EUPATILIN: A REVIEW OF ITS TRADITIONAL USES, PHARMACOLOGICAL ACTIVITIES, AND FUTURE PROSPECTS AS A THERAPEUTIC AGENT BY - POOJA BHARTI & PROF.(DR.) DHARMENDRA AHUJA
EUPATILIN: A REVIEW OF ITS TRADITIONAL USES,
PHARMACOLOGICAL ACTIVITIES, AND FUTURE PROSPECTS AS A THERAPEUTIC AGENT
AUTHORED BY - POOJA BHARTI1 & PROF.(DR.)
DHARMENDRA AHUJA2
1. Faculty of Pharmaceutical Science
Jayoti Vidyapeeth women’s university, Vedant Gyan Velly, Jharna, Jaipur
Rajasthan 303122
2. Dean & Professor, Faculty of Pharmaceutical
Science, Jayoti Vidyapeeth Women's University, Vedant Gyan Velly, Jharna, Jaipur Rajasthan 303122
Abstract
Eupatilin
(5,7-dihydroxy-3?,4?,6-trimethoxyflavone) is a pharmacologically active flavone which has been isolated
from a variety of medicinal plants. Eupatilin is known to possess various
pharmacological properties such as anti-cancer, anti-oxidant, and
anti-inflammatory. It is speculated that eupatilin could be subjected to structural
optimization for the synthesis of derivative analogs to reinforce its efficacy,
to minimize toxicity, and to optimize absorption profiles, which will
ultimately lead towards potent drug candidates. Although, reported data acclaim
multiple pharmacological activities of eupatilin but further experimentations
on its molecular mechanism of action are yet mandatory to elucidate full spectrum of its pharmacological activities. Eupatilin, a
flavonoid compound derived from the Artemisia princeps plant, has been
traditionally used in Asian medicine
for its anti-inflammatory and anti-allergic properties. From a medicinal
chemistry perspective, eupatilin's pharmacological activities can be attributed
to its unique chemical structure, which consists of a flavone
backbone with a hydroxyl group at position 5 and a methoxy
group at position
4'. This review
provides a comprehensive overview of eupatilin's traditional uses,
pharmacological activities, and future prospects as a therapeutic agent.
Keywords eupatilin; anti-cancer; anti-inflammatory; anti-oxidant; medicinal
plants; flavone
Introduction
Natural products
(NPs) have historically affirmed their worth not only as therapeutic agents but also as an important source of novel drug leads. The
term “natural products” is usually prescribed as chemical entities that are
mainly derived from the living species such as plants, microorganisms, and marine organisms. These naturally occurring
compounds are known to possess complex chemical diversity with
outstanding drug-like activities that contribute towards their multi-targeted
action. Polyphenols are secondary metabolites that are found in numerous
medicinal plants with potential anti-inflammatory, anti-cancer, anti-allergic,
anti-microbial, and anti-oxidant activities.1 Flavonoids represent
an eminent group of plant-derived polyphenols, with greater
than 8000 varying compounds (Figure 01).
Figure 01:
Biological activity of Eupatilin
Flavonoids are
associated with multiple biological effects, including antitumor,
anti-oxidation, anti-inflammation, antiviral and hepatoprotective activities,
as well as in the prevention of cardiovascular diseases. Eupatilin
(5,7-dihydroxy-3',4',6-trimethoxyflavone) is extracted from Artemisia asiatica
(A. asiatica) Nakai, and this isolated flavonoid contains pharmacologically
active ingredients. Eupatilin has been demonstrated to exert anticancer,
anti-oxidative and anti- inflammatory effects. A previous report indicated that
Stillen™ (DA-9601), produced from the ethanol extract of A. asiatica, contained
the pharmacologically active flavonoid compound eupatilin. DA-9601 demonstrated
cytoprotective effects against gastric mucosal damage and ulcerative proctitis. Eupatilin has exhibited
positive effects in the treatment of oxidant-dependent gastric disorders.2
Eupatilin, apigenin, wogonin and baicalein are all members of the same family
of flavonoids. Although
the flavones, apigenin,
wogonin and baicalein, have previously been used
in the treatment of OS, the molecular mechanisms underlying eupatilin-mediated apoptosis of the U-2 OS cell
line have remained to be elucidated. Therefore, the present study aimed to aid
the elucidation of the underlying mechanism
involved in eupatilin-induced apoptosis of U-2 OS cells. This was achieved via cytotoxicity
experiments, apoptosis studies and the analysis of changes in protein
expression associated with apoptotic cell death.3
Eupatilin (5,7-dihydroxy-3?,4?,6-trimethoxyflavone), a pharmacologically active
flavonoid mainly found in
genus Artemisia, is known to possess auspicious anti-cancer, anti-inflammatory,
anti-oxidant, neuroprotective, anti-allergic, and cardioprotective activities.
Assorted studies on eupatilin demonstrated its potent bio-activities which have
made further interest among the biologists and chemists to explore more about
this bioactive natural flavone.4
To date, no
comprehensive review has been available on the pharmacological and biological
profile of eupatilin. The present review aims to compile the available
information on eupatilin, along with signifying its current status.5 This will hopefully
help to ease the understanding of eupatilin pharmacological appearance in drug development,
thus, proposing areas where further
research work is mandatory. The literatures were screened through different e-sites;
Elsevier Science Direct,
Scopus, Springer Link, PubMed,
and other medical journals. Key words that are being used for searching are
“eupatilin”, “eupatilin and its biological activities”, “anti-cancer”, “anti-allergic”, “neuroprotective”, and
“anti-inflammatory”. 6-9
Biosynthesis of
eupatilin
Eupatilin is a
flavonoid compound that is biosynthesized in plants through the phenylpropanoid
pathway. The biosynthesis of eupatilin begins with the conversion of
phenylalanine into cinnamic acid by the
enzyme phenylalanine ammonia-lyase (PAL). Cinnamic acid is then converted into
p-coumaric acid by cinnamate 4-hydroxylase (C4H), which is subsequently
converted into p-coumaroyl-CoA by 4-coumarate: CoA ligase (4CL). The enzyme
chalcone synthase (CHS) then combines
p-coumaroyl-CoA with malonyl-CoA to form naringenin chalcone, which is converted
into naringenin by chalcone isomerase (CHI). Naringenin is then converted into eriodictyol by flavonoid 3'-hydroxylase (F3'H), and finally,
eriodictyol is converted into eupatilin by flavonoid
3',5'-hydroxylase (F3'5'H).10-12 The biosynthesis of eupatilin is
regulated by various transcription factors and enzymes that control the
expression of the genes involved in the phenylpropanoid pathway (Figure 02).
Figure 02: Biosynthesis pathway of eupatilin Method of Extraction of
Eupatilin
Eupatilin is
typically extracted from the leaves and flowers of the Artemisia princeps plant
using various solvent extraction methods. The most common method involves soaking
the plant material in a solvent such as ethanol,
methanol, or acetone,
followed by evaporation of the solvent
to leave behind a crude
extract containing eupatilin. Other methods include maceration, where the plant
material is soaked in a solvent for an extended period, and ultrasound-assisted
extraction, which uses high-frequency sound waves to enhance extraction.12
Microwave-assisted extraction,
supercritical fluid extraction, and column chromatography are also used to
extract and purify eupatilin. Finally, high-performance liquid chromatography (HPLC) is employed to separate and purify
eupatilin, resulting in a highly concentrated and purified extract (Figure 03).
Figure 03: Method
of Extraction of Eupatilin
Eupatilin and its natural sources
Eupatilin, a
bioactive flavone has been identified in many plants of family Asteraceae that
comprises ~24,000 species.
Of these, Artemisia
is a diversified genus encompassing ~500 species in the
temperate regions of Europe, Asia, and North America. It has been reported as
an affluent source of flavonoids including eupatilin. Genus
Centaurea incorporates more than 500 species, of which 45 grow instinctively in Algeria, with 7 species localized in
the Sahara. Genus Tanacetum consists of ~160 species commonly known as tansies which are widely utilized in folk medicines for various
treatment purposes. The aerial parts of T. vulgare are effective against
neuralgia, rheumatism, and migraine. It also serves
as an anthelmintic and anti-inflammatory agent. Besides them, eupatilin
has been isolated
from the plants of family
Labiatae (Lamiaceae) generally
known as “mint family”. Salvia is the most prominent genus of mint
family with more than 900 species extensively distributed all over the world.
S. plebeia is advantageous to cure asthma and skin inflammatory ailments in the
Asian countries. Eupatilin was also extracted from the leaves of C. morifolium
(Asteraceae) and L. dulcis (Verbenaceae) as modest anti-oxidative agent. The
leaves of L. dulcis are also useful as traditional medicine
for treating bronchitis and cough.13-15 Eupatilin has notable pharmacological and biological characteristics of interest for the cure of different diseases including cancer, gastritis and many others (Figure 04).
Figure 04: Chemical
Structure of Eupatilin Biological activities of
eupatilin and their mechanisms of action
The
biologically active compound “eupatilin” has been demonstrated for its broad
spectrum of pharmacological properties such as anti-cancer, anti-inflammatory, anti-oxidant, neuroprotective, anti-allergic, and
cardioprotective. Various in vivo and in vitro investigations on eupatilin have
elucidated its medicinal characteristics and mechanism of actions (figure 01).
Anti-microbial activity
Kim et al.
(1996) fractioned an extract of A. asiatica into hexane, chloroform, butanol, and
aqueous fractions, and among these fractions, the hexane fraction exerted the
highest anti- microbial activity and inhibited the growth of microorganisms
including Bacillus subtilis, Escherichia coli, Staphylococcus aureus, and
Lactobacillus plantarum at doses of 250, 500, and 750 ?g/ml, although the
inhibitory potency of this plant was weak (Kim et al., 1996). Essential oil
isolated from A. asiatica in the flowering stage with main constituents
including 1,8-cineol and selin-11-en-4a-ol exhibited anti-microbial and
anti-fungal activities against Bacillus subtilis, Staphylococcus aureus,
Escherichia coli, Pseudomonas aeruginosa, Candida albicans, Rhodotorula rubra,
and Aspergillus fumigatus with a minimum inhibitory concentration ranging from
1–3 ml/ml (Kalemba et al., 2002). Furthermore, the monoterpene alcohol
fractions showed the highest anti-microbial activity (Kalemba et al., 2002).
Compared to other plant-derived extracts or antibiotics such as
Oroxylum indicum, Camellia sinensis, and gentamicin (Sharma et al., 2012;
Sithisarn et al., 2016), however, it is suggested that A. asiatica does not
have strong anti-microbial property compared to other biological activities
such as gastroprotective effect.
Anti-cancer activity
Phytochemicals,
bioactive constituents of medicinal plants, such as polyphenols (flavonoids,
terpenoids and carotenoids) have potent competency for the cure of cancer and
thus, are anti- cancerous in nature.
Several studies on eupatilin have explained its anti-cancer property
due to its promising capacity to prompt apoptosis in different cancer cell lines encompassing gastric cancer (AGS,
MKN-1), leukemia cancer cells (HL-60), renal carcinoma (786-O), hepatocellular
cancerous cells (Huh-BAT), osteosarcoma cancer cells (U-2), glioma cancerous
cells (U87MG, LN229), and melanoma cancer cells (A375). Researches have
concluded that eupatilin generally shows synergetic effects on cell cycle
arrest, apoptosis, and numerous cell signaling pathways in distinct cancer
types.
Eupatilin and apoptosis
Naturally
occurring flavonoids have been reported as lead constituents in the development
of several chemo preventive and chemotherapeutic agents. Compiled data by the
researchers recommend that induction of apoptosis can result from varied
chemotherapeutic and chemo preventive agents. Eupatilin has capability to
stimulate apoptosis in human AGS cancerous cells by a reduction in the ratio of
Bax and Bcl-2, along with the cleavage of (poly (ADP-ribose) polymerase) PARP
and caspase-3. It has been recently approved as a cytoprotective agent by
preventing H2O2-induced apoptosis dose-dependently in
gastric cancer cells. The anti-metastatic effect of eupatilin,
isolated from A. asiatica, was also studied in MKN-1 gastric cells. Results
concluded that eupatilin effectively triggers apoptosis by enhanced expression
of caspase-3 pursued by down-regulation of pro-inflammatory cytokine-interceded metalloproteinase (MMPs) levels in a dose- and time-related
manner.16
Eupatilin, a novel anti-cancer compound, prompted apoptosis and upgraded the creation of reactive oxygen species (ROS) in renal
786-O cancer cells. The anti-proliferative potential of eupatilin in
promyelocytic HL-60 leukemia cultured cells can be dedicated to its apoptosis inducing
potential as described by distinctive nuclear condensation, release of
cytochrome c, cleavage of PARP,
and proteolytic activation of caspase-3/-7/-9, respectively. According to a study
on Huh-BAT hepatocellular cancer cells, eupatilin notably decreased bile-acid stimulated
hepatocytic apoptosis by debilitating caspase-8 cleavage in the bile duct-ligated rats. Eupatilin has potent competency to inhibit
osteosarcoma (U-2) cancer
cell growth by stimulating apoptosis and activating mitochondrial intrinsic pathway that is characterized by increased Bax/Bcl-2 proportion, decline in mitochondrial membrane potential, release
of cytochrome c, activation of caspase-3/-9,
and cleavage of PARP. Treatment with eupatilin specifically prohibited cellular
proliferation in A375 human melanoma cells via inducing apoptosis through morphological innovations of DNA fragmentation in a dose-dependent manner. Eupatilin also displayed inhibitory response on glioma cancerous cells (U87MG, LN229)
by induction of apoptosis via suppressing of Notch-1 signaling pathway.17
Eupatilin and cell cycle arrest
Eupatilin, a natural flavone,
has proficiency to prompt G2/M cell cycle phase arrest in endometrial KLE and Hec1A cancerous
cell lines in a time- and dose-dependent manner. It eloquently
prohibited mutant p53 via up-regulating p21 expression, along with the
activation of Checkpoint Chk2/ATM/Cdc2/Cdc25C pathway. Eupatilin treatment
effectively increased the expression levels of p53 and p21 cell cycle regulators
and induced G1 phase arresting in gastric AGS cancer cells. Moreover, eupatilin
medication also caused G2/M cell cycle phase arresting associated with
apoptotic cell death in U-2 and A375 cancer cell lines. Eupatilin has been
demonstrated as powerful chemo preventive agent in inhibiting EGF- stimulated
JB6 skin cell proliferation by targeting PI3K and promoting G0/G1 cell cycle
arresting through decreased expression of cyclin D1 in a dose-related manner.
In MCF10A-ras carcinoma cells, eupatilin forbided the expression of important
cell cycle organizers cyclin
B1, Cdk2, Cdc2, and cyclin D1. It
has the capacity
to up-regulate p53, p27Kip1 (Cdk inhibitors) and down-regulate p21waf1/Cip1 expressions in a time- and
concentration-dependent manner. It can be concluded that eupatilin arrests
cell cycle at G2/M phase but whether in G2 or M phase should be investigated. Furthermore,
it would be interesting to investigate the mechanism by which eupatilin
arrested cell cycle in breast cancer cells and melanomas at G0/G1
and G1/S phases as in case of other cancer cells it is generally arresting cells at G2/M phase. So, extensive studies
are yet mandatory to fully understand molecular mechanism by which
eupatilin regulates cell cycle or induces cell cycle arrest.18-20
NF-?B/STAT3 signaling pathways
and eupatilin
Flavonoids, phenyl-substituted chromones, have drawn developing consideration as effective anti- cancer agents against several cancer cell lines. Eupatilin,
a pharmacologically active flavone has
been reported to acquire anti-cancer effects via suppressing NF-?B cell
signaling pathways. Several investigations have revealed the fact that
eupatilin actively participates in the prevention of gastric cancer (MKN-1,
AGS) by decreasing NF-?B activity pursued by down-regulation of
metalloproteinase (MMP-2, MMP-9) and pro-inflammatory cytokines. Furthermore,
in MKN45 gastric cancerous cells,
eupatilin significantly prohibited STAT3-induced VEGF expression most apparently under hypoxic situations,
indicating its therapeutic potential to cure gastric cancer. Though several
studies have reported
that eupatilin induces
apoptosis via inhibition of NF-?B but whether eupatilin has direct effect
on NF-?B signaling pathway or via its up-stream signaling pathway JAK or STAT3
still needs to be investigated by researchers.21-23
Targeting PI3K/AKT
& MAPK pathways with eupatilin
Eupatilin as a pharmacological active flavone derived
from various Artemisia
species has a potential to stimulate apoptosis via MAPK and AKT
pathways in many cancer types. Eupatilin
has strong ability to induce
apoptosis in renal cancerous cells
via reactive oxygen species (ROS)- mediated phosphorylation of p38 MAPK,
ERK1/2, JNK, and inhibition of AKT/PI3K signaling cascade in a concentration-dependent manner.
Thus, eupatilin can serve as persuasive therapeutic avenue leading to the cure of
cancer. Demonstration of eupatilin as anti-cancer agent on human gastric cancer cells has declared the evidence that it works
as an inhibitory agent for the activation of AKT and ERK that are crucial constituents of cellular survival
cascades. According to a report, AKT inactivation or/and activation
of ERK1/2 may have assisted in the activation and phosphorylation of Chk2/Cdc2
checkpoint proteins causing cell cycle arrest in eupatilin treated endometrial
cancer cells. Studies on anti-proliferative potential of eupatilin against
MCF10A-ras cancer cells have uncovered the fact that blockage of cell cycle
advancement turns out to be preferable to prohibit ERK1/2 activation. 50 ?M of eupatilin effectually receded phosphorylation of AKT and ERK/MAPK
resulting in induction of apoptosis in MKN-1 cells. However, to precisely
figure out this mechanism, further experimental investigations are recommended.
In addition, there is lack of data regarding to multiple splice variants of
JNKs such as whether eupatilin targets JNK1 or JNK2 which should be addressed
by researchers.2426
Anti-inflammatory
activity
Eupatilin, a
bioactive flavone, was reported to possess anti-inflammatory potential. Oral
administration of extract inclusive of eupatilin and
quercetin-3-?-D-glucuronopyranoside meliorated the colonic injury and inflammatory response in dose-dependent manner by reducing the neutrophil activation and
oxidative stress. Treated animals exhibit decreased expression level of mucosal
myeloperoxidase, tumor necrosis factor-?, nitric oxide, and malondialdehyde and enhanced glutathione levels. Findings by researchers declared eupatilin as a suppressor of
inflammatory response and it acts via inhibiting the inflammatory modulators and NF-?B activation in endotoxin-stimulated macrophages and carrageenan-induced
inflammation in mice. Molecular mechanism underlying eupatilin associated
extenuation of inflammatory response in TNF-?-induced bronchial epithelial
inflammation lies in the inhibition of eotaxin expression and decline in the
activities of NF-?B and I?B? kinase signaling.27-28
Characterization of molecular mechanism by eupatilin in Bacteroides fragilis enterotoxin-
induced inflammatory response
suggests that eupatilin
can uncouple the association
between Hsp90 and IKK-gamma in NF-kappa B pathway, thus, this bioactive
flavone may have the ability to reduce inflammatory response. Eupatilin is an auspicious therapeutic agent against ischemia-reperfusion-induced liver injuries in mice as it can reduce apoptosis and inflammation by up-regulating B-cell lymphoma 2 protein, heat shock protein,
and down-regulating the cleaved
caspase-3 levels. Eupatilin may attune angiogenesis route, a part of anti-inflammatory response in gastric
cancer cell line by inhibition of the vascular endothelial growth factor (VEGF) and STAT3 expression. Eupatilin caused inhibition of adherence
of inflammatory cells to epithelial cells via NF-kappa
B and AKT-dependent pathways as well as targeting adhesion molecule
expression. Eupatilin, a lipophilic flavonoid, showed
notable in vivo anti-inflammatory property
in a dose-dependent way,
qualitatively comparable to hydrocortisone and intervening in conditions of efficacy amid to those of steroids and non-steroidal
anti-inflammatory drugs. Eupatilin, along with other natural compounds specifically inhibited 5-lipoxygenase: a potent mediator
of inflammatory response
and also cause the suppression of leukotriene
D4 and C4 in cultured mastocytoma cells. Eupatilin has pronounced capability to inhibit
TNF-?-induced inflammatory response via ROS/MAPK-NF-?B signaling pathway
suppression in human umbilical vein endothelial cells.29
Antioxidant activity
Eupatilin has
potential to activate sestrin-2-dependent autophagy via inhibition of oxidative stress stimulated by arachidonic acid (AA+) iron in HepG2, H4IIE, and Hepa-1c1c7 cells. It remarkably upgraded cell viability
against AA+ iron in a concentration-related manner and also prohibited
mitochondrial dysfunction and production of ROS.
In human MCF-10A epithelial cells, eupatilin blocked
TNF-?-mediated invasion and intracellular
ROS formation, along with suppressed urokinase-type plasminogen activator
(uPA) and ?-catenin
expression. Eupatilin may serve as potential therapeutic target against acute
ischemia- stimulated. kidney injury through reduced neutrophil
gelatinase-correlated lipocalin and renal damage molecule-1 expressions, serum
creatinine levels, and blood urea nitrogen level. It also enhanced the expression of Hsp70 and Bcl-2 via debilitating iNOS, caspase-3, and Bax
levels. The renoprotective effects of A. asiatica extract and eupatilin were
observed in LLC-PK1 renal epithelial cells. Results disclosed
that A. asiatica and eupatilin
cotreatment has the ability to cure cisplatin-induced kidney damage via down-regulating phosphorylated JNK and p38 protein
levels. Pretreatment with eupatilin actively blocked H2O2-mediated cell damage, ROS generation, and apoptosis via
decreasing Bax and caspase-3 expressions with increasing Bcl-2 and
phosphorylated PI3K/AKT levels in ARPE-19 retinal pigment epithelial cells.30-31
It also suppressed hypoxia/re-oxygenation (H/R)-stimulated cardio-myocyte oxidative
injury in H9c2 rat cells via down-regulating the release of lactate
dehydrogenase (LDL) and activating
AKT/GSK-3? signaling pathway. In an investigation, wound healing potential of
eupatilin was examined through inhibiting. H2O2- and FeSO4-induced cell injury
in AGS epithelial cells in a dose-dependent manner. After effects terminated
that eupatilin therapy decreased the expression of genes responsible for oxidative damage
in H2O2-treated cells such as PLAUR, HO-1 and TNFRSF10A.32
Neuroprotective activity
The multifarious array of bioactive
compounds abundantly found in natural
products plays a crucial role in the treatment and prevention
of several neurodegenerative ailments such as Parkinson’s, Alzheimer’s, and other neuronal dysfunctions. Within the recent decades, a variety of
flavonoids have been recommended to be valuable for the cure of brain-related
injuries incorporating depression and dementia.33
Eupatilin, a
bioactive flavonoid from many Artemisia species, has been shown to be
neuroprotective against global cerebral ischemia-stimulated neuronal damage in
mice by reducing a number of deteriorating neuronal cells via enhanced
expression of AKT phosphorylation. Eupatilin administration also exerted neuro
defensive effects against focal cerebral ischemia in tMCAO mice in vivo through lessening
microglial activation and number of
lba1-immunopositive cells across the ischemic
brain and suppressing NF-?B signaling
cascade functions. Stillen and its major constituent, eupatilin has
anti-depressant potential to overcome depression by up-regulating mRNA interpretation of estrogen ?-receptor and down-regulating TNF?, IL-6,
and IL-1? levels in the hippocampus, respectively.34-36
Eupatilin and its other biological activities
Among other
biological activities, eupatilin actively plays the role of an anti-diabetic
functional constituent in A. princeps and its mechanism
of action proceeds
by increasing liver and blood glucose metabolism, along with enhanced
secretion of insulin
in type 2 diabetic mice. Eupatilin
was proved to be effective for inhibiting skin aging (IC50 = 1.18 ?M)
by mobilizing peroxisome proliferator-activated receptor alpha (PPAR?). It
considerably blocked TNF?-mediated matrix metalloproteinase (MMP-2/-9) expression by reducing
NF-?B p65 nuclear translocation and down-regulating MAPK-AP-1 signaling
pathway in human HaCaT epidermal keratinocytes. Eupatilin is an adjuvant topical avenue against
atopic dermatitis (AD)-alike skin lesions and it acts via reducing
interleukin-4/-19, TNF-?, and thymic stromal lymphopoietin expressions.37-38
Eupatilin has
capability to reform cell viability via suppressing cardiomyocyte apoptosis and
triggering AKT/GSK-3? signaling pathway in H9c2 cells in vitro. The molecular mechanism
underlying this impact of eupatilin
on vascular smooth muscle contractility suggests that it has
compelling relaxation effect on agonist-activated vascular contraction via
inhibiting fluoride-induced up-regulation in pMYPTI expressions despite
endothelial function. Eupatilin is a powerful anti-atherogenic agent and its mechanism of action proceeds
via prohibiting
PDGF-BB-mediated growth and migration in human aortic smooth
muscle cells (HASMCs), along with aortic sprouting
that is expectedly stimulated through the debilitation of MKK3/6-p38
MKK4-JNK, MAPK, and PI3 K-activated pathways.39-40
In another
investigation, eupatilin eloquently stimulated relaxation effect
in human corpus cavernosal smooth muscle cells (CCSMC) through
NO-independent pathways and decreased phosphorylation of myosin
phosphate targeting subunit 1 at Thr853 of MLCP. It may act as an efficacious
versatile curative intervention for osteoporosis via prohibition of actin
rearrangement of pathogenic multinucleated cells (MNCs) and transcriptional down-regulation of NFATc1 and c-Fos of distinguishing osteoclasts (OC). Eupatilin can be
useful for treating
osteoarthritis (OA) with conspicuous antinociceptive and chondroprotective
properties. Eupatilin medication reciprocally increased extracellular matrix
formation in particular chondrocytes and suppressed oxidative
injury as well as decreased IL-1?, IL-6, iNOS, and nitrotyrosine
in the cartilage.41-43
Eupatilin may
serve as an agent for the cure of various allergic infections. In guinea pig
lung cells, eupatilin initially inhibited phosphorylation of Syk tyrosine and
then blocked downstream multisignal pathways
as well as Ca2+ influx during mast cell stimulation. Eupatilin was proved to be convenient for protection from passive cutaneous
anaphylaxis (PCA) reaction and itching behaviors stimulated by
IgE-antigen complex with IC50 of 3.4 ?M. Eupatilin is a well-known gastroprotective agent against
gastric mucosal lacerations and lowers gastrointestinal motility
by inhibiting enhanced
bowel segments of human ileum and
colon. It was also reported
as anti-ulcer agent during investigation on rat plasma, urine, bile, and liver but
accurate mechanism of action is not fully understood.44
Eupatilin showed effective anti-xanthine oxidase (XO) activity
with a Ki value of 0.13 ?g/ ml and IC50 value ranging
from 3.3 to 6.8 ?M. It strongly
exerted anti-mutagenic effects via prohibiting 3-amino-1-methyl-5H-pyrido [4,3-b] indole (Trp-P-2) metabolic activity and heterocyclic amines when
treated with Salmonella typhimurium TA98 strain. Clarification of eupatilin as an inhibitory agent on the functions of cytochrome P450 enzymes in human liver microsomes has affirmed the documentation that it vigorously inhibits CYP1A2-organized phenacetin O-deethylation and CYP2C9-catalyzed diclofenac 4-hydroxylation. Kinetic studies on liver microsomes have
reported eupatilin as a competitive inhibitor of CYP1A2 [K(i) of 2.3 ?M] and a mix-type inhibitor of CYP2C9 [K(i) of 1.6 ?M], correspondingly. It also has a potential
to prevent thrombosis via blocking
arachidonic acid (AA)-stimulated platelet aggregation and production of
serotonin and thromboxane A2 (TXA2) as well as enhancing
mobilized partial prothrombin time (PT) and
thromboplastin time (aPTT) in vitro, thus, acts an important anti-platelet and
anti-coagulant agent.45
Conclusions and future perspectives
Eupatilin, a
flavonoid compound, has been found to possess various pharmacological
properties, including anti-cancer, anti-inflammatory, anti-oxidant, neuroprotective,
anti-allergic, and cardioprotective activities. Isolated
from various plants,
including Artemisia species,
eupatilin has been shown to regulate
several cell signaling
pathways associated with proliferation, inflammation, and other ailments. The compound has been found to be an important
constituent in the seeds,
flowers, leaves, and stems of varied plants, making it a therapeutic
representative.
Accumulated
data from multitudinous studies have provided evidence for the role of
eupatilin in various types of cancer, regulating cell signaling pathways
associated with proliferation, inflammation, and other ailments. Although
eupatilin has been reported to have several pharmacological applications,
further studies are needed to uncover its safety dosage, efficacy, and
potential toxicity. Structure-activity relationship studies and preclinical
trials are necessary to fully understand its biological and pharmacological
applications.
Furthermore,
studies related to the identification of genotoxicity, reproductive toxicity,
nephrotoxicity, and hepatotoxicity need to be investigated by researchers. It
would also be worthwhile to investigate the structure-activity relationship of eupatilin in several activities, such as anti-cancer and anti-inflammatory. Eupatilin
may act as a template
for the design and synthesis of many new drugs to cure various
human ailments, but further experimentation, along with medicinal chemistry
approaches and preclinical trials, is obligatory to uncover the knowledge of
its biological and pharmacological applications and their associated mechanisms of action
for the treatment and
prevention of several diseases. Overall, eupatilin appears to be a promising
compound with potential applications in the prevention and treatment of various
diseases.
Acknowledgement
The authors
would like to express their sincere gratitude to all the researchers,
scientists, and experts whose work has contributed to the understanding of eupatilin's pharmacological properties and therapeutic potential. We also acknowledge the
support and resources provided by our institutions and laboratories, which have enabled
us to conduct this review
and share our findings
with the scientific community. Additionally, we would like to thank our
colleagues and peers for their valuable feedback, suggestions, and
encouragement. We would like to extend our special thanks to our supervisors for their guidance, mentorship, and unwavering support. Their expertise, constructive feedback,
and encouragement have been invaluable to us, and we are grateful for the
opportunity to work under their supervision.
Conflicts of Interest
The authors declare no conflicts of interest.
References
1. Siegel,
R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J.
Clin. 2020, 70, 7–30.
2. Landre, T.;
Uzzan, B.; Nicolas, P.; Aparicio, T.; Zelek, L.; Mary, F.; Taleb, C.; Guetz,
G.D. Doublet chemotherapy vs. single-agent therapy with 5FU in elderly patients
with metastatic colorectal cancer. a meta-analysis. Int. J. Color.
Dis. 2015, 30, 1305–1310.
3. Xie, Q.;
Wu, M.-Y.; Zhang, D.-X.; Yang, Y.-M.; Wang, B.-S.; Zhang, J.; Xu, J.; Zhong,
W.-D.; Hu, J.-N. Synergistic anticancer effect of exogenous wild-typep53gene
combined with 5-FU in human colon cancer resistant to 5-FUin vivo. World
J. Gastroenterol. 2016, 22, 7342–7352.
4. Webber,
E.M.; Kauffman, T.L.; O’Connor, E.; Goddard, K.A.B. Systematic review of the
predictive effect of MSI status in colorectal cancer patients undergoing
5FU-based chemotherapy. BMC Cancer 2015, 15, 156.
5. Touil, Y.;
Igoudjil, W.; Corvaisier, M.; Dessein, A.-F.; Vandomme, J.; Monte, D.; Stechly,
L.; Skrypek, N.; Langlois, C.; Grard, G.; et al. Colon Cancer Cells Escape 5FU
Chemotherapy-Induced Cell Death by Entering Stemness and Quiescence Associated
with the c-Yes/YAP Axis. Clin. Cancer Res. 2014, 20, 837–846.
6. Jegal,
K.H.; Ko, H.L.; Park, S.M.; Byun, S.H.; Kang, K.W.; Cho, I.J.; Kim, S.C.
Eupatilin induces Sestrin2-dependent autophagy to prevent oxidative
stress. Apoptosis 2016, 21, 642–656.
7. Choi,
E.-J.; Lee, S.; Chae, J.-R.; Lee, H.-S.; Jun, C.-D.; Kim, S.-H. Eupatilin
inhibits lipopolysaccharide-induced expression of inflammatory mediators in
macrophages. Life Sci. 2011, 88, 1121–1126.
8. Kim, M.-J.;
Kim, D.-H.; Na, H.-K.; Oh, T.Y.; Shin, C.-Y.; Surh, Y.-J. Eupatilin, a
Pharmacologically Active Flavone Derived from Artemisia Plants, Induces
Apoptosis in Human Gastric Cancer (AGS) Cells. J. Environ. Pathol.
Toxicol. Oncol. 2005, 24, 261–270.
9. Koroglu,
C.; Erdogan, S. Eupatilin Inhibits the Proliferation and Migration of Prostate
Cancer Cells through Modulation of PTEN and NF-?B Signaling. Anti-Cancer
Agents Med. Chem. 2021, 21, 372–382.
10. Wang, X.;
Zhu, Y.; Zhu, L.; Chen, X.; Xu, Y.; Zhao, Y.; Shao, Y.; Li, F.; Jiang, Y.; Lu,
J.; et al. Eupatilin inhibits the proliferation of human esophageal cancer TE1
cells by targeting the Akt-GSK3? and MAPK/ERK signaling cascades. Oncol.
Rep. 2018, 39, 2942–2950.
11. Wang, Y.;
Hou, H.; Li, M.; Yang, Y.; Sun, L. Anticancer effect of eupatilin on glioma
cells through inhibition of the Notch-1 signaling pathway. Mol. Med.
Rep. 2016, 13, 1141–1146.
12. Seo, H.-J.;
Surh, Y.-J. Eupatilin, a pharmacologically active flavone derived from
Artemisia plants, induces apoptosis in human promyelocytic leukemia
cells. Mutat. Res. Toxicol. Environ. Mutagen. 2001, 496,
191–198.
13. Cho, J.-H.;
Lee, J.-G.; Yang, Y.-I.; Kim, J.-H.; Ahn, J.-H.; Baek, N.-I.; Lee, K.-T.; Choi,
J.-H. Eupatilin, a dietary flavonoid, induces G2/M cell cycle arrest in human
endometrial cancer cells. Food Chem. Toxicol. 2011, 49,
1737–1744.
14. Lee, J.-Y.;
Bae, H.; Yang, C.; Park, S.; Youn, B.-S.; Kim, H.-S.; Song, G.; Lim, W.
Eupatilin Promotes Cell Death by Calcium Influx through ER-Mitochondria Axis
with SERPINB11 Inhibition in Epithelial Ovarian Cancer. Cancers 2020, 12,
1459.
15. Nam, S.Y.;
Kim, J.S.; Kim, J.M.; Lee, J.Y.; Kim, N.; Jung, H.C.; Song, I.S. DA-6034, a
Derivative of Flavonoid, Prevents and Ameliorates Dextran Sulfate
Sodium–Induced Colitis and Inhibits Colon Carcinogenesis. Exp. Biol.
Med. 2008, 233, 180–191.
16. Foty, R. A
Simple Hanging Drop Cell Culture Protocol for Generation of 3D
Spheroids. J. Vis. Exp. 2011, e2720.
17. Lim, W.;
Yang, C.; Bazer, F.W.; Song, G. Chrysophanol Induces Apoptosis of
Choriocarcinoma Through Regulation of ROS and the AKT and ERK1/2
Pathways. J. Cell. Physiol. 2016, 232, 331–339.
18. Rosa, A.;
Isola, R.; Pollastro, F.; Caria, P.; Appendino, G.; Nieddu, M. The dietary
flavonoid eupatilin attenuates in vitro lipid peroxidation and targets lipid
profile in cancer HeLa cells. Food Funct. 2020, 11, 5179–5191.
19. Storey, S.
Targeting apoptosis: Selected anticancer strategies. Nat. Rev. Drug
Discov. 2008, 7, 971–972.
20. Pelicano,
H.; Carney, D.; Huang, P. ROS stress in cancer cells and therapeutic
implications. Drug Resist. Updat. 2004, 7, 97–110.
21. Sauer, H.;
Wartenberg, M.; Hescheler, J. Reactive Oxygen Species as Intracellular
Messengers During Cell Growth and Differentiation. Cell. Physiol.
Biochem. 2001, 11, 173–186.
22. Gheytanchi,
E.; Naseri, M.; Karimi-Busheri, F.; Atyabi, F.; Mirsharif, E.S.; Bozorgmehr,
M.; Ghods, R.; Madjd, Z. Morphological and molecular characteristics of
spheroid formation in HT-29 and Caco-2 colorectal cancer cell
lines. Cancer Cell Int. 2021, 21, 204.
23. Fearon,
E.R.; Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990, 61,
759–767.
24. Jeong,
W.-J.; Ro, E.J.; Choi, K.-Y. Interaction between Wnt/?-catenin and RAS-ERK
pathways and an anti-cancer strategy via degradations of ?-catenin and RAS by
targeting the Wnt/?-catenin pathway. NPJ Precis. Oncol. 2018, 2,
5.
25. Siraj,
A.K.; Parvathareddy, S.K.; Pratheeshkumar, P.; Divya, S.P.; Ahmed, S.O.;
Melosantos, R.; Begum, R.; Concepcion, R.M.J.; Al-Sanea, N.; Ashari, L.H.; et
al. APC truncating mutations in Middle Eastern Population: Tankyrase inhibitor
is an effective strategy to sensitize APC mutant CRC To 5-FU
chemotherapy. Biomed. Pharmacother. 2020, 121, 109572.
26. Teimoori-Toolabi,
L.; Hashemi, S.; Azadmanesh, K.; Eghbalpour, F.; Safavifar, F.; Khorramizadeh,
M.R. Silencing the wild-type and mutant K-ras increases the resistance to
5-flurouracil in HCT-116 as a colorectal cancer cell line. Anti-Cancer
Drugs 2015, 26, 187–196.
27. Chun, S.Y.;
Johnson, C.; Washburn, J.G.; Cruz-Correa, M.R.; Dang, D.T.; Dang, L.H.
Oncogenic KRAS modulates mitochondrial metabolism in human colon cancer cells
by inducing HIF-1a and HIF-2a target genes. Mol. Cancer 2010, 9,
293.
28. Cristofaro,
M.; Contursi, A.; D’Amore, S.; Martelli, N.; Spaziante, A.F.; Moschetta, A.;
Villani, G. Adenomatous polyposis coli (APC)-induced apoptosis of HT29
colorectal cancer cells depends on mitochondrial oxidative
metabolism. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2015, 1852,
1719–1728.
29. Fei, X.;
Wang, J.; Chen, C.; Ding, B.; Fu, X.; Chen, W.; Wang, C.; Xu, R. Eupatilin
inhibits glioma proliferation, migration, and invasion by arresting cell cycle
at G1/S phase and disrupting the cytoskeletal structure. Cancer Manag. Res. 2019, 11,
4781–4796.
30. Zhong,
W.-F.; Wang, X.-H.; Pan, B.; Li, F.; Kuang, L.; Su, Z.-X. Eupatilin induces
human renal cancer cell apoptosis via ROS-mediated MAPK and PI3K/AKT signaling
pathways. Oncol. Lett. 2016, 12, 2894–2899.
31. Lin, P.-L.;
Wu, D.-W.; Huang, C.-C.; He, T.-Y.; Chou, M.-C.; Sheu, G.-T.; Lee, H.
MicroRNA-21 promotes tumour malignancy via increased nuclear translocation of
?-catenin and predicts poor outcome in APC-mutated but not in APC-wild-type
colorectal cancer. Carcinogenesis 2014, 35, 2175–2182.
32. Pinton, P.;
Giorgi, C.; Siviero, R.; Zecchini, E.; Rizzuto, R. Calcium and apoptosis:
ER-mitochondria Ca2+ transfer in the control of
apoptosis. Oncogene 2008, 27, 6407–6418.
33. Martucciello,
S.; Masullo, M.; Cerulli, A.; Piacente, S. Natural Products Targeting ER
Stress, and the Functional Link to Mitochondria. Int. J. Mol.
Sci. 2020, 21, 1905.
34. Schönthal,
A.H. Endoplasmic Reticulum Stress: Its Role in Disease and Novel Prospects for
Therapy. Scientifica 2012, 2012, 857516.
35. Dhanasekaran,
D.N.; Reddy, E.P. JNK signaling in apoptosis. Oncogene 2008, 27,
6245–6251.
36. Chambers,
J.; LoGrasso, P.V. Mitochondrial c-Jun N-terminal Kinase (JNK) Signaling
Initiates Physiological Changes Resulting in Amplification of Reactive Oxygen
Species Generation. J. Biol. Chem. 2011, 286, 16052–16062.
37. Tay, K.H.;
Luan, Q.; Croft, A.; Jiang, C.C.; Jin, L.; Zhang, X.D.; Tseng, H.-Y. Sustained
IRE1 and ATF6 signaling is important for survival of melanoma cells undergoing
ER stress. Cell. Signal. 2014, 26, 287–294.
38. Swart, C.;
Du Toit, A.; Loos, B. Autophagy and the invisible line between life and
death. Eur. J. Cell Biol. 2016, 95, 598–610.
39. Amaravadi,
R.; Kimmelman, A.C.; White, E. Recent insights into the function of autophagy
in cancer. Genes Dev. 2016, 30, 1913–1930.
40. Degenhardt,
K.; Mathew, R.; Beaudoin, B.; Bray, K.; Anderson, D.; Chen, G.; Mukherjee, C.;
Shi, Y.; Gélinas, C.; Fan, Y.; et al. Autophagy promotes tumor cell survival
and restricts necrosis, inflammation, and tumorigenesis. Cancer
Cell 2006, 10, 51–64.
41. Qu, X.; Yu,
J.; Bhagat, G.; Furuya, N.; Hibshoosh, H.; Troxel, A.; Rosen, J.; Eskelinen,
E.-L.; Mizushima, N.; Ohsumi, Y.; et al. Promotion of tumorigenesis by
heterozygous disruption of the beclin 1 autophagy gene. J. Clin.
Investig. 2003, 112, 1809–1820.
42. Bursch, W.;
Ellinger, A.; Gerner, C.; Fröhwein, U.; Schulte-Hermann, R. Programmed Cell
Death (PCD): Apoptosis, Autophagic PCD, or Others? Ann. N. Y. Acad.
Sci. 2006, 926, 1–12.
43. Seto, S.;
Tsujimura, K.; Horii, T.; Koide, Y. Autophagy Adaptor Protein p62/SQSTM1 and
Autophagy-Related Gene Atg5 Mediate Autophagosome Formation in Response to
Mycobacterium tuberculosis Infection in Dendritic Cells. PLoS
ONE 2013, 8, e86017.
44. Park, J.Y.;
Park, D.H.; Jeon, Y.; Kim, Y.-J.; Lee, J.; Shin, M.-S.; Kang, K.S.; Hwang,
G.S.; Kim, H.Y.; Yamabe, N. Eupatilin inhibits angiogenesis-mediated human
hepatocellular metastasis by reducing MMP-2 and VEGF signaling. Bioorganic
Med. Chem. Lett. 2018, 28, 3150–3154.
45. Park, B.B.;
Yoon, J.S.; Kim, E.S.; Choi, J.; Won, Y.-W.; Choi, J.H.; Lee, Y.Y. Inhibitory
effects of eupatilin on tumor invasion of human gastric cancer MKN-1
cells. Tumor Biol. 2013, 34, 875–885.