African Journal of
Pharmacy and Pharmacology

  • Abbreviation: Afr. J. Pharm. Pharmacol.
  • Language: English
  • ISSN: 1996-0816
  • DOI: 10.5897/AJPP
  • Start Year: 2007
  • Published Articles: 2284

Full Length Research Paper

Anxiolytic properties of Melissa officinalis and associated mechanisms of action: A review of the literature

Barbara Luisa Fermino
  • Barbara Luisa Fermino
  • Docente do Curso de Farmacia, Universidade Estadual do Centro-Oeste, UNICENTRO, Guarapuava, PR, Brasil.
  • Google Scholar
Najeh Maissar Khalil
  • Najeh Maissar Khalil
  • Docente do Curso de Farmacia, Universidade Estadual do Centro-Oeste, UNICENTRO, Guarapuava, PR, Brasil.
  • Google Scholar
Juliana Sartori Bonini*
  • Juliana Sartori Bonini*
  • Docente do Curso de Farmacia, Universidade Estadual do Centro-Oeste, UNICENTRO, Guarapuava, PR, Brasil.
  • Google Scholar
Romaiana Picada Pereira
  • Romaiana Picada Pereira
  • Departamento de Quimica, Programa de Pos-Graduacao em Quimica Aplicada, Universidade Estadual de Ponta Grossa, PR, Brasil.
  • Google Scholar
Joao Batista Teixeira da Rocha
  • Joao Batista Teixeira da Rocha
  • Departamento de Quimica, Programa de Pos-Graduacao em Bioquimica Toxicologica, Universidade Federal de Santa Maria, RS, Brasil.
  • Google Scholar
Weber Claudio Francisco Nunes da Silva
  • Weber Claudio Francisco Nunes da Silva
  • Docente do Curso de Farmacia, Universidade Estadual do Centro-Oeste, UNICENTRO, Guarapuava, PR, Brasil.
  • Google Scholar


  •  Received: 16 September 2014
  •  Published: 22 January 2015

 ABSTRACT

The anxiety disorders prevalence has significantly increased in society. These disorders can be treated with anxiolytics which, despite great efficacy, may result in several adverse side effects. Several studies have reported that anxiolytic effects result from the indirect action on the GABAergic system and mechanisms related to the cholinergic system. Melissa officinalis has been widely utilized for its sedative action and its ability to reduce agitation. Several studies using this plant in different experimental models have demonstrated its low toxicity and lack of side effects. Therefore, this study presents a literature review of the active principles responsible for the anxiolytic effect of M. officinalis and the mechanisms involved in this effect.

Key words: Melissa officinalis, lemon balm, anxiolytic action.


 INTRODUCTION

According to the World Health Organization (WHO), approximately 80% of the world population uses traditional medicine based on empirical knowledge for primary health care (Taiwo, 2007). The use of plants in traditional medicine in Brazil is popular because of the natural diversity observed in the country and low costs the deepening and expansion of studies on herbal medicines have  contributed  to  advances  in  this  usage

(Carvalho, 2011) and discovery of new drugs. Melissa officinalis was first described by Carolus Linnaeus in 1753 and initially cited in the French Pharmacopeain. It belongs to the Lamiaceae family and is a perennial lemon-scented herb in the mint family native to the Mediterranean and Southern Europe popularly known as lemon balm (Guginski, 2007){Guginski, 2007 #17;Guginski, 2007 #10}. It was introduced to North America and can be found currently in gardens androadside fields (Awad et al., 2009). This plant has been widely used because of its several therapeutic actions such as antioxidant (Dastmalchi, 2008; Pereira, 2009; Ribeiro and Bernardo-gil, 2001; Kamdem et al., 2014), anti-inflammatory, hepatoprotective (Birdane, 2007; Bolkent et al., 2005; Encalada et al., 2011), antibacterial, antifungal, antiviral, cholesterol-lowering (Bolkent et al., 2005), antitumor (Saraydin et al., 2012), anti-spas and antidepressant (L´opez et al., 2009).

The plant’s sedative properties (Cases, 2011), including a reduction of stress, agitation, and anxiety (Kennedy, 2006) have been widely explored. It is believed that these properties could be related to the citral, which is one of its most abundant secondary metabolites (Lorenzi and Matos, 2002). Thus, as M. officinalis becomes a promising alternative to the treatment of anxiety, the understanding of factors that alter the bio-synthesis of citral is critical for a safer use of this herbal medicine.

The prevalence of anxiety disorders has significantly increased in the current society, with 10 million people currently suffering with this pathology (WHO, 2002) in Brazil. Anxiety disorders are usually treated through the use of drugs known as benzodiazepines. Benzodiazepines and barbiturates are the most commonly used despite their significant drawbacks such as physical dependence, tolerance, depression, and interference with memory mechanisms (Taiwo, 2007). Therefore, the search for alternative therapies that are as effective as those in use but with reduced adverse effects is of utmost importance (Baldwin and Ajel, 2007; Kennedy and Scholey, 2004; Millan, 2003; Sinclair and Nutt, 2007; Taiwo, 2007).

Traditional medicines are important options to meet the growing needs of health care; however, there is little scientific evidence ensuring their effectiveness and safety. This study reviewed the information published in the literature about the properties of M. officinalis, with the main focus on its anxiolytic roles and the major mechanisms of action involved.


 METHODOLOGY

This study conducted an integrative literature review using articles that addressed the effects related to the anxiolytic properties of M. officinalis between 1994 and 2014 and indexed in the Scopus, Pubmed, Medline, and SciELO databases and SienceDirect. Eight articles that were published in the last eighteen years and addressed the anxiolytic activity of the plant in experimental models and one in a clinical evaluation were compared in this study. 


 RESULTS AND DISCUSSION

M. officinalis L., Lamiaceae, popularly known as melissa or  lemon  balm   is   a    perennial   herbaceous   species originated in Asia, North Africa, and Southern Europe where it is produced in large scale (Gur?ik et al., 2005; Sorensen, 2000). Melissa is reproduced through branch cuttings or imported seeds and plants (Wanderer, 2004). Melissa leaves have been used since ancient times because of its action on the digestive system, mainly due to its carminative and vermifuge properties in the stomach, and as a tonic, antiseptic, and anti-inflammatory (Bertolucci et al., 2008; Sorensen, 2000). Another important use of its leaves and branches is as a condiment (Bertolucci et al., 2008; Carvalho et al., 2005; Couto, 2006).

Citral, citronellal, and geraniol are the main medicinal and condiment constituents in M. officinalis. These constituents are found in the essential oil obtained mainly from leaves that can yield between 0.02 and 0.37% of the majority of metabolites (MoradkhanI et al., 2010); the leaves are also used in infusion to produce teas. Hydroxycinnamic acids such as rosmarinic acid, and polyphenols such as tannins and flavonoids, are other constituents of M. officinalis reported to play important pharmacological roles (MoradkhanI et al., 2010; Sorensen, 2000).

In vitro studies showed that the essential oil from ethanol extracts of M. officinalis leaves contain several metabolites: tannin and rosmarinic derivatives, caffeic acid, flavonoids, and triterpenoid acids. Lorenzi et al. (2002) reported the following compounds as the major components:  citronellal (1), citral (2), followed by β-caryophyllene (3), germancrene- D (4), ocimene (5), and citronellol (6) (Lorenzi and Matos 2002) (Figure 1).

Conversely, Allahverdiyev et al. (2004) showed that the most prevalent compounds are β–Cubebene, β–Caryophyllene (the only common compound between the studies), Sesquiterpene alcohol (C15H26O), α -Cadinol, Geranial (citral a), and Neral (citral b) (Table 1).

 

 

 

 

The different results in these two previous studies could come from the use of different extraction and identification methods; Allahverdiyev et al. (2004) used mass spectrophotometry ensuring greater reliability on the results because of the high sensitivity of this method compared to those using older identification methods. However, compounds other than those cited in the present article, such as rosmarinic acid (Boyadzhiev and Dimitrova, 2007), have been found indicating that a wide range of compounds in this plant could hinder the identification of all compounds in its composition. This could be the limiting factor for pharmacological descriptions in other articles found in the literature. 

According to Lorenzi and Matos (2002), the anxiolytic action of M. officinalis results from the interaction of limonene and citral with GABAA, one of the two ionic channels activated by the ligand responsible for the mediation of γ-aminobutyric acid (GABA), assuming a similar benzodiazepine activity in the plant through nicotinic  and   muscarinic  receptors   that   are  in  direct connection to the central nervous system (CNS). The study of Wake et al. (2000) working with M. officinalis and Valeriana officinallis to treat cholinergic receptors showed that the extract from the plant’s leaves promoted connections at different levels in the two subtypes of these receptors; the level of connections was higher in the nicotinic subtype because the concentration of this receptor in the human occipital cortex is most expressive.

GABA  is an inhibitory neurotransmitter of the central nervous system that reduces nerve impulse transmission between neurons through the hyperpolarization of postsynaptic membranes and the reduction of neuro-transmitter release into the synapse through presynaptic G-protein coupled receptor inhibition of voltage-gated Ca++ mechanisms (Weeks, 2009). The GABAergic system  is well known as a modulator of cognitive function (Lewis et al., 2008; Menzies et al., 2007) and emotional behavior (Ibarra et al., 2010; Radley et al., 2009; Thoeringer et al., 2009). In this regard,  Awad et al. (2009)  have reported that rosmarinic acid in plants works by inhibiting the enzyme GABA transaminase (GABA-T), thereby increasing the levels of the neurotransmitter GABA and consequently, reducing anxiety. However, this would only be possible in a moderate stress state, because M. officinalis is not efficient when the stress level is severe (Ibarra et al., 2010). The GABAA receptors are ionic channels that mediate the effects of GABA, producing an inhibitory action through the opening of chloride channels preventing a neuronal action potential. This is seen as the mechanism of action of diazepam and is regarded as one of the possible mechanisms of action of M. officinalis (Abuhamdah et al., 2008; Akhondzadeh et al., 2003; Kennedy et al., 2002; Wecker and Catalano, 2006).

Wake et al. (2000) reported anxiolytic effects of M. officinalis on the CNS besides the proposed mechanism of specific metabolites connections in the herb, such as limonene and citral on the GABAA neurotransmitter. These effects occur through cholinergic receptors, where muscarinic receptors produce antagonistic effects especially on the M1 receptor. This receptor is located in the nerve ganglia and front-parietal cortex and acts by mediating excitatory postsynaptic potential due to stimulation of intracellular calcium entry (Gerber et al., 2001; López et al., 2009). Wake et al. (2000) analyzed Wistar rats in traditional behavioral models such as the Y-maze, social interaction test, forced swimming, and elevated cross maze and used tea from the leaves of M. officinalis as the testing sample. These authors demonstrated anxiolytic   effects   on   the   CNS  through  the  ingestion of high doses of plant extracts and teas, which did not induce respiratory depression or depressive attenuation at the level of the CNS frame. However, Coimbra (1994) pointed out that the treatment with M. officinalis essential oil in high doses can lead to mutagenic and neurotoxic effects.

Kennedy et al. (2002) used several concentrations of Melissa’s essential oils (0.6, 1.2, and 1.8 g/kg/day) in humans to evaluate the anxiolytic action of M. officinalis, which occurs in the CNS by modulation of mood and cognitive processes. These authors reported that the most effective anxiolytic action was observed with the dose of 1.8 g/kg/day because it acted on the cholinergic system by decreasing stress and agitation in patients, thereby, confirming the proposed anxiolytic effect after ingestion of the plant’s extract.

Wake et al. (2000) indicate that the main mechanism of action of M. officinalis extracts is based on its interaction with cholinergic receptors, the acetylcholine receptor (ACh). It is pointed out that Melissa will act displacing the molecule [3H] - (N)-nicotinic nicotinic, [3H] - (N)-escopolaminica, and muscarinic receptors by increasing ACh released after nerve stimulation in a mechanism that may be involved in improving cognitive function and reducing agitation. This hypothesis is not confirmed, and details of such a mechanism are still unknown.

Nevertheless, in contrast to Kennedy et al. (2002), Abascal and Yarnell (2004) reported in a randomized, double-blind trial control study using placebo with 20 healthy volunteers, that M. officinalis promoted an improvement in attention and stress reduction with a reduced dose (300 mg/kg/day) and decreased alertness and memory loss with an increased dose (900 mg/kg/day). However, the study failed to confirm significant cholinergic action.

Akhondzadeh et al. (2003) administered M. officinalis leaf extracts (600 mg/kg/day) to a group of Alzheimer's disease patients at mild to moderate stages of the disease. The study demonstrated that this treatment during  16  weeks   resulted   in   a   significant   cognitive improvement and reduction in the agitation experienced by some patients with this disease. This study demonstrates a possible effective treatment of Alzheimer's disease with M. officinalis resulting from modulatory actions on mood and cognitive performance, and on acetylcholine receptors in the CNS, following acute administration.

Nowadays, M. officinalis is not individually used as a pharmacological treatment for any disease. It is widely used in conjunction with another plant such as in Sonhare® whose pharmacogens from Valeriana officinalis L. and M. officinalis L. have therapeutic indications in relieving sleep difficulties, tension, restlessness, and irritability. The Sonhare® has a medi-cation package and insert information sheet that do not inform which part of the plant is used or illustrate self-medication usage. Nevertheless, this is a controlled medication that is an excellent option in the prophilaxis of advanced stress (Moura, 2006).

There are few in-depth studies available reporting on the action in other systems on this subject because they failed to address pharmacological aspects. However, some can be cited such as the digestive system (Simmen et al., 2006; Schemann et al., 2006)  in which the action can be linked to gastrointestinal motility reduction (Bolkent et al., 2005). Moreover, Sadraei et al. (2003) reported anti-spasmodic effects in the ileum due to one of its major components, indicating that M. Officinalis represents a good choice of herbal treatment for spastic episodes in the gastrointestinal system.

This plant can present a protective action in the hepatic system because of the presence of phenolic compounds (Simmen et al., 2006; Schemann et al., 2006). However, a study conducted by Müzell (2006) pointed that M. officinalis displayed hepatotoxic effects after toxicity was induced in mice by Acetaminophen, resulting in inhibition or modulation of the activity of cytochromes P450 conferred by flavonoids present in the plant, and enabling these to increase or reduce the concentrations of various therapeutic drugs in plasma (Hodek et al., 2002). In the same study, Müzell (2006) covered the renal system, seeking the possibility of its protection from the plant’s activity; however, the results were unsatisfactory because the drug toxicity was intensified in this case.

Anti-inflammatory activity is also promoted by phenolic compounds present in the plant, such as flavonoids, that have the ability to inhibit the activity of monooxygenases, lipooxigenases, cyclooxygenases (Svobodová et al., 2003), oxidoredutases, and hydrolases such as the hyaluronate lyase that catalyzes the degradation of hyaluronic acid; inhibitions can be competitive in some cases or allosteric in others (Havsteen, 2002). The rosmarinic acid is among the variety of compounds cited or not by different authors, which has shown anti-inflammatory activity and inhibitory activity to 5-lipoxigense, 3R-hydroxysteroid dehydrogenase, and lipid peroxidation as reported by Nakazawa et al. (1998). This compound features astringent, antioxidant, and anti-inflammatory activity by inhibiting lipooxigenases and cyclooxygenases, antibacterial and antiviral activity, and antimutagenic effect (Pereira et al., 2005; Petersen and Simmonds, 2003).

In addition to the anti-inflammatory activity, the antiviral activity to Herpex simplex can be cited, which was reported for the first time by May and Willuhn (1978). In this context, Schnitzler et al. (2008) observed reduced viral replication activity when using M. officinalis oil in vitro.


 CONCLUSION

The use of traditional medicine presents a significant risk to public health because of the lack of knowledge regarding drug interactions and possible toxic effects. Research studies assessing the clinical efficacy of plants are necessary. Based on this literature review, it was concluded that extracts of M. officinalis have effective anxiolytic activity in reducing stress and physiological disturbances due to its direct interaction with the CNS and the cholinergic and GABAergic systems. Its mechanism of action is still controversial. Some authors suggest it is active on the GABAergic system and others on the cholinergic system. Abascal and Yarnell (2004) report no significant action on the cholinergic system while Wake et al. (2000) indicate that the main mechanism of action of M. officinalis is based on its interaction with cholinergic receptors.


 CONFLICT OF INTEREST

Authors declare that there are no conflicts of interest 



 REFERENCES

 

Abuhamdah S, Liping Huang, MSJE, Howes MJR, Ballard C, Holmes C, Burns, A, Perry EK, Lees PTFG, Chazot PL (2008). Pharmacological profile of an essential oil derived from Melissa officinalis with anti-agitation properties: focus on ligand-gated channels. J. Pharm. Pharmacol. 60:377-384.
crossref
 
Akhondzadeh S, Noroozian M, Mohammadi M, Ohadinia S, Jamshidi A, Khani M (2003). Melissa officinalis extract in the treatment of patients with mild to moderate Alzheimer's disease: a double blind, randomised, placebo controlled trial. J. Neurol. Neurosurg. Psychiatry 74:863-866.
crossref
 
Allahverdiyev A, Duran N, Ozguven M, Koltas S (2004). Antiviral activity of the volatile oils of Melissa officinalis. Against Herpes simplex virus type-2. Phytomedicine 11:657-661.
crossref
 
Awad R, Muhammad A, Durst T, Trudeau VL, Arnason JT (2009). Bioassay-guided Fractionation of Lemon Balm (Melissa officinalis L.) using an in vitro measure of GABA transaminase activity. Phytother. Res. 23(8):1075-1081.
crossref
 
Baldwin DS, Ajel K (2007). Development and evaluation of psychotropic drugs. Psychiatry 6:279-283.
crossref
 
Bertolucci S, Lameira A, Pinto J (2008). Guia das plantas medicinais. In: Lameira, A. O.; Pinto, J. E. B. P., in: Belem (Ed.), Plantas medicinais: do cultivo, manipulacao e uso a recomendacao popular pp. 199-201.
 
Birdane YEA (2007). Anti-inflamatory and nociceptive effects of Melissa officinalis L. in rodents. Rev. Med. Vet.158:75-81.
 
Bolkent S, Yanardag R, Karabulut-Bulan O, Yesilyaprak B (2005). Protective role of Melissa Officinalis L. extract on liver os hyperlipidemic rats: A morphological and biochemical study. J. Ethnopharmacol. 99:391-398.
crossref
 
Carvalho H, Cruz, F, Wiest, J (2005). Atividade antibacteriana em plantas com indicativo etnografico condimentar em Porto Alegre, RS/Brasil. Revista Brasileira de Plantas Medicinais Botucatu 7:25-32.
 
Carvalho NEA (2011). Evaluation of the genotoxic and antigenotoxic potential of Melissa officinalisinmice. Genet. Mol. Biol. 34:290-297.
crossref
 
Cases J, Ibarra A, Feuillère N, Roller M, Sukkar SG (2011). Pilot trial of Melissa officinalis L. leaf extract in the treatment of volunteers suffering from mild-to-moderate anxiety disorders and sleep disturbances. Med. J. Nutr. Metab. 4:211-218.
crossref
 
Coimbra R (1994). Manual de Fitoterapia. 2nd edn. Editora Cejup, Belem, Brazil.
 
Couto MEO (2006). Coleção de plantas medicinais, aromáticas e condimentares, Embrapa Clima Temperado. Documentos p 157.
 
Dastmalchia K, Damien Dormana HJ, Oinonena PP, Darwisd Y, Laaksoa I, Hiltunena R (2008). Chemical composition and in vitro antioxidative activity of a lemon balm (Melissa officinalis L.) extract. Food Sci. Technol. 431:391-400.
 
Encalada MA, Hoyos KM, Rehecho S, Berasategi I, de Ciriano MG, Ansorena D, Astiasarán I, Navarro-Blasco I, Cavero RY, Calvo MI (2011). Anti-proliferative effect of Melissa officinalis on human colon cancer cell line. Plant Foods Hum. Nutr. 66:328-334.
crossref
 
Gerber DJ, Sotnikova TD, Gainetdinov RR, Huang YS, Caron MG, Susumu Tonegawa S (2001). Hiperactivity, elevated dopaminergic transmission, and response to amphetamine in M1 muscarinic acetylcholine receptor deficient. Neurobiol. 98:15312-15317.
 
Guginski G (2007). Análise da atividade farmacológica do extrato etanólico obtido da Melissa officinalis. Programa de pós-graduação em farmacologia Universidade federal de santa catarina.
 
Gurčik Ľ, Dubravska R, Miklovičova J (2005). Economics of the cultivation of Salvia officinalis and Melissa officinalis. Agricural Economics, West Lafayette 51:348-356.
 
Havsteen BH (2002). The biochemistry and medical significance of the flavonoids. Pharmacol. Ther. 96:67-202.
crossref
 
Hodek P, Trefil P, Stiborova M (2002). Flavonoids-potent and versatile biologically active compounds interacting with cytocromes P450. Chem. Biol. Interact.139:1-21.
crossref
 
Ibarra A, Feuillere N, Roller M, Lesburgere E, Beracochea D (2010). Effects of chronic administration of Melissa officinalis L. extract on anxiety-like reactivity and on circadian and exploratory activities in mice. Phytomedicine 17:397-403.
crossref
 
Kamdem JP, Adeniran A, Boligon AA, Klimaczewski CV, Elekofehinti OO, Hassan W, Ibrahim M, Waczuk EP, Meinerz DF, Athayde ML (2013). Antioxidant activity, genotoxicity and cytotoxicity evaluation of lemon balm (Melissa officinalis L.) ethanolic extract: Its potential role in neuroprotection. Ind. Crops Prod.51:26-34
crossref
 
Kennedy D (2006). Anxiolytic effects of a combination of Melissa officinalis and Valeriana officinalis during laboratory induced stress. Phytother. Res. 20:96-102.
crossref
 
Kennedy DW, Scholey AB (2004). Attenuation of laboratory-induced stress in humans after acute administration of Melissa officinalis (Lemon Balm). Psychosom. Med. 66:607-613.
crossref
 
Kennedy DO, Scholey AB, Tildesley NT, Perry EK, Wesnes KA (2002). Modulation of mood and cognitive performance following acute administration of single doses of Melissa officinalis (Lemon Balm). Pharmacol. Biochem. Behav.72:953-964.
crossref
 
Lewis DA, Cho RY, Carter CS, Eklund K, Forster S, Kelly MA, Montrose D (2008). Subunit selective modulation of GABA type A receptor neurotransmission and cognition in schizophrenia. Am. J. Psychiatry 165:1585-1593.
crossref
 
López VC, MartıS, Go´mez-Serranillos MP, Carretero, ME Ja¨ger, AK, Calvo MI (2009). Neuroprotective and Neurological Properties of Melissa officinalis. Neurochem. Res. 34.
crossref
 
Lorenzi H, Matos F (2002). Plantas medicinais no Brasil: nativas e exóticas.
 
May G, Willuhn G (1978). Antiviral effect of aqueous plant extracts in tissue culture [in German; English abstract]. Arzneimittelforschung 28:1-7.
Pubmed
 
Menzies L, Ooi C, Kamath S, Suckling J, Mc Kenna P, Fletcher P, Bullmore E, Stephenson C (2007). Effects of gamma-aminobutyric acid modulating drugs on working memory and brain fucntion in patients with schizophrenia. Arch. Gen. Psychiatry 64:156-167.
crossref
 
Millan M (2003). The neurobiology and control of anxious states. Prog. Neurobiol.70:83-244.
crossref
 
Moradkhani H, Sargsyan E, Bibak H, Naseri B, Sadat-Hosseini M, Fayazi-Barjin A, Meftahizade H (2010). Melissa officinalis L., a valuable medicine plant: A review. J. Med. Plants Res. 4:2753-2759.
 
Moura LC (2006). Fitoterapia aplicada em Farmácias: Estudo dos Fitoterápicos Ansiolíticos, Sedativos e Antidepressivos, Farmácia. Escola de Saúde Pública do Ceará., Especialização em assistência farmacêutica.
 
Müzell DP (2006). Preopriedades Biológicas de Extratos de Melissa officinalis L. (Lamiaceae) em Ratos Wistar, Biociências. PUC-RS. Especialização em Biologia Molecular.
 
Nakazawa T, Ohsawa K (1998). Metabolism of Rosmarinic Acid in Rats. J. Nat. Prod. 61:993-996.
crossref
 
Pereira RP, Fachinetto R, de Souza Prestes A, Puntel RL, Santos da Silva GN, Heinzmann BM, Boschetti TK, Athayde ML, Bürger ME, Morel AF, Morsch VM, Rocha JB (2009). Antioxidant effects of different extracts from Melissa officinalis, Matricariarecutita and Cymbopogoncitratus. Neurochem. Res.34:973-983.
crossref
 
Pereira P, Tysca D, Oliveira P, Brum LFS, Picada JN, Ardenghi (2005). Neurobehavioral and genotoxic aspects of rosmarinic acid. Pharmacol. Res. 52:199-203.
crossref
 
Petersen M, Simmonds MSJ (2003). Rosmarinic acid. Phytochemistry 62:121-25.
crossref
 
Radley JJ, Gosselin KL, Sawchenko PE (2009). A discrete GABAergic-relay mediates medial prefrontal cortical inhibition of the neuroendocrine stress response. J. Neurosci. 29:7330-7340.
crossref
 
Ribeiro M, Bernardo-gil MG (2001). Melissa officinalis, L.: study of antioxidant activity in supercritical residues. J. Supercrit. Fluids 21:51-60.
crossref
 
Saraydin SU, Tuncer E, Tepe B, Karadayi S, Özer H, Åžen M, Karadayi K, Inan D, Elagöz Åž, Polat Z, Duman M, Turan M (2012). Antitumoral effects of Melissa officinalis on breast cancer in vitro and in vivo. Asian Pac. J. Cancer Prev.13:2765-2770.
crossref
 
Schemann M, Michel K, Zeller F, Hohenester B, Rühl A (2006). Region-specific effects of STW 5 (Iberogast) and its components in gastric fundus, corpus and antrum. Phytomedicine 13:90-99.
crossref
 
Schnitzler P, Schuhmacher A, Astani A, Reichling J (2008). Melissa officinalis oil affects infectivity of enveloped herpes viruses. Phytomedicine 15 :734-740.
crossref
 
Simmen U, Kelber O, Okpaneji SN, Jaeggi R, Buetler B, Weiser D (2006). Binding of STW 5 (Iberogast) and its components to intestinal 5-HT, muscarinic M3 and opioid receptors. Phytomedicine 13:51-55.
crossref
 
Sinclair L, Nutt D (2007). Anxiolytics. Psychiatry 6:284-288.
crossref
 
Sorensen J (2000). Melissa officinalis – essential oil – authenticity, production and pharmacological activity – a review. Int. J. Aromather.10:7-15.
crossref
 
Svobodová A, Psotová J, Walterová D (2003). Natural phenolics in the prevention of UV-induced skin damage. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 147(2):137-145.
crossref
 
Taiwo A (2007). Alterações Comportamentais decorrentes da administração de Mellissa officinali sem ratos, Faculdade de Ciência da Saúde. Universidade de Brasília.
 
Thoeringer C, Erhardt A, Sillaber I, Mueller M, Ohl F, Holsboer F, Keck M (2009). Long-term anxiolytic and antidepressant-like behavioural effects of tiagabine, a selective GABA transporter-1 (GAT-1) inhibitor, coincide with a decrease in HPA system activity in C57BL/6mice. J. Psychopharmacol. 24:733-743.
crossref
 
Wake G, Court J, Pickering A, Lewis R, Wilkins R, Perry E (2000). CNS acetylcholine receptor activity in European medicinal plants traditionally used to improve failing memory. J. Ethnopharmacol. 69:105-114.
crossref
 
Wanderer M (2004). Produção de mudas e rendimento de biomassa de melissa (Melissa officinalis L.) sob diferentes espaçamentos de plantas e cobertura de solos., Mestrado em Fitotecnia. Universidade Federal do Rio Grande do Sul. p 122.
 
Wecker L, Catalano G (2006). Tratamento da ansiedade e dos distúrbios do sono: In: Brody Farmacologia Humana. Elsevier 285-286.
 
Weeks BS (2009). Formulations of dietary supplements and herbal extracts for relaxation and anxiolytic action. Relarian. Med. Sci. Monit. 15:256-262.
 
WHO (2002). Monographs on selected medicinal plants. WHO Graphics 2, 351.

 




          */?>