ArticlePDF Available

Abstract

Melissa officinalis (lemon balm) which belongs to the Lamiaceae family is a known herb that has long been used in traditional medicine to treat many disorders, and several studies have been conducted to identify the plant and its healing properties. The purpose of this article is to review a series of studies on the effects of the herb extract on the function of various body organs. Due to its volatile organic compounds and active constituents such as terpenoids, flavonoids, quercetin, rutin, quercitrin, gallic acid and high antioxidant capacity, the extract of the plant can have a significant role in maintaining health and curing diseases. In this paper data have been collected from books and scientific papers published in the databases like Science Direct, Web Science, Scopus, EBSCO, Iran medex and PubMed. Search in the interval of the years 2006 to 2012 was carried out. To search for the key words Melissa officinalis, lemon balm, and balm mint were used. About 80 articles was reviewed and after rejection non-related or similar items, in the end about 50 the number was actually cited. Results indicate that the extract of the plant with its several antioxidant, anti- inflammatory, anti-pain, anti-spasmodic and anti-cancer properties as well as its cholinergic receptor activation have highly significant effects on improving behavioral symptoms, cognitive impairment, insomnia, anxiety, and stress. However, these studies have been vast and sparse, and doing more focused and extensive researches in this area is recommended.
1
Zahedan Journal of Research in Medical Sciences
Journal homepage: www.zjrms.ir
A Brief Overview of the Effects of Melissa officinalis L. Extract on the Function of
Various Body Organs
Ali Zarei,
1
Saeed Changizi-Ashtiyani,*
2
Soheila Taheri,
3
Nasser Hosseini
4
1. Young Researchers Club, Abadeh Branch, Islamic Azad University, Abadeh, Iran
2. Department of Physiology, Arak University of Medical Sciences, Arak, Iran
3. Education Development Center, Arak University of Medical Sciences, Arak, Iran
4. Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak, Iran
Article information
Abstract
Article history:
Received: 17 Feb 2014.
Accepted: 12 Mar 2014
Available online: 2 Aug 2014
ZJRMS 2015 July; 17(7): 1-6
Melissa officinalis (lemon balm) which belongs to the Lamiaceae family is a known herb
that has long been used in traditional medicine to treat many disorders, and several studies
have been conducted to identify the plant and its healing properties. The purpose of this
article is to review a series of studies on the effects of the herb extract on the function of
various body organs. Due to its volatile organic compounds and active constituents such as
terpenoids, flavonoids, quercetin, rutin, quercitrin, gallic acid and high antioxidant
capacity, the extract of the plant can have a significant role in maintaining health and
curing diseases.
In this paper data have been collected from books and scientific papers published in the
databases like Science Direct, Web Science, Scopus, EBSCO, Iran medex and PubMed.
Search in the interval of the years 2006 to 2012 was carried out. To search for the key
words Melissa officinalis, lemon balm, and balm mint were used. About 80 articles was
reviewed and after rejection non-related or similar items, in the end about 50 the number
was actually cited. Results indicate that the extract of the plant with its several antioxidant,
anti- inflammatory, anti-pain, anti-spasmodic and anti-cancer properties as well as its
cholinergic receptor activation have highly significant effects on improving behavioral
symptoms, cognitive impairment, insomnia, anxiety, and stress. However, these studies
have been vast and sparse, and doing more focused and extensive researches in this area is
recommended.
Copyright © 2015 Zahedan University of Medical Sciences. All rights reserved.
Keywords:
Blood lipids
Liver
Antioxidants
Thyroid
Melissa officinalis
*Corresponding author at:
Department of Physiology, Arak
University of Medical Sciences,
Arak, Iran.
E- mail: dr.ashtiyani@arakmu.ac.ir
Introduction
edicinal plants have had a great role in
providing health and treatment as well as
disease prevention in human communities [1,
2]. Medicinal herbs are very rich in secondary metabolites
which have profound physiological effects on the function
of mammalian tissues in health and disease conditions [3,
4]. Melissa officinalis (lemon balm) is one of these known
herbs that has been used since a very long time ago for the
treatment of many illnesses like headaches,
gastrointestinal diseases, neurological diseases and
rheumatoid [5, 6]. M. officinalis from Lamiaceae family,
(Fig. 1), with other common names like bee balm, garden
balm, melissa, melissengeist, is a perennial herbaceous
plant which grows vastly from the central and southern
Europe to Iran and central Asia. It is also cultivated
worldwide for its edible properties [7-9]. This herb has
been used extensively in traditional medicine and the
history of it goes back to more than 2,000 years ago. The
plant has been used in a variety of ways from a sedative
and mild hypnotic drug [8-10], and reducing the heart
rate, antibacterial, antiinflammatory, antivirus,
antispasmodic, antioxidant, to a neurotherapeutic agent,
peripheral analgesic, as well as a binding agent to
cholinergic receptors [7, 11].
Avicenna, the Iranian eminent physician and
philosopher introduced this plant as vitality tonic and as a
sedative drug to treat neurological disorders [11].
Figure 1. Aerial parts of Melissa officinalis
The most important ingredients in the plant are known to
be phenolic compounds, rosmarinic acid, caffeic acid,
cholinergic acid, metrilic acid; flavonoids such as
luteolin, apigenin and monoterpene derivatives; the
sesquiterpenes including beta-caryophyllene and
M
Zahedan J Res Med Sci 2015 July; 17(7): 1-6
2
germacrene; triterpenes such as oleanolic and ursolic acid;
volatile oil, and tannins [8]. The aim of present article is
to give a brief overview of the health benefits of M.
officinalis extract and is concentrated on the functions of
liver, thyroid, cancer, immune system, adipose tissue and
also plasma concentrations of some biochemical factors.
Materials and Methods
This review article is a survey of the empirical papers of
several authors who have examined the role of M.
officinalis extract in different body tissues. To conduct a
literature review, the authentic english sources in different
databases like Science Direct, PubMed, Google Scholar,
and Persian databases including: Scientific Information
Database (SID), Iran Articles Database of Medical
Sciences, Iran medex, Iranian Research Institute for
Information Science and Technology (Iran Doc), the
publications database (Magiran) and library archives, as
well as articles published in scientific journals, Persian
and English, have been used. Search in the interval of the
years 2006 to 2012 was carried out. To search for the key
words M. officinalis, lemon balm, and balm mint were
used. About 80 articles was reviewed and after rejection
non-related or similar items, in the end about 50 the
number was actually cited.
Results
The effect of M. officinalis extract on lipid profiles:
Hypertriglyceridemia is one of the most important factors
in the development of cardiovascular diseases [5]. Among
effective pharmacological agents in treating these
diseases, we can refer to statins, clofibrates and niacin.
However, in general these compounds are not effective in
reducing cholesterol concentration and are often
associated with adverse effects and drug toleration, [12-
19]. Therefore, all efforts have been concentrated on
prevention strategies like phytochemical diet.
According to recent studies, M. officinalis essential oil
has significant metabolic effects in vivo. Terpenoids are
among the essential compounds in herbal oils that induce
hypolipidemic effects by inhibiting liver biosynthesis and
formation of cholesterol nucleus in bile. Studies have
shown that the essential oil compounds of Plantago
asiatica inhibit mRNA and protein expression of HMG-
COA reductase in HePG
2
cells and C57BL/6 cells in rats
[19].
Although the lipid-lowering mechanism of M. officinalis
is not clearly known, studies have shown that regular and
daily drinking of M. officinalis tea may improve the
metabolic parameters like cholesterol and triglycerides in
humans [20]. In addition, M. officinalis has the potential
to inhibit hypercholesterolemia, to reduce serum lipid
concentrations and lipid peroxidation in the liver of
hypercholesterolemic rats [21]. Evidence show that herbal
oils, including M. officinalis oils have various
pharmacological effects which are mainly related to
volatile terpenoids such as geranial, cineol and caffeic
acid [5].
In a study on rats with hypercholesterolaemia, Changizi-
Ashtiyani et al. have shown that M. officinalis and
barberry extract can reduce serum cholesterol, low-
density lipoprotein (LDL) and triglyceride. However, the
hypolipidemic properties of alcoholic extract of M.
officinalis are more than those of barberry root which are
most likely related to the antioxidant properties of M.
officinalis and its effect on increasing thyroid hormone
[22].
Jun et al. in a study on the impact of M. officinalis
extract on reducing plasma triglyceride levels has shown
that this is due to the presence of quercetin compounds in
the plant and its possible effect on the inhibition of lipid
peroxidation [5].
M. officinalis essential oil also has anti diabetic
properties and improves glucose tolerance and adjusts the
expression of the genes involved in hepatic
gluconeogenesis studies by Chang et al. have shown that
daily uptake of M. officinalis at low doses can cause
hypoglycemia by increasing glucose uptake and its
metabolism in the liver, as well as by gluconeogenesis
inhibition [23].
Study of Zarei et al. on the effect of the M. officinalis
extract on the activity of liver enzymes in
hypercholesterolemic rats showed that the enzymes level
in hypercholesterolemic group receiving the extract
reduced [24]. Various reasons have been proposed for the
increase in liver enzymes in hypercholesterolemic rats.
For example one theory says that the accumulation of
lipids in the liver raises a pathological state causing
fibrous changes and eventually cirrhosis and hepatic
dysfunction. Following these disorders, levels of liver
enzymes, particularly ALT increases [25]. On the other
hand, hyperlipidemia will also stimulate the production of
free radicals [26].
The impact of M. officinalis extract on reducing liver
enzymes is known to be due to its powerful antioxidant
properties. This plant contains phenolic compounds,
which are among the most important antioxidant agents.
These compounds, especially flavonoids have a
protective effect on liver against damages caused by free
radicals because of their inhibitory effect on the
cytochrome system. Flavonoids can also protect the cells
against glutathione depletion by increasing the capacity of
the antioxidant enzymes (glutathione reductase, oxidase
and catalase) [24].
In another study, Bolkent et al. examined the effect of
M. officinalis extract on hyperlipidemic rats' liver in
which liver cholesterol, total lipid, lipid peroxidation, and
liver enzymes reduced while the glutathione levels
increased [21].
The effect of M. officinalis extract on the function of
pituitary-thyroid axis: The results of the study done by
Zarei et al. on the impact of M. officinalis extract on the
function of pituitary-thyroid axis in rats with
hypercholesterolemia showed increased thyroid hormone
levels and reduced levels of thyroid stimulating hormone
(TSH) [27]. Initially M. officinalis extract increases the
secretion of TRH and TSH and consequently it increases
the amount of T
3
and T
4
, this increase in T
3
and T
4
can
The effects of Melissa officinalis Zarei A et al.
3
finally reduce TSH level through exerting a negative
feedback effect. Studies show that some neuromodulators
and neurotransmitters control the neurons which secret
TRH hormone in the hypothalamus. Some of these
neurotransmitters, such as catecholamines (epinephrine,
norepinepherine, serotonin and dopamine) have an
increasing role and some, such as interleukin-1 (IL
1
) and
gamma aminobutyric acid (GABA) have a decreasing role
[27, 28].
On one hand, the flavonoid compounds in the plant
preserve and conserve catecholamines by inhibiting the
monoamine oxidase enzyme (MAO) and on the other
hand, by inhibiting the cyclooxygenase enzyme, they
reduce the production of prostaglandins and inflammatory
cytokines in response to inflammatory stimuli. In this way
they can also exert their anti-inflammatory effects [29].
M. officinalis extract has two mechanisms on GABA: one
is its GABAergic property to inhibit GABA and the other
one is its cholinergic property which inhibits
acetylcholinesterase, and enhances the capabilities of
brain cholinergic receptors [27, 30].
According to studies, GABA inhibits and reduces TRH
secretion. So, if GABA is inhibited, TRH will increase
and subsequently the secretion of thyroid hormones will
increase too. Studies also indicate that the relation
between fat and leptin, is direct, but fats and thyroid
hormone levels have an inverse and significant relation.
Since M. officinalis extract can reduce blood lipid levels,
it is most likely that at least part of this effect is exerted
by increasing thyroid hormone [22].
Antioxidant effects of M. officinalis: M. officinalis has
powerful antioxidant effects and these effects probably
are exerted through the rosmarinic acid and the
benzodioxole present in the extract. The antioxidant
effects of these compounds are up to ten times stronger
than the effects of those of vitamins B and C. In this way,
Melissa officinalis, like vitamin C, can moderate the
neurotoxic effects of chemical drugs [7, 31].
In addition, compounds such as acid linoleic acid,
carnosic acid, urosolicacidare are also present in the
extract all of which have antioxidant properties.
M. officinalis compounds are able to bind to
acetylcholine; moreover, they contain an inhibitory effect
on the acetylcholinesterase (AChE) enzyme and thus are
able to improve cognitive functions like memory [32].
Pereira et al. has also shown that the antioxidant activity
of phenolic compounds in the plant extract is mostly
because of rutin, quercitrin, garlic acid, and quercetin.
The highest antioxidant properties of compounds belong
to quercetin and then to garlic acid, quercitrin and rutin
respectively. In this study it has been shown that
M. officinalis extract has a protective function against the
oxidative damage caused by different peroxidative agents.
These agents can cause lipid peroxidation in a number of
ways. Therefore, this herbal extract can inhibit the
production of chemically active species in their early
stages, or later, it may block lipid peroxidation through
various processes. Finally, the researchers state that
M. officinalis extract can prevent neurological diseases
associated with oxidative stress [33].
In another study, Martins et al. have tested the effect of
the M. officinalis extract on oxidative stress induced by
manganese (Mn). Manganese is an essential element for
biological systems, but its increased level may lead to
neurodegenerative diseases. Although the mechanism of
this neurotoxicity is not fully clear, the oxidative stress
has a key role in the development of these diseases. In this
study, it has been shown that that manganese increases the
level of thiobarbituric acid reactive substances (TBARS)
as a marker of oxidative stress in hippocampus and the
striatum. In that study the amount of the marker in
animals treated with the extract had decreased, too [34].
Analgesic and anti-inflammatory effects of Melissa
officinalis: Using herbs as pain relief has a long history in
the world of medicine. One of these plants is
M. officinalis whose analgesic effect is not dose-
dependent. The analgesic effect is likely done by opioid
receptors. Acute analgesic effect of this extract does not
differ much from that of morphine and aspirin. However,
its chronic analgesic properties are less than morphine and
aspirin. So, the peripheral analgesic function of the extract
seems to be less considerable than the central analgesic
mechanism [35].
The anti-nociceptive and anti-inflammatory effects of
M. officinalis are attributed to the rosmarinic acid and
flavonoids and terpenoids present in the extract. Probably
flavonoids have a more effective role by facilitating
prostaglandin synthesis. The analgesic activity of
flavonoids is through moderating opioidergic mechanism
[35, 36]
Drozd and Anuszewska have examined the effect of
M. officinalis extract on the immune system response in
rats. They have found that this effect is comparable to the
effect of levamisole, which is known for its effect on the
immune system. Aqueous extract of M. officinalis is
effective on both blood and cellular responses [37].
M. officinalis oil extract derived from the leaves of the
plant contains nerol (30.44%), citral (27.03%), isopolcule
(22.02%), cariophiline (2.29%), oxide carolyn (1.24%)
and citronella (1.06%). In animal models its clear and
strong anti-inflammatory and analgesic effects has been
shown in comparison to those of a standard analgesic and
anti-inflammatory drug (indomethacin) [38]. A
phytochemical study on M. officinalis suggests that
among its phenolic compounds, rosmarinic acid and galic
acid have the highest and the lowest concentrations
respectively [39].
Anxiolytic effect of M. officinalis extract: Anxiety
disorders are the most common mental disorders with the
prevalence of 10-30 percent. Results of several studies
indicate that the aqueous extract of M. officinalis with the
dose of 5 mg/kg has anxiolytic effect, whereas at higher
doses it has a sedative effect in rats. So, the anxiolytic
effects are dose dependent and may be applied through
opioid receptors [40].
In another study conducted to examine the effect of
traditional herbals on neurological disorders, including
Alzheimer's, epilepsy and depression, it is concluded that
the M. officinalis extract does not have any role in the
serotonin transport; however, it is involved in AChE
Zahedan J Res Med Sci 2015 July; 17(7): 1-6
4
activity. Moreover, M. officinalis extract has a moderate
affinity to gamma-aminobutyric acid receptor (GABA).
So it seems that the anxiolytic properties of M. officinalis
may be due to binding to GABA type receptors [41].
In some European countries, the herb extract is used as
pain reliever and relaxant especially when there is
disruption in the first stage of sleep by some unpleasant
stressful factors [42]. In another study on the effect of the
extract on people with Alzheimer's, it is found that M.
officinalis extract can reduce agitation in Alzheimer's
patients. The administration of the citronella, taken from
the extract, has caused sound sleep and has reduced
muscle tone in people with sleep and neurological
disorders. M. officinalis extract has also resulted in a
significant improvement in insomnia, irritability,
headaches, and heart disease in mentally ill patients [40].
Ibarra et al. study on the effects of chronic administration
of M. officinalis extract on anxiety reactions and circadian
activities has shown that the use of this extract reduces
anxiety like reactivities. Because the herb extract contains
significant amounts of rosmarinic acid, oleanolic acid,
ursolicacid, and triterpenoids, it is most likely that these
compounds inhibit GABA transport activity and increase
GABA level in brain [43, 44].
Akhondzadeh et al. study in which M. officinalis extract
was administrated to patients with mild to moderate
Alzheimer's for 4 months has shown a significant
improvement in their behavioral and cognitive symptoms
and a decrease in anxiety and apprehension. The
incidence of these effects is likely due to the stimulating
function of acetylcholine receptors present in the plant.
Similarly, Perry et al. confirm the healing effects of
M. officinalis on memory disorders which are due to its
cholinergic activity. In this respect, the plant is put in the
same rank as Gingo [45].
The effect of the extract on the nervous system:
Cerebral ischemia by causing metabolic disorders leads to
neuronal death. Researchers in a study on the effect of
M. officinalis on death inducing hypoxia, in cultured
cortical neurons in vitro and in the ischemic hippocampus
in vivo (rat hippocampus) have shown that using the oil
extract significantly protects neurons in hypoxia culture.
M. officinalis extract significantly decreases the activity
of caspase 3 and of TUNEL-positive cells (cells that are
located in the CA1 region of the hippocampus).
M. officinalis also inhibits the production of
malondialdehyde (MDA), and reduces antioxidant
capacity in hippocampus. mRNA levels of
proinflammatory cytokines, TNFX and IL
1
B and hypoxia
inducible factor-1α (HIF-lα) have shown a significant
increase after ischemia. The administration of
M. officinalis extract inhibits the expression of HIF-lα.
Studies have also shown that M. officinalis extract can be
used as a protective agent in several neurologic disorders
associated with cerebral ischemia [46].
Hassanzadeh et al. study suggests that the protective
effect of M. officinalis plant against apoptosis is resulted
from the impact of methylenedioxy-methylamphetamine
(MDMA) on nerve cells, part of which is probably
because of the removal of free radicals and amino oxidase
inhibitors. The in vitro study they conducted on neuronal
cells showed that daily use of M. officinalis extracts
within 3 weeks led to cell proliferation, neuroblast
differentiation and integration and increase of GABA
[47].
M. officinalis effect on Alzheimer's disease, seizures
and epilepsy: Alzheimer's disease is a degenerative
neurological disease which is believed to cause dementia.
Dementia is characterized by a progressive loss of
cognitive powers, which leads to social or occupational
disability [48]. M. officinalis has sedative and relaxing
effects and in its homogenate notably binds to brain from
nicotinic and muscarinic acetylcholine receptors and also
inhibits the effects of acetylcholine esterase enzyme.
Thus, by regulating the cholinergic system it is helpful in
Alzheimer's treatment and adjusting mood and cognitive
processes [7].
Epilepsy includes a group of disorders caused by
abnormal electrical activity in the brain. Epileptic attacks
may appear as seizure, convulsion, or other nervous
disorders (in sensory, cognitive, and affective functions).
The ingredients in M. officinalis plant imitate nicotine
effect in the body and accelerate starting time and
increases the duration of the attack. M. officinalis plant is
effective as a pretreatment in modulating the seizure
symptoms caused by the injection of pentylenetetrazol
(PTZ) in rats [48].
Anti tumor effects of M. officinalis extract: Saraydin
et al. studied the effect of M. officinalis extract on
cytotoxicity of breast cancer cell lines (MCF-7, MDA-
468, and MDA-MBA-231). The results showed that the
extract contained active cytotoxicity against all 3 cancer
cell lines. Protein expression of caspase-7 and (TUNEL)
positive cells in a group of rats treated with the extract
was much more than those of the control group, while Ki-
67 expression had reduced. In addition, in vitro studies
indicated that inhibition of tumor volume in the group
treated with the extract group compared to the control
group of rats reduced by 40%. Finally, we can say that M.
officinalis extracts has an anti tumor potential against
breast cancer [49]. de Carvalho et al. also showed that the
M. officinalis extract had anti mutagenic or antigeotaxis
properties [50]
.
Discussion
M. officinalis plant is one of the oldest and most known
herbaceous aromatic plants, and has been used in different
forms such as oil extracts, aqueous extract, and applying
ointment and compress. There are a variety of active
ingredients in the plant which make its antioxidant,
sedative, neuroprotective, anti-anxiety and hypnotic
properties possible. Its metabolic interventions occur
when the plant extract acts to protect the liver and reduce
the amount of lipid profiles and influence the thyroid
hormone function. The presence of active and effective
antioxidants, especial ability to inhibit the production of
free radicals, as well as cytotoxic and anti-mutagenic
effects, have given a unique feature to the plant. It seems
that this ancient medicinal herb has still enormous
The effects of Melissa officinalis Zarei A et al.
5
potential that lies ahead of keen researchers to conduct a
lot of biological research.
Acknowledgements
The present article summarized and updated the latest
finding about Melissa officinalis. This was done as part of
the research project no: 90-123-12 in Arak University of
Medical Sciences. We would like to appreciate the kind
help we received from research deputy of the university.
Authors’ Contributions
All authors declare that they have no conflict of interest.
Conflict of Interest
The authors declare no conflict of interest.
Funding/Support
Arak University of Medical Sciences.
References
1. Changizi-Ashtiyani S, Najafi H, Jalalvandi S, et al.
Protective effects of Rosa canina L fruit extracts on renal
disturbances induced by reperfusion injury in rats. Iran J
Kidney Dis. 2013; 7(4): 290-8.
2. Changizi-Ashtiyani S, Zohrabi M, Hassanpoor A, et al.
Oral administration of the aqueous extract of
Rosmarinusofficinalis in rats before renal reperfusion
injury. Iran J Kidney Dis. 2013; 7(5): 367-75.
3. Taheri S, Zarei A, Changizi-Ashtiyani S, et al. Evaluation
of the effects of hydroalcoholic extract of Berberis
vulgaris root on the activity of liver enzymes in male
hypercholesterolemic rats. Avicenna J Phytomed. 2012;
2(3): 153.
4. Zarei A, Shariati M, Shekar-Forosh S, et al. [The effect of
Physalisalkekengi extract on the physiologic function of
organ tissues] Persian. Arak Med Univ J. 2012; 15(66):
94-104.
5. Jun HJ, Lee JH, Jia Y, et al. Melissa officinalis essential
oil reduces plasma triglycerides in human apolipoprotein
E2 transgenic mice by inhibiting sterol regulatory
element-binding protein-1c-dependent fatty acid synthesis.
J Nutr. 2012; 142(3): 432-40.
6. Wichtl M. Herbal drugs and phytopharmaceuticals.
Germany: Medpharm Press; 2004.
7. Ghayoor N, Rasouli B, Afsharian M, et al. [The protective
effects of Melissa officinalis leaves usage on learning
disorder induced by lead acetate administration during pre
and postnatal periods in rats] Persian. Arak Med Univ J.
2010; 13(1): 97-104.
8. Rasmussen P. Lemon balm. J Prim Health Care. 2011;
3(2): 165-166.
9. Chen XK, Yang Q, Smith G, et al. Environmental lead
level and pregnancy-induced hypertension. Environ Res.
2006; 100(3): 424-30.
10. NourEddine D, Miloud S, Abdelkader A. Effect of lead
exposure on dopaminergic transmission in the rat brain.
Toxicology. 2005; 207(3): 363-68.
11. Naghibi F, Mosadegh M, Mohammadi-Motamed S, et al.
[Labiatae family in folk medicine in Iran: From
ethnobotany topharmacology] Persian. Iran J Pharm Res.
2005; 4(2): 63-79.
12. Mahmoodi M. [Experimental study to evolution the
pretreatment of Melissa officinalis extract against by
phenyltetrazol in Wistar rats] Persian. J Kerman Univ Med
Sci. 2001; 8(2): 88-94.
13. Shekar-Forosh S, Changizi-Ashtiyani S, Akbarpour B, et
al. [The effect of Physalis alkekengi alcoholic extract
onthyroid hormones concentrations in rats] Persian.
Zahedan J Res Med Sci. 2012; 13(9): 1-7.
14. Stohler R, Keller U, Riesen WF. Effects of simvastatin
and fenofibrate on serum lipoproteins and apolipoproteins
in primary hypercholesterolaemia. Eur J Clin Pharmacol.
1989; 37(2): 199-203.
15.
Drexel H. Statins, fibrates, nicotinic acid, cholesterol
absorption inhibitors, anion-exchange resins, Omega-3
fatty acids: Which drugs for which patients? Fundam Clin
Pharmacol. 2009; 23(6): 687-92.
16. Blane GF. Comparative toxicity and safety profile of
fenofibrate and other fibric acid derivatives. Am J Med.
1987; 83(5B): 26-36.
17. Morgan JM, Capuzzi DM, Guyton JR. New extended-
release niacin (Niaspan): Efficacy, tolerability, and safety
in hypercholesterolemic patients. Am J Cardio. 1998;
82(12): 29-41.
18. Verge`s B. Fenofibrate therapy and cardiovascular
protection in diabetes: recommendations after field. Curr
Opin Lipidol. 2006; 17(16): 653 -8.
19. Chung MJ, Park KW, Kim KH, et al. Asian plantain
(Plantago asiatica) essential oils suppress 3- hydroxy-3-
methyl-glutaryl-co-enzyme A reductase expression in
vitro and in vivo and show hypocholesterolaemic
properties in mice. Br J Nutr. 2008; 99(1): 67-75.
20. Jun HJ, Lee JH, Jia Y, et al. Melissa officinalis essential
oil reduces plasma triglycerides in human apolipoprotein
E2 transgenic mice by inhibiting sterol regulatory
element-binding protein-1C-dependent fatty acid
synthesis. J Nutr. 2012; 142(3): 432-40.
21. Bolkent S, Yanardag R, Karabulut-Bulan O and
Yesilyaprak B. Protective role of Melissa officinalis L.
extract on liver of hyperlipidemic rats: A morphological
and biochemical study. J Ethnopharmacol. 2005; 99(3):
391-8.
22. Changizi-Ashtiyani S, Zarei A, Taheri S, et al. A
comparative study of hypolipidemic activities of the
extracts of Melissa officinalis and Berberis vulgaris in
rats. J Med Plants. 2013; 12(47): 38-47.
23. Chung MJ, Cho SY, Bhuiyan MJ, et al. Anti-diabetic
effects of lemon balm (Melissa officinalis) essential oil on
glucose- and lipid-regulating enzymes in type 2 diabetic
mice. Br J Nutr. 2010; 104(2): 180-8.
24. Zarei A, Changizi-Ashtiyani S, Taheri S, et al.
Comparison between effects of different doses of Melissa
officinalis and atorvastatin on the activity of liver enzymes
in hypercholesterolemia rats. Avicenna J Phytomed. 2013;
4(1): 15-23.
25. Murray RK, Rodwell VW, Bender D, editors.
Harpersillustrated biochemistry. 29
th
ed. USA: McGraw-
Hill Press; 2012: 260-286.
26. Nazari A, Delfan B, Shahsavari G. [The effect of Satureja
khuzestanica on triglyceride, glucose, creatinine and
alkaline phosphatase activity in rat] Persian. J Shahrekord
Univ Med Sci. 2005; 7(2): 1-8.
27. Zarei A, Changizi-Ashtiyani S, Sokhandani M, et al. The
comparison between the effects of the alcoholic extract of
Melissa officinalis and atorvastatin on serum levels of
Zahedan J Res Med Sci 2015 July; 17(7): 1-6
6
thyroid hormones in hypercholesterolemia male rats.
Zahedan J Res Med Sci. 2013; 15(8): 6-12.
28. Gharib-Naseri MK, Handali S, Hoseini H. Antispasmodic
activity of Physalisalkekengi L. leaf hydroalcoholic
extract on rat ileum. J Med Plants. 2007; 23(3): 340-349.
29. Vaez G, Tavasoli Z, Ranjbar-Bahadori S. [Study on the
different dosages of Elaeagnusangustifolia aqueous extract
with and without morphine on the antinociceptive rate in
mice] Persian. Pejouhesh. 2011; 35(1): 27-33.
30. Hossini E, Sadeghi H, Daneshi A. [Evaluation of hydro-
alcoholic extract of peganumharmala on pituitary-thyroid
hormonesin adult male rats] Persian. Yasuj Univ Med Sci.
2010; 14(4): 23-30.
31. Dastmalchi K, Damien-Dorman HJ, Oinonen PP, et al.
Chemical composition and in vitro antioxidative activity
of a lemon balm (Melissa officinalis L.) extract. Food Sci
Technol Bull. 2008; 41(3): 391-400.
32. Rostami S, Momeni Z, Behnam-Rassouli M, et al.
[Comparison of antioxidant effect of Melissa officinalis
leaf and vitamin C in lead acetate induced learning deficits
in rat] Persian. Sci Res J Shahed Univ. 2010; 17(86):47-
54.
33. Pereira RP, Fachinetto R, de Souza-Prestes A, et al.
Antioxidant effects of different extracts from Melissa
officinalis, Matricariarecutita and Cymbopogoncitratus.
Neurochem Res. 2009; 34(5): 973-83.
34. Martins EN, Pessano NT, Leal L, et al. Protective effect of
Melissa officinalis aqueous extract against Mn-induced
oxidative stress in chronically exposed mice. Brain Res
Bull. 2012; 87(1): 74-9.
35. Miladi-Gorgi H, Vafaee A, Rashidipoor A, et al. [The role
of opioid receptors on anxiolytic effects of the aqueous
extract of Melissa officinalis in mice] Persian. Razi J Med
Sci. 2005; 12(47): 145-153.
36. Anjaneyulu M, Chopra K. Quercetin, a bioflavonoid,
attenuates thermal hyperalgesia in a mouse model of
diabetic neuropathic pain. Prog Neuropsychopharmacol
Biol Psychiatry. 2003; 27(6): 1001-1005.
37. Drozd J, Anuszewska E. The effect of the Melissa
officinalis extract on immune response in mice. Acta Pol
Pharm. 2003; 60(6): 467-70.
38. Bounihi A, Hajjaj G, Alnamer R, et al. In vivo potential
anti-inflammatory activity of Melissa officinalis L.
essential oil. Adv Pharmacol Sci. 2013; 2013: 101759.
39. Arceusz A, Wesolowski M. Quality consistency
evaluation of Melissa officinalis L. commercial herbs by
HPLC fingerprint and quantitation of selected phenolic
acids. J Pharm Biomed Anal. 2013; 83: 215-20.
40. Ulbricht C, Brendler T, Gruenwald J, et al. Lemon balm
(Melissa officinalis L): An evidence-based systematic
review by the natural standard research collaboration. J
Herbal Pharmacotherapy. 2005; 5(4): 71-114.
41. Salah SM, Jager AK. Screening of traditionally used
Lebanese herbs for neurological activities. J
Ethnopharmacol. 2005; 97(1): 145-9.
42. Soulimani R, Fleurentin J, Mortijer F, et al. Neurotropic
action of the hydroalcoholic extract of Melissa officinalis
in the mouse. Planta Med. 1991; 57(2): 105-9.
43. Ibarra A, Feuillere N, Roller M, et al. 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.
44. Awad R, Levac D, Cybulska P, et al. Effects of
traditionally used anxiolytic botanicals on enzymes of the
gamma-aminobutyric acid (GABA) system. Can J Physiol
Pharmacol. 2007; 85(9): 933-942.
45. Akhondzadeh S, Noroozian M, Mohammadi M, et al.
Melissa officinalis extract in the treatment of patients with
mild to moderate Alzheimer's disease. J Neurol Neurosurg
Psychiatry. 2003; 74(7): 863-6.
46. Bayat M, Azami-Tameh A, Ghahremani MH, et al.
Neuroprotective properties of Melissa officinalis after
hypoxic-ischemic injury both in vitro and in vivo. Daru.
2012; 20(1): 42.
47. Hassanzadeh G, Pasbakhsh P, Akbari M, et al.
Neuroprotective properties of Melissa officinalis L. extract
against ecstasy-induced neurotoxicity. Cell J. 2011; 13(1):
25-30.
48. Bhat JU, Nizami Q, Aslam M, et al. Antiepileptic activity
of the whole plant extract of Melissa officinalis in Swiss
albino mice. Int J Pharm Sci Res. 2012; 3(3): 886-889.
49. Saraydin SU, Tuncer E, Tepe B, et al. Antitumoral effects
of Melissa officinalis on breast cancer in vitro and in vivo.
Asian Pac J Cancer Prev. 2012; 13(6): 2765-70.
50. de Carvalho NC, Correa-Angeloni MJ, Leffa DD, et al.
Evaluation of the genotoxic and antigenotoxic potential of
Melissa officinalis in mice. Genet Mol Biol. 2011; 34(2):
290-7.
Please cite this article as: Zarei A, Changizi-Ashtiyani S, Taheri S, Hosseini N. A brief overview of the effects of Melissa officinalis L.
extract on the function of various body organs. Zahedan J Res Med Sci. 2015; 17(7): 1-6.
... According to published studies, the antinociceptive & anti inflammatory action of Melissa officinalis extract is attributed to flavonoids and terpenoids, specifically rosmarinic acid (RA). Melissa officinalis (MO) compounds inhibit the cyclooxygenase enzyme, which decreases the production of prostaglandins and inflammatory cytokines in response to inflammatory stimuli, as well as the monoamine oxidase enzyme, which prevents catecholamine degradation (Zarei et al., 2015). The flavonoids quercetin, luteolin, and apigenin, phenolic components of MO with immune-modulating activities can decrease inflammation and may prevent cardiac tissue remodeling in animal models of myocarditis (Milenković et al., 2010;Wu et al., 2020). ...
Article
Full-text available
Melissa officinalis is used for its therapeutic properties all over the world in the prevention of numerous diseases. It has several bioactive compounds viz. flavonoids, terpenoids, phenolic acids, tannins, and essential oil. Triterpenes (ursolic acid and oleanolic acid), phenolic acids (rosmarinic acid, caffeic acid, and chlorogenic acid), flavonoids (quercetin, rhamnocitrin, and luteolin), and volatile chemicals (geranial, neral, citronellal, and geraniol) etc. The biological researches indicate that Melissa officinalis essential oil and extracts include active components that determine a variety of pharmacological actions with potential medical applications. This review sought to summarize Melissa officinal's botanical traits, traditional uses, phytochemicals, and its pharmacological actions Review Article 9 which indicate the effective use of Melissa officinalis as an alternate treatment option for combating numerous disorders due to the presence of vast amount of phytochemical constituents contributing to its various therapeutic potential.
... Aerial Parts of Melissa officinalis L[19]. ...
Article
Full-text available
Melissa officinalis, an herb known for its bioactive compounds, was subjected to aqueous extraction, with the resultant extracts employed for treatment on A549 and BEAS-2B cell lines. Gas chromatography analysis of the aqueous extract revealed prominent constituents, including palmitic acid, stearic acid, linoleic acid, oleic acid, palmitoleic acid, and linolenic acid. Through MTT colorimetric assays, the IC50 values for the aqueous extracts were determined as 32.84 and 58.60 for A549 and BEAS-2B cell lines, respectively, after 48 hours of exposure. Notably, the aqueous extracts exhibited significant growth inhibition on A549 and BEAS-2B cells, with notable effect at higher concentrations (100 µg/mL). To delve deeper, a protein-protein interaction network analysis was conducted to identify pivotal targets governing biological processes and pathways. Subsequently, network pharmacology was employed to delineate the pathways involved in studying anticancer properties in Melissa officinalis Extract. Molecular docking simulations were carried out to estimate the binding affinities between the extracts and the identified hub targets. The analysis pinpointed ALOX5, PTGES, and CYP2C19 as the most promising targets through protein-protein interaction assessment. KEGG pathway analysis corroborated the potential of these compounds to modulate cancer-associated pathways, including large-and small-cell lung cancer. Furthermore, molecular docking simulations underscored the high binding affinities of linolenic acid, oleic acid, and stearic acid against cancer-associated targets, suggesting a promising strategy for treating lung cancer.
... Flavonoids are known for their powerful antioxidant activities and are also present in M extract. 12 Hypericum perforatum, or St. John's wort (H), is another medicinal plant of great importance that has been used for thousands of years. This perennial herb belongs to the Hypericacea family and is native to temperate regions of the world. ...
Article
Full-text available
Objectives This study aimed to formulate a novel herbal mixture of Hypericum perforatum (H) and Melissa officinalis (M) and evaluate its toxicity, microbial load, and phytochemical content. Materials and Methods Total flavonoids were measured using the AlCl3/NaNO2 complex formation method and colorimetric assay. The quercetin content of the herbal mixture was determined by reverse-phase high-performance liquid chromatography. The in vitro and in vivo safety of the herbal formulations were analyzed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and acute oral toxicity analysis in the rat model, respectively. Results The formulated extract (HM), compared with the standard rutin extract, had a total flavonoid content of 15.29 ± 0.64 mg rutin per mL sample. Reverse-phase high-performance liquid chromatography revealed a quercetin content of 0.187 mg/mL. Microbial tests for Escherichia coli, Pseudomonas aeruginosa,Staphylococcus aureus, and Salmonella spp. were negative. Colony counts for total aerobic microbial and yeast and mold counts were 10 in each case. The MTT assay (with up to about 5% v/v HM extract) using the NIH/3T3 cell line revealed no cell toxicity in the range of concentrations tested. Acute oral toxicity was tested in the Wistar rat model, and the LD50 was 695.2 ± 7.5 mg/kg. The dry weight of the HM extract was 38.1 mg/mL. Conclusion Preliminary results proved the safety of the HM herbal mixture, with its toxicity and microbial load within the limits of accepted guidelines allowable for use in clinical trials.
... It can also be used to treat neurosis, nervous excitability and anxiety-and depression in mice exposed to restraint stress [9]. MO is also regarded to be useful for people with Alzheimer's disease and has therapeutic potential in mood and cognitive performance regulation [10]. Furthermore, it was found that MO ameliorated hepatic and renal dysfunction in rats intoxicated with lead [11], liver fibrosis in the non-alcoholic fatty liver disease model [12] as well as its chemo-preventive effect against HCC in rats [13]. ...
Article
Full-text available
Background Melissa officinalis (MO) is a well-known medicinal plant species used in the treatment of several diseases; it is widely used as a vegetable, adding flavour to dishes. This study was designed to evaluate the therapeutic effect of MO Extract against hyperthyroidism induced by Eltroxin and γ-radiation. Methods Hyperthyroidism was induced by injecting rats with Eltroxin (100 µg/kg/ day) for 14 days and exposure to γ-radiation (IR) (5 Gy single dose). The hyperthyroid rats were orally treated with MO extract (75 mg/kg/day) at the beginning of the second week of the Eltroxin injection and continued for another week. The levels of thyroid hormones, liver enzymes and proteins besides the impaired hepatic redox status and antioxidant parameters were measured using commercial kits. The hepatic gene expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its inhibitor Kelch-like ECH-associated protein-1(Keap-1) in addition to hepatic inflammatory mediators including tumor necrosis factor-α (TNF- α), Monocyte chemoattractant protein-1 (MCP-1) and fibrogenic markers such as transforming growth factor-beta1 (TGF-β1) were determined. Results MO Extract reversed the effect of Eltroxin + IR on rats and attenuated the thyroid hormones. Moreover, it alleviated hyperthyroidism-induced hepatic damage by inhibiting the hepatic enzymes’ activities as well as enhancing the production of proteins concomitant with improving cellular redox homeostasis by attenuating the deranged redox balance and modulating the Nrf2/Keap-1 pathway. Additionally, MO Extract alleviated the inflammatory response by suppressing the TNF- α and MCP-1 and prevented hepatic fibrosis via Nrf2-mediated inhibition of the TGF-β1/Smad pathway. Conclusion Accordingly, these results might strengthen the hepatoprotective effect of MO Extract in a rat model of hyperthyroidism by regulating the Nrf-2/ Keap-1 pathway.
... The first difference was the appearance of another α anomeric proton at 4.93 of rhamnose moiety and doublet of its CH3 group at 1.04 which was confirmed by the downfield shift of X-type, doublet of (H-3'5') at 7.04. Based on the information presented above and a comparison to previously published data of isoflavone isolated from Salvia triloba 31 (6), nepetin -7-methyl ether 35 (7), luteolin 36 (8), apigenin 37 (9) together with two phenolic acids; Caffeic acid 38 (10) and Chlorogenic acid 39 (11). They were identified by CoPC against authentic samples. ...
Article
Full-text available
Abstract: Phytochemical investigation of 70% aqueous methanol extract of leaves of lemon balm (Melissa officinalisL., Lamiaceae) was subjected to different chromatographic separation techniques. The structures of isolated compounds were determined based on spectral data (UV, 1H-NMR, 13CNMR, and mass spectrometry). Also, assessment of the antioxidant, anticonvulsant, anti-inflammatory as well as antimicrobial activities was determined using different techniques. Result: three isoflavones;4'- methoxy genistein Biochanin A] (1),6, 8 dimethoxy- biochanin A -7 - O- α - L- rhamnopyranosyl (1'''-6'') - O -  - D- glucopyranoside (2), isoflavone 6, 8dimethoxy 7-O- - D rutinoside, 4'-O- α- L-rhamnopyranoside (3).Besides six known flavonoids; hyperin (4), luteolin 7-O- rhamnoside(5), scutellarein – 4',7 dimethyl ether(6), nepetin - 7- methyl ether(7),luteolin(8),and apigenin (9),in addition to caffeic acid(10), chlorogenic acid (11) isolated for the first time from Melissa officinalis. The examined extract revealed significant anticonvulsant activity, effective in inhibition of both acute & chronic inflammation. Scutellarein – 4',7-dimethyl ether (6) showed remarkable inhibition against fungi and exhibited the highest antibacterial effect against both Gram +ve and Gram -ve bacteria. The above evidence suggests that Melissa officinalis leaves are a good supply of natural isoflavones that can be utilized to avoid convulsion, and inflammation, as well as an antioxidant and antimicrobial supplement.
Article
Background: Alzheimer's disease (AD) is a progressive neurodegenerative condition characterized by the gradual decline of cognitive abilities, primarily caused by impairments in the cholinergic system. AD is diagnosed based on the presence of specific pathological features, in-cluding senile plaques, neurofibrillary tangles, and the loss of neurons and synapses. Despite on-going efforts, the etiology of AD remains unclear, and there is a significant lack of effective treatments to meet the medical needs of affected individuals. The complex nature of AD, involv-ing multiple factors, presents challenges in the development of potential therapies. Numerous ob-stacles hinder the achievement of optimal pharmacological concentration of promising molecules for AD treatment. These obstacles include the presence of the blood-brain barrier (BBB), which restricts the entry of therapeutic agents into the brain, as well as issues related to poor bioavaila-bility and unfavorable pharmacokinetic profiles. Unfortunately, many therapeutically promising compounds have failed to overcome these hurdles and demonstrate efficacy in treating AD. Background Alzheimer’s disease (AD) is a progressive neurodegenerative disease that is manifested by depleted cognitive abilities resulted due to cholinergic impairments. AD is further diagnosed with pathological hallmarks including senile plaques, neurofibrillary tangles and neuronal and synaptic death. With constant efforts, few therapeutic targets and interventions have been identified but AD is still a disease with unclear etiopathology and unmet medical needs. The multifactorial nature of AD poses difficulties to develop a potential treatment. Unfortunately, large numbers of therapeutically efficient molecules for the treatment of AD failed to attain optimal pharmacological concentration due to numerous hurdles such as the presence of blood-brain barrier (BBB), poor bioavailability, or pharmacokinetic profile. Methods: The PEGylated chitosan nanoconjugate was developed and evaluated for delivery of anti-Alzheimer natural extract of Salvia officinalis and Melissa officinalis to the brain. The nano-conjugates (S-PCN and M-PCN) were developed by ionic gelation technique. Result: The nanoconjugates (S-PCN and M-PCN) were evaluated for various optical and in-vitro parameters. MTT assay on UCSD229i-SAD1-1 human astrocytoma cells indicated IC50 values of 0.42, 0.49, 0.67, and 0.75 μM for S-PCN, M-PCN formulations, and free Salvia officinalis and Melissa officinalis extracts, respectively. The In vitro assessments using cell lines have confirmed the improved uptake and distribution of nanoconjugates compared to free extracts. These findings were validated through confocal microscopy and apoptosis assays, revealing a substantial in-crease in the accumulation of nanoconjugates within the brain. The targeting potential OF M- PCN over S-PCN was found to be 2-fold significant. Method 1. Sample Preparation - Crude drug Salvia officinalis and Melissa officinalis, plants were collected from the botanical gardens of Warangal and Tirupathi and authenticated.The two plants, 1 g each, were crushed (using a lab mill) for 1 min, to obtain the corresponding powder. The extraction powder was performed as described in previous reports, via addition of 100 mL boiling water to 1 g of plant powder and after 5 min, the extract was filtered through a 0.45 mm filter. This procedure was optimized to obtain the highest potential activity of these plants. After the crude plant sedimentation, samples were filtered and maintained at 80 ͦ C, for freeze-drying procedures (Heto Holten A/S Drywinner, Allerød, Denmark). Then, solutions of 1% (w/v) of freeze-dried powder were dissolved in methanol for analytical evaluation other activity tests. Before injections, samples were filtered again through a 0.45 mm filter. 2. Preparation of PEGylated Chitosan Nanoparticles - Ionotropic gelation technique was employed for the synthesis of chitosan, encapsulating whole Salvia officinalis and Melissa officinalis extract separately. Accurately weighed 100 mg of Salvia officinalis and Melissa officinalis extract and 0.4% w/v of Chitosan were dissolved in 1% v/v aqueous glacial acetic acid (GAA) solution. Drop wise addition of 0.4% Sodium tripolyphosphate solution (TPP) was performed in drug polymer solution at the rate of 2ml/min (12 ml TPP in 20 ml drug polymer solution). The obtained particles dispersion were sonicated using a probe sonicator (S-4000; Misonix, Farmingdale, NY) at medium amplitude (50%) for 5 min to obtain nano sized particles. The dispersion was then filtered through a 0.2 um hydrophilic filter (Minsart, Sartorius) for isolation of smaller nanosize particles in order to achieve maximum transportation at targeted site. The nano sized particles, thus obtained were carefully purified by ultrafiltration (Amicon 8200 with a millipore PBMK membrane, MWCO 300000) against double distilled water at optimal temperature. The ultrafiltration facilitates elimination of residual of unreacted solvent and unbound drug. For the PEgylation process, accurately 50 mL of 0.3 % chitosan nanoparticles were added into polyethylene glycol (PEG) solution with a ratio of 3:1 and stirred at 500 rpm for 1 h. Further, dispersion was applied to the mixture for 60 seconds to produce homogeneous PEG-Chitosan nanoparticles. Result The formation of the PEGylated chitosan nanoparticles entrapping natural extract Salvia officinalis and Melissa officinalis ensued impulsively upon combination of the pawn anion TPP into the consistent Chitosan polymer solutions. Nanoparticle formation resulted from the ionic interactions between the negative charge ion TPP and the positively charged amino groups of Chitosan. The ratio of CS/TPP was optimized to attained stable dispersion and formation of nanosize particles. Preliminary experiments were performed in order to identify the optimal concentrations of CS and TPP for NP formation. The process parameters along with formulation parameters were thoroughly optimized for the achievement of physiochemical and thermal stable nanoparticles. The obtaining nano size particles were broadly characterized as either a clear solution, an opalescent suspension displaying a tyndall effect (NPs), or aggregate. 1 Particle size, Zeta Potential and Morphology The results achieved from the zeta sizer measurement displayed very distinct size of prepared S-PCN and M-PCN formulations ranging 150-250 nm (Figure 1- a & b). The nano size of the S-PCN, M-PCN formulations displayed decent encapsulation of extract in the polymer matrix due to the formulation and process optimization. The surface charge of both nanoformulation S-PCN, M-PCN was found to be -10.89 mV and -16.21 mV respectively (Figure 1- e & f) demonstrated negative charge nature of both formulation. The negative charge of formulation showed better stability and optimum candidature for enhance brain targeting. The pH of S-PCN, M-PCN formulations was measured as 6.9 ± 0.01 which play a vital role in nearly neutral microenvironment delivery for efficient brain targeting. The pH facilitate targeting mechanism act as the key element for the onsite degradation of the polymer matrix. This polymeric degradation activation mechanism enhanced the drug release at a controlled rate resulting into the desired therapeutic potential. 2 DLS Analysis The DLS outcomes again nanosize range dispersion of prepared S-PCN and M-PCN nanoformulation. The size distribution pattern of both nanoformulation is some identical to each other exhibiting size range of 160-240 nm for S-PCN and 150-230 nm for M-PCN formulation. The optimal nanosize range of both nanoformulation demonstrated the enhanced brain delivery and onsite targeting which efficiently comply the size of cells and its micro-environment. The DLS investigations showed diverse size distribution of and dispersion pattern. The PDI exhibited by S-PCN and M-PCN was found to be of 0.271 ± 0.08 and 0.259 ± 0.11. The DLS results showed enhance stability with even size distribution pattern of prepared nanoparticles between 100-500 nm (Figure 1-c & d). This nanosize stable pattern facilitates enhance diffusion of prepared nanoparticles across the blood brain barriers leading to optimal pharmacological potential during brain targeting. Therefore, it can be unswervingly state out that both the nanoformulations exhibited optimal and stable nano dispersal features for the operative brain targeting against Alzheimer management in clinical platform. 3 Transmission Electron Microscopy (TEM) The TEM analysis showed very discrete particles size exhibiting oval shape nanoparticles of both nanoformulation. The size revealed by TEM analysis for S-PCN and M-PCN was ranging 100-250 nm validating DLS measurement zeta sizer analysis (Figure 2- a & b). The formation of nanoparticles by entrapping natural extract showed better crosslinking between polymer and cross linker avoiding unwanted leakage. Also the aggregation of nanoparticles was found negligible showing better PEGylation process of chitosan boundaries. The TEM outcomes displayed suitable nano carrier system for the effective brain delivery, revealing decent BBB infiltration appearance of both nanoformulation. 4 Scanning Electron Microscopy (SEM) The SEM analysis significantly the results obtained by zeta sizer and TEM assay showing fine particles formation with spherical shape and smooth morphology. The SEM images noteworthy validates the sharp oval boundaries of both nanoformulation exhibiting better PEGylation process. The SEM images also clarifies no sign of clusters formation of agglomeration of particles showing significant PEG outer layer. The SEM analysis exhibiting size range of 150-250 nm again qualitatively validating the TEM, and zeta-sizer analysis and confirming the ideal brain targeting delivery characteristics of both S-PCN and M-PCN (Figure 2 – c & d). 5 In-vitro drug release studies In vitro drug release data of Salvia officinalis and Melissa officinalis extract associated with PEGylated nanoformulations is demonstrated in figure 3- a & b. The drug release pattern from both the nanoformulation S-PCN and M-PCN at different pH (4.0 & 7.4) exhibited a non-linear release profile characterized by a relatively faster initial drug release during the first 3-4 h, followed by slower release in later period. The two pH range was provided to deeply evaluate the effect of nanoformulations for better brain targeting and onsite delivery. The biphasic drug release pattern was observed by both nanoformulation with initial bursting of nanoparticles in early 1-8 h followed by slow release in 24 h. The in-vitro drug release studies suggested that initially both S-PCN and M-PCN provided burst release of drug extract at pH 4.0. The drug release was found to be 89.45 ± 3.67 % at 6h, 91.42 ± 2.11 at 8 h, 90.26 ± 1.84 % at 6 h and 95.67 ± 2.20 % at 8 h for S-PCN and M-PCN, respectively. On the contrary at pH 7 the drug release was significantly (P < 0> S-PCN. 6 In vitro cellular uptake The capacity of cellular targeting and intracellular transport of developed nanoformulation S-PCN and M-PCN evaluated and measured by using UCSD229i-SAD1-1 human astrocytoma cells line. The human astrocytoma cells line are imperative part of BBB and broadly engaged for the examination of brain delivery. The developed S-PCN and M-PCN showed noteworthy cellular acceptance and circulation compared to the free drug extract of Salvia officinalis and Melissa officinalis when evaluated by CLSM analysis. The CLSM signals for the developed S-PCN and M-PCN were resilient and sharp with enhance absorbance when treated with Rhodamine B isothiocyanate (RITC) compared to the free drug extract of Salvia officinalis and Melissa officinalis suspension on incubation for 12 h (Figure 4). In addition, the confocal laser scanning microscopic intense fluorescence signals displayed by nanoformulations showed the clear sign of vesicular localization of nanoparticles demonstrating enhance endocytic pathway progression. The CLSM signals showed by M-PCN samples treated UCSD229i-SAD1-1 human astrocytoma cells showed sharp red fluorescence signal around the cell nucleus when compared to the cells treated S-PCN incubated at 4 h and 12 h of time periods which is found enhanced and significant. The results of CLSM intensity examination showed 2 folds enhance cellular uptake and resilience in-vitro by M-PCN compared to S-PCN on the brain cell membranes. The S-PCN and M-PCN treated cells were also quantitatively observed inductively attached with the plasma optical emission (ICP-OE) spectrometry for 12 h of incubation. The results efficiently inveterate that the around ~45% of M-PCN and ~33% of S-PCN nanoformulation have pointedly traversed into the BBB layer, validated by the transwell assay at basolateral side. The free drug extracts showed scanty diffusion across BBB via UCSD229i-SAD1-1 human astrocytoma cells of ~16% signifying non-significant intracellular transport and penetrating efficiency due to early adsorption at cell membrane restricting direct diffusion to the cells (Figure 3c). Overall, at different incubation time interval, the cell uptake and transportation capability of M-PCN was remarkable compared to S-PCN with strong fluorescent adverts bereft of any morphological difference in cell lines, resulting in enhanced brain targeting efficiency. 7 In vitro cytotoxicity assay The MTT assay was employed for the investigation of developed M-PCN and S-PCN toward UCSD229i-SAD1-1 human astrocytoma cells. The MTT assay qualitatively showed significant anti-proliferation capability of nanoformulations in 24h of incubation. The investigations showed sharp cell viability of 100% and 10% by control Normal control (saline solution) and negative control group (Triton X 100 surfactant solution) respectively. The developed S-PCN and M-PCN showed notable cell viability of 96%, 89%, 76% & 65% and 98%, 90%, 80% & 71% at different concentration (0.1, 1, 10 and 20 μg/mL of individual concentration) on 24 h of incubation (Figure 3d). Whereas free drug extract of Salvia officinalis and Melissa officinalis showed cell viability of 96%, 88%, 68%, & 48% and 95%, 86%, 69% & 52% respectively on 24 h of incubation. The MTT investigation established non-significant cell cytotoxicity by different samples in 24h of incubation showing nonlinear relationship between incubation time and anti-proliferation efficiency. The MTT results clearly displayed significant cell viability of nanoformulation over free drug extract in 24 h of incubation expressing biologically safe brain targeting efficiency with negligible toxicity on human astrocytoma cells. The enhance cell viability showed by developed S-PCN and M-PCN is due to better physiochemical compatibility between nanocomposite resulting in efficient cellular transport and brain delivery. On inter-comparison of nanoformulation the cell viability of M-PCN is greater than S-PCN with less cell cytotoxicity at higher concentration. The inter-comparison results showed better endocytosis and resilience of M-PCN which is found statistically significant when analyzed by student’s T test. Overall the cell toxicity examinations clearly expounds that the developed nanocomposite may be used as novel drug carrier encapsulating natural extract for the treatment of brain diseases as targeted delivery system. 8 Apoptosis assay The Apoptosis investigation showed by free drug extract, S-PCN and M-PCN and verified striking apoptosis at all concentrations. The developed S-PCN and M-PCN showed inherent apoptosis compared to the free drug extract. It has been noted out that both S-PCN and M-PCN showed mitochondrial apoptosis phenomenon or death activator by provoking cell surface receptor. By activating cell surface receptor the activation of caspase cascade establishes optimum cell death which results in desired apoptosis process. The apoptosis index of free drug were found to be 0.39 and 0.42 for Salvia officinalis and Melissa officinalis respectively whereas the S-PCN and M-PCN showed apoptotic index of 0.66 and 0.79 respectively. The nanoformulation showed significant apoptosis action compared to plain free natural extract which is nearly two folds more and found significant (*P<0.01) (Figure 5). The chief cause for better apoptosis of nanoformulation over free drug extract is the nanosized particles, causing quick onsite drug transportation, sufficient distribution and better release. On inter-comparison of S-PCN and M-PCN the apoptosis potential is significant showed by m-PCN compared to S-PCN when analyzed by student T test. Overall PEgylation of chitosan nanoparticles facilitates better circulation of nanoparticles in brain microenvironment causing extended release and negligible drug toxicity resulting in better brain targeting against Alzheimer disease. Conclusion: Based on the findings, it can be inferred that biodegradable PEGylated chitosan nanoconjugates hold promise as effective nano-targeting agents for delivering anti-Alzheimer drugs to the brain. The incorporation of PEGylated chitosan nanoparticles in this approach demonstrates enhanced delivery capabilities, ultimately leading to improved therapeutic out-comes. Other Characterization 1 Particle size, Zeta potential, pH and Morphology The developed S-PCN, M-PCN particle size and surface charge was measured by Malvern Zetasizer 3000 particle size and zeta potential analyzer (Malvern Instruments, Bedfordshire, UK). The Zeta potential of S-PCN, M-PCN was examined by smearing the principle of electrophoretic movement of particles in an applied electrical field. The concentration of both S-PCN, M-PCN formulation was attuned at 0.01% w/v by distilled water or in 0.01 M sodium chloride solution for potential assessment. The pH was calculated by using a digital pH meter (HI-TECH WATER TECH. New Delhi, India). The pH meter was first calibrated using buffer tablet, the pH meter was dipped in a beaker comprising S-PCN and M-PCN nanoformulations on post calibration. The nanoformulations, evaluation was triplicated and the measurement was repeated thrice with an average value along with SD was reported. 2 Dynamic Light Scattering (DLS) The S-PCN and M-PCN nanoformulations was examined for the Dynamic Light Scattering (DLS) investigating mean diameter and PDI by employing Brook-heaven BI 9000 AT instrument (Brookheaven Instrument Corporation, USA). The DLS examination was measured for the more distinct and significant evaluation of both S-PCN and M-PCN nanoformulations. The DLS evaluation were done at wavelength 417 and 215 nm for S-PCN and M-PCN nanoformulations receptively at temperature of 25°C. 3.3 Transmission Electron Microscopy (TEM) The TEM of both S-PCN and M-PCN nanoformulations was measured by using Hitachi H-7500 TEM analyzer. TEM metaphors were obtained to visualize the shape and structure of nanoformulaion. The S-PCN and M-PCN nanoformulations were coated with 2.5% w/v of phosphor-tungstic acid (PTA) solution and placed in a copper disc grid. The grid was then desiccated in 60 watt LED lamp (Philips, India Ltd) and was finally placed into the disc holder and scanned for TEM evaluation. 4 Scanning Electron Microscopy (SEM) The morphology and structure of prepared S-PCN and M-PCN nanoformulations were analyzed by SEM, Nova Nano SEM 450, Germany. Before the SEM assessment, the formulations were lyophilized by using freeze dry lyophilizer (REMI, New Delhi, India). The dried formulations were then placed on a SEM stub by using dual adhesive tape at 50mA 5-10 minutes via sputter (KYKY SBC-12, Beijing, China). A SEM aided with secondary electron detector was engaged to get the digital images of the developed S-PCN and M-PCN nanoformulations. 5 Entrapment Efficiency (EE): EE plays essential part in transporting the bioactive to the targeted site at detailed therapeutic dose in order to get the anticipated therapeutic value. To measure the EE, both the nanoformulations were centrifuged at 10000 rpm for 5 minutes to obtain the pellets. The collected supernatant was carefully diluted with PBS of pH 7 and the drug content was determined spectrophotometrically by using UV spectrophotometer (Schimadzu, Japan) at 317 nm and 215 nm for S-PCN and M-PCN nanoformulation respectively against a blank solvent. The EE can be measured by using the following formula: EE= weight of drug in nanoformulation / initial weight of drug taken x 100 6 In vitro Drug Release studies The release of from both S-PCN and M-PCN nanoformulations was tracked to predict the diffusion and kinetic behavior of the nanosystems for desired therapeutic efficiency. For release studies, both S-PCN and M-PCN obtained after centrifugation were suspended in 10 mL of a phosphate buffered saline (PBS) solution, pH 7.4. This nanoparticle suspension was transferred to clean Eppendorf’s tube and placed in a water bath at 37 °C under stirring. After 0.5, 1, 2, 4, 6, and 24 h, samples were collected from the bath and centrifuged at 14 000 rpm for 5 min (BOECO, Hamburg, Germany). Supernatants were analyzed by UV spectroscopy and used to calculate the amount of drug released from the nanoparticles over the specified time. Triplicate samples were analyzed at each time. 7 Cell Line studies 7.1 Cell Culture and Seeding The Human UCSD229i-SAD1-1 human astrocytoma cells line was obtained from NCCs Pune and was conserved in Dulbecco’s modified Eagles Medium. The cell line was then supplemented with 10
Chapter
Essential oils are complex, volatile compounds synthesized by plants. They are highly valuable secondary metabolites of plants with a wide range of applications in various sectors such as food, beverage, pharmaceutical, medicine, cosmetics, perfumery and agricultural industries with their various and valuable biological activities. In this chapter, the biological applications of essential oils will be discussed based on their bioactive properties. First, the introduction will cover commonly used and well-researched essential oils, their main components, bioactivities, application areas, and toxicities. Then, the biological activities of essential oils, namely antimicrobial, antioxidant, anti-inflammatory, anticarcinogenic, analgesic, anxiolytic and antidepressant, antihyperglycemic, and insecticidal activities will be discussed in a very comprehensive way. While giving this information, how these biological activities of essential oils are applied to solving problems encountered in the past and today, and in which application areas they benefit humanity will be mentioned. The mechanisms of biological activities of essential oils will be discussed by making use of previous studies on these subjects. At the end of this chapter, information about the previously encountered and possible toxic effects of essential oils will be given.
Article
Full-text available
Background:Noticing the side effects associated with chemical drugs, using natural medicinal plants has gained more prominence recently. Physalis alkekengi extract is a medicinal plant belonging to Solanaceae family which similar to most drugs used in traditional medicine, despite possessing a multitude of medicinal properties, has not received sufficient attention. The aim of the present study was to briefly review the effects of Physalis alkekengi extract on the concentration of thyroid hormones, blood cholesterol, some plasma biochemical factors, liver function, immune system, and sexual hormones. Due to the extensive usage of Physalis alkekengi extract in traditional medicine, determining its advantages and possible side effects is of great physiologic and pharmacologic significance. Physalis alkekengi extract due to the presence of such effective substances as alkaloids, lycopene, glucocorticoids, alcoholic agents, and a large amount of vitamin C as well as antioxidant properties can play a significant role in changes in body homeostasis. This study dealt with the function and effect of Physalis alkekengi extract on different body organs through using proper keywords and extensive online search through electronic databases and credible sources. The results of this mini-review showed that Physalis alkekengi extract can bring about various significant changes in different body organs that have not been properly recognized. Therefore, further and more extensive studies should be done on this plant.
Article
Full-text available
Background: Hyperlipidemia is a heterogeneous group of disorders characterized by an excess of lipids in the bloodstream Objective: Given previous studies on barberry and Melissa officinalis extracts, this study aims at comparing hypolipidemic activities of Melissa officinalis extract and Berberis vulgaris. Methods: For the purpose of this study, 64 Wistar rats were selected and divided into 8 groups (n=8). The control group was administered with ordinary diet; the sham group was administered with high-fat diet and intraperitoneally 0.2 ml/dl of the extract solvent (normal saline); and similarly, experimental groups received minimal, moderate and maximum dosages of barberry and Melissa officinalis extracts. The treatment group's was given high-fat diet for 21 days. After this period, blood samples were taken and the gathered data were analyzed using SPSS software. Results: The amount of cholesterol, triglyceride and LDL were increased in the model group compared to the control group, whereas the same substances were decreased significantly in the group receiving the extract compared to the model group (p<0.05). Conclusion: Hypolipidemic properties of alcohol extracts of Melissa officinalis are more effective than those of Berberis vulgaris. Moreover, it should be noted that it is rather the antioxidant properties of Melissa officinalis and their effects on the increase in thyroid hormones as well as the presence of alkaloid compounds, such as berberine in Berberis vulgaris, that inhibits cholesterol synthesis and enables its excretion.
Article
Full-text available
Melissa officinalis(MO) is a herb which grows in different parts of Iran. In previous studies, it was reported that 10% ethanolic extract of the aerial parts of MO, i.e. leaf and stem, could bring about antinociceptive, anticonvulsive, and sedative effects on rats. This study was designed to evaluate anxiolytic effects of different doses of the aqueous decoction extract of MO and also the role of opioid receptors. The subjects of this study were ninety-three male mice whose weights ranged from 25-30gr. Different doses of the extract(5, 10, 25, 50mg/kg-IP) were injected to four separated groups of ten(case groups) and water(10ml/kg-IP) was injected to the control group including 10 mice. A pilot study was also carried out on 15 mice. In order to evaluate the role of opioid receptors on anxiolytic effects of the aqueous decoction extract of MO, the rest of the subjects were divided into 4 groups including seven each. Then Naloxone(2mg/kg) and the extract of MO(5 mg/kg) were injected to them. To increase their activity and curiosity, animals were put inside a box with black walls for 5 min. Then animals were transfered to the elevated plus maze at adjusted intervals and their anxiety reactions including enterance numbers and time spent in open arm were recorded in 5 min. Results indicated that injection of 5mg of the extract reduced anxiety reactions. In comparison to control group, case group animals had both more numbers of entrance and more time spent in open arm(P<0.01). However, higher doses of MO reduced entrance numbers and also time spent in open arm, that is, they had hypnotic effects. Naloxone reduced anxiolytic effects of low doses of Melissa officinalis extract(5mg/kg). It is concluded that the aqueous extract of MO plays an important role in fear, anxiety and drowsiness so that the lowest dose(5mg/kg) produces anxiolytic effects and higher doses exert hypnotic ones. It is also probable that it works through opioid receptors.
Article
Full-text available
Objectives: Hyperlipidemia can cause a variety of diseases such as atherosclerosis, diabetes, and fatty liver which is followed by increased liver enzymes. Since Berberis vulgaris (B. vulgaris) root possesses antioxidant properties, the present study was conducted to investigate the effect of its extract on the activity of liver enzymes in rats. Materials and Methods: In this experimental study, sixty Wistar rats were selected and allocated to six groups of ten each. The control group received a normal diet and the sham group received a fatty diet while the other groups including experimental groups received a fatty diet and the alcoholic extract of B. vulgaris at minimum (75 mg/kg), moderate (150 mg/kg), and maximum (300 mg/kg) doses by intraperitoneal injection (i.p.) or oral atorvastatin (10 mg /kg) with a fatty diet. At the end of this 21-day period, blood samples were drawn and the levels of the intended factors were measured. Data were analyzed using SPSS software version 11.5. Results: The comparison of the obtained results showed that the levels of alanine transaminase (ALT) and alkaline phosphatase (ALP) enzymes in the sham group that only received fatty food increased (p≤0.05), whereas in the treatment groups receiving B. vulgaris extract as well as in the group receiving Atorvastatin, these enzymes significantly decreased; however, no significant changes were observed in aspartate transaminase (AST) levels. Conclusion: Noticing the antioxidant properties of B. vulgaris root extract and its effects on reducing the activity of liver enzymes, the extract of this plant can be a good choice for improving the function of liver.
Article
Full-text available
Objectives: Liver is one of the most sensitive tissues to oxidant damage. Hence, the present study was conducted to compare the effects of Melissa officinalis (MO) extract and Atorvastatin on the activity of liver enzymes in rats. Materials and Methods: In this experimental study, 60 male Wistar rats were selected and allocated to six groups (n=10). The control group received a normal diet, sham group received a fatty diet while other groups received a fatty diet and the alcoholic extract of MO, at minimum (25 mg/kg), moderate (50 mg/kg), and maximum (75 mg/kg) doses (i.p.). The last group received Atorvastatin (10 mg/kg) through gavage with a fatty diet over a 21-day period. At the end of this 21-day period, blood samples were drawn and the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP), activity of liver enzymes as well as cholesterol in the samples were measured. Results: The obtained results showed that the activity of liver enzymes in the treatment groups receiving MO extract and the group receiving Atorvastatin decreased significantly. MO extract reduced the level of liver enzymes. Conclusion: Therefore, further studies for obtaining a better understanding of the mechanism of effect of MO for treating liver diseases are recommended.
Article