS: absolute bioavailability and dose proportionality. RSC Adv. 2017;7(15):9057–9063. doi:10.1039/C6RA28237G

145. Yang JH, Zhang L, Li JS, et al. Enhanced oral bioavailability and prophylactic effects on oxidative stress and hepatic damage of an oil solution containing a rosmarinic acid–phospholipid complex. J Funct Foods. 2015;19:63–73. doi:10.1016/j.jff.2015.09.013

146. Huerta-Madroñal M, Caro-León J, Espinosa-Cano E, Aguilar MR, Vázquez-Lasa B. Chitosan – rosmarinic acid conjugates with antioxidant, anti-inflammatory and photoprotective properties. Carbohydr Polym. 2021;273:118619. doi:10.1016/j.carbpol.2021.118619

147. Zhang J, Wang L, Tan W, Li Q, Dong F, Guo Z. Preparation of chitosan-rosmarinic acid derivatives with enhanced antioxidant and anti-inflammatory activities. Carbohydr Polym. 2022;296:119943. doi:10.1016/j.carbpol.2022.119943

148. Blažević T, Reznicek G, Ding L, et al. Short Chain (≤C4) Esterification increases bioavailability of rosmarinic acid and its potency to inhibit vascular smooth muscle cell proliferation. Front Pharmacol. 2021:11. doi:10.3389/fphar.2020.609756.

149. Yang JH, Mao KJ, Huang P, Ye YJ, Guo HS, Cai BC. Effect of piperine on the bioavailability and pharmacokinetics of rosmarinic acid in rat plasma using UPLC-MS/MS. Xenobiotica. 2018;48(2):178–185. doi:10.1080/00498254.2017.1292564

150. Gad HA, Ishak RAH, Labib RM, Kamel AO. Ethyl lauroyl arginate-based hydrophobic ion pair complex in lipid nanocapsules: a novel oral delivery approach of rosmarinic acid for enhanced permeability and bioavailability. Int J Pharm. 2023;630:122388. doi:10.1016/j.ijpharm.2022.122388

151. Fachel FNS, Schuh RS, Veras KS, et al. An overview of the neuroprotective potential of rosmarinic acid and its association with nanotechnology-based delivery systems: a novel approach to treating neurodegenerative disorders. Neurochem Int. 2019;122:47–58. doi:10.1016/j.neuint.2018.11.003

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.