ArticlePDF AvailableLiterature Review

Daphnetin: A bioactive natural coumarin with diverse therapeutic potentials

Frontiers
Frontiers in Pharmacology
Authors:

Abstract

Daphnetin (DAP), a coumarin derivative extracted from Daphne species, is biologically active phytochemical with copious bioactivities including anti-inflammatory, anti-oxidant, neuroprotective, analgesic, antipyretic, antimalarial, anti-bacterial, anti-arthritic, neuroprotective, hepatoprotective, nephroprotective and, anti-cancer activities. It is reported to interact with multiple cellular mediators and signaling pathways to provide protection against neurodegeneration and arthritis. This review focuses on sources, synthesis, structure activity relationship, and various bioactivities of DAP. Neuroprotective and anti-inflammatory action of DAP is additionally aided by its modulation of the JNK-MAPK, JAK-STAT, and TLR-4/NF-κB signaling pathways. Although, the part of its anti-arthritic effect is mediated through immunoregulation, antioxidant and anti-inflammatory actions via regulation of NF-κB, MAPK and MMP signaling pathways, the anticancer action of DAP is mediated due to inhibition of Akt/ NF-Kb, MAPK signaling pathways, and the activation of Keap1-Nrf2 pathway. It is devoid of any organ toxicity and mortality as well as mutagenicity, mucosal irritation and sensitization reactions. Based on a review of the literature, DAP has a promising pharmacological and safety profiles and can be employed as a pharmaceutical moiety to treat a variety of illnesses. The current review intends to provide an in-depth focus on pharmacological activity and phytoanalytical approaches of DAP.
Daphnetin: A bioactive natural
coumarin with diverse
therapeutic potentials
Maira Javed
1
, Ammara Saleem
1
*, Anne Xaveria
1
and
Muhammad Furqan Akhtar
2
*
1
Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University
Faisalabad, Faisalabad, Pakistan,
2
Riphah Institute of Pharmaceutical Sciences, Riphah International
University, Lahore Campus, Lahore, Pakistan
Daphnetin (DAP), a coumarin derivative extracted from Daphne species, is
biologically active phytochemical with copious bioactivities including anti-
inammatory, anti-oxidant, neuroprotective, analgesic, anti-pyretic, anti-
malarial, anti-bacterial, anti-arthritic, neuroprotective, hepatoprotective,
nephroprotective, and anti-cancer activities. A wide range of studies have
been conducted exploring the signicance and therapeutic potential of DAP.
This study reviewed various databases such as NCBI, PubMed, Web of Science,
Scopus and Google Scholar for published research articles regarding the
sources, synthesis, and various bioactivities of DAP using different key
words, including but not limited to pharmacological activities,”“sources,
neuroprotective effect,”“synthesis,”“cancer,”“anti-inammatory effectof
daphnetin.Furthermore, this review encompasses both in-vivo and in-vitro
studies on DAP for treating various diseases. A comprehensive review of the
literature revealed that the DAP had a promising pharmacological and safety
prole, and could be employed as a pharmaceutical moiety to treat a variety of
illnesses including microbial infections, cancer, arthritis, hepatic damage,
inammation and neurological anomalies. The current review intends to
provide an in-depth focus on all pharmacological activities and therapeutic
approaches for the pharmaceutical and biomedical researchers.
KEYWORDS
daphnetin, neuroprotective, anti-inammatory, anti-bacterial, psoriasis
1 Introduction
Phytochemicals are secondary metabolites that naturally exist in plants. These are
categorized into various groups based on their chemical structure (Martinez et al., 2017).
Coumarin is a naturally occurring secondary metabolite and benzopyrone derivative. This
was one of the rst metabolites to be identied in the 1930s, and found in a variety of
plant species (Archbold et al., 2011;Xu et al., 2011;Amin et al., 2014). A signicant
number of researches have been conducted to identify individual compounds, and
develop procedures for their detection, synthesis, effectiveness and toxicity (Sovrlić
and Manojlović, 2017). Since then, over 50 coumarins have been discovered in
OPEN ACCESS
EDITED BY
Mansour Sobeh,
Mohammed VI Polytechnic University,
Morocco
REVIEWED BY
Gabin Bitchagno,
Mohammed VI Polytechnic University,
Morocco
Latifa Bouissane,
Université Sultan Moulay Slimane,
Morocco
*CORRESPONDENCE
Ammara Saleem,
ammarasaleem@gcuf.edu.pk,
amarafurqan786@hotmail.com
Muhammad Furqan Akhtar,
furqan.pharmacist@gmail.com
SPECIALTY SECTION
This article was submitted to
Ethnopharmacology,
a section of the journal
Frontiers in Pharmacology
RECEIVED 13 July 2022
ACCEPTED 09 September 2022
PUBLISHED 29 September 2022
CITATION
Javed M, Saleem A, Xaveria A and
Akhtar MF (2022), Daphnetin: A
bioactive natural coumarin with diverse
therapeutic potentials.
Front. Pharmacol. 13:993562.
doi: 10.3389/fphar.2022.993562
COPYRIGHT
© 2022 Javed, Saleem, Xaveria and
Akhtar. This is an open-access article
distributed under the terms of the
Creative Commons Attribution License
(CC BY). The use, distribution or
reproduction in other forums is
permitted, provided the original
author(s) and the copyright owner(s) are
credited and that the original
publication in this journal is cited, in
accordance with accepted academic
practice. No use, distribution or
reproduction is permitted which does
not comply with these terms.
Frontiers in Pharmacology frontiersin.org01
TYPE Review
PUBLISHED 29 September 2022
DOI 10.3389/fphar.2022.993562
Daphne species. Depending on their structure, there are simple,
dimeric or trimeric coumarins. Daphne contains various
coumarins including: daphnetin, DAP-8-glucoside, daphnin,
esculin, umbelliferone, and acetil-umbelliferon (Manojlović
et al., 2012;Sovrlićet al., 2015). Rutarensin, daphnoretin,
daphneretusin-A, dimethyl-daphnoretin-7-O-glucoside are
categorized among dimeric coumarins while trimeric
coumarin metabolites i.e., daphneretusin B, and triumbellin
were also identied in Daphne species (Mansoor et al., 2013).
This review elaborates sources, pharmacological activities as well
as toxicity of daphnetin (DAP) so as to nd and explore its
therapeutic potential and promote the drug development.
DAP i.e., 7, 8-dihydroxycoumarin is generally an odorless
and tasteless white or off-white powder that is freely soluble in
ethanol, methanol and dimethyl-sulfoxide while slightly
soluble in water (Shan et al., 2011). It has a molecular
weight of 178.14 g/mole and melting point 262.0°C(Liao
et al., 2013). It shows high solubility and permeability, and
is metabolized by phase 1 reaction through CYP3A4 with a
short half-life of 15 min. It exhibits poor bioavailability and
absorbs through the intestine by passive diffusion. It is
metabolized to methyl, glucuronide and sulfonate
conjugated metabolites (Yang et al., 1999;Du et al., 2009;
Shan, 2009;Liang et al., 2010;Chen, 2011;Zhang et al., 2014;
Liang et al., 2015;Liang et al., 2017;Xia et al., 2018a). Some
studies on DAP metabolism focused on glucuronidation, but
other studies provided glimpse of other conjugated
metabolites such as sulfonation, and methylation. In
comparison to glucuronidation and sulfonation, the
methylation pathway demonstrated a higher clearance rate
(Liang et al., 2016).
The DAP is derived from different Daphne species. Daphne is
a genus comprising 70 to 95 species of perennial and evergreen
shrubs of Thymelaeaceae family that is indigenous to India,
Europe, and North Africa. These plants are renowned for
their fragrant owers and brilliantly colored fruit (Riveiro
et al., 2010). DAP-8-glucoside is derived from D. odora in
which it is formed from DAP-7-glucoside (Ueno and Saito,
1976;Halda et al., 1998). Other sources of DAP include D.
gnidium (isolated from the leaves and stems), D. mezereum
(synthesized from shoots), D. giraldii, D. Koreana Nakai,D.
tangutica and D. oleoides. Seventeen compounds including DAP
were isolated from D. oleoides (Brown, 1986;Riaz et al., 2016;
Han et al., 2020;Khouchlaa et al., 2021). D. pedunculata leaves
and stems are also sources of DAP (Moshiashvili et al., 2020)as
shown in Figure 1.E. lathyris Linnaeus, ethnically known as
Euphorbia semenin East Asia, is also a source of coumarins
including DAP. Previously, it was reported that simple
coumarins including daphnetin, esculetin, esculin etc. Had
been isolated and identied from the seeds of E. semen
(Masamoto et al., 2003;Zhu et al., 2018). Different sources of
DAP are shown in Figure 1.
This review focusses on the DAP-based treatment and
prevention of diseases which are gradually receiving special
FIGURE 1
Different sources of daphnetin.
Frontiers in Pharmacology frontiersin.org02
Javed et al. 10.3389/fphar.2022.993562
attention due to underlying exceptional properties of DAP. In
this context, an overview of DAPs signicance as an essential
phytochemical and its intriguing uses have been presented and
addressed. Relevant data were collected using various search
engines such as Google scholar, PubMed, Web of Science,
NCBI and Scopus by using the various search terms such as
daphnetin,”“Structure activity of daphnetin,”“sources of
daphnetin,”“Synthesis of daphnetin,”“Traditional uses of
Daphne species,”“isolation,”“physical properties,
pharmacology of Daphentin”“hepatoprotective,
neuroprotective,”“anti-inammatory,”“anti-arthriticand
anti-cancerand toxicity of daphnetin,etc.
2 synthesis of Daphnetin
DAP, naturally occurring or synthesized, has oxygen-
containing heterocycles with a characteristic benzo-α-
pyrone framework (Xia et al., 2018b). It is biosynthesized
from shikimate pathway from L-Tyrosine and
L-Phenylalanine (Norma Francenia, 2019). Previously, it
was synthesized when pyrogallol and malic acid were
heated in concentrated H
2
SO
4
under nitrogen presence. It
is also synthesized from umbelliferone by hydroxylation as
shown in Figure 2 (NDong et al., 2003;Pan et al., 2017;Wang
et al., 2020a).
FIGURE 2
Synthesis of daphnetin.
FIGURE 3
Pharmacological and therapeutic targets of Daphnetin.
Frontiers in Pharmacology frontiersin.org03
Javed et al. 10.3389/fphar.2022.993562
TABLE 1 Pharmacological activities of Daphnetin.
Pharmacological
action
In-vivo/
In -vitro
study
Cell line/
Animal
Method Dose Molecular mechanism Effects/Targets References
Anti-stress In-vivo/in-
vitro
Kunming mice
Cortical neurons
from SD rat brains
Unpredictable stressor 2 and 8 mg/kg GRs in spatial learning and memory improves the
cognitive decits caused by chronic stress
Liao et al. (2013)
Neuroprotective In-vivo E18 C57BI/6 mice NMDA induced
excitotoxicity
20 and 40 mg/kg × NR2B-containing NMDA
receptors
× apoptosis × calcium overload Yang et al. (2014)
Hepatoprotective In-vivo In-
vitro
male C57BL/6 mice Oxidative stress induced
hepatotoxicity
20, 40 and
80 mg/kg
Keap1-Nrf2/ARE-Trx-
1ASK1/JNK, P53 protein
t-BHP in HepG2 cells Nrf2/Trx-1 GSH, ROS Lv et al. (2020)
Helicobacter Pylori
infection
In-vitro 20 H. pyloristrains
isloted from gastric
antrum
6.25 or
12.5 μg/ML
DNA damage,
recA ×membrane changes
bab A, urel transcription and H. pylori adhesion to
GES-1 cell line
Wang et al. (2019)
Lung protection In-vitro/
In-vivo
Mice Endotoxin induced Lung
injury
5, 10 mg/kg × activation of macrophage and
human alveolar epithelial cells,
induced TNFAIP3 pro-
inammatory cytokines
NF-Kb related signal pathway, anti-inammatory
potential
Yu et al. (2014b)
In-vivo C57BL/6 mice L-arginine 2-4 mg/kg i.p IL-6, TNF-α, MPO JAK-2,
STAT-3
inltration and cytokine secretion in inammatory
cells
Yang et al. (2021c)
Rheumatoid arthritis In-vivo Rats Freunds complete
adjuvant induced arthritis
2.25 and
4.5 mg/kg
IL-1, TNF-αand MIF paw swelling and arthritic scores × inammatory cells
inltration and articular cartilage degeneration
(Gao et al., 2008;Yao
et al., 2011;Tu et al.,
2012)
In-vivo Female Wistar rats Collagen induced arthritis 1 and 4 mg/kg Foxp3 Th1/Th2/Th17 Yao et al. (2011)
activity of Th17 RORγt, NF-
KB, CD77 IL-10 Tregs
paw swelling ×hyperplasia of synovial, destruction
and degeneration of chondrocytes Modulate balance of
Th17 and Tregs
Osteoporosis In-vivo In-
vitro
Sprague Dawley
rats MC3T
3-
E
1
pre
osteoblasts
Dexamethasone induced 1 and 4 mg/kg activate Wnt/GSK-3β/ßcatenin
signaling pathway
body weight gain, bone mineral content and
microstructure parameters
Wang et al. (2020b)
osteoblast proliferation, differentiation and
mineralization
Hepatoprotective In-vivo Mice Lipopolysaccharide/
D-galactosamine induced
liver failure
20, 40, 80 mg/kg × Inos × COX-2 autophagy
pro-autophagy protein
expression
ALT, AST pro-inammatory cytokines MDA
myeloperoxidase GSH, SOD level × MAPK, NF-kβ,
NLRP3
Lv et al. (2018)
Liver cancer In-vivo Huh7 and SK-
HEP-1
0, 5, 10, 50 and
100 µM
G1 phase arrest ×cell viability Liu et al. (2022)
×tumorigenesis
cell apoptosis
×Wnt/ßcatenin signaling
(Continued on following page)
Frontiers in Pharmacology frontiersin.org04
Javed et al. 10.3389/fphar.2022.993562
TABLE 1 (Continued) Pharmacological activities of Daphnetin.
Pharmacological
action
In-vivo/
In -vitro
study
Cell line/
Animal
Method Dose Molecular mechanism Effects/Targets References
Nephroprotective In-vivo C57BL/6 mice Cisplatin induced
nephrotoxicity
2.5,5,10 mg/kg TNF- α,IL-1β, ROS, MDA ×NF-kB signaling pathway activate Nrf2 pathway Zhang et al. (2018)
BUN, creatinine
renal injury
inammation, oxidative stress, apoptosis
In-vivo WT and Nrf2 mice Cisplatin induced
nephrotoxicity
20-40 mg/kg SOD, GSH, SIRT1, SIRT6, HO-
1, Nrf2, NQO1 MDA, MPO
BUN, creatinine renal injury inammation,
oxidative stress, apoptosis
Fan et al. (2020)
Diabetic nephropathy In-vivo mesangial cells High Glucose induced 0, 10, 20, 40 μMNrf2 ×p-Akt ×p-p65 ROS, MDA TNF- α,IL-1βIL-6, bronectin
collagen IV SOD activity cell proliferation
Xu et al. (2019)
Cerebral Ischemia/
Reperfusion injury
In -vivo C57BL/6mice Middle cerebral artery
occlusion
5, 10, 20 mg/kg TNF- α,IL-1β, IL-6, TLR4 ×TLR4/NF-kβIkBαdegradation neural cell
apoptosis
Liu et al. (2016a)
In-vitro Hippocampal
neuron
Reoxygenation induced
lung injury
10, 20 and 40 µm Nrf2 HO1 × oxidative stress and neuronal apoptosis Zhi et al. (2019)
Ischemic brain injury In-vitro HT22 cells glutamate induced toxicity
in hippocampal HT22 cell
5, 10, 25, 50, 75,
and 100 μM/L
×NF-kB pathway SOD, GSH ×TLR4/NF-kB pathway Du et al. (2014)
Microglial activation In-vivo Murine microglia Intracellular signal
transduction
0160 µm ×iNOS ×COX-2 TNF-α,IL-1β, IL-6 NO, ×microglial activation
Th17 development ×NF-kB × MAPK × IKK /IkB PI-
3K/Akt
Yu et al. (2014a)
Psoriasis In-vitroIn-
vitro
HaCaT human
keratinocytes in
Mice
imiquimod induced
psoriasis like skin lesion
50-100 mg/ cm IL-1β, IL-6, IL-8, IL-17A, TNF-
α, IL23A, MCP-1
pathway ×p65 phosphorylation ×nuclear translocation
erythema scaling, epidermal hyperplasia,
inammatory cells inltration
Gao et al. (2020)
Cell proliferation and
Estrogenicity
In-vivo In-
vitro
MCF-7 cells Female
mice
17.5, 35, 70,
140 mg/kg
Cyclin D1 p27 GO phase G1 phase S phase G2 phase M phase
cyclin/CDK2 cyclin D1/CDK4 ×cyclin D1
Jiménez-Orozco et al.
(2011)
Leukemia In-vivo Albino Wistar rats Benzene induced 12.5, 25,
50 mg/kg
sphingosine1-phosphate
receptor-1 SGOT Cytochrome
P450 CYP2E1
NF-kB Hematological parameter nucleated bone
marrow cells megakaryocyte , SOD, GSH MDA, 8-
OhdG albumin, total protein BUN, bilirubin
prothrombin time
Pei et al. (2021)
Ovarian cancer In-vitro A2780 5, 10, 20,
40 μg/ml
p-Akt p-mTOR p-AMPK,
LC3 II, p62
ROS production ×cell proliferation apoptosis,
autophagy, Blood count, Hemoglobin
proinammatory cytokines
Fan et al. (2021a)
Human renal cell
carcinoma
In-vitro A-498 cells 10 and 50 µm p38 MAP kinase cytokeratin
8 and 18
MAPK Signaling pathway ×ERK1/ERK2 pathway ×S
phase transition ×DNA synthesis
Finn et al. (2004)
(Continued on following page)
Frontiers in Pharmacology frontiersin.org05
Javed et al. 10.3389/fphar.2022.993562
TABLE 1 (Continued) Pharmacological activities of Daphnetin.
Pharmacological
action
In-vivo/
In -vitro
study
Cell line/
Animal
Method Dose Molecular mechanism Effects/Targets References
Corneal inammation and
neovascularization
In-vivo Male ICR mice Alkali burn (10-20 μmol/L)
DAP
eyedrops, q.i.d
HUVECs STAT3, ERK, AKT ×corneal inammation (VEGF-A) and
neovascularization (TLR4/NLRP3)
Yang et al. (2022)
In various tumors In-vivo Female in bred
BDF1 C57Bl/6
S180 sarcoma, MXT breast
adenocarcino ma,
C26 colon carcinoma
10,20 and
40 mg/kg
p38 MAP kinase cytokeratin
8 and 18 pro-apoptotic
caspase-3
×mitogenic pathway Cyclin D1 ×S phase ×Akt/ NF-kβ
pathway ×proliferation
Jiménez-Orozco et al.
(2020)
In-vivo Murine Osteosarcoma LM8 cells 30-60 µm RhoA Cdc
42
intracellular stress bers and lopodia Fukuda et al. (2016)
Mitochondrial
dysfunction and cell death
In-vitro C57Bl/6 mice Tert-butyl hydroperoxide 2.5,5,10 µg/ml HO-1, SOD NADPH, NQO1,
GCLM GCLC, BCl2 Bax,
Caspase 3
×ROS production ×cytochrome c release,
NLRP3 activation Nrf2 pathway activate JNK
and ERK
Lv et al. (2017)
CFA induced
inammatory pain
In-vivo Murine CFA 4 and 8 mg/kg spinal pro-inammatory
cytokines
×spinal glial activation × NF-kβpathway
Nrf2 pathway/HO-1 signaling pathway
Yang et al. (2021a)
Inammatory bowel
disease
In-vivo Mice Fecal transplantation 16, 8, 4 mg/kg T reg cells development Th 17 cell differentiation Ji et al. (2019)
Lipid metabolism In-vitro HepG2 cells 5, 20 and 50 µm PNPLA3 TG Liu et al. (2019)
Insulin resistance In-vitro HepG2 cells 20 and 50 µm pAKT/AKT P13K glucose uptake Liu et al. (2019)
Oxidative stress In-vitro HepG2 cells 5, 20, and 50 µm CYP2E1 and CYP4A Nrf2 oxidative stress Liu et al. (2019)
Angiogenesis In-vivo Rat TNF and VEGF induced 9.375900 µM c-Src, FAK, ERK1/2, Akt,
VEGFR2, iNOS, MMP2
× angiogenesis ×migration ×invasion ×tube
formation × NF-kβpathway ×TNF-αinduced IkBα
degradation ×translocation of the NF- kβp65 protein
apoptosis
Kumar et al. (2016b)
Abbreviations: Inhibits; : Upregulates, Increase; : Downregulates, Decrease; CUS, chronic unpredictable stress; GRs: Glucocorticoid receptors; ALF, acute liver failure; APAP, acetaminophen; ASK1, Apoptosis signaling-regulating kinase 1; AREs,
Antioxidant response elements; HO-1, Heme oxygenase-1; JNK, c-Jun N-terminal kinase; NF-κB, Nuclear factor-kappaB; Nrf2, Nuclear factor erythroid 2-related factor; 2NLRP3, Nucleotide-binding domain-like receptor protein 3 ROS, reactive oxygen
species; Trx-1, Thioredoxin-1; Txnip, Thioredoxin-interacting protein; PALI, pancreatic acute lung injury; JAK-2, Janus kinase-2; STAT-3, Signal transducer and activator of transcription 3; VEGFR2, Vascular endothelial growth factor 2; iNOS, inducible
nitric oxide synthase.
Frontiers in Pharmacology frontiersin.org06
Javed et al. 10.3389/fphar.2022.993562
3 Pharmacological activities of
Daphnetin
The DAP has been used to treat coagulation disorders,
various skin diseases, rheumatoid arthritis (RA), cancer,
lumbago, and fever (Tu et al., 2012;Wang et al., 2013). It
exhibited numerous pharmacological activities, including
analgesic, anti-pyretic (Singh et al., 2021a), anti-arthritic, anti-
inammatory, anti-oxidant (Qi et al., 2016;Lv et al., 2018), anti-
proliferative (Fylaktakidou et al., 2004;Kostova et al., 2011), anti-
bacterial (Cottigli et al., 2001), neuroprotective (Qi et al., 2016),
cardio-protective, nephroprotective, stroke, coagulation
disorders, ischemic brain injury, hepatoprotective and anti-
cancer activities (Pinto and Silva, 2017;Zhang et al., 2018;
Boulebd and Khodja, 2021) as mentioned in Figure 3. It has
been used for treating RA and coagulation disorders for long
duration without signicant toxic effects (Du et al., 2014).
Pharmacological actions of DAP are summarized in Table 1.
3.1 Neuroprotective action
Nerve cells interact with one another to carry out various
physiological functions. Any communication breakdown in the
brain, even in a solitary area, can impair the operation of other
brain regions and is the main factor in catastrophic neurological
illnesses or neurodegenerative disorders of the central and peripheral
nervous systems. Progressive neuronal degeneration causes
temporary or permanent sensory loss in a variety of
neurodegenerative disorders. The reactive oxygen species (ROS)
and inammatory signaling molecules i.e., tumor necrosis factor
alpha (TNF-α), and interleukin (IL)-6 are the key contributors to
neurodegenerative disorders (Qi et al., 2016;Chitnis and Weiner,
2017;Boulebd and Khodja, 2021). For neuroprotective action, DAP
reduces Toll-like receptor-4 (TLR-4), nuclear factor-ĸβ(NF-ĸβ), and
other pro-inammatory cytokines. It also inhibits JAK/STAT
phosphorylation which is responsible for the increase of pro-
inammatory cytokines and enzymes, culminating in the
reduction in COX-2 and inducible nitric oxide synthase (iNOS)
levels. It signicantly enhances the Nrf-2 expression. DAP is
reported to enhance Heat shock protein (HSP)-70 by
downregulating the expression of NF-ĸβand mitogen-activated
protein kinase (MAPK) at the molecular level, causing the
enzymes to regulate neuronal apoptosis by increasing or
decreasing the phosphorylation of pro-apoptotic proteins (Bax/
Bad) and an anti-apoptotic protein (Bcl-2) (Singh et al., 2021a).
3.1.1 Cognition and memory
DAP has shown signicant potential to prevent memory loss
and cognition. It inhibited apoptosis and calcium overload
induced by down-regulating NR2B-containing N-methyl-D-
aspartate (NMDA) receptors as well as calcium accumulation
which was activated by glutamate and caused excitation of
neurons. DAP exhibited neuroprotective properties by
preventing NMDA-induced neuronal cell loss and regulating
the balance of Bcl-2 and Bax expression in cortisol neurons of
mice (Yang et al., 2014).
The neuroprotective effect of DAP has been reported in the
posterior cerebral artery occlusion (MCAO)/reperfusion mice
model. The DAP (1 mg/kg) showed a substantial reduction in
cerebral infarct volume (Du et al., 2014). It has a neuroprotective
effect in stressed mice on microglial activation and its subsequent
inammatory response. The pre-incubation with DAP
dramatically inhibited TNF-αand IL-1 production in
lipopolysaccharide (LPS) or ß-amyloid activated BV2 cells.
The MAPK and protein kinase B (Akt) pathways play a
negative role in the anti-inammatory action of DAP.
Furthermore, pre-treatment with Wortmannin, a PI-3 k/Akt
inhibitor, resulted in a signicant decrease in LPS-induced
TNF-αand nitric oxide generation in BV2 cells,
demonstrating an opposing role of the MAPK/Akt pathway in
mediating anti-inammatory effect of DAP. It also reduced the
expression of NF-κB to promote neuroprotection (Yu et al.,
2014a). The neuroprotective action of DAP is presented in
Figure 4.
In another study, the neuroprotective effect in mice was
studied by utilizing the middle cerebral artery occlusion
(MCAO)/R model. It reduced the extent of the MCAO/
R-induced cerebral infarct, neuronal apoptosis, and brain IL-
1β, IL-6, TLR-4/NF-kB, and TNF-αlevels in the cerebral cortex
(Vivier et al., 2016). Lei Shen and his coworkers showed that
DAP had reduced endotoxin lethality in a mouse model of LPS-
induced endotoxemia and suppressed the inammatory response
to LPS in Raw264.7 cells by inhibiting ROS generation, JAK1 and
JAK2, and enhancing suppression of STAT1 and
STAT3 phosphorylation, and ultimately prevented the
STAT1 and STAT3 transport in the nucleus (Shen et al., 2017).
Several preclinical researches have revealed that DAP had
improved spatial memory and depressed behavior. The chronic
unexpected stress mice model was used to assess the effect of
DAP. It improved the Chronic Unpredictable Stress (CUS)
impaired spatial memory as indicated by mouse performance
in the Morris water maze test. DAP (2 and 8 mg/kg) injection
reduced the immobilization time in a forced swim test compared
to the CUS-treated group, conrming the involvement of DAP in
improving spatial memory and restoring depressive behavior in
mice (Liao et al., 2013).
3.1.2 Stroke
Cerebral ischemia is caused by impaired blood ow and is
accompanied by an inammatory reaction, release of cytokines,
and inammatory mediators that play a pivotal role in the
development of stroke (Iadecola and Alexander, 2001). The
TLR4 is highly induced after reperfusion injury. In an earlier
study, DAP was used for treating cerebral ischemia/reperfusion
injury and it was found to exhibit neuroprotective and anti-
inammatory effect by inhibiting TLR4/NF-kβpathway,
Frontiers in Pharmacology frontiersin.org07
Javed et al. 10.3389/fphar.2022.993562
alleviating the production of inammatory cytokines and neural
cell apoptosis (Liu et al., 2016a). DAP (at 5, 10, 25, 50, 75, and
100 μM/L) displayed dose-dependent neuroprotective action in
glutamate-induced toxicity in hippocampal HT22 cells and
ischemic brain injury by restoring reduced glutathione (GSH)
and superoxide dismutase (SOD) (Du et al., 2014).
It was further found that DAP suppressed oxidative stress
and cell apoptosis in hippocampal neurons. It increased nuclear
translocation of nuclear factor erythroid 2 (Nrf2) and HO-1
expression in neurons exposed to reoxygenation-induced cell
injury at 10, 20, and 40 µM doses. DAP inhibited oxidative stress
and neuronal apoptosis by activating Nrf2/HO-1 signaling
pathway (Zhi et al., 2019).
3.2 Hepatoprotective action
Excessive generation of ROS causes signicant damage to cell
membrane phospholipids, proteins and DNA leading to a variety
of disorders (Blair, 2008;Mendes-Braz and Martins, 2018;Zhao
et al., 2018). DAP reduced hepatotoxicity caused by tert-butyl
hydroperoxide (t-BHP) and acetaminophen through the
modulating Nrf2/Trx-1 pathway. It effectively prevented
t-BHP-induced hepatotoxicity by regulating Nrf2/Trx-
1 pathway in HepG2 cells. Moreover, it inhibited ASK1/JNK
activation and decreased the acute liver failure (ALF),
cytochrome C, and Bax mitochondrial translocation, all of
which concomitantly restored the mitochondrial function. It is
also found that DAP inhibited inammatory reactions in the liver
by inactivating the thioredoxin-interacting protein (Txnip)/
NLRP3 inammasome. It also improved the Nrf2 nuclear
translocation and Trx-1 expression (Lv et al., 2020). It
inhibited MAPK, NF-kβ, nucleotide binding domain like
receptor protein 3 (NLRP3) and decreased the pro-
inammatory cytokines in acute liver failure (ALF) (Lv et al.,
2018). The hepatoprotective mechanism of DAP is given in
Figure 4.
A study reported that the DAP had reduced the lipid
accumulation within the hepatocytes by regulating PI3K
expression and pAKT/AKT levels. Moreover, it decreased
the insulin resistance by promoting the hepatocellular
glucose uptake through upregulating the expression of
Nrf2. In addition, DAP reduced the level of ROS in
hepatocytes by downregulating the expression of
CYP2E1 and CYP4A (Liu et al., 2019). In an earlier study,
DAP improved carbon tetrachloride (CCL4)-induced
biochemical changes. It decreased the CCL4 induced-lipid
peroxidation and boosted the antioxidant defense system.
DAP induced nuclear translocation of Nrf2 related factor 2 to
induce the expression of hydroxyl ion. Thus, DAP prevented
the hepatotoxicity induced by oxidative stress by activating
Nrf2-mediated hydroxyl ion expression (Mohamed et al.,
2014).
FIGURE 4
The molecular interaction of multiple mediators implicated in the neuroprotective effect of daphnetin. TLR-4: toll-like receptor -4; NF-ĸβ:
nuclear factor-ĸβ; Janus kinase: JAK/STAT; COX-2: cyclooxygenase -2; iNOS: inducible nitric oxide; Nrf-2: Nuclear factor erythroid 2-related factor;
MAPK: mitogen activated protein kinase; Bcl-2: pro-apoptotic proteins; Bax/Bad; anti-apoptotic protein; ROS: reactive oxygen species.
Frontiers in Pharmacology frontiersin.org08
Javed et al. 10.3389/fphar.2022.993562
3.3 Effect on heavy metal and endotoxin
induced lung injury
It is found that DAP exhibited a phenomenal anti-oxidant in
arsenic-induced cytotoxicity in human lung epithelial cells. DAP
(at 2.5, 5, 10 g/ml) progressively shielded Beas-2B cells from
NaAsO2-induced apoptosis as well as arsenic cytotoxicity via
Nrf2-dependent pathway and increased GSH level (Lv et al.,
2019).
Endotoxin is an important toxin precipitating lung injury
and is responsible for increased serum concentration of all
cytokines and growth factors (Rojas et al., 2005). DAP at
5 and 10 mg/kg provided considerable protection from
endotoxin-induced acute lung injury in mice by inhibiting
the activation of macrophages and human alveolar epithelial
cells through reducing the production of inammatory
mediators, induction of TNF-αinduced protein 3
(TNFAIP3) and decreasing the expression of iNOS and
NF-κB to attenuate inammation. It also downregulated
the phosphorylation of MAPKs including p38, extracellular
signal regulated kinase (ERK), and JNK kinases (Yu et al.,
2014b).
3.5 Anti-bacterial action
DAP was investigated for anti-bacterial activity against
Helicobacter pylori; a Gram-negative bacterium that usually
colonizes stomach causing gastritis and peptic ulcers (Zali,
2011). DAP showed virtuous activity against multidrug
resistant (MDR) H. pylori via enhancing DNA damage,
phosphatidylserine (PS) translocation, and recA expression
while downregulating blood group antigen binding adhesion
(babA) and urease l (urel) with the decrease in the attachment
of H. pylori to GES-1 cells with minimal inhibitory concentration
(MIC) from 25 to 100 μg/ml (Wang et al., 2019); (Walker and
Crabtree, 1998). It also showed antibacterial activity by
destroying cell wall and preventing membrane coherence of
Pseudomonas uorescens and Shewanella putrefaciens with
MIC of 0.16 and 0.08 mg/ml respectively (Liu et al., 2020). In
another study, the effect of DAP on the Ralstonia solanacearum
was investigated in which it was found that DAP had exhibited
strongest anti-bacterial effect due to the presence of
hydroxylation at C6, C7, or C8 which increased its anti-
bacterial effect by destructing the bio-membrane against R.
solanacearum with the MIC at 64 mg/L (Yang et al., 2016).
DAP (at 10 mg/kg i.p.) was used to treat bacterial
pneumonia caused by methicillin-resistant Staphylococcus
aureus (MRSA) in C57BL/6 mice. It protected against
inammation, tissue damage, and stimulated the mTOR-
dependent autophagy pathway, which resulted in the
increased bactericidal activity of macrophages by
suppressing ROS production (Zhang et al., 2019).
3.6 Anti-malarial action
Malaria is one of the major fatal diseases, affecting around
1 million people worldwide and leading to death (Hu et al., 2018).
DAP and its two derivatives, DAP78 and DAP79, have
demonstrated anti-malarial activity against Plasmodium
falciparum; nevertheless, DAP functions as an iron chelator,
and its anti-malarial potency decreased signicantly with time,
leading its chelating action to be abolished (Huang et al., 2006).
In another investigation, DAP was found to have a high iron
chelating activity when compared to the potent iron-chelator
desferroxamine B at different dosages (Mu et al., 2002). It caused
50% inhibition of
3
H-hypoxanthine incorporation by P.
falciparum at 25 and 40 µM. DAP did not immediately
generate superoxide under in-vitro conditions, therefore it is
not considered an oxidant. However, during in-vivo studies, it
signicantly prolonged the survival of mice infected with P. yoelli
(Yang et al., 1992). Wang et al. (2000) reported the schizontocidal
activity of DAP by using P. falciparum FCC1 strain in-vitro. The
in-vivo activity was evaluated against P. berghei in Anka mice at
the dosage of 10100 mg/kg/day which demonstrated positive
outcome.
3.7 Anti-inammatory and anti-arthritic
actions
The excess endogenous production of ROS leads to oxidative
stress due to decreased concentration of GSH, SOD, and
increased level of malondialdehyde (MDA). Oxidative stress
also causes activation of the NF-κB pathway. This pathway
controls the release of different cytokines by directing the
expression of number of pro-inammatory cytokines,
inhibiting the apoptosis proteins (IAPS) and COX-2 which
leads to inammation. For anti-inammatory action, DAP
inhibits these pathways.
Adjuvant-induced arthritis is an autoimmune disorder
characterized by chronic inammation of joints that exhibits
the same pathological response as that of RA (Connor et al.,
1995). Various pro-inammatory mediators play a signicant
role in the pathogenesis of this disorder (Barsante et al., 2005).
DAP signicantly attenuated the poly-arthritis by suppressing
the production of pro-inammatory cytokines (IL-1 and TNF-α)
(Gao et al., 2008).
In another study, DAP alleviated the inammation and
pathological changes in the joint tissue, synovial hyperplasia,
and chondrocyte degeneration in collagen-induced arthritis
(CIA) in female rats at 1 and 4 mg/kg by restoring the
expression of Th1/Th2/Th17 type cytokines, Foxp3, IL-17, IL-
6, TGF- β, IL-4, and IFN- γ(Tu et al., 2012). In another study, it
inhibited the proliferation of broblast-like synoviocytes (FLS) in
rats with CIA and induced apoptosis by suppressing PI3k/AKT/
MTOR signaling pathway at 0-60 μg/ml (Deng et al., 2020). In
Frontiers in Pharmacology frontiersin.org09
Javed et al. 10.3389/fphar.2022.993562
another study, DAP was combined with B cell lymphoma
2 targeted small interfering RNA (si-Bcl2) on broblast-like
synoviocytes (FLS) in rats with CIA by downregulation of
Bcl2. When si-Bcl2 was combined with DAP (40 μg/ml), it
increased the effect via promoting apoptosis on RAFLS and
by reducing the expression of Bcl2 and STAT3 (Chen et al.,
2018).
In a previous study, DAP reduced the serum level of Th17,
Th2, and Th1 type cells and upregulated the levels of Tregs in
arthritis rats at 1 and 4 mg/kg. It also decreased RORγt, NF-kB,
and CD77 in joint tissue while increased the expression of Foxp
3
and IL-10. Thus it modulated the balance of Tregs and Th
17
cells
and is considered to be an effective agent in the treatment of CIA
in rats (Yao et al., 2011).
Zhang et al. (2020) reported the chondroprotective effect of
DAP against osteoarthritis. DAP (at 12, 24 and 48 ng/ml)
profoundly protected chondrocytes of rabbits by averting IL-
1β, -6,-12, MMP3,-9, and -13 and decreasing the caspase-3 and
BAX while increasing BCL-2. In another study, DAP exhibited
anti-arthritic action by demethylation of pro-apoptotic genes in
synovial cells (FasL and P53). For this purpose, MTT analysis was
performed on CIA-treated rat synovial cells to determine the
inhibitory effect of DAP and DNA methyltransferase inhibitor
drug (5-aza-dc) in the range of 1.25-40 μg/ml. It inhibited cell
growth in synovial cells in a dose and time dependent manner
(Shu et al., 2014). Zheng et al. reported anti-arthritic action of
DAP using collagen induced FLS. It escalated caspase 3, 8, and 9,
Bax, FasL, and cytochrome c (Cyt-c) with the reduction in Bcl-2
and enhanced the Cyt-c discharge from mitochondria to the
cytosol (Zheng et al., 2020). In another study, DAP at 4 and
8 mg/kg inhibited spinal glial activation in murine mice
provoked by CFA. It also decreased the expression of pro-
inammatory cytokines. It inhibited the NF-κβ pathway and
activated the Nrf2/HO-
1
signaling pathway (Yang et al., 2021a).
3.8 Osteoporosis
Glucocorticoids are effective agents in treating inammatory
and autoimmune diseases. While its long-term usage results in
osteoporosis. DAP at 1 and 4 mg/kg exhibited the therapeutic
action against dexamethasone induced osteoporosis in male rats
by restoring bone mineral content, microstructure parameters,
and bone turnover. In-vitro, it promoted osteoblast proliferation,
differentiation, and mineralization in pre-osteoblasts by
activating Wnt/GSK-3β/βcatenin signaling pathway (Wang
et al., 2020b).
3.9 Multiple sclerosis
Multiple sclerosis (MS) is an inammatory and
neurodegenerative illness that is identied by projected
inammation, axonal injury, and demyelination. DAP
(8 mg/kg) has exhibited an immune-regulatory role in
autoimmune encephalomyelitis in the murine model used for
MS (Leung et al., 2011). Autoimmune encephalomyelitis is a
demyelinating inammatory illness of the central nervous system
caused in experimental animals by an immune response to
myelin epitopes (Fletcher et al., 2010). T cells attract
macrophages, microglia, and astrocytes which release
inammatory mediators such as nitric oxide (NO), and ROS.
DAP therapy lowered the level of pro-inammatory cytokines,
induced heme oxygenase-1 (HO-1), decreased the level of MDA,
and displayed anti-inammatory and neuroprotective effects in
mice at 8 mg/kg (Wang et al., 2020c). In a previous study, DAP
administered for 28 days mitigated the encephalomyelitis in mice
via suppressing the activation, maturation, and antigen-
presenting capability of Dendritic cells, and regulated NF-κB
signaling (Wang et al., 2016).
3.10 Systemic lupus erythematosus
Li et al. reported the anti-inammatory potential of DAP in
the NZB/WF1 systemic lupus erythematosus (SLE) murine
model. In the SLE-prone NZB/W F1 mice, DAP (at 5 mg/kg)
treatment enhanced the survival rates, reduced renal damage and
blood urea nitrogen levels, and lowered the serum autoantibody
production. Furthermore, its therapy signicantly reduced the
serum levels of TNF-αand IL-6, inhibited NF-kB activity,
lowered the nuclear factor of activated T-cell protein
production, and increased the A20 protein expression in SLE-
prone NZB/W F1 mice. Finally, DAP reduced the inammation
in the NZB/WF1 murine SLE model via NF-κB suppression
mediated by A20 overexpression (Li et al., 2017).
3.11 Anti-psoriasis action
Psoriasis is a chronic inammatory disease of the skin
characterized by excessive proliferation, abnormal
differentiation of keratinocytes, and inltration of
inammatory cells into the epidermis and dermis.
Hyperproliferation of keratinocytes and extreme inammatory
response play a pivotal role in its pathogenesis. Cytokines
secreted by immune cells cause keratinocytes
hyperproliferation which produces pro-inammatory
cytokines to potentiate inammatory response. A previous
study showed the anti-psoriatic activity of DAP in HaCaT
keratinocytes mouse which occurred through the
downregulation of inammatory cytokines and suppression of
NF-κB signaling pathway (Gao et al., 2020). DAP also decreased
the epidermal hyperplasia and inltration of inammatory cells
in imiquimod induced skin lesions in mice. In another research,
DAP above 40 μM caused a decrease in cell viability in human
Frontiers in Pharmacology frontiersin.org10
Javed et al. 10.3389/fphar.2022.993562
HaCaT keratinocytes by upregulation of IL-1, -6, -8, TNF-α, and
IL-23A while inhibiting P65 phosphorylation and nuclear
translocation. Additionally, it improved the inammation,
erythema, scaling, and epidermal thickness of psoriatic mice
(Gao et al., 2020).
3.12 Anticancer action
DAP is known for anticancer potential against leukemia,
ovary, kidney, colon, and liver cancers. Uncontrolled
proliferation and suppression of apoptosis lead to cancer.
Mitogen pathways are responsible for regulating apoptosis.
DAP is a protein kinase inhibitor; therefore, it signicantly
suppresses this pathway and acts as an anti-proliferative
agent. It also acts at different phases of the cell cycle. DAP
inactivates Akt/NF-κB (an anti-apoptotic pathway), JNK,
MAPK, and ERK pathways that are responsible for causing
cancer. DAP activated Keap1-Nrf2 pathway that protected the
cell against oxidative stress by activating transcription of several
cytoprotective genes thus helping to combat cancer (Figure 4)
(Jiménez-Orozco et al., 2020). In a previous study, effect of DAP
(at 2.5, 5, and 10 μg/ml) on tert-butyl hydroperoxide (t-BHP)
induced mitochondrial dysfunction and cell death in C57B1/
6 mice and RAW 264.7 cells revealed that the DAP suppressed
the production of ROS by stimulating various anti-oxidant genes
and activating Nrf2 pathway that protected the body against
oxidative damage. Activation of Nrf2 pathway suppressed
NLRP3 activation, thus inhibiting the activation of caspases
and release of pro-inammatory cytokines. In this way, DAP
protected the body from cell death and mitochondrial
dysfunction (Lv et al., 2017).
In another study, anti-proliferative properties of DAP in
cancer cells were reported. DAP inhibited migration and
invasion of highly metastatic murine osteosarcoma LM8 cells.
It reduced the intracellular stress bers and lopodia. It also
decreased the expressions of RhoA and Cdc
42
(Fukuda et al.,
2016).
3.12.1 Kidney cancer
The human renal cell carcinoma (RCC) accounts for up to
90% of kidney cancers due to the alterations of the genes
responsible for controlling cell division (Motzer et al., 1996;
Hsieh et al., 2017). MAPKs pathway causes activation of
transcription factors which in turn regulate gene expression,
thus controlling the cell growth, differentiation, and
proliferation. ERK pathway also controls the proliferation and
differentiation and survival of cells. DAP prevented the RCC
proliferation by inhibiting ERK/MAPK pathway and upregulated
the differentiation mediated by p38 MAP kinase. It also
suppressed the G
1
to S phase transition by inhibiting DNA
synthesis at 10 and 50 µM in AQ-498 cells (Finn et al., 2004).
The anticancer mechanism of DAP is shown in Figure 4.
p38 MAP kinase is intrinsically involved in mediating the
effect of DAP in A-498 cells. Moreover, DAP is involved in
promoting the cellular maturation and is considered to be a new
less toxic approach for treating poorly differentiated RCC (Finn
et al., 2004).
3.12.2 Ovarian cancer
Ovarian cancer is the sixth most common cancer among
European women (Colombo et al., 2006). Autophagy, apoptosis,
and ROS production can trigger cell death and help to treat
cancer. AMPK/Akt/mTOR pathway is associated with autophagy
and apoptosis. In an earlier study, DAP exhibited the anticancer
potential in A2780 xenograft tumor model against ovarian cancer
at 0, 5, 10, 20, and 40 μg/ml in-vitro and 30 mg/kg in-vivo by
inducing cell death, increasing ROS production, inducing
autophagy, and inhibiting the cell proliferation (Fan et al.,
2021a).
3.12.3 Leukemia
Benzene is a chemical present in the atmosphere that can
cause different types of leukemia. Exposure to the vapors of
benzene leads to oxidative damage, inammatory responses,
changes in cell cycle progression, and DNA damage (Huff,
2007). In a benzene-induced leukemia study, treatment of rats
with DAP at 12.5, 25, and 50 mg/kg caused an increased blood
count, and hemoglobin concentration, reduced the level of
inammatory mediators, and inhibited ROS production to
retard cancer progression (Pei et al., 2021).
3.12.4 Liver cancer
A previous study reported the therapeutic potential of DAP
against liver cancer. Hepatocellular carcinoma (HCC) was
induced in Wistar rats by diethyl nitrosamine (DEN)
(200 mg/kg) and its effect was enhanced by phenobarbital for
4 weeks. DAP (at 10, 20, and 30 mg/kg) repressed the
biochemical parameters with enhanced levels of GSH,
glutathione S-transferase (GST), SOD and CAT while
decreasing the level of MDA. It also reduced the
inammatory markers such as COX-2, NF-κB, prostaglandin
(PGE2), IL-1β, IL-6, and TNF-αin treated rats (Li et al., 2022).
In another study, DAP inhibited the progression of
hepatocellular carcinoma in Huh7 and SK-HEP-1 cell lines.
DAP suppressed the cell viability and tumorigenesis,
promoted the apoptosis of cells, and induced the arrest the
cells in G1 phase dose-dependently which were rescued by
SKL 2001, an activator of Wnt/β-catenin signaling. Thus,
DAP exerted an antitumor role through the inactivation of
Wnt/β-catenin signaling (Liu et al., 2022).
3.12.5 Breast cancer
Cell proliferation and estrogenicity lead to the tumor
development in breast. DAP acts at different phases of cell
cycle thus controlling cell proliferation and tumor
Frontiers in Pharmacology frontiersin.org11
Javed et al. 10.3389/fphar.2022.993562
development. Cyclin D1 is a major protein for the initiation of
cell cycle and proliferation of cells. In a previous study, DAP
suppressed cyclin D1, thereby preventing the proliferation in
MCF-7 cells. It is also a protein kinase inhibitor which leads to
the inhibition of proliferation. It did not possess estrogenic
activity (Jiménez-Orozco et al., 2011).
In another study. DAP inhibited p-AKT which reduced NF-
κB in mammary cancer. It was considered to be an effective agent
in the treatment of mammary cancer in rats by suppressing the
Nrf-2-Keap
1
pathway and NF- κB expression (Kumar et al.,
2016a).
3.13 Nephroprotective action
The DAP showed nephroprotective effect against
cisplatin-induced nephrotoxicity by suppressing the NF-κB
signaling pathway and activating the Nrf2 pathway when
C57BL/6miceweretreatedwithDAPat2.510 mg/kg. It
decreased the blood urea nitrogen and creatinine levels along
with the reduction of ROS (Zhang et al., 2018). Another study
stated that the DAP (at 40 mg/kg) restored the weight loss,
blood urea, kidney index, and creatinine levels in cisplatin-
induced acute nephrotoxicity. It remarkably increased sirtuins
(SIRT1, SIRT6) and Nrf2 with an increased SOD and GSH
levels, and the reduction in MDA and MPO levels in wild-type
mice (Fan et al., 2020). The nephroprotective mechanism of
DAP is shown in Figure 4. A study also reported the preventive
potential of DAP in diabetic nephropathy in mesangial cells at
10-40 µM by preventing cell proliferation, protection against
oxidative stress and inammation by targeting Nrf2/keap1,
and Akt/NF-kB inammatory pathways (Shen et al., 2017;Xu
et al., 2019).
DAP protected the mice from gentamicin-induced
nephrotoxicity at 40 mg/kg by preventing renal injury and
decreasing cell damage. It upregulated the expression of Nrf2,
and antioxidant enzymes such as HO-1, NQ0
1
, GCLC and
GCLM (Fan et al., 2021b).
3.14 Other actions of Daphnetin
Song et al. investigated the potential effect of DAP as
immunosuppressive agent in BALB/c mice using a 100 μl
emulsion comprising of 100 μg OVA as prototype antigen.
DAP (at 5, 10, and 20 mg/kg i.p.) downregulated the OVA-
specic antibody IgG subclasses IgG1 and IgG2b and reduced the
growth of Th1 and Th2 cytokines as well as restrained in-vivo
splenocytes proliferation (Song et al., 2021).
It was reported that the pretreatment with DAP (at 1, 10, 20,
and 40 µM) improved the cell viability in rat insulinoma (INS-1)
cells that were previously exposed to streptozotocin (STZ) as
compared to INS-1 cells (Negative control). It also improved the
insulin secretion in the INS-1 cells. Thus, the antidiabetic effect of
DAP relied on insulin stimulating, and antiapoptotic actions
(Vinayagam and Xu, 2017).
DAP (at 416 mg/kg) considerably improved the
experimental colitis by suppressing the colonic inammation,
improving colonic integrity, and restoring the immune and
metabolic homeostasis. It increased the abundance of short-
chain fatty acid producing microbiota of gut that were
responsible for the increased development of T
reg
cells and
the reduced pro-inammatory T
h
17 cell differentiation (Ji et al.,
2019).
Previously, it was found that the DAP inhibited melanin
biosynthesis by suppressing the expression of microphthalmia
associated transcription factor responsible for melanogenesis,
and also inhibited melanogenic enzymes such as tyrosine and
tyrosine-related proteins in B16F10 cells. DAP downregulated
the phosphorylation of kinases such as PKA, ERK, mitogen and
stress activated protein kinase (MSK)-1 and cAMP response
element binding protein (CREB). It inhibited the melanin
synthesis, and exhibited the anti-pigmentation activity by
modulating PKA/CREB, and ERK/MSK1/CREB pathways
(Nam et al., 2022).
DAP possesses analgesic action. In a previous study, DAP at
10 mg/kg averted reserpine induced bromyalgia (chronic pain
syndrome along with depression) in mice. DAP effectively
averted bromyalgia by downregulating monoamine oxidase-
A (MAO-A), glutamate level, IL-1β, and TNF-αwhile elevating
the GSH, dopamine, serotonin and norepinephrine levels (Singh
et al., 2021b).
A previous study reported that DAP reduced Toll-like
receptor-4 (TLR4) expression and suppressed the activation of
the NF-κB signaling pathway in acute pancreatitis showing its
potential to avert pancreatitis (Liu et al., 2016b). DAP as an
emulsion [locus bean gum (0.5%) and sodium alginate (1.5%)] is
used in the food industry as an additive, preservative and
packaging material. It is used as a preservative because of
antimicrobial and antioxidant properties (Liu et al., 2021;
Cheng, 2022).
Wen et al. investigated the percutaneous absorption of DAP
in rat abdominal skin using various chemical enhancers. The
experiment was performed using isopropyl myristate as a vehicle
while other enhances of O-acylmenthol derivatives were
synthesized from which only M-LA was explored to improve
the DAP permeation. Its effects were also pronounced when DAP
was used with span 80 (Wen et al., 2009).
Yang and his coworkers investigated the effect of DAP on
ischemia repurfusion (I/R) injury. DAP (at 2.5, 5,10, and
20 mg/kg) decreased the myocardial I/R injury with improved
cardiac function in treated cells. It reduced the apoptosis,
oxidative stress, and inammation both under in-vitro and in-
vivo experiments. It also reduced the risk of ventricular
arrhythmias by downregulation of TLR4, MyD88, and NF-κβ
in I/R mice (Yang et al., 2021b).
Frontiers in Pharmacology frontiersin.org12
Javed et al. 10.3389/fphar.2022.993562
In another study, DAP exhibited anti-angiogenic properties
through inhibition of different stages of angiogenesis such as
migration, invasion, and tube formation. It suppressed the NF-
κB pathway, TNF-αinduced IκBαdegradation and translocation
of the NF- κB-p65 protein. It signicantly decreased the
expression of c-Src, FAK, ERK1/2, Akt, VEGFR2, Inos, and
MMP2 as well as induced apoptosis (Kumar et al., 2016b).
DAP also exhibited the protection against LPS induced
inammatory bone destruction in murine osteolysis model. It
inhibited RANKL-induced osteoclast differentiation, fusion and
bone resorption. It inhibited the activation of ERK and
NFAT
c
1 signaling cascade, so it has the potential to be used
for the treatment of inammatory osteolysis (Wu et al., 2019).
Zhou and Zhang et al. investigated the in-vitro integration of
DAP-Cu (II) complex with calf thymus DNA (ctDNA). The
ndings exhibited that the DAP and Cu
2+
exerted synergistic
pharmacological actions (Zhou et al., 2016).
4 Toxicity studies
A previous study reported the Minimum inhibitory
concentration (MIC of DAP (25 μg/ml) against highly resistant H.
pylori isolated from human gastric antrum. Using the Cell Counting
Kit-8 (CCK-8), the sub-minimum inhibitory concentration (MIC) of
DAP was studied in GES-1 cells. DAP was well tolerated by GES-1
cells, and there was insignicant difference on cytotoxic effect of DAP
by culture media conditions (Wang et al., 2019). Furthermore, the
acute toxicity of DAP was evaluated using the Bacterial reverse
mutation assay (Ames test), which revealed no genetic toxicity.
Bone marrow micronucleus test revealed that DAP) had no effect
on mouse bone marrow cells at various concentrations (0.75, 1.5, and
3 mg/kg). DAP was prepared for an acute skin allergy test at a nal
concentration of 4 mg/ml, indicating that it is non-allergenic. A local
mucosa stimulation test was done on the oral mucosa of rabbits which
revealed no oral mucosa ulceration, erosion, erythema and irritation
caused by DAP.
In mice, the maximal oral toxic dose of DAP was greater than
100 mg/kg (Nanzhen et al., 2018). Hippocampal HT-22 cell line was
used with 5 mM glutamate and different concentrations of DAP.
After 12 h of incubation, 100 mΜDAP protected HT-22 cells in
concentration dependent manner against glutamate toxicity (Du
et al., 2014). BV2 microglia were used and treated with 0-160 μM
concentrations of DAP which showed insignicant change in cell
survival rate (Yu et al., 2014a). Indeed, the in-vitro and in-vivo
studies conrmed that DAP was devoid of any signicant toxicity at
pharmacologically relevant concentrations.
5 Structure activity relationship
There is a series of DAP derivatives ranging from 1-22 that
showed moderate inhibitory or activating action on GPCRs
resultantly responsible for copious pharmacological activities.
The activity of GPCRs depends on the chemical alteration of
hydroxyl groups at the C-7, C-8, and C-3/C-4 positions of DAP
(Satô and Hasegawa, 1969;Wang et al., 2020a). The C-7 and C-8
substituents were generated through phenolic O-acylation/
O-alkylation (nucleophilic acyl/alkyl substitution), whereas the
C-3 and C-4 substituents were formed using Pechmann
condensation. DAP 2, 3, 4, 5, 15, 16, 18, 19, and 20 cause
moderate activation on GPCRs while 3-5, and 19 exhibit
profound activation with EC50 of 1.181.91 nM (Wang et al.,
2020a).
Substitution at C-3 or C-4 of DAP produces different
derivatives. The anti-oxidant activities of different DAP
derivatives were evaluated. The catechol group was considered
a key pharmacophore for the anti-oxidant activity. The
introduction of electron-withdrawing hydrophilic group at the
C-4 position increased the anti-oxidant potential but it was not
observed with C-3 substitution. Introduction of the hydrophobic
phenyl group produced negative effect on the anti-oxidant
activity at C-3 and C-4. The 4-carboxymethyl DAP exerted
the most powerful anti-oxidant activity. It also displayed
strong metabolic stability (Dar et al., 2015;Xia et al., 2018b).
6 Discussion
The inclusion of multiple research investigations on
pharmacological mechanisms of DAP in treating numerous
chronic conditions was a main emphasis of the current study.
Consequently, the review offers a comprehensive outline of the
prospective medicinal uses of this phytochemical. The current study
uncovered the pharmacological and therapeutic benets of DAP for
human health. Several medicinal plants are rich source of bioactive
compounds which exhibit numerous pharmacological activities,
minimal side effects, and the potential source of novel drugs for
the treatment of diseases. Many drugs currently available in the
market have either been directly or indirectly derived from the
traditional plants. By reviewing the available information, it was
determined that DAP was a bioactive constituent with a variety of
effects against bacterial microorganisms, inammation, malarial
parasite, viral infections, cardiovascular diseases, rheumatoid
arthritis, kidney disorders, cerebral disorders, various cancers,
lung infections, melanogenesis, bowel diseases, oxidative stress,
diabetes mellitus, and others. However, its impact against
different cancer types are particularly important in the therapy of
numerous malignant diseases.
DAP is a simple coumarin derivative with a variety of
therapeutic benets in preclinical research and mostly isolated
from the Daphne genus.Thus, there are areas to work on the
isolation from other genus and synthesis in laboratory.
Furthermore, identication of various intermediate metabolites
may broaden the range of biologically active compounds to be
tested for various ailments.
Frontiers in Pharmacology frontiersin.org13
Javed et al. 10.3389/fphar.2022.993562
In numerous preclinical researches, DAPs neuroprotective
effects have been thoroughly documented. Studies have
demonstrated the protective effect of DAP against ischemic/
reperfusion injury, and spatial memory impairment caused by
CUS, NMDA-induced excitotoxicity, glutamate-excited HT-22
cells, as well as cerebral ischemia (Liao et al., 2013;Du et al., 2014;
Yang et al., 2014;Liu et al., 2016a;Berman and Bayati, 2018). The
neuroprotective effect can be signicantly achieved by the
modication of the TLR-4/NF-κB, HSP70, JAK/STAT, and
Nrf-2/HO
1
downstream pathways (Figure 4). DAPs potential
as a neuroprotective compound could further be supported in
preclinical research by examining its impact on the Aβamyloid,
tau, Parkinsons, and Huntingtin proteins.
In cancer, cells divide uncontrollably and metastasize other
tissues. The ndings of current review indicated that the DAP
was pharmacologically effective against different type of cancers
including cancers of kidney, liver, ovary and leukemia via
inhibiting the proliferation and promotion of apoptosis
making it a viable adjuvant in the treatment of cancer.
The inammation and oxidative stress are increased in severe
joint inammatory conditions such as osteoarthritis and RA. The
upsurge of pro-inammatory cytokines, NF-κB,
myeloperoxidase, iNOS, NOS, COX-2, and other mediators
worsen the disease (Prasad et al., 2021). Thus, the analysis of
previous studies indicated that the DAP was effective in retarding
the progression of RA and other inammatory diseases even at
low doses via inhibiting the pro-inammatory cytokines, NF-κB
and MMP levels, and restored the protein expressions.
The DAP and derivatives were effective against several
bacterial infections as well as different malarial parasites via
suppressing the metabolic functions of these microbes as
demonstrated by diverse in-vitro and in-vivo studies. Such
studies are need to be extended to antibiotic resistant
microbes so as to treat and prevent drug resistant infections.
The hepatoxicity could be due to infections, malignant diseases
and drug therapy. Previous investigations indicated that the DAP
had antioxidant potential and inhibited lipid and protein
oxidation, decreased ROS, and pro-inammatory cytokines to
prevent and treat hepatotoxicity (Khan et al., 2019).
Several intriguing pharmacological investigations on DAP
against different diseases have outlined the mechanisms
supporting the use of DAP as supplementary therapy. Further
research is needed on long-term toxicity, information on
potential medication interactions and its effect as adjuvant
therapy against chronic diseases. To further support the
clinical value in medical practice, supplementary clinical trials
are also required. Previous studies show that DAP is a suitable
candidate for the drug development.
7 Conclusion and future perspectives
This review provides pertinent information regarding the
pharmacological aspects of DAP to explore its hidden potential
as it targets various molecular and cellular pathways to combat
numerous inammatory disorders, infectious diseases,
neurological disorders, hepatotoxicity, nephrotoxicity,
psoriasis, diabetic nephropathy, leukemia, and other cancers.
DAP exhibited no mutagenic effect, allergenic action,
sensitization, mucosal irritation, erythema, and mortality in
toxicity studies. The information summarized above will be
used for the development of an effective formulation for the
treatment of various ailments without signicant adverse/toxic
effects.
On the basis of literature reviewed, it has been found that
DAP exhibited remarkable pharmacological prole and it could
be used as a treatment or adjuvant for the treatment of different
disorders. Thus, still its biosynthesis and structure activity
relationship should be critically analyzed. The effect of DAP
on H. pylori was investigated but other microorganisms causing
gastrointestinal infections should be investigated to reduce their
impact on human and animal health. The development of
nanoformulations of DAP need attention to enhance its
therapeutic effect and half-life. The synergistic effect of DAP
with commercially available drugs should be studied to
enhancing their effects in treating various diseases. As DAP
attenuates the activation of microglia that plays a crucial role
in the pathogenesis of multiple neurodegenerative diseases, this
evidence suggests the possibility of DAP as treatment option for
Huntington and Parkinsons disease. Further research should be
implicated to explore its various bioactivities and mechanisms.
Toxicity study of DAP and its derivatives should be conducted in
detail to assure their safety in human and animals (Lu et al.,
2021).
Author contributions
AS and MA: Conceptualization, visualization, review,
supervision, analysis, review, and editing. MJ, AX: Literature
search, collection of data, writing original draft.
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could
be construed as a potential conict of interest.
Frontiers in Pharmacology frontiersin.org14
Javed et al. 10.3389/fphar.2022.993562
Publishers note
All claims expressed in this article are solely those of the
authors and do not necessarily represent those of their afliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or
claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
References
Amin, K. M., Abdel Gawad, N. M., Abdel Rahman, D. E., and El Ashry, M. K. M.
(2014). New series of 6-substituted coumarin derivatives as effective factor Xa
inhibitors: Synthesis, in vivo antithrombotic evaluation and molecular docking.
Bioorg. Chem. 52, 3143. doi:10.1016/j.bioorg.2013.11.002
Archbold, J. K., Flanagan, J. U., Watkins, H. A., Gingell, J. J., and Hay, D. L.
(2011). Structural insights into RAMP modication of secretin family G protein-
coupled receptors: implications for drug development. Trends Pharmacol. Sci. 32
(10), 591600. doi:10.1016/j.tips.2011.05.007
Barsante, M. M., Roffe, E., Yokoro, C. M., Tafuri, W. L., Souza, D. G., Pinho, V.,
et al. (2005). Anti-inammatory and analgesic effects of atorvastatin in a rat model
of adjuvant-induced arthritis. Eur. J. Pharmacol. 516 (3), 282289. doi:10.1016/J.
EJPHAR.2005.05.005
Berman, T., and Bayati, A. (2018). What are neurodegenerative diseases and how
do they affect the brain? Front. Young Minds 6. doi:10.3389/frym.2018.00070
Blair, I. A. (2008). DNA adducts with lipid peroxidation products. J. Biol. Chem.
283 (23), 1554515549. doi:10.1074/jbc.R700051200
Boulebd, H., and Khodja, I. A. (2021). A detailed DFT-based study of the free
radical scavenging activity and mechanism of daphnetin in physiological
environments. Phytochemistry 189, 112831. doi:10.1016/j.phytochem.2021.112831
Brown, S. A., Biosynthesis of daphnetin in Daphne mezereum L. Z. für
Naturforsch. C, 1986. 41(3), 247252. doi:10.1515/znc-1986-0301
Chen, X., Kuang, N., Zeng, X., Zhang, Z., Li, Y., Liu, W., et al. (2018). Effects of
daphnetin combined with Bcl2-siRNA on antiapoptotic genes in synovial
broblasts of rats with collagen-induced arthritis. Mol. Med. Rep. 17 (1),
884890. doi:10.3892/mmr.2017.8008
Chen, L. (2011). Effects of Glycyrrhiza uralensis on rat intestinal absorption and
metabolism of daphnetin. Nanjing: Nanjing University of Chinese Medicine.
Cheng, H. (2022). Characterization of sodium alginatelocust bean gum lms
Reinforced with daphnetin Emulsions for the Development of active packaging.
Polymers 14, 731. doi:10.3390/polym14040731
Chitnis, T., and Weiner, H. L. (2017). CNS inammation and neurodegeneration.
J. Clin. Invest. 127 (10), 35773587. doi:10.1172/JCI90609
Colombo, N., Van Gorp, T., Parma, G., Amant, F., Gatta, G., Sessa, C., et al.
(2006). Ovarian cancer. Crit. Rev. Oncol. Hematol. 60 (2), 159179. doi:10.1016/j.
critrevonc.2006.03.004
Connor, J. R., Manning, P. T., Settle, S. L., Moore, W. M., Jerome, G. M., Webber,
R. K., et al. (1995). Suppression of adjuvant-induced arthritis by selective inhibition
of inducible nitric oxide synthase. Eur. J. Pharmacol. 273 (1), 1524. doi:10.1016/
0014-2999(94)00672-T
Cottigli, F., Loy, G., Garau, D., Floris, C., CasuM.Pompei, R., et al. (2001).
Antimicrobial evaluation of coumarins and avonoids from the stems of Daphne
gnidium L. Phytomedicine 8 (4), 302305. doi:10.1078/0944-7113-00036
Dar, M. Y., Ara, T., and Akbar, S. (2015). Isolation of daphnetin 8-methyl ether
from Daphne oleoides and its anti-bacterial activity. J. Phytopharm. 4 (4), 224226.
doi:10.31254/phyto.2015.4407
Deng, H., Zheng, M., Hu, Z., Zeng, X., Kuang, N., and Fu, Y. (2020). Effects of
daphnetin on the autophagy signaling pathway of broblast-like synoviocytes in
rats with collagen-induced arthritis (CIA) induced by TNF-α.Cytokine 127, 154952.
doi:10.1016/j.cyto.2019.154952
Du, Q., Di, L. Q., Shan, J. J., Liu, T. S., and Zhang, X. Z. (2009). Intestinal
absorption of daphnetin by rats single pass perfusion in situ.Yao xue xue bao = Acta
Pharm. Sin. 44 (8), 922926. PMID: 20055163.
Du, G., Tu, H., Li, X., Pei, A., Chen, J., Miao, Z., et al. (2014). Daphnetin, a natural
coumarin derivative, provides the neuroprotection against glutamate-induced
toxicity in HT22 cells and ischemic brain injury. Neurochem. Res. 39 (2),
269275. doi:10.1007/s11064-013-1218-6
Fan, X., Wei, W., Huang, J., Peng, L., and Ci, X. (2020). Daphnetin attenuated
cisplatin-induced acute nephrotoxicity with enhancing antitumor activity of
cisplatin by upregulating SIRT1/SIRT6-nrf2 pathway. Front. Pharmacol. 11,
579178. doi:10.3389/fphar.2020.579178
Fan, X., Xie, M., Zhao, F., Li, J., Fan, C., Zheng, H., et al. (2021a). Daphnetin
triggers ROS-induced cell death and induces cytoprotective autophagy by
modulating the AMPK/Akt/mTOR pathway in ovarian cancer. Phytomedicine.
82, 153465. doi:10.1016/j.phymed.2021.153465
Fan, X., Gu, W., Gao, Y., Ma, N., Fan, C., and Ci, X. (2021b). Daphnetin
ameliorated GM-induced renal injury through the suppression of oxidative
stress and apoptosis in mice. Int. Immunopharmacol. 96, 107601. doi:10.1016/j.
intimp.2021.107601
Finn, G. J., Creaven, B. S., and Egan, D. A. (2004). Daphnetin induced
differentiation of human renal carcinoma cells and its mediation by
p38 mitogen-activated protein kinase. Biochem. Pharmacol. 67 (9), 17791788.
doi:10.1016/j.bcp.2004.01.014
Fletcher, J. M., Lalor, S. J., Sweeney, C. M., TubridyN.and Mills, K. H. G. (2010).
T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin.
Exp. Immunol. 162 (1), 111. doi:10.1111/j.1365-2249.2010.04143.x
Fukuda, H., Nakamura, S., Chisaki, Y., Takada, T., Toda, Y., Murata, H., et al.
(2016). Daphnetin inhibits invasion and migration of LM8 murine osteosarcoma
cells by decreasing RhoA and Cdc42 expression. Biochem. Biophys. Res. Commun.
471 (1), 6367. doi:10.1016/j.bbrc.2016.01.179
Fylaktakidou, K. C., Hadjipavlou-Litina, D. J., Litinas, K. E., and Nicolaide s, D. N.
(2004). Natural and synthetic coumarin derivatives with anti-inammatory/
antioxidant activities. Curr. Pharm. Des. 10 (30), 38133833. doi:10.2174/
1381612043382710
Gao, Q., Shan, J., Di, L., Jiang, L., and Xu, H. (2008). Therapeutic effects of
daphnetin on adjuvant-induced arthritic rats. J. Ethnopharmacol. 120 (2), 259263.
doi:10.1016/j.jep.2008.08.031
Gao, J., Chen, F., Fang, H., Mi, J., Qi, Q., and Yang, M. (2020). Daphnetin inhibits
proliferation and inammatory response in human HaCaT keratinocytes and
ameliorates imiquimod-induced psoriasis-like skin lesion in mice. Biol. Res. 53,
48. doi:10.1186/s40659-020-00316-0
Halda, J. J., Horáček, L., and Panarotto, P. (1998). Some taxonomic problems in the
genus Daphne L. Czechia: Okresní muzeum Orlických hor.
Han, S., Li, L.-z., and Song, S.-j. (2020). Daphne giraldii Nitsche (Thymelaeaceae):
Phytochemistry, pharmacology and medicinal uses. Phytochemistry 171, 112231.
doi:10.1016/j.phytochem.2019.112231
Hsieh, J. J., Purdue, M. P., Signoretti, S., Swanton, C., Albiges, L., Schmidinger, M.,
et al. (2017). Renal cell carcinoma. Nat. Rev. Dis. Prim. 3 (1), 1700917019. doi:10.
1038/nrdp.2017.9
Hu, X.-L., Gao, C., Xu, Z., Liu, M. L., Feng, L. S., and Zhang, G. D. (2018). Recent
development of coumarin derivatives as potential antiplasmodial and antimalarial
agents. Curr. Top. Med. Chem. 18 (2), 114123. doi:10.2174/
1568026618666171215101158
Huang, F., Tang, L. H., Yu, L. Q., Ni, Y. C., Wang, Q. M., and Nan, F. J. (2006). In
vitro potentiation of antimalarial activities by daphnetin derivatives against
Plasmodium falciparum. Biomed. Environ. Sci. 19 (5), 367370. PMID: 17190189.
Huff, J. (2007). Benzene-induced cancers: Abridged history and occupational
health impact. Int. J. Occup. Environ. Health 13 (2), 213221. doi:10.1179/oeh.2007.
13.2.213
Iadecola, C., and Alexander, M. J. (2001). Cerebral ischemia and inammation.
Curr. Opin. Neurol. 14 (1), 8994. doi:10.1097/00019052-200102000-00014
Ji, J., Ge, X., Chen, Y., Zhu, B., Wu, Q., Zhang, J., et al. (2019). Daphnetin
ameliorates experimental colitis by modulating microbiota composition and Treg/
Th17 balance. FASEB 33(8), 93089322. doi:10.1096/fj.201802659RR
Jiménez-Orozco, F. A., Rosales, A. A. R., Vega-Lopez, A., Dominguez-Lopez, M.
L., Garcia-Mondragon, M. J., Maldonado-Espinoza, A., et al. (2011). Differential
effects of esculetin and daphnetin on in vitro cell proliferation and in vivo
estrogenicity. Eur. J. Pharmacol. 668 (1), 3541. doi:10.1016/j.ejphar.2011.06.024
Jiménez-Orozco, F. A., Ranđelović, I., Hegedüs, Z., Vega-Lopez, A., and
Martínez-Flores, F. (2020). In vitro anti-proliferative effect and in vivo
antitumor action of daphnetin in different tumor cells. Cir. Cir. 88 (6),
765771. doi:10.24875/ciru.20000197
Frontiers in Pharmacology frontiersin.org15
Javed et al. 10.3389/fphar.2022.993562
Khan, H., Ullah, H., and Nabavi, S. M. (2019). Mechanistic insights of
hepatoprotective effects of curcumin: Therapeutic updates and future prospects.
Food Chem. Toxicol. 124, 182191. doi:10.1016/j.fct.2018.12.002
Khouchlaa, A., El Menyiy, N., Guaouguaou, F. E., El Baaboua, A., Char, S.,
Lakhdar, F., et al. (2021). Ethnomedicinal use, phytochemistry, pharmacology, and
toxicology of Daphne gnidium: A review. J. Ethnopharmacol. 275, 114124. doi:10.
1016/j.jep.2021.114124
Kostova, I., Bhatia, S., Grigorov, P., BalkanSky, S., Parmar, V. S., Prasad, A. K.,
et al. (2011). Coumarins as antioxidants. Curr. Med. Chem. 18 (25), 39293951.
doi:10.2174/092986711803414395
Kumar, A., Jha, S., and Pattanayak, S. P. (2016a). Daphnetin ameliorates 7, 12-
dimethylbenz[a]anthracene-induced mammary carcinogenesis through Nrf-2-
Keap1 and NF-κB pathways. Biomed. Pharmacother. 82, 439448. doi:10.1016/j.
biopha.2016.05.028
Kumar, A., Sunita, P., Jha, S., and Pattanayak, S. P. (2016b). Daphnetin inhibits
TNF-αand VEGF-induced angiogenesis through inhibition of the IKK s/IκBα/NF-
κB, Src/FAK/ERK 1/2 and Akt signalling pathways. Clin. Exp. Pharmacol. Physiol.
43 (10), 939950. doi:10.1111/1440-1681.12608
Leung, G., Sun, W., Zheng, L., BrookeS, S., TullyM.and Shi, R. (2011). Anti-
acrolein treatment improves behavioral outcome and alleviates myelin damage in
experimental autoimmune encephalomyelitis mouse. Neuroscience 173, 150155.
doi:10.1016/j.neuroscience.2010.11.018
Li, M., Shi, X., Chen, F., and Hao, F. (2017). Daphnetin inhibits inammation in
the NZB/W F1 systemic lupus erythematosus murine model via inhibition of NF-κB
activity. Exp. Ther. Med. 13 (2), 455460. doi:10.3892/etm.2016.3971
Li, T., Yang, G., Hao, Q., Zhang, X., and Zhang, X. (2022). Daphnetin ameliorates
the expansion of chemically induced hepatocellular carcinoma via reduction of
inammation and oxidative stress. J. Oleo Sci. 71 (4), 575585. doi:10.5650/jos.
ess21415
Liang, S.-C., Ge, G. B., Liu, H. X., Zhang, Y. Y., Wang, L. M., Zhang, J. W., et al.
(2010). Identication and characterization of human UDP-
glucuronosyltransferases responsible for the in vitro glucuronidation of
daphnetin. Drug Metab. Dispos. 38 (6), 973980. doi:10.1124/dmd.109.030734
Liang, S.-C., Ge, G. B., Xia, Y. L., Zhang, J. W., Qi, X. Y., Tu, C. X., et al. (2015). In
vitro evaluation of the effect of 7-methyl substitution on glucuronidation of
daphnetin: metabolic stability, isoform selectivity, and bioactivity analysis.
J. Pharm. Sci. 104 (10), 35573564. doi:10.1002/jps.24538
Liang, S.-C., Xia, Y. L., Hou, J., Ge, G. B., Zhang, J. W., He, Y. Q., et al. (2016).
Methylation, glucuronidation, and sulfonation of daphnetin in human hepatic
preparations in vitro: Metabolic proling, pathway comparison, and bioactivity
analysis. J. Pharm. Sci. 105 (2), 808816. doi:10.1016/j.xphs.2015.10.010
Liang, S., Ge, G. b., Xia, Y. l., Qi, X. y., Wang, A. x., Tu, C. x., et al. (2017). In vitro
metabolism of daphnetin in rat liver S9 fractions. Yao Xue Xue Bao 52(2), 291295.
PMID: 29979523
Liao, M. J., Lin, L. F., Zhou, X., Zhou, X. W., Xu, X., Cheng, X., et al. (2013).
Daphnetin prevents chronic unpredictable stress-induced cognitive decits.
Fundam. Clin. Pharmacol. 27 (5), 510516. doi:10.1111/j.1472-8206.2012.01049.x
Liu, J., Chen, Q., Jian, Z., Shao, L., Jin, T., Zhu, X., et al. (2016a). Daphnetin
protects against cerebral ischemia/reperfusion injury in mice via inhibition of
TLR4/NF-κB signaling pathway. Biomed. Res. Int. 2016, 2816056. doi:10.1155/
2016/2816056
Liu, Z., Liu, J., Zhao, K., Shi, Q., Zuo, T., Wang, G., et al. (2016b). Role of
daphnetin in rat severe acute pancreatitis through the regulation of TLR4/NF-
[Formula: see text]B signaling pathway activation. Am. J. Chin. Med. 44 (01),
149163. doi:10.1142/S0192415X16500105
Liu, Y., Liao, L., Chen, Y., and Han, F. (2019). Effects of daphnetin on lipid
metabolism, insulin resistance and oxidative stress in OA-treated HepG2 cells. Mol.
Med. Rep. 19 (6), 46734684. doi:10.3892/mmr.2019.10139
Liu, W., Mei, J., and Xie, J. (2020). Elucidating Antibacterial activity and
mechanism of daphnetin against Pseudomonas uorescens and Shewanella
putrefaciens. J. Food Qual. 2011, 110. doi:10.1155/2020/6622355
Liu, W., Mei, J., and Xie, J. (2021). Effect of locust bean gum-sodium alginate
coatings incorporated with daphnetin emulsions on the quality of Scophthalmus
maximus at refrigerated condition. Int. J. Biol. Macromol. 170, 129139. doi:10.
1016/j.ijbiomac.2020.12.089
Liu, C., Liu, H., Pan, J., Lin, R., Chen, Y., and Zhang, C. (2022). Daphnetin inhibits
the survival of hepatocellular carcinoma cells through regulating Wnt/β-catenin
signaling pathway. Drug Dev. Res. 83, 952. doi:10.1002/ddr.21920
Lu, Y., Pang, J., Wang, G., Hu, X., Li, X., Li, G., et al. (2021). Quantitative
proteomics approach to investigate the antibacterial response of Helicobacter pylori
to daphnetin, a traditional Chinese medicine monomer. RSC Adv. 11 (4),
21852193. doi:10.1039/D0RA06677J
Lv, H., Liu, Q., Zhou, J., Tan, G., Deng, X., and Ci, X. (2017). Daphnetin-mediated
Nrf2 antioxidant signaling pathways ameliorate tert-butyl hydroperoxide (t-BHP)-
induced mitochondrial dysfunction and cell death. Free Radic. Biol. Med. 106,
3852. doi:10.1016/j.freeradbiomed.2017.02.016
Lv, H., Fan, X., Wang, L., Feng, H., and Ci, X. (2018). Daphnetin alleviates
lipopolysaccharide/d-galactosamine-induced acute liver failure via the inhibition of
NLRP3, MAPK and NF-κB, and the induction of autophagy. Int. J. Biol. Macromol.
119, 240248. doi:10.1016/j.ijbiomac.2018.07.101
Lv, X., Li, Y., Xiao, Q., and Li, D. (2019). Daphnetin activates the Nrf2-dependent
antioxidant response to prevent arsenic-induced oxidative insult in human lung
epithelial cells. Chem. Biol. Interact. 302, 93100. doi:10.1016/j.cbi.2019.02.001
Lv, H., Zhu, C., Wei, W., Lv, X., Yu, Q., Deng, X., et al. (2020). Enhanced Keap1-
Nrf2/Trx-1 axis by daphnetin protects against oxidative stress-driven
hepatotoxicity via inhibiting ASK1/JNK and Txnip/NLRP3 inammasome
activation. Phytomedicine 71, 153241. doi:10.1016/j.phymed.2020.153241
Manojlović, N. T., Maskovic, P., Vasiljevic, P., Jelic, R., Juskovic, M., Sovrlic, M.,
et al. (2012). HPLC Analysis, antimicrobial and antioxidant activities of Daphne
cneorum L. Hem. Ind. 66 (5), 709716. doi:10.2298/HEMIND120114029M
Mansoor, F., Anis, I., Ali, S., Choudhary, M. I., and Shah, M. R. (2013). New
dimeric and trimeric coumarin glucosides from Daphne retusa Hemsl. Fitoterapia
88, 1924. doi:10.1016/j.tote.2013.03.029
Martinez, K. B., Mackert, J. D., and McIntosh, M. K. (2017). Chapter 18 -
polyphenols and intestinal health,in Nutrition and functional foods for healthy
aging. Editor R. R. Watson (Cambridge: Academic Press), 191210. doi:10.1016/
B978-0-12-805376-8.00018-6
Masamoto, Y., Ando, H., Murata, Y., Shimoishi, Y., Tada, M., and Takahata, K.
(2003). Mushroom tyrosinase inhibitory activity of esculetin isolated from seeds of
Euphorbia lathyris L. Biosci. Biotechnol. Biochem. 67 (3), 631634. doi:10.1271/bbb.
67.631
Mendes-Braz, M., and Martins, J. O. (2018). Diabetes mellitus and liver surgery:
the effect of diabetes on oxidative stress and inammation. Mediat. Inamm. 2018,
2456579. doi:10.1155/2018/2456579
Mohamed, M. R., Emam, M. A., Hassan, N. S., and Mogadem, A. I. (2014).
Umbelliferone and daphnetin ameliorate carbon tetrachloride-induced
hepatotoxicity in rats via nuclear factor erythroid 2-related factor 2-mediated
heme oxygenase-1 expression. Environ. Toxicol. Pharmacol. 38 (2), 531541.
doi:10.1016/j.etap.2014.08.004
Moshiashvili, G., Tabatadze, N., and Mshvildadze, V. (2020). The genus Daphne:
A review of its traditional uses, phytochemistry and pharmacology. Fitoterapia 143,
104540. doi:10.1016/j.tote.2020.104540
Motzer, R. J., Bander, N. H., and Nanus, D. M. J. N. E. J. o. M. (1996). Renal-cell
carcinoma. N. Engl. J. Med. 335 (12), 865875. doi:10.1056/NEJM199609193351207
Mu, L., Wang, Q., and Ni, Y. (2002). In vitro antimalarial effect of daphnetin
relating to its iron-chelating activity. Zhongguo ji Sheng Chong xue yu ji Sheng
Chong Bing za zhi= Chin. J. Parasitol. Parasit. Dis. 20 (2), 8385. PMID: 12567565.
Nam, G., An, S. K., Park, I. C., Bae, S., and Lee, J. H. (2022). Daphnetin inhibits α-MSH-
induced melanogenesis via PKA and ERK signaling p athways in B16F10 melanoma cells.
Biosci. Biotechnol. Biochem. 86 (5), 596609. doi:10.1093/bbb/zbac016
Nanzhen, K., Jieying, W., Wenwei, Z., and Xiaoping, Z. (2018). Toxicological
studies of daphnetin. Pharmacogn. Mag. 14 (58), 561. doi:10.4103/pm.pm_523_17
NDong, C., Anzellotti, D., Ibrahim, R. K., Huner, N. P. A., and Sarhan, F. (2003).
Daphnetin methylation by a novel O-methyltransferase is associated with cold
acclimation and photosystem II excitation pressure in rye. J. Biol. Chem. 278 (9),
68546861. doi:10.1074/jbc.M209439200
Norma Francenia, S.-S. (2019). Shikimic acid pathway in biosynthesis of phenolic
compounds. Vienna, Austria: IntechOpen. doi:10.5772/intechopen.83815
Pan, L., Li, X., Jin, H., Yang, X., and Qin, B. (2017). Antifungal activity of
umbelliferone derivatives: Synthesis and structure-activity relationships. Microb.
Pathog. 104, 110115. doi:10.1016/j.micpath.2017.01.024
Pei, Q., Hu, P., Zhang, H., Li, H., Yang, T., and Liu, R. (2021). Daphnetin exerts an
anticancer effect by attenuating the pro-inammatory cytokines. J. Biochem. Mol.
Toxicol. 35 (6), 18. doi:10.1002/jbt.22759
Pinto, D. C., and Silva, A. (2017). Anticancer natural coumarins as lead
compounds for the discovery of new drugs. Curr. Top. Med. Chem. 17 (29),
31903198. doi:10.2174/1568026618666171215095750
Prasad, S., Kulshreshtha, A., Lall, R., and Gupta, S. C. (2021). Inammation and
ROS in arthritis: management by ayurvedic medicinal plants. Food Funct. 12 (18),
82278247. doi:10.1039/D1FO01078F
Qi, Z., Qi, S., Gui, L., Shen, L., and Feng, Z. (2016). Daphnetin protects oxidative
stress-induced neuronal apoptosis via regulation of MAPK signaling and
HSP70 expression. Oncol. Lett. 12 (3), 19591964. doi:10.3892/ol.2016.4849
Frontiers in Pharmacology frontiersin.org16
Javed et al. 10.3389/fphar.2022.993562
Riaz, M., Saleem, A., Siddique, S., Khan, B. A., Nur-e-Alam, M., Shahzad-ul-
Hussan, S., et al. (2016). Phytochemistry of Daphne oleoides. Nat. Prod. Res. 30 (8),
880897. doi:10.1080/14786419.2015.1092146
Riveiro, M. E., De KimpeN.Moglioni, A., Vazquez, R., MonczorF.Shayo, C., et al.
(2010). Coumarins: old compounds with novel promising therapeutic perspectives.
Curr. Med. Chem. 17 (13), 13251338. doi:10.2174/092986710790936284
Rojas, M., Woods, C. R., Mora, A. L., Xu, J., and Brigham, K. L. (2005).
Endotoxin-induced lung injury in mice: structural, functional, and biochemical
responses. Am. J. Physiol. Lung Cell. Mol. Physiol. 288 (2), L333L341. doi:10.1152/
ajplung.00334.2004
Satô, M., and Hasegawa, M. (1969). Conversion of daphnin to daphnetin-8-
glucoside in Daphne odora. Phytochemistry 8 (7), 12111214. doi:10.1016/S0031-
9422(00)85559-4
Shan, J. (2009). Studies on oral absorption and metabolism of the main active
constituents of Zushima (D). Nanjing: Nanjing University of Chinese Medicine.
Shan, J., Zhu, H., Geng, T., Zhang, L., and Ding, A. (2011). Determination of
equilibrium solubility and apparent oil/water partition coefcient of Daphnetin.
J. Nanjing Univ. Traditional Chin. Med. 35, 31443146.
Shen, L., Zhou, T., Wang, J., Sang, X., Lan, L., Luo, L., et al. (2017). Daphnetin
reduces endotoxin lethality in mice and decreases LPS-induced inammation in
Raw264.7 cells via suppressing JAK/STATs activation and ROS production.
Inamm. Res. 66 (7), 579589. doi:10.1007/s00011-017-1039-1
Shu, K., Kuang, N., Zhang, Z., Hu, Z., Zhang, Y., Fu, Y., et al. (2014). Therapeutic
effect of daphnetin on the autoimmune arthritis through demethylation of
proapoptotic genes in synovial cells. J. Transl. Med. 12 (1), 287. doi:10.1186/
s12967-014-0287-x
Singh, L., Singh, A. P., and Bhatti, R. (2021a). Mechanistic interplay of various
mediators involved in mediating the neuroprotective effect of daphnetin.
Pharmacol. Rep. 73, 12201229. doi:10.1007/s43440-021-00261-z
Singh, L., Kaur, A., Singh, A. P., and Bhatti, R. (2021b). Daphnetin, a natural
coumarin averts reserpine-induced bromyalgia in mice: modulation of MAO-A.
Exp. Brain Res. 239 (5), 14511463. doi:10.1007/s00221-021-06064-1
Song, B.-C., Jiang, M. M., Zhang, S., Ma, H., Liu, M., Fu, Z. R., et al. (2021).
Immunosuppressive activity of daphnetin on the humoral immune responses in
ovalbumin-sensitized BALB/c mice. Immunopharmacol. Immunotoxicol. 43 (2),
171175. doi:10.1080/08923973.2021.1872618
Sovrlić, M. M., and Manojlović, N. T. (2017). Plants from the genus Daphne: A
review of its traditional uses, phytochemistry, biological and pharmacological
activity. Serbian J. Exp. Clin. Res. 18 (1), 6980. doi:10.1515/sjecr-2016-0024
Sovrlić, M., Vasiljevic, P., Juskovic, M., Maskovic, P., and Manojlovic, N. (2015).
Phytochemical, antioxidant and antimicrobial proles of extracts of Daphne alpina
(Thymelaeaceae) L leaf and twig from Mt Kopaonik (Serbia). Trop. J. Pharm . Res. 14
(7), 12391248. doi:10.4314/TJPR.V14I7.17
Tu, L., Li, S., Fu, Y., Yao, R., Zhang, Z., Yang, S., et al. (2012). The therapeutic
effects of daphnetin in collagen-induced arthritis involve its regulation of Th17 cells.
Int. Immunopharmacol. 13 (4), 417423. doi:10.1016/j.intimp.2012.04.001
Ueno, K., and Saito, N. (1976). Daphnetin, isolated from Daphne odora. Acta
Crystallogr. Sect. B 32 (3), 946948. doi:10.1107/S0567740876004275
Vinayagam, R., and Xu, B. (2017). 7, 8-Dihydroxycoumarin (daphnetin) protects
INS-1 pancreatic β-cells against streptozotocin-induced apoptosis. Phytomedicine
24, 119126. doi:10.1016/j.phymed.2016.11.023
Vivier, D., Chen, Q., Jian, Z., Xiong, X., Shao, L., Jin, T., et al. (2016). Daphnetin
protects against cerebral ischemia/reperfusion injury in mice via inhibition of
TLR4/NF-κB signaling pathway. Biomed. Res. Int. 59, 28160562816335. doi:10.
1155/2016/2816056
Walker, M., and Crabtree, J. E. (1998). Helicobacter pylori infection and the
pathogenesis of duodenal ulceration. Ann. N. Y. Acad. Sci. 859 (1), 96111. doi:10.
1111/j.1749-6632.1998.tb11114.x
Wang, Q. M., Ni, Y. C., Xu, Y. Q., Ha, S. H., and Cai, Y. (2000). The schizontocidal
activity of daphnetin against malaria parasites in vitro and in vivo.Zhongguo ji sheng
chong xue yu ji sheng chong bing za zhi = Chin. J. Parasitol. Parasit. Dis. 18 (4),
204206. PMID: 12567659.
Wang, Y., Li, C. F., Pan, L. M., and Gao, Z. L. (2013). 7, 8-Dihydroxycoumarin
inhibits A549 human lung adenocarcinoma cell proliferation by inducing apoptosis
via suppression of Akt/NF-κB signaling. Exp. Ther. Med. 5 (6), 17701774. doi:10.
3892/etm.2013.1054
Wang, D., Lu, Z., Zhang, H., Jin, S. F., Yang, H., Li, Y. M., et al. (2016). Daph netin
alleviates experimental autoimmune encephalomyelitis via regulating dendritic cell
activity. CNS Neurosci. Ther. 22 (7), 558567. doi:10.1111/cns.12537
Wang, G., Pang, J., Hu, X., Nie, T., Lu, X., Li, X., et al. (2019). Daphnetin: A novel
anti-Helicobacter pylori agent. Int. J. Mol. Sci. 20 (4), 850. doi:10.3390/
ijms20040850
Wang, Y., Wang, J., Fu, Z., Sheng, R., Wu, W., Fan, J., et al. (2020a ). Syntheses and
evaluation of daphnetin derivatives as novel G protein-coupled receptor inhibitors
and activators. Bioorg. Chem. 104, 104342. doi:10.1016/j.bioorg.2020.104342
Wang, Y., Chen, J., Chen, J., Dong, C., Yan, X., Zhu, Z., et al. (2020b). Daphnetin
ameliorates glucocorticoid-induced osteoporosis via activation of Wnt/GSK-3β/β-
catenin signaling. Toxicol. Appl. Pharmacol. 409, 115333. doi:10.1016/j.taap.2020.
115333
Wang, D., Zhu, B., Liu, X., Han, Q., Ge, W., Zhang, W., et al. (2020c). Daphnetin
ameliorates experimental autoimmune encephalomyelitis through regulating heme
oxygenase-1. Neurochem. Res. 45 (4), 872881. doi:10.1007/s11064-020-02960-0
Wen, Z., Fang, L., and He, Z. (2009). Effect of chemical enhancers on
percutaneous absorption of daphnetin in isopropyl myristate vehicle across rat
skin in vitro.Drug Deliv. 16 (4), 214223. doi:10.1080/10717540902836715
Wu, Z., Wu, H., Li, C., Fu, F., Ding, J., Shao, S., et al. (2019). Daphnetin attenuates
LPS-induced osteolysis and RANKL mediated osteoclastogenesis through
suppression of ERK and NFATc1 pathways. J. Cell. Physiol. 234 (10),
1781217823. doi:10.1002/jcp.28408
Xia, Y.-L., Dou, T. Y., Liu, Y., Wang, P., Ge, G. B., and Yang, L. (2018a). In vitro
evaluation of the effect of C-4 substitution on methylation of 7, 8-
dihydroxycoumarin: metabolic prole and catalytic kinetics. R. Soc. Open Sci. 5
(1), 171271. doi:10.1098/rsos.171271
Xia, Y., Chen, C., Liu, Y., Ge, G., Dou, T., and Wang, P. (2018b). Synthesis and
structure-activity relationship of daphnetin derivatives as potent antioxidant agents.
Mol. (Basel, Switz. 23 (10), 2476. doi:10.3390/molecules23102476
Xu, W. C., Shen, J. G., and Jiang, J. Q. (2011). Phytochemical and biological
studies of the plants from the genus Daphne. Chem. Biodivers. 8 (7), 12151233.
doi:10.1002/cbdv.201000117
Xu, K., Guo, L., Bu, H., and Wang, H. (2019). Daphnetin inhibits high glucose-
induced extracellular matrix accumulation, oxidative stress and inammation in
human glomerular mesangial cells. J. Pharmacol. Sci. 139 (2), 9197. doi:10.1016/j.
jphs.2018.11.013
Yang, Y.-Z., Ranz, A., Pan, H. Z., Zhang, Z. N., Lin, X. B., and Meshnick, S. R.
(1992). Daphnetin: a novel antimalarial agent with in vitro and in vivo activity. Am.
J. Trop. Med. Hyg. 46 (1), 1520. doi:10.4269/ajtmh.1992.46.15
Yang, E. B., Zhao, Y. N., Zhang, K., and Mack, P. (1999). Daphnetin, one of
coumarin derivatives, is a protein kinase inhibitor. Biochem. Biophys. Res. Commun.
260 (3), 682685. doi:10.1006/bbrc.1999.0958
Yang, L., Yang, Q., Zhang, K., Li, Y. J., Wu, Y. M., Liu, S. B., et al. (2014).
Neuroprotective effects of daphnetin against NMDA receptor-mediated
excitotoxicity. Molecules 19 (9), 1454214555. doi:10.3390/molecules190914542
Yang, L., Ding, W., Xu, Y., Wu, D., Li, S., Chen, J., et al. (2016). New insights into
the antibacterial activity of hydroxycoumarins against Ralstonia solanacearum.
Molecules 21 (4), 468. doi:10.3390/molecules21040468
Yang, Y., Sheng, Q., Nie, Z., Liu, L., Zhang, W., Chen, G., et al. (2021a). Daphnetin
inhibits spinal glial activation via Nrf2/HO-1/NF-κB signaling pathway and
attenuates CFA-induced inammatory pain. Int. Immunopharmacol. 98, 107882.
doi:10.1016/j.intimp.2021.107882
Yang, F., Jiang, X., Cao, H., Shuai, W., Zhang, L., Wang, G., et al. (2021b).
Daphnetin preconditioning decreases cardiac injury and susceptibility to
ventricular arrhythmia following ischaemia-reperfusion through the TLR4/
MyD88/NF-?b signalling pathway. Pharmacology 106 (7-8), 369383. doi:10.
1159/000513631
Yang, S., Song, Y., Wang, Q., Liu, Y., Wu, Z., Duan, X., et al. (2021c). Daphnetin
ameliorates acute lung injury in mice with severe acute pancreatitis by inhibiting the
JAK2STAT3 pathway. Sci. Rep. 11 (1), 11491. doi:10.1038/s41598-021-91008-6
Yang, T., Wang, X., Guo, L., Zheng, F., Meng, C., Zheng, Y., et al. (2022).
Daphnetin inhibits corneal inammation and neovascularization on a mouse model
of corneal alkali burn. Int. Immunopharmacol. 103, 108434. doi:10.1016/j.intimp.
2021.108434
Yao, R., Fu, Y., Li, S., Tu, L., Zeng, X., and Kuang, N. (2011). Regulatory effect of
daphnetin, a coumarin extracted from Daphne odora, on the balance of Treg and
Th17 in collagen-induced arthritis. Eur. J. Pharmacol. 670 (1), 286294. doi:10.
1016/j.ejphar.2011.08.019
Yu, W., Wang, H., Ying, H., Yu, Y., Chen, D., Ge, W., et al. (2014a). Daphnetin
attenuates microglial activation and proinammatory factor production via
multiple signaling pathways. Int. Immunopharmacol. 21 (1), 19. doi:10.1016/j.
intimp.2014.04.005
Yu, W.-w., Lu, Z., Zhang, H., Kang, Y. h., Mao, Y., Wang, H. h., et al. (2014b).
Anti-inammatory and protective properties of daphnetin in endotoxin-induced
lung injury. J. Agric. Food Chem. 62 (51), 1231512325. doi:10.1021/jf503667v
Zali, M. R. (2011). Facing resistance of H. pylori infection. Gastroenterol. Hepat ol.
Bed Bench 4 (1), 311.
Frontiers in Pharmacology frontiersin.org17
Javed et al. 10.3389/fphar.2022.993562
Zhang, W., Di, L. q., Li, J. S., Shan, J. j., Kang, A., Qian, S., et al. (2014). The effects
of Glycyrrhizae uralenis and its major bioactive components on pharmacokinetics
of daphnetin in Cortex daphnes in rats. J. Ethnopharmacol. 154 (3), 584592. doi:10.
1016/j.jep.2014.03.047
Zhang,L.,Gu,Y.,Li,H.,Cao,H.,Liu,B.,Zhang,H.,etal.(2018).Daphnetinprotects
against cisplatin-induced nephrotoxicity by inhibiting inammatory and oxidative
response. Int. Immunopharmacol. 65, 402407. doi:10.1016/j.intimp.2018.10.018
Zhang,W.,Zhuo,S.,He,L.,Cheng,C.,Zhu,B.,Lu,Y.,etal.(2019).Daphnetinprevents
methicillin-resistant Staphylococcus aureus infection by inducing autophagic response.
Int. Immunopharmacol. 72, 195203. doi:10.1016/j.intimp.2019.04.007
Zhang, X., Yao, J., Wu, Z., Zou, K., Yang, Z., Huang, X., et al. (2020).
Chondroprotective and antiarthritic effects of Daphnetin used in vitro and in
vivo osteoarthritis models. Life Sci. 240, 116857. doi:10.1016/j.lfs.2019.116857
Zhao, H., Jiang, Z., Chang, X., Xue, H., Yahefu, W., and Zhang, X. (2018). 4-
Hydroxyphenylacetic acid prevents acute APAP-induced liver injury by increasing
phase II and antioxidant enzymes in mice. Front. Pharmacol. 9, 653. doi:10.3389/
fphar.2018.00653
Zheng, M., Kuang, N., Zeng, X., Wang, J., Zou, Y., and Fu, Y. (2020). Daphnetin
induces apoptosis in broblast-like synoviocytes from collagen-induced arthritic
rats mainly via the mitochondrial pathway. Cytokine 133, 155146. doi:10.1016/j.
cyto.2020.155146
Zhi, J., Duan, B., Pei, J., Wu, S., and Wei, J. (2019). Daphnetin protects
hippocampal neurons from oxygen-glucose deprivationinduced injury. J. Cell.
Biochem. 120 (3), 41324139. doi:10.1002/jcb.27698
Zhou, X., Zhang, C., Zhang, G., and Liao, Y. (2016). Intercalation of the
daphnetinCu (II) complex with calf thymus DNA. RSC Adv. 6 (7), 54085418.
doi:10.1039/C5RA22274E
Zhu, A., Zhang, T., and Wang, Q. (2018). The phytochemistry, pharmacokinetics,
pharmacology and toxicity of Euphorbia semen. J. Ethnopharmacol. 227, 4155.
doi:10.1016/j.jep.2018.08.024
Frontiers in Pharmacology frontiersin.org18
Javed et al. 10.3389/fphar.2022.993562
... Daphnetin (DAP) is an active ingredient extracted from Daphne Koreana Nakai, also known as Zushimazin [41]. It is the first new drug in China and is chemically named 7,8-dihydroxycoumarin. ...
Article
Full-text available
Esophageal cancer (ESC) is a malignant tumor that originates from the mucosal epithelium of the esophagus and is part of the digestive tract. Although the exact pathogenesis of ESC has not been fully elucidated, excessive oxidative stress is an important characteristic that leads to the development of many cancers. Abnormal expression of several proteins and transcription factors contributes to oxidative stress in ESCs, which alters the growth and proliferation of ESCs and promotes their metastasis. Natural compounds, including alkaloids, terpenes, polyphenols, and xanthine compounds, can inhibit reactive oxygen species production in ESCs. These compounds reduce oxidative stress levels and subsequently inhibit the occurrence and progression of ESC through the regulation of targets and pathways such as the cytokine interleukins 6 and 10, superoxide dismutase, the NF-+ACY-kappa+ADs-B/MAPK pathway, and the mammalian Nrf2/ARE target pathway. Thus, targeting tumor oxidative stress has become a key focus in anti-ESC therapy. This review discusses the potential of Natural products (NPs) for treating ESCs and summarizes the application prospects of oxidative stress as a new target for ESC treatment. The findings of this review provide a reference for drug development targeting ESCs. Nonetheless, further high-quality studies will be necessary to determine the clinical efficacy of these various NPs.
... In order to enhance the efficacy of drug discovery, a pragmatic approach involves commencing the process with natural bioactive substances derived from medicinal plants or alternative natural origins. Coumarins, which were initially extracted from melilot flowers and tonka beans, have been extensively explored for their therapeutical applications, encompassing anti-inflammatory [39], anticancer [40], antiviral [41], antimicrobial [42], antioxidant [43], and anticoagulant [44]. Warfarin is widely recognized as a prominent pharmaceutical agent derived from coumarin [45]. ...
Article
Full-text available
In the present study, we explored the potential of coumarin-based compounds, known for their potent anticancer properties, by designing and synthesizing a novel category of 8-methoxycoumarin-3-carboxamides. Our aim was to investigate their antiproliferative activity against liver cancer cells. Toward this, we developed a versatile synthetic approach to produce a series of 8-methoxycoumarin-3-carboxamide analogues with meticulous structural features. Assessment of their antiproliferative activity demonstrated their significant inhibitory effects on the growth of HepG2 cells, a widely studied liver cancer cell line. Among screened compounds, compound 5 exhibited the most potent antiproliferative activity among the screened compounds (IC50 = 0.9 µM), outperforming the anticancer drug staurosporine (IC50 = 8.4 µM), while showing minimal impact on normal cells. The flow cytometric analysis revealed that compound 5 induces cell cycle arrest during the G1/S phase and triggers apoptosis in HepG2 cells by increasing the percentage of cells arrested in the G2/M and pre-G1 phases. Annexin V-FITC/PI screening further supported the induction of apoptosis without significant necrosis. Further, compound 5 exhibited the ability to activate caspase3/7 protein and substantially inhibited β-tubulin polymerization activity in HepG2 cells. Finally, molecular modelling analysis further affirmed the high binding affinity of compound 5 toward the active cavity of β-tubulin protein, suggesting its mechanistic involvement. Collectively, our findings highlight the therapeutic potential of the presented class of coumarin analogues, especially compound 5, as promising candidates for the development of effective anti-hepatocellular carcinoma agents. Supplementary Information The online version contains supplementary material available at 10.1186/s13065-023-01063-5.
... The interest in these compounds stems from their potential therapeutic effects, and researchers continue to explore their potential applications in medicine and healthcare. For example, Garg et al. (Garg et al., 2020) reported esculetin (esculin) and daphnetin among the notable biologically active coumarins, displaying bioactivities such as neuroprotection, hepatoprotection (Javed et al., 2022), anti-inflammatory, antioxidant (Kılıç, 2022) and anticancer (Desam & Al-Rajab, 2022) effects. Upon examining the relationship between the number of publications and years, a linear regression showed an R 2 value of 0.7812, indicating a direct connection between the two variables. ...
Chapter
This chapter focuses on antineoplastic lead molecules identified from plants and microbial sources. It discusses the various natural products used in the chemotherapy. The first part of the chapter emphasizes the use of plants in traditional medicinal practices and how modern science has used their potential extractives in treating cancer. The major antineoplastic molecules from plant sources are classified into phenolics, terpenoids, and alkaloids, some of which have been successfully employed to control neoplasms. The second part focuses on antineoplastic lead molecules derived from microbes, particularly bacteria and fungi. These microorganisms produce a diverse array of bioactive compounds with potential anticancer properties. The molecules are classified into antibiotics, statins, enzymes, and enzyme inhibitors, among others. The chapter highlights the promising potential of natural molecules, both from plants and microbes, in targeting key proteins involved in complex cancer pathogenesis. These molecules offer unique chemical structures and mechanisms of action that make them viable candidates for developing novel antineoplastic drugs. Overall, the chapter serves as a valuable resource for researchers and professionals in the field of drug discovery, providing insights into the potential of natural sources for identifying effective antineoplastic agents to combat cancer.
Article
Full-text available
Cancer is a complex disease resulting from abnormal cell growth that is induced by a number of genetic and environmental factors. The tumor microenvironment (TME), which involves extracellular matrix, cancer-associated fibroblasts (CAF), tumor-infiltrating immune cells and angiogenesis, plays a critical role in tumor progression. Cyclic adenosine monophosphate (cAMP) is a second messenger that has pleiotropic effects on the TME. The downstream effectors of cAMP include cAMP-dependent protein kinase (PKA), exchange protein activated by cAMP (EPAC) and ion channels. While cAMP can activate PKA or EPAC and promote cancer cell growth, it can also inhibit cell proliferation and survival in context- and cancer type-dependent manner. Tumor-associated stromal cells, such as CAF and immune cells, can release cytokines and growth factors that either stimulate or inhibit cAMP production within the TME. Recent studies have shown that targeting cAMP signaling in the TME has therapeutic benefits in cancer. Small-molecule agents that inhibit adenylate cyclase and PKA have been shown to inhibit tumor growth. In addition, cAMP-elevating agents, such as forskolin, can not only induce cancer cell death, but also directly inhibit cell proliferation in some cancer types. In this review, we summarize current understanding of cAMP signaling in cancer biology and immunology and discuss the basis for its context-dependent dual role in oncogenesis. Understanding the precise mechanisms by which cAMP and the TME interact in cancer will be critical for the development of effective therapies. Future studies aimed at investigating the cAMP-cancer axis and its regulation in the TME may provide new insights into the underlying mechanisms of tumorigenesis and lead to the development of novel therapeutic strategies.
Article
Full-text available
Idiopathic pulmonary fibrosis (IPF) is a chronic and refractory interstitial lung disease. Although there is no cure for IPF, the development of drugs with improved efficacy in the treatment of IPF is required. Daphnetin, a natural coumarin derivative, has immunosuppressive, anti-inflammatory, and antioxidant activities. However, its antifibrotic effects have not yet been elucidated. In this study, we investigated the antifibrotic effects of daphnetin on pulmonary fibrosis and the associated molecular mechanism. We examined the effects of daphnetin on splenocytes cultured in Th17 conditions, lung epithelial cells, and a mouse model of bleomycin (BLM)-induced pulmonary fibrosis. We identified that daphnetin inhibited IL-17A production in developing Th17 cells. We also found that daphnetin suppressed epithelial-to-mesenchymal transition (EMT) in TGF-β-treated BEAS2B cells through the regulation of AKT phosphorylation. In BLM-treated mice, the oral administration of daphnetin attenuated lung histopathology and improved lung mechanical functions. Our findings clearly demonstrated that daphnetin inhibited IL-17A and EMT both in vitro and in vivo, thereby protecting against BLM-induced pulmonary fibrosis. Taken together, these results suggest that daphnetin has potent therapeutic effects on lung fibrosis by modulating both Th17 differentiation and the TGF-β signaling pathway, and we thus expect daphnetin to be a drug candidate for the treatment of IPF.
Article
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a human virus that burst at Wuhan in China and spread quickly over the world, leading to millions of deaths globally. The journey of this deadly virus to different mutant strains is still ongoing. The plethora of drugs and vaccines have been tested to cope up this pandemic. The herbal plants and different spices have received great attention during pandemic, because of their anti-inflammatory, and immunomodulatory properties in treating viruses and their symptoms. Also, it has been shown that nano-formulation of phytochemicals has potential therapeutic effect against COVID-19. Furthermore, the plant derived compound nano-formulation specifically increases its antiviral property by enhancing its bioavailability, solubility, and target-specific delivery system. This review highlights the potentiality of herbal plants and their phytochemical against SARS-CoV-2 utilizing different mechanisms such as blocking the ACE-2 receptors, inhibiting the main proteases, binding spike proteins and reducing the cytokine storms.
Chapter
Malaria, one of the “big three” killer diseases, is a major public health problem as well as a major cause of mortality and morbidity worldwide. The goals of the global malaria elimination program mainly include (1) to reduce malaria case incidence by at least 90% by 2030; (2) to reduce malaria mortality rates by at least 90% by 2030; (3) to eliminate malaria in at least 35 countries by 2030; and (4) to prevent a resurgence of malaria in all malaria-free countries. Currently, the malaria elimination strategy mainly depends on chemotherapy, and the currently available antimalarials are mainly divided into four categories according to the chemical structure and mechanism of drug actions, including arylamino alcohol compounds, 8-aminoquinolines, antifolate compounds, artemisinins, and other agents. Artemisinin, a sesquiterpene lactone extracted from the Chinese medicinal herb A. annua, has been recommended as the first-choice treatment for malaria by the World Health Organization. However, the rapid emergence of artemisinin resistance in malaria parasites urges the development of novel treatments during the stage toward malaria elimination in the world. Previous studies have shown the effectiveness and safety of traditional Chinese medicines for the treatment of parasitic diseases, including malaria. In this chapter, we discuss other traditional Chinese medicines that show potential value for malaria treatment except artemisinins. The crude extracts and pure compounds from ginger, garlic, the Asteraceae family, Bupleuri radix and Daphne spp. have shown in vitro and in vivo anti-Plasmodium activities, and some have been tested for their antimalarial actions in malaria patients. Further randomized, controlled clinical trials to examine the efficacy and safety of traditional Chinese medicine for the treatment of malaria and to unravel the underlying mechanisms seem justified.
Article
Full-text available
Hepatocellular carcinoma (HCC) is the 5th most common cancer disease and the 3rd cause of cancer related disease. Oxidative stress and inflammatory reactions are increases due to the expansion of hepatic cancer. Daphnetin is a well-known antioxidant and anti-inflammatory drug. The current experimental study was exploring the chemoprotective effect of daphnetin against diethylnitrosamine (DEN) induced HCC in rats and scrutinizing the possible mechanism. In this experimental study, Swiss Wistar rats were used for the current protocol and intraperitoneal injection of DEN (200 mg/kg) and phenobarbital (8 mg/kg) were used for the induction and progression of HCC and after induction the HCC, the rats were received the oral administration of different doses of daphnetin. Body weight was estimated at regular time intervals. Macroscopical evaluation was done at the end of the experimental study for the confirmation of hepatic nodules. Hepatic markers, antioxidant and inflammatory mediators were estimated in the serum of experimental rats. Daphnetin treatment successfully attenuated the hepatic injury induced by DEN/Pb as shown by the suppressed the levels of biochemical parameters including alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (T-Bil) and total protein (TP). Daphnetin significantly (p < 0.001) enhanced the level of glutathione (GSH), glutathione S-transferase (GST), superoxide dismutase (SOD), catalase (CAT) and decreased the malonaldehyde (MDA) level. Daphnetin treatment significantly altered the level of phase I and phase II enzymes and also significantly (p < 0.001) decreased the level of interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α); inflammatory mediators include cyclooxygenase-2 (COX-2), nuclear kappa B factor (NF-κB) and prostaglandin (PGE2). Collectively, we can say that daphnetin suggestively suppressed the hepatic cancer via suppression of antioxidant and inflammatory reactions. graphical abstract Fullsize Image
Article
Full-text available
In this study, we characterized an active film made of sodium alginate (SA)—locust bean gum (LBG) containing daphnetin-based film. Physicochemical characteristics, as well as antioxidant and antibacterial properties, were investigated. The results showed that the addition of a low concentration of daphnetin increased the flexibility of SA–LBG cling film, leading to an improvement in elongation at break and tensile strength. As the daphnetin content increased, solubility, brightness and transparency of the cling film decreased, and the moisture permeability increased. The antioxidant capacity and antibacterial activity of films with daphnetin were improved compared to those of the basal film. In addition, the cling film formed by adsorption had higher bacterial (Shewanella putrefaciens and Pseudomonas fluorescens) inhibition and antioxidant activity rates than direct film formation. The results indicate that the combination of daphnetin in SA–LBG film provides an active film with antioxidant and antibacterial properties, with potential for the development of food-grade packaging material.
Article
Full-text available
Severe acute pancreatitis (SAP) is often associated with pulmonary inflammation leading to acute lung injury. Daphnetin, a natural coumarin derivative, has been reported to exert anti-inflammatory effects. Here, we explored the effect and possible mechanism of daphnetin in a mouse model of SAP-associated lung injury induced by an intraperitoneal injection of l -arginine. The severity of pancreatic and lung injury is determined by histology and its score. Immunostaining of inflammatory and apoptotic cells was used to demonstrate lung tissue inflammation and apoptosis; ELISA analysis of serum and tissue cytokine levels; and western blotting and immunohistochemical staining for the activated Janus kinase 2 (JAK2)–signal transducer and activator of transcription protein 3 (STAT3) signalling pathway in lung tissues. Daphnetin pretreatment significantly reduced SAP-induced pancreatic and lung tissue damage, reduced interleukin-6 and tumour necrosis factor-α concentrations in both serum and lung tissues, reduced serum amylase and myeloperoxidase activities, and reduced macrophage (CD11b) and neutrophil (Ly6G) infiltration and cell apoptosis in the lung tissue. Moreover, SAP-induced phosphorylation of JAK2 and STAT3 in the lung tissue was also significantly diminished by the daphnetin pretreatment. These results indicated that daphnetin reduces SAP-associated lung tissue damage, likely by inhibiting the activation of JAK2–STAT3 signalling.
Article
Daphnetin is a dehydroxylated derivative of coumarin isolated from Daphne species. However, the effect of daphnetin on melanogenesis has not been elucidated. This study aims to investigate the inhibitory effect of daphnetin on melanogenesis in α-melanocyte stimulating hormone (α-MSH)-treated B16F10 cells and its potential mechanism. Melanin content analysis and cellular tyrosinase activity assay showed that daphnetin inhibited melanin biosynthesis in α-MSH-treated B16F10 cells. Immunoblotting and qRT-PCR also indicated that daphnetin suppressed the expression of microphthalmia-associated transcription factor, a mastering transcription factor of melanogenesis and its downstream melanogenic enzymes including tyrosinase and tyrosinase-related proteins. Moreover, daphnetin downregulated the phosphorylation of PKA, ERK, MSK1, and CREB. Additionally, daphnetin inhibited melanin synthesis in UVB-irradiated HaCaT conditioned medium system suggesting that daphnetin has potential as an antipigmentation activity in a physiological skin condition. Our data propose that daphnetin inhibits melanogenesis via modulating both the PKA/CREB and the ERK/MSK1/CREB pathways.
Article
Evidence has demonstrated that Daphnetin has antiangiogenesis activity, indicating it might be a new multi‐targeted medication for cancer therapy. Here, we aimed to reveal Daphnetin role in hepatocellular carcinoma (HCC) progression and the underlying mechanism. Huh7 and SK‐HEP‐1, two human HCC cell lines were used in this study. MTT (3‐(4,5‐Dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide), colony formation, flow cytometry, and tumor‐bearing experiments were applied to evaluate the effects of different concentrations of Daphnetin on cell viability, apoptosis, cell cycle, and in vivo tumor formation, respectively. Real‐time PCR (Polymerase Chain Reaction)and western blotting were applied to measure the mRNA and protein levels of β‐catenin. We observed that Daphnetin inhibited cell viability and tumorigenesis, promoted cell apoptosis, and induced a G1 phase arrest in a dose‐dependent manner in both Huh7 and SK‐HEP‐1 cells, which were rescued by SKL2001, an activator of the Wnt/β‐catenin signaling. Taken together, this study reveals that Daphnetin exerts an antitumor role in HCC through the inactivation of Wnt/β‐catenin signaling.
Article
Alkali burn is a significant contributor to corneal injury. Alkali burn-induced corneal inflammation often causes vision loss due to corneal neovascularization. Daphnetin (DAP) has been studied for its anti-inflammatory and antiangiogenic properties with encouraging results. Driven by those encouraging results, we sought to explore the effects of DAP in treating alkali burn-induced corneal inflammation and neovascularization and its mechanism of action. We found that the angiogenesis processes of human umbilical vein endothelial cells (HUVECs) induced by vascular endothelial growth factor A (VEGF-A) were primarily attenuated by treatment with DAP, including proliferation, migration, and tube formation. Treatment of DAP significantly suppressed the VEGF-A-induced protein expression of VEGF receptor2 (VEGFR2), as well as the activation of downstream signal transducer and activator of transcription 3 (STAT3), AKT, and extracellular signal-regulated kinase (ERK) signaling. In the mouse corneal alkali burn model, the inflammatory cell infiltrations and neovascularization in the cornea caused by alkali burn were inhibited by 10 µM DAP eye drops. Alkali burn-induced corneal protein expression of VEGF-A, VEGFR2, phosphorylated (p-)STAT3, p-AKT, and p-ERK in corneal tissue were reduced mainly by DAP. Moreover, the upregulation of inflammatory caused by alkali burn in the pathological process was significantly neutralized by DAP. Mechanistically, the inflammatory response could be alleviated by DAP in the way of inhibiting the expression levels of TLR4, p-NF-κB, NLRP3, ASC, Cleaved-caspase-1 (p20), mature-IL-1β (p17), and N-GSDM. In conclusion, our findings confirmed that the corneal inflammation and neovascularization caused by alkali burn could be inhibited by DAP in vitro and in vivo, elucidating the underlying mechanisms of its protective effects. DAP may have tremendous therapeutic potential for the treatment of corneal alkali burn.
Article
The aim of the present work was to isolate and identify secondary metabolites of Daphne oleoides, which was not phytochemically analysed in this important geographical region of the world until now, followed by the determination of anti-bacterial activity. An important plant coumarin daphnetin 8-methyl ether (1)[7-hydroxy8-methoxy coumarin] not previously reported from this species was isolated from the methanolic extract of aerial parts of Daphne oleoides, along with already reported 4-ethoxy benzoic acid (2)and 4-hydroxy benzoic acid(3) by repeated column chromatography. The structures of these compounds were elucidated on the basis of 1H, 13C NMR and MS analysis. The compound 1 was evaluated for its anti-bacterial potential which showed moderate activity.
Article
Daphnetin (7, 8-dihydroxycoumarin, DAPH), a coumarin derivative isolated from Daphne odora var., recently draws much more attention as a promising drug candidate to treat neuroinflammatory diseases due to its protective effects against neuroinflammation. However, itscontribution to chronic inflammatory pain is largely unknown. In the current work, we investigated the effects of DAPH in a murine model of inflammatory pain induced by complete Freund's adjuvant (CFA) and its possible underlying mechanisms. Our results showed that DAPH treatment significantly attenuated mechanical allodynia provoked by CFA. A profound inhibition of spinal glial activation, followed by attenuated expression levels of spinal pro-inflammatory cytokines, was observed in DAPH-treated inflammatory pain mice. Further study demonstrated that DAPH mediated negative regulation of spinal NF-κB pathway, as well as its preferential activation of Nrf2/HO-1 signaling pathway in inflammatory pain mice. This study, for the first time, indicated that DAPH might preventthe development of mechanical allodynia in mice with inflammatory pain. And more importantly, these data provide evidence for the potential application of DAPH in the treatment of chronic inflammatory pain.
Article
Chronic joint inflammatory disorders like osteoarthritis and rheumatoid arthritis, which are manifested by joint dysfunction, show an upsurge in inflammation and oxidative stress. Although conventional anti-arthritic drugs are being used to relieve pain from arthritic symptoms, they usually cause severe side effects. Traditionally used Ayurvedic medicinal plants are a promising alternative for the management of arthritic symptoms, as they are safe and effective. Ayurvedic medicinal plants improve arthritic symptoms by reducing joint tenderness, joint pain, swelling, bone and cartilage damage, and increasing knee flexion, walking distance and sports activities. These beneficial effects of Ayurvedic medicinal plants on arthritis are mediated through various cellular mechanisms including inhibition of the inflammatory markers NF-κB, cytokines, adipokines, PGE2, NO, iNOS, COX-2, and MMPs and induction of antioxidant status by decreasing free radicals, lipid peroxidation, and myeloperoxidase, and increasing antioxidant enzymes, Nrf2, and HO-1. Thus, a strategy requires using these Ayurvedic medicinal plants to treat arthritis. This article describes the status of inflammation and oxidative stress in arthritic conditions. We also provide evidence that Ayurvedic medicinal plants and their bioactive components are highly effective in improving arthritic symptoms.
Article
Daphnetin, a biologically active coumarin derivative found in plants of the genus Daphne, is a potent antioxidant phenolic compound. The present work describes the mechanisms and kinetics of the HO, NO, HOO, and NO2 scavenging activities of daphnetin in physiological environments using quantum chemistry calculations. The main antiradical mechanisms have been studied: formal hydrogen transfer (FHT), sequential electron transfer proton transfer (SETPT), sequential proton loss electron transfer (SPLET), and radical adduct formation (RAF). Besides its good HO scavenging activity in physiological environments, daphnetin is expected to exhibit good HOO and NO2 scavenging activities in water with koverall = 1.51 × 10⁷ and 4.79 × 10⁸ M⁻¹s⁻¹, respectively. The FHT mechanism decides the HO scavenging activity in aqueous solution, as well as HO, HOO, and NO2 scavenging activities in lipid media, while SPLET is the primary mechanism in water for HOO and NO2 scavenging activities. The theoretical predictions were found to be in good agreement with the available experimental data, which supports the reliability of the calculations.