ArticlePDF AvailableLiterature Review

Pharmacological, Neurochemical, and Behavioral Mechanisms Underlying the Anxiolytic- and Antidepressant-like Effects of Flavonoid Chrysin

Authors:

Abstract and Figures

Chrysin (5,7-dihydroxyflavone) is a flavonoid isolated from plants, such as Passiflora coerulea, Passiflora incarnata, and Matricaria chamomilla. This natural molecule exerts diverse pharmacological effects, which includes antioxidant, anti-inflammatory, anti-cancer, neuroprotective, and anti-apoptotic effects. Additionally, in brain structures, such as the hippocampus, prefrontal cortex, raphe nucleus, and striatum, involved in the physiopathology of anxiety and depression disorders, several neuropharmacological activities, including the activation of neurotransmitter systems (GABAergic, serotonergic, dopaminergic, and noradrenergic), neurotrophic factors, such as brain-derived neurotrophic factor and the nerve growth factor, and some signaling pathways are affected. The results showed that the anxiolytic and antidepressant-like effects of chrysin occurs through its interaction with specific neurotransmitter systems, principally the GABAergic and the serotonergic, and activation of other neurotrophic factors. However, it is not possible to discard the antioxidant and anti-inflammatory activities of chrysin while producing its anxiolytic- and antidepressant-like effects. Although these results have been obtained principally from pre-clinical research, they consistently demonstrate the potential therapeutic use of flavonoid chrysin as an anxiolytic and antidepressant agent. Therefore, this flavonoid could be considered as a promising novel therapy for anxiety and depression disorders.
Mechanism of action of the flavonoid chrysin potentially involved in its anxiolytic-like effects. (A) It has been confirmed that chrysin produces its anxiolytic-like effect through its action on the GABAA/benzodiazepine receptor complex producing configurational changes in the receptor and regulating the opening of the Cl − ion channel [14,15,17,24,35]; which may produce inhibitory effects in the GABAergic system associated with its anxiolytic-like effects. These effects can be blocked by specific antagonists of the GABAA receptor such as picrotoxin, bicuculline and flumazenil [14]. (B) Probably, anti-oxidant effects of chrysin could be involved in its anxiolytic-like effects. Chrysin significantly reduces ROS by inhibiting the production of NO, NT, and NOX4 [29]. These effects reduce the oxidative stress and reduces the neuronal damage. Additionally, chrysin reduces the activity of Bax, caspase-9 and caspase-3, while increasing the production of Bcl-2, thereby reducing the damage of DNA and inhibiting apoptotic processes [60,61]; which reduces the neuronal death. (C) Additionally, the anti-inflammatory effects of chrysin could contribute in its anxiolyticlike effects, considering that it may reduce the inflammatory response by inhibiting the signaling pathway NF-κB/IKK-β [27,28]. Chrysin may attenuate the expression of NF-κB that participates as transcriptional factors at nuclear level, binding to genes that induce neuro-inflammation process. Chrysin also inhibits the production of pro-inflammatory cytokines like IL-1β and IL-6, in addition to suppressing the production of proinflammatory mediators like TNF-α, PGE2 and COX-2 [27,28,32]. These effects could reduce neuro-inflammation associated with the anxiety-like behavior. ROS = reactive oxygen species; NO = nitric oxide; NT = nitrotyrosine; NOX4 = NADPH oxidase; O2¯ = superoxide; Green circles = chlorine ions; SOD = superoxide dismutase; GSH = reduced glutathione; CAT = catalase; GPx = glutathione peroxidase; Bcl-2 = anti-apoptotic protein of the subfamily Bcl-2; Bax = pro-apoptotic protein of the subfamily Bax; NF-κB = nuclear factor kappa B; IKK-β = inhibitor of nuclear factor kappa-B; TNF-α = tumor necrosis factor-α; IL-1β = interleukin-1β; IL-6 = interleukin-6; PGE2 = prostaglandins E2; COX-2 = cycloxygenase-2. (Figure was prepared by the authors).
… 
Content may be subject to copyright.
Molecules 2022, 27, x. https://doi.org/10.3390/xxxxx www.mdpi.com/journal/molecules
Review
Pharmacological, Neurochemical, and Behavioral Mechanisms
Underlying the Anxiolytic- and Antidepressant-Like Effects of
Flavonoid Chrysin
Juan Francisco Rodríguez-Landa 1,2, *, León Jesús German-Ponciano 1, Abraham Puga-Olguín 3
and Oscar Jerónimo Olmos-Vázquez 1
1 Laboratorio de Neurofarmacología, Instituto de Neuroetología, Universidad Veracruzana,
Xalapa 91190, Mexico; lgerman@uv.mx (L.J.G.-P.); oscarplsstahp1@gmail.com (O.J.O.-V.)
2 Facultad de Química Farmacéutica Biológica, Universidad Veracruzana, Xalapa 91000, Mexico
3 Unidad de Salud Integrativa, Centro de EcoAlfabetización y Diálogo de Saberes, Universidad Veracruzana,
Xalapa 91090, Mexico; abpuga@uv.mx
* Correspondence: juarodriguez@uv.mx
Abstract: Chrysin (5,7-dihydroxyflavone) is a flavonoid isolated from plants, such as Passiflora co-
erulea, Passiflora incarnata, and Matricaria chamomilla. This natural molecule exerts diverse pharma-
cological effects, which includes anti-oxidant, anti-inflammatory, anti-cancer, neuroprotective, and
anti-apoptotic effects. Additionally, in brain structures such as hippocampus, prefrontal cortex, ra-
phe nucleus, and striatum, involved in the physiopathology of anxiety and depression disorders,
several neuropharmacological activities, including the activation of neurotransmitter systems (GA-
BAergic, serotonergic, dopaminergic, and noradrenergic), neurotrophic factors such as brain-de-
rived neurotrophic factor and the nerve growth factor, and some signaling pathways are affected.
The results showed that the anxiolytic and antidepressant-like effects of chrysin occurs through its
interaction with specific neurotransmitter systems, principally the GABAergic and the serotonergic,
and activation of other neurotrophic factors. However, it is not possible to discard the anti-oxidant
and anti-inflammatory activities of chrysin while producing its anxiolytic- and antidepressant-like
effects. Although these results have been obtained principally from pre-clinical research, they con-
sistently demonstrate the potential therapeutic use of flavonoid chrysin as an anxiolytic and anti-
depressant agent. Therefore, this flavonoid could be considered as a promising novel therapy for
anxiety and depression disorders.
Keywords: antidepressant; anxiolytic; chrysin; flavonoid; natural medicine; neuropharmacology
1. Introduction
Throughout the development of pharmacology, the study of natural molecules has
been of special importance to identify secondary metabolites from plants and to stimulate
the discovery and design of new drugs for the treatment of diseases in humans [1]. The
multidisciplinary study of diverse molecules extracted from plants, isolated, and chemi-
cally characterized, has permitted the identification of the mechanism of action involved
in its beneficial and toxic effects [2]. The use of molecular, biochemical, pharmacological,
histological, and behavioral techniques has helped develop multi-target drugs for the pre-
vention and treatment of diverse diseases, particularly those that have recently increased
in association with social dynamics, lifestyle, and environmental factors, such as neuro-
psychiatric and stress-related disorders [3].
Polyphenols are a group of molecules with multiple beneficial effects on health. Its
anti-oxidant effects beneficially impact the physiological process of the organism [4].
Among polyphenols, flavonoids have been widely studied to develop complementary
Citation: Rodríguez-Landa, J.F.; Ger-
mán-Ponciano, L.J.;
Puga-Olguín, A.; Olmos-Vázquez,
O.J. Pharmacological, Neurochemi-
cal, and Behavioral Mechanisms Un-
derlying the
Anxiolytic- and Antidepressant-Like
Effects of Flavonoid Chrysin.
Molecules 2022, 27, x.
https://doi.org/10.3390/xxxxx
Academic Editor(s):
Received: date
Accepted: date
Published: date
Publisher’s Note: MDPI stays neu-
tral with regard to jurisdictional
claims in published maps and institu-
tional affiliations.
Copyright: © 2022 by the authors.
Submitted for possible open access
publication under the terms and con-
ditions of the Creative Commons At-
tribution (CC BY) license (https://cre-
ativecommons.org/licenses/by/4.0/).
Molecules 2022, 27, x FOR PEER REVIEW 2 of 18
therapeutic strategies for the treatment of metabolic, cardiovascular, neuropsychiatric dis-
orders and cancer [57]. In vitro and in vivo studies on the pharmacological action of the
flavonoid chrysin have identified multiple effects of it on different systems in the organ-
ism [810], including the central nervous system (CNS). Chrysin exerts a neuropharma-
cological effect in brain structures such as the amygdala, hippocampus (HP), prefrontal
cortex (PFC), and raphe nucleus, which are involved in the physiopathology of several
neuropsychiatric disorders, such as anxiety and depression [1114]. The effects include
activation of the GABAergic system by modulating the GABAA/benzodiazepine receptor
complex [1417]; changes in serotonin levels and modification in the expression of their
receptors such as 5-HT1A and 5-HT2A in the raphe nucleus and HP [13,18]; and restoration
of dopamine and noradrenaline levels in the CNS [12,19]. In addition, chrysin activates
neurotrophic factors that increase brain-derived neurotrophic factor (BDNF) and nerve
growth factor (NGF) levels, thereby activating signaling pathways in the brain [11,20].
However, anti-inflammatory, anti-apoptotic, and neuroprotective effects should be con-
sidered as potential mechanisms of action involved in their anxiolytic- and antidepres-
sant-like effects, considering that neuroinflammation and apoptotic processes are in-
volved in the pathophysiology of anxiety and depression disorders [21].
In this review, we describe, analyze, and discuss the scientific results from pre-clini-
cal research that report the pharmacological, neurochemical, and behavioral mechanisms
that could underlie the anxiolytic and antidepressant effects of flavonoid chrysin, which
is considered as a promising therapy for anxiety and depression disorders. In addition,
we propose the incorporation of chrysin as a potential anxiolytic and antidepressant drug
in future scientific research, not only for its classical actions on neurotransmission sys-
tems, but also for its anti-oxidant and anti-inflammatory effects, and the activation of neu-
rotrophic factors and the associated signaling pathways. This may contribute to the de-
velopment of specific therapies for anxiety and depression disorders according to their
etiology.
2. Generalities of the Flavonoid Chrysin
Flavonoids are polyphenolic compounds that are present in plants. They produce
pharmacological actions in the peripheral and CNS [9]. They can cross the blood-brain
barrier and interact with several neurotransmission systems and, thereby activating sig-
naling pathways in specific brain structures involved in the physiopathology of anxiety
and depression disorders [10]. In particular, the flavonoid chrysin (5,7-dihydroxyflavone)
has been studied for its anti-oxidant properties; however, its neuropharmacological effects
in specific brain structures involved in the physiopathology of several neuropsychiatric
disorders such as anxiety and depression need to be studied [9,10,14].
Chrysin (Figure 1) has a backbone structure that consists of a fused A and C rings,
and a phenyl B ring, which is attached to the second position of ring C and shares the
basic structure of the flavones, with an additional hydroxyl group at the fifth and seventh
positions of the A ring. The potential of chrysin to act as a free radical scavenger has been
attributed to the presence of these hydroxyl groups [20,22], and it has been suggested that
these functional groups represent the main site of action of this flavonoid to produce a
great variety of pharmacological activities and therapeutic effects. It has the potential to
be used as an alternative in the treatment of metabolic, cardiovascular, and neuropsychi-
atric disorders [10]. In addition, the presence of hydroxyl groups in the backbone of chry-
sin has been associated with its anxiolytic-like effects [23]. Chrysin, but not the flavone
backbone, decreases anxiety-like behavior in rats and zebrafish, suggesting that the pres-
ence of hydroxyl groups in its basic structure is indispensable for producing anxiolytic-
like effects in pre-clinical research [23].
Molecules 2022, 27, x FOR PEER REVIEW 3 of 18
Figure 1. Basic structure of flavones showing fused A and C rings, and phenyl B rings with corre-
sponding numbering system (left figure). Structure of the flavonoid chrysin, 5,7-dihydroxyflavone
(right figure).
Chrysin, either isolated from plants like Passiflora coerulea, Passiflora incarnata, and
Matricaria chamomilla, or even as a synthetic drug, produces anxiolytic- and antidepres-
sant-like effects. These effects involve several activation of neurotransmission systems
and signaling pathways, including the serotonergic and GABAergic systems, and the ac-
tivation of neurotrophic factors, such as BDNF and NGF. It is likely that the activation of
anti-inflammatory and anti-oxidant signaling pathways may also be involved in these ef-
fects. Although these effects have been principally evaluated in pre-clinical research
[14,1618,2426], they show the potential therapeutic use of chrysin for anxiety, depres-
sion, and other neuropsychiatric disorders.
3. Biochemical and Pharmacological Activity of Flavonoid Chrysin
Diverse flavonoids, including chrysin, exert significant anxiolytic- and antidepres-
sant-like activities in mammals and non-mammals, similar to those produced by clinically
effective anxiolytic and antidepressant drugs [5,10,13]. These effects have been related to
its neurochemical activity on the CNS that impact neuronal functioning (Table 1).
Table 1. Some neurochemical effects produced by flavonoid chrysin potentially involved in its an-
xiolytic- and antidepressant-like effects.
Activity
Chrysin Treatment
Effects
Reference
Anti-oxidant
20 mg/kg/30 days, p.o.
TBARS, lipid hydroperoxides, conjugated
dienes tissue, circulatory levels
SOD, CAT, GPx, Gsr activity, GSH, GSTs, vit-
amin C and vitamin E levels in ethanol-induced
toxicity in rats
[22]
50 mg/kg/12 days, i.p.
GHS levels and CAT and SOD activity in
heart homogenate in male rats
[27]
30 and 60 mg/kg/28 days,
p.o.
NO and GHS levels, GSHPx, CAT, and SOD
activity in rat heart homogenate
[28]
60 mg/kg/28 days, p.o
8-OHdG, TBARS levels
GSH, CAT, NO levels
[29]
1 and 10 mg/kg/60 days,
p.o.
SOD, CAT and GPx activity in PFC and HP of
aged mice
[20]
1.25, 2.5, and 5 µM/30 min
exposure
ROS formation in neuronal SH-SY5Y and mi-
croglial THP-1 cells in vitro
[30]
10, 30, and 100 mg/kg/44
days, p.o.
Protects against aluminum-induced oxidative
stress by restored LPO levels and SOD and
CAT activity in cortex and HP of male Swiss
mice
[30]
Molecules 2022, 27, x FOR PEER REVIEW 4 of 18
Anti-inflammatory
7.50, 4.75, and 120.90 µM,
18 h exposure
NO, PGE2 and TNF-α biosynthesis
in CLP-induced RAW 264.7 cells
[31]
30 mg/kg/2 weeks, i.p.
ALT and AST activity
TNF-α and IL-1β levels
IL-10 and adiponectin in high-fat feeding
mice
[32]
25 and 50 mg/kg/12 days,
i.p.
NF-κB, iNOS, COX-2, and TNF-α expression
in heart homogenate of DOX-induced cardio-
toxicity mice
[27]
30 and 60 mg/kg/28 days,
p.o.
PPAR-γ and TGF-β expression
NF-κBp65 and IKK-β expression
and TNF-α level in heart homogenate of isopro-
terenol-induced myocardial injury rats
[28,29]
5 and 20 mg/kg/28 days,
p.o.
TNF-α, IL-1β and IL-6 levels in PFC and HP
of chronically stressed mice
[18]
5 mg/kg/1 h before LP, i.p.
AST and TNF-α serum levels in septic mice
survival
[33]
5 µM/24 h exposure
iNOS, IL-1β, and TNF-α expression in micro-
glial THP-1 cells exposed to LPS
[30]
GABAergic/BZD
3 µM, 60 min exposure
Acts as competitive ligand for central BZD site
in bovine cerebral cortical membranes in vitro
[15]
13 µM, 60 min exposure
Acts as competitive ligand for peripheral BZD
binding site in rat kidneys
membranes in vitro
[15]
1 mg/kg, i.p.
Activates the GABAA/BZD receptor complex in
male CF1 mice
[24]
1 mg/kg, i.p.
Activates the GABAA/BZD receptor complex in
male Sprague-Dawley rats
[17]
0.62 µM, 2 h exposure
Acts as competitive ligand for central BZD site
in synaptosomal fractions of rat brain in vitro
[34]
10 and 30 µM, 30 s expo-
sure
Modulates the activity of Cl ion channel in the
GABAA receptor expressed in Xenopus oocytes
in vitro
[35]
2 mg/kg, i.p.
Anxiety-like behavior by modulating Cl ion
channel in the GABAA receptor of cycling fe-
male rats
[36]
2 mg/kg, i.p.
Depression-like behavior by modulating
GABA-binding site in the GABAA receptor of
ovariectomized female rats
[37]
0.5 µg/rat, i.h.
Anxiety-like behavior by modulating
GABAA/BZD receptor complex in the dorsal
hippocampus of cycling female rats
[14]
Serotonergic
5 and 20 mg/kg, p.o.
5-HT levels and 5-HIAA/5-HT ratio in HP of
chronic stressed mice
[18]
20 mg/kg/28 days, p.o.
5-HT levels in PFC and HP in female mice
with hypothyroidism
[12]
50 mg/kg twice a day per 4
days, p.o.
5-HT levels in the striatum
of the rat brain
[38]
10 and 30 mg/kg/2 weeks,
p.o.
5-HT spinal levels
[39]
Molecules 2022, 27, x FOR PEER REVIEW 5 of 18
5-HIAA/5-HT ratio in male mice with experi-
mental neuropathy
5 mg/kg/28 days, i.p.
5-HT1A receptor expression in
the dorsal raphe
5-HT1A and 5-HT2A in the hippocampus of
male rats
[13]
Dopaminergic
10 mg/kg/28 days, p.o
DA striatal levels in mice
[19]
50, 100 and 200 mg/kg/ 5
days, p.o.
DA levels in striatum of mice treated
with 1-methyl-1,2,3,6-tetrahidropidine
[40]
20 mg/kg/28 days, p.o.
DA levels in PFC and HP in a hypothyroid-
ism model in female mice
[12]
Noradrenergic
50, 100 and 150 mg/kg, i.p.
NE serum levels in rats with pain
induced by formalin
[41]
20 mg/kg/28 days, p.o.
No effects
[12]
Anti-apoptotic
25 and 50 mg/kg/12 days,
i.p.
Bax, caspase-3, and cytochrome c activity
Bcl-2 expression in rat heart tissue extract
[27]
30 and 60 mg/kg/28 days,
p.o.
Bcl-2 expression
Bax and caspase-3 activity
[29]
5 and 20 mg/kg/28 days,
p.o.
Caspase-3 and caspase-9 activity in HP and
PFC of chronically stressed mice
[18]
25, 50 and 100 mg/kg/3
days, p.o.
Apoptotic index in cerebral cortex
and HP of rats with traumatic brain injury
[42]
Neuroendocrine
5 and 20 mg/kg/28 days,
p.o.
Corticosterone plasma levels in chronically
stressed mice
[11]
5 and 20 mg/kg/28 days,
p.o.
CRH and ACTH in chronically stressed mice
[18]
50, 100 and 150 mg/kg, i.p
Corticosterone serum levels in rats with pain
induced by formalin
[41]
Neurotrophic
5 and 20 mg/kg/28 days,
p.o.
BDNF and NGF levels in PFC and HP in
chronically stressed mice
[11]
1 and 10 mg/kg/60 days,
p.o.
BDNF levels in HP and PFC in aged mice
[20]
10 mg/kg/28 days, p.o.
BDNF and NGF levels in striatum in a Parkin-
son’s disease model in mice
[19]
20 mg/kg/28 days, p.o.
BDNF and NGF in HP and PFC in mice sub-
jected to a hypothyroidism model
[43]
TBARS = thiobarbituric acid reactive substance; SOD = superoxide dismutase; CAT = catalase; GPx
= glutathione peroxidase; Gsr = glutathione reductase; GSTs = glutathione-S-transferase; GSH = re-
duced glutathione; NO = nitric oxide; GSHPx = plasma glutathione peroxidase; 8-OHdG = 8-hy-
droxy-2-deoxyguanosine; PFC = prefrontal cortex; HP = hippocampus; ROS = reactive oxygen spe-
cies; LPO = lipid peroxidation; PGE2 = prostaglandin E 2; TNF-α = tumor necrosis factor-α; CLP =
cecal ligation and puncture procedure; ALT = alanine aminotransferase; AST = aspartate ami-
notransferase; IL-1β = interleukin 1 beta; IL-10 = interleukin 10; NF-κBp65 = nuclear transcription
116 factor kappa B heterodimer; iNOS = inducible nitric oxide synthase; COX- 2 = cyclooxygenase-
2; DOX = doxorubicin; PPAR-γ = peroxisome proliferator-activated receptor-gamma; TGF-β = trans-
forming growth factor-beta; IKK-β = inhibitor of nuclear factor kappa-B kinase subunit beta; IL-6 =
interleukin-6; LPS = lipopolysaccharide; BZD = benzodiazepine; GABA = gamma-aminobutyric
acid; 5-HT = 5-hydroxytriptamina, serotonin; 5-HIAA = 5-hydroxyindoleacetic; 5-HT1A = serotonin
1A receptor; 5-HT2A = serotonin 2A receptor; DA = dopamine; NE = noradrenaline; BAX = pro-apop-
totic protein of the subfamily Bax; Bcl-2 = B-cell lymphoma-2; CRH = corticotropin-releasing hor-
mone; ACTH = adrenocorticotropic hormone; BDNF = brain-derived neurotrophic factor; NGF =
nerve growth factor; = increase; = decrease; p.o. = per oral route; i.p. = intraperitoneal injection;
i.h. = intrahippocampal microinjection.
Molecules 2022, 27, x FOR PEER REVIEW 6 of 18
3.1. Action of Chrysin on Neurotransmission Systems
Chrysin was the first monoflavonoid reported as a specific ligand for central and pe-
ripheral benzodiazepine-binding sites [15]. It was identified when chrysin inhibited the
[3H] flunitrazepam binding in synaptosomal membranes of the bovine cerebral cortex.
Similarly, chrysin also displaced the binding of [3H]Ro 5-4864, a potent ligand for the pe-
ripheral benzodiazepine receptor, to kidney membranes in a mixed, competitive and non-
competitive manner. Interestingly, intracerebroventricular microinjection of chrysin pre-
vents tonic-clonic seizures induced by pentylenetetrazol [15]. This effect was blocked by
Ro 15-1788, a central benzodiazepine receptor antagonist, suggesting that this flavonoid
exerts some of its pharmacological actions on the benzodiazepine-binding site of the
GABAA receptor, enhancing the activation of GABAergic neurotransmission [15]. In sup-
port of this, in vitro studies have reported that chrysin and other flavonoids modulate the
chloride ion channel in the GABAA receptor in a manner different from benzodiazepines
[35].
Additionally, chrysin can also activate other neurotransmitter systems involved in
the physiopathology of several neuropsychiatric disorders, including anxiety and depres-
sion. Chrysin at 10 mg/kg for 28 days restored dopamine, homovanillic acid, and 3,4-di-
hydroxyphenylacetic acid levels in the stratum of male C57B/6J mice, in which a depletion
of this neurotransmitter was induced by 6-hidroxidopamine [19]. In addition, chrysin at
50, 100 and 200 mg/kg for 5 days prevented the decrease in dopamine in the striatum
produced by the injection of 1-methyl-a,2,3,6-tetrahidropidine in C57BL/6J mice [40]. In
contrast, chrysin at 20 mg/kg for 28 days reversed the decrease in dopamine and serotonin
levels in the PFC and HP of female mice subjected to a hypothyroidism model [12]. Chry-
sin at 50 mg/kg for 4 days and chrysin 330 mg/kg for 2 weeks significantly increased
serotonin levels in mice and rat brains [38,39]; while chrysin 50, 100, and 150 mg/kg atten-
uated the increased noradrenaline and corticosterone levels in male Wistar rats with pain
induced by formalin [41].
It has been shown that the flavonoid chrysin has a positive impact on several neuro-
transmitter systems that participate in the etiology of anxiety and depression; however, it
is also capable of modifying several signaling pathways and activating neurotrophic pro-
cesses in the CNS, which could be related to its anxiolytic- and antidepressant-like effects.
3.2. Anti-Oxidant Activity of Chrysin
Psychiatric disorders have been linked to alterations in the activity of anti-oxidant
enzymes in the CNS, leading to an increase in the production of reactive oxygen species
(ROS) and, in the first instance, induction of oxidative stress processes [44]. Oxidative
stress leads to an imbalance between the level of anti-oxidants and production of ROS,
which can generate long-term alterations such as damage to neuronal membranes with
potential activation of apoptosis processes associated with the increase of ROS [45]. These
alterations, induced by oxidative stress, are involved in the etiology of anxiety and de-
pression disorders [46,47]. Chrysin has the capacity to modulate the nitric oxide (NO)
pathway, which contributes to the production of ROS, in both in vivo and in vitro assays
[31,33]. In a diabetic rat model, chrysin 60 mg/kg for 28 days inhibited oxidative stress by
restoring the alterations in NO, glutathione (GSH), catalase (CAT), thiobarbituric acid re-
active substances (TBARS) levels, along with the restoration of the activity of superoxide
dismutase (SOD), nitrotyrosine (NT), and NADPH oxidase 4 (Nox4) [29]. The same effect
was produced by the administration of chrysin at 20 mg/kg for 30 days, exerting a reduc-
tion in ethanol-induced toxicity, as evidenced by decreasing markers of oxidative stress
in organs such as the liver, kidney, and heart of rats [22]. In addition, chrysin 50 mg/kg
restored the levels of oxidative stress markers such as GSH, malondialdehyde (MDA),
CAT, SOD, and ROS under doxorubicin-induced acute cardiotoxicity in rats to normal
values [27]. The anti-oxidant effects of chrysin described above occur in the peripheral
organs of metabolically challenged (diabetic) or chemically treated (ethanol and
Molecules 2022, 27, x FOR PEER REVIEW 7 of 18
doxorubicin) animals; which is important because diabetes and alcohol intake is associ-
ated with oxidative stress and inflammation [48,49], and may predispose to anxiety and
depression disorders [50,51]. Interestingly, it has been previously reported in preclinical
and clinical studies that reduced oxidative stress at the peripheral level is associated with
a decrease in anxiety and depression symptoms [5255]. In this way, it is possible that
peripheral and central anti-oxidant effects of chrysin could contribute to its anxiolytic and
antidepressant effects; however, this aspect needs to be explored further.
In support, daily administration of chrysin at 1 and 10 mg/kg for 60 days restored the
activity of SOD and CAT in the HP and PFC of aged mice and reduced oxidative stress by
decreasing ROS and inversely increasing BDNF levels in these brain structures [20]. This
last data is important because a reduction in anxiety-like behaviors in the elevated plus
maze (EPM) test was related to high levels of BDNF, which was accompanied by greater
expression of TrkB and BDNF mRNA in brain structures such as the HP, PFC, and amyg-
dala [56]. Interestingly, the anxiolytic-like activity produced by diazepam has been asso-
ciated with the restoration of normal levels of anti-oxidant enzymes, such as SOD and
CAT, as well as a reduction in nitrite concentration in the mouse brain [57]. Therefore,
alterations in peripheral and central enzymatic anti-oxidant systems can generate oxida-
tive stress and contribute to anxiety- and depression-like behaviors. In this way, it is pos-
sible that these alterations related to oxidative stress may be restored by the flavonoid
chrysin and contribute to its anxiolytic- and antidepressant-like effects, without discard-
ing the participation of other neurochemical processes in the CNS.
3.3. Anti-Inflammatory and Anti-Apoptotic Activity of Chrysin
In vitro studies with chrysin showed attenuated release of pro-inflammatory media-
tors by inhibiting prostaglandins E2 (PGE2) and tumor necrosis factor-α (TNF-α) from
lipopolysaccharide-induced RAW 264.7 cells [31]. Chrysin regulates inflammatory and
apoptotic signaling pathways by attenuating the effects of nuclear factor-κB (NF-κB) ex-
pression, inhibition of nuclear factor kappa-B kinase (IKK-β), TNF-α, interleukin-(IL-
1β), interleukin-6 (IL-6), and cycloxygenase-2 (COX-2) [27,28,32]. In addition, in a mouse
model of polymicrobial sepsis induced by cecal ligation, the administration of chrysin at
5 mg/kg reduced TNF-α levels [33]. In addition, chrysin at 50 and 60 mg/kg for 12 to 28
days in models of myocardial injury in diabetic rats or doxorubicin-induced acute cardi-
otoxicity exerts anti-inflammatory effects. These effects were associated with the inhibi-
tion of TNF-α and NF-κB/IKK-β expression, as well as with the reduced apoptosis due to
increase in Bcl-2 (anti-apoptotic protein) expression and decrease in the expression of pro-
apoptotic markers such as Bax and caspase-3 [27,28]. These effects of chrysin at the pe-
ripheral level and in different models of inflammation support its potential anti-inflam-
matory and anti-apoptotic activities. This is important because peripheral and central pro-
inflammatory processes have been involved in anxiety development and depression dis-
orders [58], while some anxiolytic effects of drugs have been related to stable functioning
of COX-2 in the infralimbic and prelimbic cortex, HP, and ventral tegmental area [59]. In
addition, anxiolytic- and antidepressant-like effects produced while exercising are due to
the increase in Bcl-2 and the reduction of Bax and caspase-3 in the dorsal raphe nucleus of
rats evaluated in the EPM and forced swim test (FST) without observing neuronal death
[60,61]. In contrast, a reduction in Bcl-2 and an increase in Bax in the frontal cortex are
related to anxiety-like behavior and other stress-related disorders [60]. Therefore, chrysin
can produce anxiolytic- and antidepressant-like effects by exerting beneficial effects on
these biochemical systems.
Similarly, chrysin at 5 and 20 mg/kg for 28 days produced significant changes in neu-
rochemical factors that are implicated in the reduction of pro-inflammatory cytokines in
mice subjected to the chronic unpredictable mild stress model (CUMS) [18]. Furthermore,
chrysin also prevented the increase in concentrations of corticotropin-releasing hormone
(CRH) and adrenocorticotropic hormone (ACTH) in plasma, while the levels of TNF-α,
IL-1β, and IL-6 in the PFC and HP were reduced by both doses of chrysin [18]. Similarly,
Molecules 2022, 27, x FOR PEER REVIEW 8 of 18
under these conditions, restoration of caspase-3 and caspase-9 activity in the HP and PFC
was observed. All these changes were associated with a reduction in depression-like be-
haviors, reaching values similar to those reported in unstressed mice [18].
It is necessary to highlight that mice and rats that displayed lower anxiety-like be-
havior in the EPM test showing lower expression of pro-inflammatory cytokines, includ-
ing TNF-α, IL-1β, and IL-6 in the HP, frontal cortex, and serum [44,62]. However, a con-
trary effect was reported in animals showing high anxiety-like behavior. Additionally,
chrysin can regulate the hypothalamic-pituitary-adrenal (HPA) axis and physiological re-
sponses to stressful situations [18]. Adequate regulation of the HPA axis combined with
the lower production and release of corticosterone has been associated with the adequate
coping with stress and low risk of developing neuropsychiatric disorders [63,64].
Altogether, these results suggest that neurochemical changes produced by flavonoid
chrysin, in addition to suppressive effects on inflammatory and oxidative processes, could
contribute to its anxiolytic- and antidepressant-like effects [42,65]. These results support
the idea that chrysin could be considered a potential candidate for ameliorating anxiety
and depression symptoms in humans.
3.4. Effects of Chrysin on Gut-Microbiota
Currently, there is growing evidence proposing that dysregulation in the composi-
tion of the gut-microbiota is related to the pathophysiology of anxiety and depression
disorders due to its interaction with neuroimmune, neuroendocrine, and neural pathways
[66,67]. These pathways are part of the braingutmicrobiota axis that may modulate
brain development and function, which also impacts on behavior. For example, a fecal
microbiota sample from depressed patients when transferred by oral gavage to a micro-
biota-deficient rat model produced depressive-like behaviors in the recipient animal [68].
Contrarily, fecal microbiota coming from intact rats transplanted to chronically stressed
rats significantly reduced depression- and anxiety-like behaviors and anhedonia, which
was associated with the suppressed activation of glial cells and the NLRP3 inflammasome
in the brain [69]. Interestingly, polyphenols may regulate the composition of the gut-mi-
crobiota, promoting beneficial effects on the intestinal microbiota and inhibiting the pro-
liferation of harmful bacteria, thereby maintaining intestinal health and ameliorating
some neurological and neuropsychic disorders [70,71]; this is important because a correct
structure of gut microbiota in addition to an anti-inflammatory effect of diverse mole-
cules, including polyphenols, has been associated with its antidepressant-like effects [72].
In the specific case of chrysin, its beneficial effects in modulating the structural alteration
of the gut microbiota in mice have been explored [73]. Chrysin 10 mg/kg for 28 days at-
tenuated the increase in pro-inflammatory cytokines and the intestinal damage induced
by LPS in C57BL6/J mice, which was associated with adequate gut microbiota structure
[74]. Likewise, chrysin can modulate intestinal inflammation in Caco2 cells stimulated
with IL-1B, improving intestinal absorption and metabolic stability [75], which could be
related with a normal function of the gut microbiota; this is important because anti-in-
flammatory effects of several molecules at the peripheral and central level have been as-
sociated with a reduction of anxiety and depression-like behavior [58]. In this sense, it is
possible that the regulation of the gut microbiota by polyphenols such as chrysin may
open new perspectives to explore the effects of chrysin on the brain-gut-microbiota axis
and its potential relationship with its anxiolytic- and antidepressant-like effects.
4. Anxiolytic-Like Effects of Flavonoid Chrysin
In 1994, Wolfman et al. reported an anxiolytic-like effect of chrysin in mice. A single
dose of chrysin at 1 mg/kg significantly increased the time spent in open arms of the EPM
[24]. In the light/dark box (LDB), increased time was spent in the illuminated compart-
ment [17], and in both cases the effects were similar to that produced by diazepam. These
behavioral effects are considered to be associated with anxiolytic-like effects in pre-clinical
Molecules 2022, 27, x FOR PEER REVIEW 9 of 18
research. Chrysin, but not diazepam, is devoid of motor effects related to sedation [24];
and this may represent the advantage of chrysin over benzodiazepines such as diazepam
in the treatment of anxiety disorders [76]. Interestingly, in male Sprague-Dawley rats, the
anxiolytic-like effects of chrysin at 1 and 2 mg/kg in LDB were blocked by previous ad-
ministration of flumazenil [17,77], an antagonist of the benzodiazepine binding site in the
GABAA receptor. Additionally, acute administration of chrysin (1 mg/kg) produced anxi-
olytic-like effects in male Wistar rats [78] and CD-1 male mice [79] evaluated in the EPM.
Similarly, anxiolytic-like effects of chrysin in mammals (mice and rats) have also been
reported in non-mammalian organisms (zebrafish). Chrysin at 1 mg/kg decreased anxiety-
like behavior in rats and zebrafish, similar to diazepam [23]; however, treatment with a
flavone backbone at 1 mg/kg was devoid of anxiolytic-like effects in both rats and
zebrafish, suggesting that the presence of hydroxyl groups in its basic structure could be
indispensable to produce anxiolytic-like effects [23].
Anxiety symptoms in women are associated with a reduction in steroid hormones
such as estradiol and progesterone, and its reduced metabolite allopregnanolone in the
peripheral and CNS, which may occur pre-menstruation, post-partum, and transition to
menopause stage [80,81]. These steroid hormones may modulate several neurotransmis-
sion systems, such as the serotoninergic, noradrenergic, dopaminergic, and GABAergic
[82]; therefore, some of these hormones have been proposed as novel groups of anxiolytic
drugs for treating particular anxiety and depression disorders associated with reduced
concentrations of steroid hormones [83]. It has recently been proposed that the flavonoid
chrysin mimics some of the pharmacological effects of neurosteroids in female rats [37].
Anxiety-like behaviors in female rats significantly increase during metestrus-diestrus
phase of the ovarian cycle, which is associated with low concentration of steroid hormones
[84]; this phase is considered an equivalent of the premenstrual period in women [85].
Interestingly, chrysin at 2 mg/kg, similar to diazepam at 2 mg/kg, prevents anxiety-like
behavior that naturally occurs during metestrus-diestrus phase in female rats evaluated
in the EPM and LDB. This effect can be blocked by a previous injection of picrotoxin [36].
In support, microinjection of chrysin at 0.5 μg in the dorsal HP prevented anxiety-like
behavior that naturally occurs during diestrus, which was blocked by previous injection
of picrotoxin, bicuculline, and flumazenil, indicating that the GABA/benzodiazepine re-
ceptor complex in the dorsal HP mediates the anxiolytic-like effects of this flavonoid [86].
Interestingly, this same effect on anxiety-like behavior during diestrus was prevented by
microinjection of neurosteroid allopregnanolone at 0.5 μg into the dorsal HP, which was
blocked by picrotoxin, bicuculline, and flumazenil in the EPM [86]. In contrast, in a surgi-
cal menopause model in rats characterized by high anxiety-like behavior associated with
a permanent reduction of steroid hormones, chrysin at 2 and 4 mg/kg and diazepam at 1
mg/kg, reversed this anxiety-like behavior, which was blocked by a previous injection of
picrotoxin [26]. The fact that picrotoxin, bicuculline, and flumazenil prevented the anxio-
lytic-like effect of different doses of chrysin supports the idea that its pharmacological
effects are established on the GABA/benzodiazepine receptor complex, as occurs with
clinically effective GABAergic anxiolytic drugs and several neurosteroids, such as allo-
pregnanolone [87], but does not produce the typical sedative effects of benzodiazepines
[24]. However, we cannot discard the possibility of other neurotransmitter systems’ par-
ticipation and the anti-inflammatory and anti-oxidant effects in different regions of the
brain due to the anxiolytic-like effects of chrysin (Figure 2). Specific studies are required
to support or discard this possibility.
Molecules 2022, 27, x FOR PEER REVIEW 10 of 18
Figure 2. Mechanism of action of the flavonoid chrysin potentially involved in its anxiolytic-like
effects. (A) It has been confirmed that chrysin produces its anxiolytic-like effect through its action
on the GABAA/benzodiazepine receptor complex producing configurational changes in the receptor
and regulating the opening of the Cl ion channel [14,15,17,24,35]; which may produce inhibitory
effects in the GABAergic system associated with its anxiolytic-like effects. These effects can be
blocked by specific antagonists of the GABAA receptor such as picrotoxin, bicuculline and fluma-
zenil [14]. (B) Probably, anti-oxidant effects of chrysin could be involved in its anxiolytic-like effects.
Chrysin significantly reduces ROS by inhibiting the production of NO, NT, and NOX4 [29]. These
effects reduce the oxidative stress and reduces the neuronal damage. Additionally, chrysin reduces
the activity of Bax, caspase-9 and caspase-3, while increasing the production of Bcl-2, thereby reduc-
ing the damage of DNA and inhibiting apoptotic processes [60,61]; which reduces the neuronal
death. (C) Additionally, the anti-inflammatory effects of chrysin could contribute in its anxiolytic-
like effects, considering that it may reduce the inflammatory response by inhibiting the signaling
pathway NF-κB/IKK-β [27,28]. Chrysin may attenuate the expression of NF-κB that participates as
transcriptional factors at nuclear level, binding to genes that induce neuro-inflammation process.
Chrysin also inhibits the production of pro-inflammatory cytokines like IL-1β and IL-6, in addition
to suppressing the production of proinflammatory mediators like TNF-α, PGE2 and COX-2
[27,28,32]. These effects could reduce neuro-inflammation associated with the anxiety-like behavior.
ROS = reactive oxygen species; NO = nitric oxide; NT = nitrotyrosine; NOX4 = NADPH oxidase; O2¯
= superoxide; Green circles = chlorine ions; SOD = superoxide dismutase; GSH = reduced glutathi-
one; CAT = catalase; GPx = glutathione peroxidase; Bcl-2 = anti-apoptotic protein of the subfamily
Bcl-2; Bax = pro-apoptotic protein of the subfamily Bax; NF-κB = nuclear factor kappa B; IKK-β =
inhibitor of nuclear factor kappa-B; TNF-α = tumor necrosis factor-α; IL-1β = interleukin-1β; IL-6 =
interleukin-6; PGE2 = prostaglandins E2; COX-2 = cycloxygenase-2. (Figure was prepared by the
authors).
5. Antidepressant-Like Effects of Flavonoid Chrysin
Few studies have explored the antidepressant-like effects of chrysin; however, their
results are promising. Filho et al. [18] reported that chrysin at 5 and 20 mg/kg for 28 days
increased sucrose consumption and decreased immobility in the tail suspension test (TST)
in female mice C57B/6J exposed to CUMS, which is considered to have antidepressant-
like effects in pre-clinical research. This effect was also associated with an increase in ser-
otonin, BDNF, and NGF levels, and decreased pro-inflammatory levels of cytokines such
Molecules 2022, 27, x FOR PEER REVIEW 11 of 18
as TNF-α, IFN-γ, IL-1β, and IL-6 in the HP and PFC of C57B/6J mice [11,18]. Additionally,
chrysin at 20 mg/kg for 14 days produced an antidepressant-like effect in the FST in male
mice C57B/6J subjected to depression induced by olfactory bulbectomy. This effect was
associated with decreased pro-inflammatory cytokines (i.e., TNF-α, IFN-γ, IL-1β, IL-6),
kynurenine (KYN, a metabolite resulting from serotonin degradation), and indolamine-2,
3-dyoxigenase (IDO, enzyme responsible for serotonin metabolism) activity, besides pro-
ducing an increase in BDNF and serotonin levels in HP [25]. Interestingly, chrysin at 1, 5
and 10 mg/kg for 28 days produced antidepressant-like effects in the FST in male Wistar
rats [13]. In addition, chrysin at 1 and 5 mg/kg for 28 days significantly reduced 5-HT1A
receptor expression in the raphe nucleus and increased it in HP, whereas 5-HT2A receptor
expression was increased in HP [13]. These effects were similar to those produced by the
antidepressant fluoxetine at 1 mg/kg for 28 days. In another study, chrysin at 20 mg/kg
for 28 days produced antidepressant-like effects in the TST and FST in female C57BL/6
mice exposed to a depression model induced by hypothyroidism, which were associated
with increased serotonin and dopamine levels in the HP [12].
As previously mentioned, a reduced concentration of ovarian hormones in women,
during their transition to menopause, increases the risk of developing anxiety and depres-
sion symptoms [88]. Interestingly, using a surgical menopause model in Wistar rats, it was
reported that chrysin at 1 mg/kg reversed depression-like behavior in the FST; this effect
was similar to that produced by neurosteroids progesterone at 1 mg/kg and allopregnano-
lone at 1 mg/kg [37]. The effects of chrysin and neurosteroids were blocked by previous
administration of bicuculline, a selective competitive antagonist of the binding site of γ-
aminobutyric acid in the GABAA receptor, which supports the idea that activation of the
GABAergic system participates in the antidepressant-like effect of chrysin, as has been
reported with neurosteroids [89].
Based on the results described above, we suggest that the mechanism of action un-
derlying the antidepressant-like effect of chrysin involves multiple neurochemical pro-
cesses, such as the activation of neurotransmitter systems, anti-inflammatory and anti-
oxidant processes, and the activation of neurotrophic factors (Figure 3); however, further
exploration is required to improve our understanding of these mechanisms underlying
the antidepressant-like effects of chrysin, and to explore its effects in controlled clinical
trials.
Molecules 2022, 27, x FOR PEER REVIEW 12 of 18
Figure 3. Possible mechanisms of action involved in the antidepressant-like effect of chrysin. (A)
The flavonoid chrysin can modulate ERα and ERβ of membrane, which triggers the MAPK/ERK1/2
signaling pathway involved in phosphorylation and subsequently CREB activation (CREB), which
promotes the increase of BDNF levels ( BDNF) [11,19,20,43]; which can further activate the
MAPK/ERK1/2 signaling by the TrkB interaction [56]. The above-mentioned pathway also promotes
an increase of TpOH expression (TpOH) and serotonin levels (Serotonin) resulting in the antide-
pressant-like effect [12,25]. (B) Furthermore, chrysin can decrease the pro-inflammatory cytokine
levels (TNF-α, IL-1β, IL-6, IFN-γ), which inhibits IDO activity (IDO) improving serotonergic
neurotransmission and producing its antidepressant-like effect [25]. ER = estrogen receptor; MAPK
= mitogen-activated-protein-kinases; CREB = cAMP response element binding; BDNF = brain de-
rived neurotrophic factor; TrkB = tropomyosinreceptor kinase B; TpOH = tryptophan-hydroxylase;
TNF-α = tumor necrosis factor-α; IL-1β = interleukin 1 beta; IL-6 = interleukin 6; IFN-γ = interferon
gamma; IDO = indoleamine 2,3-dioxygenase; ERK1/2 = extracellular signal-regulated kinase 1 and
2; KYN = kynurenine. (Figure was prepared by the authors).
6. Future Considerations
It is noteworthy that human studies focused specifically on the anxiolytic and anti-
depressant properties of the flavonoid chrysin are nonexistent. Despite this, the results
from pre-clinical studies are promising and support the potential therapeutic effects of
chrysin, which are similar to those produced by clinically effective anxiolytic and antide-
pressants drugs. However, unlike other drugs chrysin does not produce side effects on
motor activity associated with sedation. These results from in vitro and in vivo studies in
preclinical research support the feasibility to evaluate the potential anxiolytic and
Molecules 2022, 27, x FOR PEER REVIEW 13 of 18
antidepressant effects of the flavonoid chrysin in human patients. In the future, it could
play a role in developing new pharmacological strategies to ameliorate the symptoms of
anxiety and depression disorders in patients nonrespondent to conventional antidepres-
sant and anxiolytic drugs.
For many years, increased monoamine availability in the synaptic cleft has been hy-
pothesized as the mechanism underlying the therapeutic effects of clinically effective an-
tidepressant drugs [90]. A wide range of antidepressant drugs has been developed based
on this hypothesis [90]. This is a relevant key that supports the potential antidepressant
actions of chrysin, considering that this flavonoid can increase serotoninergic and dopa-
minergic neurotransmission, which is associated with its antidepressant-like effect [18,25].
In addition, activation of the GABAergic system by this flavonoid supports its potential
anxiolytic properties.
Currently, it is known that an increase in BDNF levels is one of the main effects of
antidepressant drugs and significantly contributes to their therapeutic effects [91]. Ac-
cordingly, a relatively recent meta-analysis found higher serum BDNF concentrations in
patients with major depression disorder after 4 to 12 weeks of antidepressant treatment
with a selective serotonin reuptake inhibitor and a selective noradrenaline recapture in-
hibitor [92]. Similarly, treatment with vortioxetine (an inhibitor of the serotonin trans-
porter; 515 mg over 4 weeks) increased the plasma BDNF levels in patients with major
depression disorder, based on their basal values [93]. This is important, considering that
chrysin is similar to conventional antidepressants which increases the concentration of
this neurotrophin in pre-clinical research [11].
Neuroinflammation has also been shown to play a crucial role in the risk of neuro-
psychiatric disorders, including anxiety and depression [58,65]. In agreement with this,
some meta-analyses have reported that administration of clinically effective antidepres-
sant drugs, such as selective serotonin reuptake inhibitors, selective noradrenaline
reuptake inhibitors, and tricyclic antidepressants drugs, decrease the pro-inflammatory
cytokine levels (IL-1β, IL-6, and TNFα) in patients diagnosed with major depression dis-
order [9496]. Pre-clinical research has also demonstrated that chrysin can reduce pro-
inflammatory cytokine levels [11,18,25], which are positively correlated with depression-
like behavior. Similarly, oxidative stress has been shown to be involved in the risk of anx-
iety and depression disorders [46,47], and the anti-oxidant effects of chrysin identified in
pre-clinical research may contribute to its anxiolytic and antidepressant effects.
Finally, as mentioned above, the different neurochemical changes associated with
chrysin treatment may play a significant role in the establishment of anxiolytic- and anti-
depressant-like effects of chrysin, which could be helpful in the treatment of particular
groups of patients. Therefore, in future studies, it will be important to explore the anxio-
lytic and antidepressant effects of chrysin in particular groups of subjects, considering the
etiology of anxiety and depression symptoms. This could help identify specific groups of
patients in which chrysin could be used as an alternative for the treatment of anxiety and
depression disorders, where their etiology could be related to changes in steroid hor-
mones, neurotransmitters, oxidative stress, or neuro-inflammatory processes. It could
permit the evaluation of therapeutic effects of chrysin in human patients, as it has oc-
curred, for example, with the neurosteroid allopregnanlone [83,87,97,98]; and the mecha-
nism of action underlying its anxiolytic and antidepressant effects is shared with those
produced by the flavonoid chrysin.
7. Conclusions
Chrysin exerts diverse pharmacological effects on the peripheral and CNS. Its action
is in particular on some neurotransmitter systems, by activating neurotrophic factors, reg-
ulating biomarkers of oxidative stress, and regulating inflammatory and apoptotic signal-
ing pathways, which contributes to the anxiolytic- and antidepressant-like effects of this
flavonoid. Although these effects have been evaluated principally in mice and rats, the
results are solid and convincing, and could contribute to clinical evaluation of its potential
Molecules 2022, 27, x FOR PEER REVIEW 14 of 18
anxiolytic and antidepressant effects in particular groups of patients in a short time. In
summary, chrysin is a natural molecule that could become a novel and promising com-
plementary therapy for anxiety and depression disorders in humans.
Author Contributions: J.F.R.-L. and L.J.G.-P. conceived the idea of the paper and developed its
structure. All authors selected and discussed the material to be included in the paper. A.P.-O. and
O.J.O.-V. wrote the first draft of the manuscript. All authors have read and agreed to the published
version of the manuscript.
Funding: This study is part of SIREI project no. DGI: 266502021159 to JFR-L. This research was par-
tially funded by Sistema Nacional de Investigadores: Exp. 32753 (J.F.R-L.), 84949 (L.J.G.-P.), 153293
(A.P.-O.).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare that the research was conducted in the absence of any
commercial or financial relationships that could be construed as a potential conflict of interest.
References
1. Ferreira, M.J.U. Natural products in drug discovery and human health. Phytochem. Rev. 2021, 20, 14.
https://doi.org/10.1007/s11101-020-09736-y.
2. Tresina, P.S.; Selvam, M.S.; Rajesh, A.; Doss, A.; Mohan, V.R. Natural products in drug discovery: Approaches and develop-
ment. J. Pharm. Res. Int. 2021, 33, 93110. http://doi.org/10.9734/jpri/2021/v33i35A31879.
3. Dzobo, K. The role of natural products as sources of therapeutic agents for innovative drug discovery. Ref. Module Biomed. Sci.
2021, B978-0-12-820472-6.00041-4. https://doi.org/10.1016/B978-0-12-820472-6.00041-4.
4. Bertelli, A.; Biagi, M.; Corsini, M.; Baini, G.; Cappellucci, G.; Miraldi, E. Polyphenols: From theory to practice. Foods 2021, 10,
2595. https://doi.org/10.3390/foods10112595.
5. German-Ponciano, L.J.; Rosas-Sánchez, G.U.; Rivadeneyra-Domínguez, E.; Rodríguez-Landa, J.F. Advances in the preclinical
study of some flavonoids as potential antidepressant agents. Scientifica 2018, 2018, 2963565.
https://doi.org/10.1155/2018/2963565.
6. Fernández-Demeneghi, R.; Rodríguez-Landa, J.F.; Guzmán-Gerónimo, R.I.; Acosta-Mesa, H.G.; Meza-Alvarado, E.; Vargas-Mo-
reno, I.; Herrera-Meza, S. Effect of blackberry juice (Rubus fruticosus L.) on anxiety-like behaviour in Wistar rats. Int. J. Food Sci.
Nutr. 2018, 70, 856867. https://doi.org/10.1080/09637486.2019.1580680.
7. Safe, S.; Jayaraman, A.; Chapkin, R.S.; Howard, M.; Mohankumar, K.; Shrestha, R. Flavonoids: Structure-function and mecha-
nisms of action and opportunities for drug development. Toxicol. Res. 2021, 37, 147162. https://doi.org/10.1007/s43188-020-
00080-z.
8. Garg, A.; Chaturvedi, S. A comprehensive review on chrysin: Emphasis on molecular targets, pharmacological actions and bio-
pharmaceutical aspects. Curr. Drug Targets. 2022, 23, 420436. https://doi.org/10.2174/1389450122666210824141044.
9. Mishra, A.; Mishra, P.S.; Bandopadhyay, R.; Khurana, N.; Angelopoulou, E.; Paudel, Y.N.; Piperi, C. Neuroprotective potential
of chrysin: Mechanistic insights and therapeutic potential for neurological disorders. Molecules 2021, 26, 6456.
https://doi.org/10.3390/molecules26216456.
10. Talebi, M.; Talebi, M.; Farkhondeh, T.; Kopustinskiene, D.M.; Simal-Gandara, J.; Bernatoniene, J.; Samarghandian, S. An up-
dated review on the versatile role of chrysin in neurological diseases: Chemistry, pharmacology, and drug delivery approaches.
Biomed. Pharm. 2021, 141, 111906. https://doi.org/10.1016/j.biopha.2021.111906.
11. Filho, C.B.; Jesse, C.R.; Donato, F.; Giacomeli, R.; Del Fabbro, L.; da Silva Antunes, M.; de Gomes, M.G.; Goes, A.T.R.; Boeira,
S.P.; Prigol, M.; et al. Chronic unpredictable mild stress decreases BDNF and NGF levels and Na+,K+-ATPase activity in the
hippocampus and prefrontal cortex of mice: Antidepressant effect of chrysin. Neuroscience 2015, 289, 367380.
https://doi.org/10.1016/j.neuroscience.2014.12.048.
12. Bortolotto, V.C.; Pinheiro, F.C.; Araujo, S.M.; Poetini, M.R.; Bertolazi, B.S.; de Paula, M.T.; Meichtry, L.B.; de Almeida, F.P.; de
Freitas Couto, S.; Jesse, C.R.; et al. Chrysin reverses the depressive-like behavior induced by hypothyroidism in female mice by
regulating hippocampal serotonin and dopamine. Eur. J. Pharmacol. 2018, 822, 7884. https://doi.org/10.1016/j.ejphar.2018.01.017.
13. Germán-Ponciano, L.J.; Rosas-Sánchez, G.U.; Ortiz-Guerra, S.I.; Soria-Fregozo, C.; Rodríguez-Landa, J.F. Effects of chrysin on
mRNA expression of 5-HT1A and 5-HT2A receptors in the raphe nuclei and hippocampus. Rev. Bras. Farmacog. 2021, 31, 352360.
https://doi.org/10.1007/s43450-021-00164-3.
14. Rodríguez-Landa, J.F.; Hernández-López, F.; Martínez-Mota, L.; Scuteri, D.; Bernal-Morales, B.; Rivadeneyra-Domínguez, E.
GABAA/benzodiazepine receptor complex in the dorsal hippocampus mediates the effects of chrysin on anxiety-like behaviour
in female rats. Front. Behav. Neurosci. 2022, 15, 789557. https://doi.org/10.3389%2Ffnbeh.2021.789557.
Molecules 2022, 27, x FOR PEER REVIEW 15 of 18
15. Medina, J.H.; Paladini, A.C.; Wolfman, C.; Levi de Stein, M.; Calvo, D.; Diaz, L.E.; Peña, C. Chrysin (5,7-di-OH-flavone), a nat-
urally-occurring ligand for benzodiazepine receptors, with anticonvulsant properties. Biochem. Pharmacol. 1990, 40, 22272231.
http://doi.org/10.1016/0006-2952(90)90716-x.
16. Salgueiro, J.B.; Ardenghi, P.; Dias, M.; Ferreira, M.B.; Izquierdo, I.; Medina, J.H. Anxiolytic natural and synthetic flavonoid
ligands of the central benzodiazepine receptor have no effect on memory tasks in rats. Pharmacol. Biochem. Behav. 1997, 58, 887-
891. https://doi.org/10.1016/s0091-3057(97)00054-3.
17. Zanoli, P.; Avallone, R.; Baraldi, M. Behavioral characterisation of the flavonoids apigenin and chrysin. Fitoterapia 2000, 71,
S117S123. https://doi.org/10.1016/S0367-326X(00)00186-6.
18. Filho, C.B.; Jesse, C.R.; Donato, F.; Del Fabbro, L.; de Gomes, M.G.; Goes, A.T.R.; Souza, L.C.; Giacomeli, R.; Antunes, M.; Lu-
chese, C.; et al. Neurochemical factors associated with the antidepressant-like effect of flavonoid chrysin in chronically stressed
mice. Eur. J. Pharmacol. 2016, 791, 284296. https://doi.org/10.1016/j.ejphar.2016.09.005.
19. Goes, A.T.; Jesse, C.R.; Antunes, M.S.; Ladd, F.V.L.; Ladd, A.A.L.; Luchese, C.; Paroul, N.; Boeira, S.P. Protective role of chrysin
on 6-hydroxydopamine-induced neurodegeneration a mouse model of Parkinson's disease: Involvement of neuroinflammation
and neurotrophins. Chem. Biol. Interact. 2018, 279, 111120. https://doi.org/10.1016/j.cbi.2017.10.019.
20. Souza, L.C.; Antunes, M.S.; Filho, C.B.; Del Fabbro, L.; de Gomes, M.G.; Goes, A.T.; Donato, F.; Prigol, M.; Boeira, S.P.; Jesse,
C.R. Flavonoid chrysin prevents age-related cognitive decline via attenuation of oxidative stress and modulation of BDNF levels
in aged mouse brain. Pharmacol. Biochem. Behav. 2015, 134, 2230. https://doi.org/10.1016/j.pbb.2015.04.010.
21. Feiger, J.A.; Snyder, R.L.; Walsh, M.J.; Cissne, M.; Cwiek, A.; Al-Momani, S.I.; Chiou, K.S. The role of neuroinflammation in
neuropsychiatric disorders following traumatic brain injury: A systematic review. J. Head Trauma Rehabil. 2022, in press.
https://doi.org/10.1097/HTR.0000000000000754.
22. Sathiavelu, J.; Senapathy, G.J.; Devaraj, R.; Namasivayam, N. Hepatoprotective effect of chrysin on prooxidant-antioxidant sta-
tus during ethanol-induced toxicity in female albino rats. J. Pharm. Pharmacol. 2009, 61, 809817.
https://doi.org/10.1211/jpp.61.06.0015.
23. German-Ponciano, L.J.; Costa, B.P.D.; Feitosa, L.M.; dos Santos Campos, K.; da Silva Chaves, S.N.; Cueto-Escobedo, J.; Max-
imino, C. Chrysin, but not flavone backbone, decreases anxiety-like behavior in animal screens. Neurochem. Int. 2020, 140,
104850. https://doi.org/10.1016/j.neuint.2020.104850.
24. Wolfman, C.; Viola, H.; Paladini, A.; Dajas, F.; Medina, J.H. Possible anxiolytic effects of chrysin, a central benzodiazepine
receptor ligand isolated from Passiflora Coerulea. Pharmacol. Biochem. Behav. 1994, 47, 14. http://doi.org/10.1016/0091-
3057(94)90103-1.
25. Filho, C.B.; Jesse, C.R.; Donato, F.; Del Fabbro, L.; Gomes de Gomes, M.; Rossito Goes, A.T.; Souza, L.C.; Boeira, S.P. Chrysin
promotes attenuation of depressive-like behavior and hippocampal dysfunction resulting from olfactory bulbectomy in mice.
Chem. Biol. Interact. 2016, 260, 154162. https://doi.org/10.1016/j.cbi.2016.11.005.
26. Rodríguez-Landa, J.F.; Hernández-López, F.; Cueto-Escobedo, J.; Herrera-Huerta, E.V.; Rivadeneyra-Domínguez, E.; Bernal-
Morales, B.; Romero-Avendaño, E. Chrysin (5,7-dihydroxyflavone) exerts anxiolytic-like effects through GABAA receptors in a
surgical menopause model in rats. Biomed. Pharmacother. 2019, 109, 23872395. https://doi.org/10.1016/j.biopha.2018.11.111.
27. Mantawy, E.M.; El-Bakly, W.M.; Esmat, A.; Badr, A.M.; El-Demerdash, E. Chrysin alleviates acute doxorubicin cardiotoxicity
in rats via suppression of oxidative stress, inflammation and apoptosis. Eur. J. Pharmacol. 2014, 728, 107118.
https://doi.org/10.1016/j.ejphar.2014.01.065.
28. Rani, N.; Bharti, S.; Bhatia, J.; Tomar, A.; Nag, T.C.; Ray, R.; Arya, D.S. Inhibition of TGF-β by a novel PPAR-γ agonist, chrysin,
salvages β-receptor stimulated myocardial injury in rats through MAPKs-dependent mechanism. Nutr. Metab. 2015, 12, 11.
https://doi.org/10.1186/s12986-015-0004-7.
29. Rani, N.; Bharti, S.; Bhatia, J.; Nag, T.C.; Ray, R.; Arya, D.S. Chrysin, a PPAR-γ agonist improves myocardial injury in diabetic
rats through inhibiting AGE-RAGE mediated oxidative stress and inflammation. Chem. Biol. Interact. 2016, 250, 5967.
https://doi.org/10.1016/j.cbi.2016.03.015.
30. Campos, H.M.; da Costa, M.; da Silva Moreira, L.K.; da Silva Neri, H.F.; Branco da Silva, C.R.; Pruccoli, L.; Dos Santos, F.C.A.;
Costa, E.A.; Tarozzi, A.; Ghedini, P.C. Protective effects of chrysin against the neurotoxicity induced by aluminium: In Vitro and
In Vivo studies. Toxicology 2022, 465, 153033. https://doi.org/10.1016/j.tox.2021.153033.
31. Harasstani, O.A.; Moin, S.; Tham, C.L.; Liew, C.Y.; Ismail, N.; Rajajendram, R.; Harith, H.H.; Zakaria, Z.A.; Mohamad, A.S.;
Sulaiman, M.R.; et al. Flavonoid combinations cause synergistic inhibition of proinflammatory mediator secretion from lipo-
polysaccharide-induced RAW 264.7 cells. Inflamm. Res. 2010, 59, 711721. https://doi.org/10.1007/s00011-010-0182-8.
32. Feng, X.; Qin, H.; Shi, Q.; Zhang, Y.; Zhou, F.; Wu, H.; Ding, S.; Niu, Z.; Lu, Y.; Shen, P. Chrysin attenuates inflammation by
regulating M1/M2 status via activating PPARγ. Biochem. Pharmacol. 2014, 89, 503514. https://doi.org/10.1016/j.bcp.2014.03.016.
33. Harasstani, O.A.; Tham, C.L.; Israf, D.A. Kaempferol and chrysin synergies to improve septic mice survival. Molecules 2017, 22,
92. https://doi.org/10.3390/molecules22010092.
34. Wang, H.; Hui, K.M.; Chen, Y.; Xu, S.; Wong, J.T.; Xue, H. Structure-activity relationships of flavonoids, isolated from Scutellaria
baicalensis, binding to benzodiazepine site of GABAA receptor complex. Planta Med. 2002, 68, 10591062.
https://doi.org/10.1055/s-2002-36357.
35. Goutman, J.D.; Waxemberg, M.D.; Doñate-Oliver, F.; Pomata, P.E.; Calvo, D.J. Flavonoid modulation of ionic currents mediated
by GABAA and GABAc receptors. Eur. J. Pharmacol. 2003, 461, 7987. https://doi.org/10.1016/s0014-2999(03)01309-8.
Molecules 2022, 27, x FOR PEER REVIEW 16 of 18
36. Rodríguez-Landa, J.F.; Guillén-Ruiz, G.; Hernández-López, F.; Cueto-Escobedo, J.; Rivadeneyra-Domínguez, E.; Bernal-Mo-
rales, B.; Herrera-Huerta, E.V. Chrysin reduces anxiety-like behavior through actions on GABAA receptors during metestrus-
diestrus in the rat. Behav. Brain Res. 2021, 397, 112952. https://doi.org/10.1016/j.bbr.2020.112952.
37. Cueto-Escobedo, J.; Andrade-Soto, J.; Lima-Maximino, M.; Maximino, C.; Hernández-López, F.; Rodríguez-Landa, J.F. Involve-
ment of GABAergic system in the antidepressant-like effects of chrysin (5, 7-dihydroxyflavone) in ovariectomized rats in the
forced swim test: Comparison with neurosteroids. Behav. Brain Res. 2020, 386, 112590. https://doi.org/10.1016/j.bbr.2020.112590.
38. Haider, M.; Salman, M.; Kaushik, P.; Bharadwaj, N.; Aggarwal, N.B.; Tabassum, H.; Parvez, S. Chrysin ameliorates 3 nitro-
propinoic acid induced neurotoxicity targeting behavioural, biochemical and histological alterations. Int. J. Neurosci. 2020, 19.
https://doi.org/10.1080/00207454.2020.1821677.
39. Wu, J.; Wang, Y.; Cui, W.; Zhou, W.; Zhao, X. 5-HT1A receptor-mediated attenuation of heat hyperalgesia and mechanical allo-
dynia by chrysin in mice with experimental mononeuropathy. Reg. Anesth. Pain Med. 2020, 45, 610619.
https://doi.org/10.1136/rapm-2020-101472.
40. Krishnamoorthy, A.; Sevanan, M.; Mani, S.; Balu, M.; Balaji, S.; Ramajayan, P. Chrysin restores MPTP induced neuroinflamma-
tion, oxidative stress and neurotrophic factors in an acute Parkinson’s disease mouse model. Neurosci. Lett. 2019, 709, 134382.
https://doi.org/10.1016/j.neulet.2019.134382.
41. Farkhondeh, T.; Samarghandian, S.; Azimin-Nezhad, M.; Samini, F. Effect of chrysin on nociception in formalin test and serum
levels of noradrenalin and corticosterone in rats. Int. J. Clin. Exp. Med. 2015, 8, 2465.
42. Rashno, M.; Ghaderi, S.; Nesari, A.; Khorsandi, L.; Farbood, Y.; Sarkaki, A. Chrysin attenuates traumatic brain injury-induced
recognition memory decline, and anxiety/depression-like behaviors in rats: Insights into underlying mechanisms. Psychophar-
macology 2020, 237, 16071619. https://doi.org/10.1007/s00213-020-05482-3.
43. Bortolotto, V.C.; Araujo, S.M.; Pinheiro, F.C.; Poetini, M.R.; Meichtry, L.B.; Fronza, M.G.; Boeira, S.P.; Savegnago, L.; Prigol, M.
Chrysin restores memory deficit in hypothyroidism mice: Behavioral, neurochemical and computational approaches involving
the neurotrophinergic system. J. Psychiatr. Res. 2021, 144, 225233. https://doi.org/10.1016/j.jpsychires.2021.10.018.
44. Gargouri, B.; Bhatia, H.S.; Bouchard, M.; Fiebich, B.L.; Fetoui, H. Inflammatory and oxidative mechanisms potentiate bifenthrin-
induced neurological alterations and anxiety-like behavior in adult rats. Toxicol. Lett. 2018, 294, 7386.
https://doi.org/10.1016/j.toxlet.2018.05.020.
45. Gandhi, S.; Abramov, A.Y. Mechanism of oxidative stress in neurodegeneration. Oxid. Med. Cell Longev. 2012, 2012, 428010.
https://doi.org/10.1155/2012/428010.
46. Smaga, I.; Niedzielska, E.; Gawlik, M.; Moniczewski, A.; Krzek, J.; Przegaliński, E.; Pera, J.; Filip, M. Oxidative stress as an
etiological factor and a potential treatment target of psychiatric disorders. Part 2. Depression, anxiety, schizophrenia and au-
tism. Pharmacol. Rep. 2015, 67, 569580. https://doi.org/10.1016/j.pharep.2014.12.015.
47. Bhatt, S.; Nagappa, A.N.; Patil, C.R. Role of oxidative stress in depression. Drug Discov. Today 2020, 25, 12701276.
https://doi.org/10.1016/j.drudis.2020.05.001.
48. Di Sarno, R.; Brigida, A.; Caprio, G.G.; Ciardiello, D.; Dallio, M.; Sangineto, M.; Fagoonee, S.; Abenavoli, L.; Luzza, F.; Gravina,
A.G.; et al. Critical review on the use and abuse of alcohol. When the dose makes the difference. Minerva Med. 2020, 111, 344
353. https://doi.org/10.23736/S0026-4806.20.06584-2.
49. Bhatti, J.S.; Sehrawat, A.; Mishra, J.; Sidhu, I.S.; Navik, U.; Khullar, N.; Kumar, S.; Bhatti, G.K.; Reddy, P.H. Oxidative stress in
the pathophysiology of type 2 diabetes and related complications: Current therapeutics strategies and future perspectives. Free
Radic. Biol. Med. 2022, 184, 114134. https://doi.org/10.1016/j.freeradbiomed.2022.03.019.
50. Hovatta, I.; Juhila, J.; Donner, J. Oxidative stress in anxiety and comorbid disorders. Neurosci. Res. 2010, 68, 261275.
Htpps://doi.org/10.1016/j.neures.2010.08.007.
51. Sarwar, H.; Rafiqi, S.I.; Ahmad, S.; Jinna, S.; Khan, S.A.; Karim, T.; Qureshi, O.; Zahid, Z.A.; Elhai, J.D.; Levine, J.C.; et al. Hy-
perinsulinemia associated depression. Clin. Med. Insights Endocrinol. Diabetes. 2022, 15, 11795514221090244.
Htpps://doi.org/10.1177/11795514221090244.
52. Bouayed, J.; Rammal, H.; Younos, C.; Soulimani, R. Positive correlation between peripheral blood granulocyte oxidative status
and level of anxiety in mice. Eur. J. Pharmacol. 2007, 564, 146149. https://doi.org/10.1016/j.ejphar.2007.02.055.
53. Salim, S.; Sarraj, N.; Taneja, M.; Saha, K.; Tejada-Simon, M.V.; Chugh, G. Moderate treadmill exercise prevents oxidative stress-
induced anxiety-like behavior in rats. Behav. Brain Res. 2010, 208, 545552. https://doi.org/10.1016/j.bbr.2009.12.039.
54. Kaufmann, F.N.; Gazal, M.; Mondin, T.C.; Cardoso, T.A.; Quevedo, L.Á .; Souza, L.D.; Ghisleni, G. Cognitive psychotherapy
treatment decreases peripheral oxidative stress parameters associated with major depression disorder. Biol. Psychol. 2015, 110,
175181. https://doi.org/10.1016/j.biopsycho.2015.08.001.
55. Moccia, M.; Capacchione, A.; Lanzillo, R.; Carbone, F.; Micillo, T.; Perna, F.; Brescia Morra, V. Coenzyme Q10 supplementation
reduces peripheral oxidative stress and inflammation in interferon-β1a-treated multiple sclerosis. Ther. Adv. Neurol. Disord.
2019, 12, 1756286418819074. https://doi.org/10.1177%2F1756286418819074.
56. Nakagawa, Y.; To, M.; Saruta, J.; Yamamoto, Y.; Yamamoto, T.; Shimizu, T.; Kamata, Y.; Matsuo, M.; Tsukinoki, K. Effect of
social isolation stress on saliva BDNF in rat. J. Oral Sci. 2019, 61, 516520. https://doi.org/10.2334/josnusd.18-0409.
57. Khurana, K.; Bansal, N. Lacidipine attenuates caffeine-induced anxiety-like symptoms in mice: Role of calcium-induced oxido-
nitrosative stress. Pharmacol. Rep. 2019, 71, 12641272. https://doi.org/10.1016/j.pharep.2019.07.008.
Molecules 2022, 27, x FOR PEER REVIEW 17 of 18
58. Risbrough, V.B.; Vaughn, M.N.; Friend, S.F. Role of inflammation in traumatic brain injury-associated risk for neuropsychiatric
disorders: State of the evidence and where do we go from here. Biol. Psychiatry. 2022, 91, 438448. https://doi.org/10.1016/j.bi-
opsych.2021.11.012.
59. Zorzo, C.; Méndez-López, M.; Méndez, M.; Arias, J.L. Adult social isolation leads to anxiety and spatial memory impairment:
Brain activity pattern of COx and c-Fos. Behav. Brain Res. 2019, 365, 170177. https://doi.org/10.1016/j.bbr.2019.03.011.
60. Park, S.S.; Park, H.S.; Kim, C.J.; Baek, S.S.; Kim, T.W. Exercise attenuates maternal separation-induced mood disorder-like be-
haviors by enhancing mitochondrial functions and neuroplasticity in the dorsal raphe. Behav. Brain Res. 2019, 372, 112049.
https://doi.org/10.1016/j.bbr.2019.112049.
61. Zhang, L.X.; Levine, S.; Dent, G.; Zhan, Y.; Xing, G.; Okimoto, D.; Kathleen Gordon, M.; Post, R.M.; Smith, M.A. Maternal dep-
rivation increases cell death in the infant rat brain. Brain Res. Dev. Brain Res. 2002, 133, 111. https://doi.org/10.1016/S0926-
6410(01)00118-5.
62. Wang, Q.; Dong, X.; Wang, Y.; Liu, M.; Sun, A.; Li, N.; Lin, Y.; Geng, Z.; Jin, Y.; Li, X. Adolescent escitalopram prevents the
effects of maternal separation on depression- and anxiety-like behaviours and regulates the levels of inflammatory cytokines in
adult male mice. Int. J. Dev. Neurosci. 2017, 62, 3745. https://doi.org/10.1016/j.ijdevneu.2017.07.007.
63. Vega-Rivera, N.M.; Fernández-Guasti, A.; Ramírez-Rodríguez, G.; Estrada-Camarena, E. Acute stress further decreases the ef-
fect of ovariectomy on immobility behavior and hippocampal cell survival in rats. Psychoneuroendocrinology 2013, 38, 14071417.
https://doi.org/10.1016/j.psyneuen.2012.12.008.
64. Wang, Z.; Gu, J.; Wang, X.; Xie, K.; Luan, Q.; Wan, N.; Zhang, Q.; Jiang, H.; Liu, D. Antidepressant-like activity of resveratrol
treatment in the forced swim test and tail suspension test in mice: The HPA axis, BDNF expression and phosphorylation of
ERK. Pharmacol. Biochem. Behav. 2013, 112, 104110. https://doi.org/10.1016/j.pbb.2013.10.007.
65. Abd Al Haleem, E.N.; Ahmed, H.I.; El-Naga, R.N. Lycopene and chrysin through mitigation of neuroinflammation and oxida-
tive stress exerted antidepressant effects in clonidine-induced depression-like behavior in rats. J. Diet Suppl. 2021, 120.
https://doi.org/10.1080/19390211.2021.1988797.
66. Bear, T.L.; Dalziel, J.E.; Coad, J.; Roy, N.C.; Butts, C.A.; Gopal, P.K. The role of the gut microbiota in dietary interventions for
depression and anxiety. Adv. Nutr. 2020, 11, 890907. https://doi.org/10.1093/advances/nmaa016.
67. Sonali, S.; Ray, B.; Ahmed Tousif, H.; Rathipriya, A.G.; Sunanda, T.; Mahalakshmi, A.M.; Rungratanawanich, W.; Essa, M.M.;
Qoronfleh, M.W.; Chidambaram, S.B.; et al. Mechanistic insights into the link between gut dysbiosis and major depression: An
extensive review. Cells 2022, 11, 1362. https: //doi.org/10.3390/cells11081362.
68. Kelly, J.R.; Borre, Y.; O’Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Dinan, T.G. Transferring the blues: Depression-associated
gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109118.
https://doi.org/10.1016/j.jpsychires.2016.07.019.
69. Rao, J.; Qiao, Y.; Xie, R.; Lin, L.; Jiang, J.; Wang, C.; Li, G. Fecal microbiota transplantation ameliorates stress-induced depression-
like behaviors associated with the inhibition of glial and NLRP3 inflammasome in rat brain. J. Psychiatr. Res. 2021, 137, 147157.
https://doi.org/10.1016/j.jpsychires.2021.02.057.
70. Hervert-Hernández, D.; Goñi, I. Dietary polyphenols and human gut microbiota: A review. Food Rev. Int. 2011, 27, 154169.
https://doi.org/10.1080/87559129.2010.535233.
71. Hong, M.; Zhang, R.; Liu, Y.; Wu, Z.; Weng, P. The interaction effect between tea polyphenols and intestinal microbiota: Role
in ameliorating neurological diseases. J. Food Biochem. 2022, 46, e13870. https://doi.org/10.1111/jfbc.13870.
72. Zhou, N.; Gu, X.; Zhuang, T.; Xu, Y.; Yang, L.; Zhou, M. Gut microbiota: A pivotal hub for polyphenols as antidepressants. J.
Agric. Food Chem. 2020, 68, 60076020. https://doi.org/10.1021/acs.jafc.0c01461.
73. Cheng, N.; Chen, S.; Liu, X.; Zhao, H.; Cao, W. Impact of schisandra chinensis bee pollen on nonalcoholic fatty liver disease and
gut microbiota in high fat diet induced obese mice. Nutrients 2019, 11, 346. https://doi.org/10.3390/nu11020346.
74. Wu, W.T.; Wang, Z.H. Effects of chrysin on intestinal inflammation and gut microbiota in lipopolysaccharides-induced mice.
FASEB J. 2019, 33, 764768. https://doi.org/10.1096/fasebj.2019.33.1_supplement.764.8.
75. Wen, X.; Walle, T. Methylated flavonoids have greatly improved intestinal absorption and metabolic stability. Drug Metab.
Dispos. 2006, 34, 17861792. https://doi.org/10.1124/dmd.106.011122.
76. Paladini, A.C.; Marder, M.; Viola, H.; Wolfman, C.; Wasowski, C.; Medina, J.H. Flavonoids and the central nervous system:
From forgotten factors to potent anxiolytic compounds. J. Pharm. Pharmacol. 1999, 51, 519526.
https://doi.org/10.1211/0022357991772790.
77. Brown, E.; Hurd, N.S.; McCall, S.; Ceremuga, T.E. Evaluation of the anxiolytic effects of chrysin, a Passiflora incarnata extract, in
the laboratory rat. AANA J. 2007, 75, 333337.
78. Germán-Ponciano, L.J.; Puga-Olguín, A.; Rovirosa-Hernández, M.J.; Caba, M.; Meza, E.; Rodríguez-Landa, J.F. Differential ef-
fects of acute and chronic treatment with the flavonoid chrysin on anxiety-like behavior and Fos immunoreactivity in the lateral
septal nucleus in rats. Acta Pharm. 2020, 70, 387397. https://doi.org/10.2478/acph-2020-0022.
79. Ognibene, E.; Bovicelli, P.; Adriani, W.; Saso, L.; Laviola, G. Behavioral effects of 6-bromoflavanone and 5-methoxy-6,8-dibro-
moflavanone as anxiolytic compounds. Prog. Neuropsychopharmacol. Biol. Psychiatry 2008, 32, 128134.
https://doi.org/10.1016/j.pnpbp.2007.07.023.
80. Albert, K.; Pruessner, J.; Newhouse, P. Estradiol levels modulate brain activity and negative responses to psychosocial stress
across the menstrual cycle. Psychoneuroendocrinology 2015, 59, 1424. https://doi.org/10.1016/j.psyneuen.2015.04.022.
Molecules 2022, 27, x FOR PEER REVIEW 18 of 18
81. Rocca, W.A.; Grossardt, B.R.; Geda, Y.E.; Gostout, B.S.; Bower, J.H.; Maraganore, D.M.; de Andrade, M.; Melton, L.J. 3rd Long-
term risk of depressive and anxiety symptoms after early bilateral oophorectomy. Menopause 2018, 25, 12751285.
https://doi.org/10.1097/GME.0000000000001229.
82. Giannini, A.; Caretto, M.; Genazzani, A.R.; Simoncini, T. Neuroendocrine changes during menopausal transition. Endocrines
2021, 2, 405416. https://doi.org/10.3390/endocrines2040036.
83. Pinna, G. Allopregnanolone, the neuromodulator turned therapeutic agent: Thank you, next? Front. Endocrinol. 2020, 11, 236.
https://doi.org/10.3389/fendo.2020.00236.
84. Puga-Olguín, A.; Rodríguez-Landa, J.F.; Rovirosa-Hernández, M.J.; Germán-Ponciano, L.J.; Caba, M.; Meza, E.; Guillén-Ruiz,
G.; Olmos-Vázquez, O.J. Long-term ovariectomy increases anxiety- and despair-like behaviors associated with lower Fos im-
munoreactivity in the lateral septal nucleus in rats. Behav. Brain Res. 2019, 360, 185195. https://doi.org/10.1016/j.bbr.2018.12.017.
85. Lovick, T.A. GABA in the female brainoestrous cycle-related changes in GABAergic function in the periaqueductal grey mat-
ter. Pharmacol. Biochem. Behav. 2008, 90, 4350. https://doi.org/10.1016/j.pbb.2007.12.014.
86. Rodríguez-Landa, J.F. Considerations of timing post-ovariectomy in mice and rats in studying anxiety- and depression-like
behaviors associated with surgical menopause in women. Front. Behav. Neurosci. 2022, 16, 829274.
https://doi.org/10.3389/fnbeh.2022.829274.
87. Paul, S.M.; Pinna, G.; Guidotti, A. Allopregnanolone: From molecular pathophysiology to therapeutics. A historical perspective.
Neurobiol. Stress 2020, 12, 100215. https://doi.org/10.1016/j.ynstr.2020.100215.
88. Georgieva, I.; Lepping, P.; Bozev, V.; Lickiewicz, J.; Pekara, J.; Wikman, S.; Lantta, T. Prevalence, new incidence, course, and
risk factors of PTSD, depression, anxiety, and panic disorder during the CoViD-19 pandemic in 11 countries. Healthcare 2021, 9,
664. https://doi.org/10.3390/healthcare9060664.
89. Zhukov, D.A.; Vinogradova, E.P. Neurosteroids and depression. Neurochem. J. 2021, 15, 240246.
https://doi.org/10.1134/S1819712421030144.
90. Perez-Caballero, L.; Torres-Sanchez, S.; Romero-López-Alberca, C.; González-Saiz, F.; Mico, J.A.; Berrocoso, E. Monoaminergic
system and depression. Cell Tissue Res. 2019, 377, 107113. https://doi.org/10.1007/s00441-018-2978-8.
91. Rana, T.; Behl, T.; Sehgal, A.; Srivastava, P.; Bungau, S. Unfolding the role of BDNF as a biomarker for treatment of depression.
J. Mol. Neurosci. 2021, 71, 20082021. https://doi.org/10.1007/s12031-020-01754-x.
92. Zhou, C.; Zhong, J.; Zou, B.; Fang, L.; Chen, J.; Deng, X.; Zhang, L.; Zhao, X.; Qu, Z.; Lei, Y.; et al. Meta-analyses of comparative
efficacy of antidepressant medications on peripheral BDNF concentration in patients with depression. PLoS ONE 2017, 12,
e0172270. https://doi.org/10.1371/journal.pone.0172270.
93. Sagud, M.; Nikolac Perkovic, M.; Vuksan-Cusa, B.; Maravic, A.; Svob Strac, D.; Mihaljevic Peles, A.; Zivkovic, M.; Kusevic, Z.;
Pivac, N. A prospective, longitudinal study of platelet serotonin and plasma brain-derived neurotrophic factor concentrations
in major depression: Effects of vortioxetine treatment. Psychopharmacol 2016, 233, 32593267. https://doi.org/10.1007/s00213-016-
4364-0.
94. Hannestad, J.; Dellagioia, N.; Bloch, M. The effect of antidepressant medication treatment on serum levels of inflammatory
cytokines: A meta-analysis. Neuropsychopharmacol 2011, 36, 2452. https://doi.org/10.1038/npp.2011.132.
95. Köhler, C.A.; Freitas, T.H.; Stubbs, B.; Maes, M.; Solmi, M.; Veronese, N.; de Andrade, N.Q.; Morris, G.; Fernandes, B.S.; Brunoni,
A.R.; et al. Peripheral alterations in cytokine and chemokine levels after antidepressant drug treatment for major depressive
disorder: Systematic review and meta-analysis. Mol. Neurobiol. 2018, 55, 41954206. https://doi.org/10.1007/s12035-017-0632-1.
96. Więdłocha, M.; Marcinowicz, P.; Krupa, R.; Janoska-Jaździk, M.; Janus, M.; Dębowska, W.; Mosiołek, A.; Waszkiewicz, N.; Szulc,
A. Effect of antidepressant treatment on peripheral inflammation markers-A meta-analysis. Prog. Neuro-Psychopharmacol. Biol.
Psychiatry 2018, 80, 217226. https://doi.org/10.1016/j.pnpbp.2017.04.026.
97. Patatanian, E.; Nguyen, D.R. Brexanolone: A novel drug for the treatment of postpartum depression. J. Pharm. Pract. 2020,
0897190020979627. https://doi.org/10.1177%2F0897190020979627.
98. Patterson, R.; Krohn, H.; Richardson, E.; Kimmel, M.; Meltzer-Brody, S.A. Brexanolone treatment program at an academic med-
ical center: Patient selection, 90-day posttreatment outcomes, and lessons learned. J. Acad. Consult. Liaison Psychiatry 2022, 63,
1422. https://doi.org/10.1016/j.jaclp.2021.08.001.
... The rationale for using allopregnanolone as a pharmacological control of antidepressantlike activity was because this substance reduces immobility in the FST through its action on the GABA A receptor, which may be blocked by antagonists such as picrotoxin and bicuculline [28,31,32]. Chrysin also exerts anxiolytic and antidepressant-like effects through its action on the GABA A receptor, and can also be blocked by picrotoxin and bicuculine [28,33]. Moreover, both chrysin and allopregnanolone activate some neurotrophic factors (BDNF and NGF) and the serotonergic system. ...
... This effect could be related to chrysin-induced changes in the level of metabotropic serotonergic receptors such as 5-hydroxytryptamine subtype 1A (5-HT 1A ) receptor [29], and in the production of serotonin. The establishment of long-term therapeutic effects could also involve changes in neuronal plasticity associated with the production of neurotrophic factors such as BDNF and NGF in structures involved with depression [65], such as the prefrontal cortex and the hippocampus [33,66,67]. Thus, it appears likely that 5 mg/kg chrysin induces neuronal plastic changes of long duration, as reported by other authors with fluoxetine [68,69]. ...
... In summary, we propose that chrysin at higher doses may activate other neurotransmission systems (serotonergic, dopaminergic and norepinephrinergic) to increase the production of neurotrophic factors such as BDNF and NGF [14,15,29,71]. Additionally, chrysin exerts antioxidant and anti-inflammatory effects on the central nervous system (CNS), which may also contribute antidepressant effects [33], as occurs with conventional antidepressant drugs. This may explain why the effects of chrysin were not blocked by the GABA A receptor antagonist and highlights a crucial point in psychopharmacology. ...
Article
Background: The flavonoid chrysin produces rapid and long-lasting anxiolytic- and antidepressant-like effects in rats. However, it is not known whether low and high doses of chrysin produce differential anti-immobility effects through the Gamma-Aminobutyric Acid sub-type A (GABAA) receptor. The goal of this work was therefore to compare low and high doses of chrysin for their effects on depression-like behavior in a longitudinal study. Moreover, chrysin was compared with the serotonergic fluoxetine and Gamma-Aminobutyric Acid (GABA)ergic allopregnanolone, and its involvement with the GABAA receptor after chronic treatment was also investigated. Methods: Male Wistar rats were assigned to five groups (n = 8 each): vehicle, 1 mg/kg chrysin, 5 mg/kg chrysin, 1 mg/kg fluoxetine, and 1 mg/kg allopregnanolone. In the first experiment, treatments were injected daily and the effects on locomotor activity and the forced swim test were evaluated at 0, 1, 14, and 28 days of treatment, and 48 h after the final treatment. In the second experiment, similar groups were treated for 28 days with injection of 1 mg/kg picrotoxin to investigate the role of the GABAA receptor. Depending on the experimental design, one- and two-way analysis of variance (ANOVA) tests were used for statistical analysis, with p < 0.05 set as the criteria for significance. Results: In both experiments, the treatments did not alter locomotor activity. However, low and high doses of chrysin, allopregnanolone, and fluoxetine gradually produced antidepressant-like effects in the forced swim test, and maintained this effect for 48 h post-treatment, except with low dose chrysin. Picrotoxin blocked the antidepressant-like effects produced by low dose chrysin, but did not affect those produced by high dose chrysin, allopregnanolone, or fluoxetine. Conclusions: The differential antidepressant-like effects caused by low and high doses of chrysin are time-dependent. Low dose chrysin produces a rapid antidepressant-like effect, whereas high dose chrysin produces a delayed but sustained the effect, even 48 h after withdrawal. The effect with high dose chrysin was similar to that observed with allopregnanolone and fluoxetine. The mechanism for the antidepressant-like effect of low chrysin appears to be GABAergic, whereas the effect of high dose chrysin may involve other neurotransmission and neuromodulation systems related to the serotonergic system.
... In vitro studies have shown reduction in ROS formation and decrease in NO, PGE2, and TNF-α biosynthesis as well as attenuation of IL-1β and NF-kβ expression. In addition, chrysin allosterically modulates the GABA receptor through the benzodiazepine-binding site, both in vitro and in vivo studies [47,48]. ...
Article
Full-text available
The brain-derived neurotrophic factor (BDNF) plays a crucial role during neuronal development as well as during differentiation and synaptogenesis. They are important proteins present in the brain that support neuronal health and protect the neurons from detrimental signals. The results from the present study suggest BDNF expression can be increase up to ~8-fold by treating the neuroblastoma cells SHSY-5Y with an herbal extract of Oroxylum indicum (50 μg/mL) and ~5.5-fold under lipopolysaccharides (LPS)-induced inflammation conditions. The Oroxylum indicum extract (Sabroxy) was standardized to 10% oroxylin A, 6% chrysin, and 15% baicalein. In addition, Sabroxy has shown to possess antioxidant activity that could decrease the damage caused by the exacerbation of radicals during neurodegeneration. A mode of action of over expression of BDNF with and without inflammation is proposed for the Oroxylum indicum extract, where the three major hydroxyflavones exert their effects through additive or synergistic effects via five possible targets including GABA, Adenoside A2A and estrogen receptor bindings, anti-inflammatory effects, and reduced mitochondrial ROS production.
... Primary screening was done based on the title and abstract by the 2 researchers (SS, FZ); 303 articles were excluded, thus 31 were chosen for the final screening process. From the remaining 31, 21 were eliminated after the full-text screening process for the following reasons: Four were animal studies [24][25][26][27]; 1 was a study protocol [28]; 3 used duplicated datasets [29][30][31]; 5 were irrelevant to the subject [21,[32][33][34][35]. Eight studies were reviews [36][37][38][39][40][41][42][43]. The inclusion process of this systematic review is presented in Figure 1. ...
Article
Full-text available
Anxiety disorder is a prevalent psychiatric issue that affects 4.05% of the global population. As complementary and alternative medicine gains popularity, many individuals with anxiety symptoms seek herbal remedies. This systematic review aims to explore the sedative efficacy of chamomile as an herbal medicine for anxiety treatment. Our search was conducted in PubMed, Google Scholar, and Scopus databases until August 2023. Among 389 papers found, after removing duplicates and irrelevant papers, 10 clinical trials investigating the effect of oral consumption of chamomile on anxiety were included. Two researchers independently completed all steps, including the screening process and data extraction. Out of the 10 articles selected, 9 studies have concluded that chamomile is effective in reducing anxiety. Even though, the exact mechanism of chamomile's anxiolytic action is not well understood, evidence suggests that its active compounds, including apigenin, may modulate the function of the hypothalamic-pituitary-adrenocortical axis by affecting neurotransmitter pathways. This systematic review showed that chamomile potentially has an anxiolytic effect. In addition, due to the side effects of drugs used to treat anxiety disorders, the use of chamomile seems to be effective and less dangerous.
... Additionally, there are many research studies reported on the strong correlation between the antioxidant effect of medicinal plants and their flavonoid content [12,19,22,[24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41][42], especially this Asian water lily plant [8,9,12,18,19,22]. Our previous work investigated flavonoid profiles from both stamen and perianth ethanolic extracts of this medicinal plant at the population level to study the antioxidant potential of Asian water lily populations throughout Thailand using difference assays to detecting different antioxidant mechanisms [9]. ...
Article
Full-text available
Our previous study investigated the major flavonoids and antioxidant potential of Asian water lily (Nymphaea lotus L., family Nymphaeaceae) stamens and perianth extracts. Quercetin-3-O-rhamnoside (Que-3-Rha) and kaempferol-3-O-galactoside (Kae-3-Gal) were reported as the two most prominent flavonoids found in these extracts. Many flavonoids have been reported on the skin anti-aging effect that are useful for cosmeceutical/phytopharmaceutical application. However, Que-3-Rha and Kae-3-Gal occurring in this medicinal plant have not yet been evaluated for their ability to inhibit skin-aging enzymes. Therefore, this study aimed (1) to assess the enzyme inhibitory activity of Que-3-Rha and Kae-3-Gal, and (2) to conduct molecular modeling of these compounds against critical enzymes involved in skin aging such as collagenase, elastase, and tyrosinase. In vitro enzymatic assays demonstrated that both of the two most prominent flavonoids exhibited moderate to good inhibitory activity toward these enzymes. These experimental findings were supported by molecular docking analysis, which indicated that Que-3-Rha and Kae-3-Gal showed superior binding affinity to the target enzymes compared to the positive controls. Additionally, computational predictions suggested favorable skin permeability and no severe toxicity for both compounds. The results from molecular dynamic (MD) simulation revealed that all the complexes remained stable during the 200 ns MD simulation. Structural analyses and binding free energy calculations also supported the inhibitory potential of these two flavonoids against skin-aging enzymes. In conclusion, this study provides valuable insights into the anti-aging potential of the two major flavonoids occurring in this medicinal plant, paving the way for further development of cosmeceutical/phytopharmaceutical products targeting skin aging.
... It exhibits anti-carcinogenic, anti-estrogenic, anti-hyperlipidemic, anti-inflammatory, antiangiogenic, anti-oxidant, antibacterial, anti-allergic, anti-diabetic, and pro-apoptotic characteristics (Kasala et al., 2015;Mani and Natesan, 2018). Like other bioflavonoids, chrysin has been involved in the protection against cancer, stroke, and coronary heart disease as well as neuropsychiatric and stress-related disorders (Rodriguez-Landa et al., 2022). Chrysin is a poorly water soluble drug with consequent low intestinal absorption, which in turn negatively affects its oral bioavailability (being approximately 0.003 %) (Gao et al., 2021). ...
... Flavonoids are the most abundant type of polyphenols and have been investigated as complementary therapeutics for a variety of diseases [12]. Chrysin, also known as 5,7-dihydroxyflavone, is found in a variety of plants, such as propolis, bitter melon, and walnut pellicle; it exerts antioxidant, anti-inflammatory, and anti-apoptotic effects, but has demonstrated toxicity and safety issues [13,14]. CM1, a hydroxyethyl derivative of chrysin, is generated by the exposure of chrysin methanolic solution to ionising radiation and has an anti-inflammatory effect on primary dendritic cells and dextran sodium salt (DSS)-induced colitis in mice [15]. ...
Article
Full-text available
Sepsis, a leading cause of death worldwide, is a harmful inflammatory condition that is primarily caused by an endotoxin released by Gram-negative bacteria. Effective targeted therapeutic strategies for sepsis are lacking. In this study, using an in vitro and in vivo mouse model, we demonstrated that CM1, a derivative of the natural polyphenol chrysin, exerts an anti-inflammatory effect by inducing the expression of the ubiquitin-editing protein TNFAIP3 and the NAD-dependent deacetylase sirtuin 1 (SIRT1). Interestingly, CM1 attenuated the Toll-like receptor 4 (TLR4)-induced production of inflammatory cytokines by inhibiting the extracellular-signal-regulated kinase (ERK)/MAPK and nuclear factor kappa B (NF-κB) signalling pathways. In addition, CM1 induced the expression of TNFAIP3 and SIRT1 on TLR4-stimulated primary macrophages; however, the anti-inflammatory effect of CM1 was abolished by the siRNA-mediated silencing of TNFAPI3 or by the genetic or pharmacologic inhibition of SIRT1. Importantly, intravenous administration of CM1 resulted in decreased susceptibility to endotoxin-induced sepsis, thereby attenuating the production of pro-inflammatory cytokines and neutrophil infiltration into the lung compared to control mice. Collectively, these findings demonstrate that CM1 has therapeutic potential for diverse inflammatory diseases, including sepsis.
... The anxiolytic and antidepressant effect of chrysin is mainly related to the interaction with the GABAergic and serotonergic systems, and the activation of neurotrophic factors, e.g. brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) [23]. The aqueous extract of P. incarnata L. (800 mg/kg) also proved effective in inhibiting the increased motor activity induced by nicotine administration [24]. ...
... Flavonoids are most abundant type of polyphenols and have been investigated as an essential constituents of complementary therapeutic strategies in a variety of human health and diseases [11]. Flavonoid chrysin, also known as 5,7-dihydroxyflavone, has been found in a variety of plants, such as propolis, bitter melons and walnut pellicle and exerts various biological functions including antioxidant, antiinflammatory, and anti-apoptotic activity, but has limitations in toxicity and safety [12,13]. We previously identified that CM1, a hydroxyethyl derivative of chrysin, is produced by the exposure of chrysin methanolic solution to ionizing radiation and exhibited anti-inflammatory effects in primary dendritic cells and dextran sodium salt (DSS)-induced colitis in mice [14]. ...
Preprint
Full-text available
Human sepsis is one of the leading cause of death worldwide and is known to be a harmful damaging host inflammatory condition, primarily caused by endotoxin released by gram-negative bacteria. Despite antibiotics administration being widely used to treat disease, effective targeted therapeutic strategies for the sepsis are still lacking. Here, we demonstrate that CM1, a derivative of natural polyphenol chrysin, exerts anti-inflammatory activity by inducing the ubiquitin-editing protein TNFAIP3 and NAD-dependent deacetylase sirtuin 1 (SIRT1). We found that CM1 attenuated Toll-like receptor 4 (TLR4)-induced the generation of inflammatory cytokines by inhibiting the extracellular-signal-regulated kinase (ERK)/MAPK and nuclear factor kappa B (NF-κB) signaling pathway. In addition, the treatment of macrophages with CM1 induced the expression of TNFAIP3 and SIRT1 in TLR4-stimulated primary macrophages, however the anti-inflammatory properties of CM1 was disappeared by siRNA silencing of TNFAPI3 or by the genetic and pharmacologic inhibition of SIRT1. Importantly, intravenously administration of CM1 resulted in the decreased susceptibility to endotoxin-induced sepsis, leading to inhibition of the generation of proinflammatory cytokines and neutrophils infiltration into lung than control mice. Collectively, these findings suggest that CM1 has the potential to be a treatment candidate for diverse inflammatory diseases including sepsis.
Article
Background: Chrysin is a natural flavonoid with several demonstrated neuro-pharmacological effects in brain areas related to anxiety. However, the intra-hypothalamic molecular mechanisms underlying the anxiolytic effects of chrysin remain unclear. Objectives: The current study revealed the effects of chrysin on hypothalamic corticotrophin-releasing hormone (CRH) and calcitonin gene-related peptide (CGRP) gene expression levels in a rat model of stress. Methods: Thirty male Wistar rats weighing 200 ± 10 g were divided into six groups for this investigation. Acute restraint stress was induced in the animals for 2 hours. Intact or stress-induced rats received 20 or 40 µg of chrysin via the third cerebral ventricle, respectively. Open-field and forced swimming tests were performed to evaluate stress-related behaviors. Hypothalamic samples were then removed, and real-time polymerase chain reaction (PCR) was used to measure relative gene expression. Results: The results showed that in the rats receiving chrysin, CRH and CGRP gene expression levels were significantly decreased compared to the stress group. Additionally, chrysin injection reduced anxiogenic behaviors. Conclusions: Chrysin decreased the expression of hypothalamic CRH and CGRP genes in stressed rats.
Article
Full-text available
Purpose The early stage of this study verified that a turmeric extract (TUR) including 59% curcumin (CU), 22% demethoxycurcumin (DMC), and 18% bisdemethoxycurcumin (BDMC), could enhance the stability of CU and had greater antidepressant potential in vitro. The objective of the study was to develop a nano-delivery system containing TUR (TUR-NE) to improve the pharmacokinetic behavior of TUR and enhance its antidepressant effect. Methods The antidepressant potential of TUR was explored using ABTS, oxidative stress-induced cell injury, and a high-throughput screening model. TUR-NE was fabricated, optimized and characterized. The pharmacokinetic behaviors of TUR-NE were evaluated following oral administration to normal rats. The antidepressant effect of TUR-NE was assessed within chronic unpredictable mild stress model (CUMS) mice. The behavioral and biochemical indexes of mice were conducted. Results The results depicted that TUR had 3.18 and 1.62 times higher antioxidant capacity than ascorbic acid and CU, respectively. The inhibition effect of TUR on ASP+ transport was significantly enhanced compared with fluoxetine and CU. TUR-NE displayed a particle size of 116.0 ± 0.31 nm, polydispersity index value of 0.121 ± 0.007, an encapsulation rate of 98.45%, and good release and stability in cold storage. The results of pharmacokinetics indicated the AUC(0-t) of TUR-NE was 8.436 and 4.495 times higher than that of CU and TUR, while the Cmax was 9.012 and 5.452 times higher than that of CU and TUR, respectively. The pharmacodynamic study confirmed that the superior antidepressant effect of TUR-NE by significantly improving the depressant-like behaviors and elevating the content of 5-hydroxytryptamine in plasma and brain in CUMS mice. TUR-NE showed good safety with repeated administration. Conclusion TUR-NE, which had small and uniform particle size, enhanced the bioavailability and antidepressant effect of TUR. It could be a promising novel oral preparation against depression.
Article
Full-text available
Hyperinsulinemia promotes fat accumulation, causing obesity. Being an inflammatory state, obesity can induce further inflammation and is a risk factor for HPA (hypothalamic pituitary axis) dysregulation through hypercortisolism-related hyperglycemia. In another hypothesis, the sympathetic nervous system (SNS) plays a significant role in the regulation of hormone secretion from the pancreas such as an increase in catecholamines and glucagon as well as a decrease in plasma insulin levels, a disruption on SNS activity increases insulin levels, and induces glycogenolysis in the liver and lipolysis in adipose tissue during hypoglycemia. Hyperglycemia-hyperinsulinemia exacerbates inflammation and increases the oxidative stress along with regulating the levels of norepinephrine in the brain sympathetic system. Increased inflammatory cytokines have also been shown to disrupt neurotransmitter metabolism and synaptic plasticity which play a role in the development of depression via inhibiting serotonin, dopamine, melatonin, and glutamate signaling. An increased level of plasma insulin over time in the absence of exercising causes accumulation of lipid droplets in hepatocytes and striated muscles thus preventing the movement of glucose transporters shown to result in an increase in insulin resistance due to obesity and further culminates into depression. Further hyperinsulinemia-hyperglycemia condition arising due to exogenous insulin supplementation for diabetes management may also lead to physiological hyperinsulinemia associated depression. Triple therapy with SSRI, bupropion, and cognitive behavioral therapy aids in improving glycemic control, lowering fasting blood glucose, decreasing the chances of relapse, as well as decreasing cortisol levels to improve cognition and the underlying depression. Restoring the gut microbiota has also been shown to restore insulin sensitivity and reduce anxiety and depression symptoms in patients.
Article
Full-text available
Depression is a highly common mental disorder, which is often multifactorial with sex, genetic, environmental, and/or psychological causes. Recent advancements in biomedical research have demonstrated a clear correlation between gut dysbiosis (GD) or gut microbial dysbiosis and the development of anxiety or depressive behaviors. The gut microbiome communicates with the brain through the neural, immune, and metabolic pathways, either directly (via vagal nerves) or indirectly (via gut- and microbial-derived metabolites as well as gut hormones and endocrine peptides, including peptide YY, pancreatic polypeptide, neuropeptide Y, cholecystokinin, corticotropin-releasing factor, glucagon-like peptide, oxytocin, and ghrelin). Maintaining healthy gut microbiota (GM) is now being recognized as important for brain health through the use of probiotics, prebiotics, synbiotics, fecal microbial transplantation (FMT), etc. A few approaches exert antidepressant effects via restoring GM and hypothalamus–pituitary–adrenal (HPA) axis functions. In this review, we have summarized the etiopathogenic link between gut dysbiosis and depression with preclinical and clinical evidence. In addition, we have collated information on the recent therapies and supplements, such as probiotics, prebiotics, short-chain fatty acids, and vitamin B12, omega-3 fatty acids, etc., which target the gut–brain axis (GBA) for the effective management of depressive behavior and anxiety.
Article
Full-text available
Systemic injections of the flavonoid chrysin (5,7-dihydroxyflavone) exert anxiolytic-like effects in ovariectomised and cycling female rats through actions on gamma-aminobutyric acid-A (GABA A ) receptors; however, it is unknown if chrysin directly acts on brain structures that are involved in regulating emotional processes, such as the hippocampus. The present study evaluated the effects of intrahippocampal microinjections of 0.25, 0.5, and 1 μg of chrysin on anxiety-like behaviour in the elevated plus maze (EPM) and locomotor activity test (LAT) in female rats in proestrus and dioestrus. Similar doses of the neurosteroid allopregnanolone were used as a reference GABAergic anxiolytic drug. The participation of the GABA A /benzodiazepine receptor complex was evaluated by administering the antagonists picrotoxin, bicuculline and flumazenil. In proestrus, 0.5 and 1 μg of chrysin and allopregnanolone induced anxiogenic-like behaviour. In dioestrus, chrysin, and allopregnanolone (0.5 μg) induced anxiolytic-like effects. Picrotoxin, bicuculline and flumazenil prevented the effects of chrysin and allopregnanolone in both proestrus and dioestrus. None of the treatments significantly affected locomotor activity. These results indicate that the GABA A /benzodiazepine receptor complex in the dorsal hippocampus regulates the effects of chrysin on anxiety-like behaviour, similar to the actions of allopregnanolone. The divergent effects of treatments across the oestrous cycle phases suggest complex interactions between GABA A receptors and compounds with an anxiolytic potential.
Article
Full-text available
Background: The importance of polyphenols in human health is well known; these compounds are common in foods, such as fruits, vegetables, spices, extra virgin olive oil and wine. On the other hand, the different factors that modulate the biological activity of these compounds are less well known. Conceptualization of the work: In this review we took into account about 200 relevant and recent papers on the following topics: "polyphenols bioavailability", "polyphenols matrix effect", "food matrix effect", "polyphenols-cytochromes interaction", after having reviewed and updated information on chemical classification and main biological properties of polyphenols, such as the antioxidant, anti-radical and anti-inflammatory activity, together with the tricky link between in vitro tests and clinical trials. Key findings: the issue of polyphenols bioavailability and matrix effect should be better taken into account when health claims are referred to polyphenols, thus considering the matrix effect, enzymatic interactions, reactions with other foods or genetic or gender characteristics that could interfere. We also discovered that in vitro studies often underrate the role of phytocomplexes and thus we provided practical hints to describe a clearer way to approach an investigation on polyphenols for a more resounding transfer to their use in medicine.
Article
Full-text available
Chrysin, a herbal bioactive molecule, exerts a plethora of pharmacological effects, including anti-oxidant, anti-inflammatory, neuroprotective, and anti-cancer. A growing body of evidence has highlighted the emerging role of chrysin in a variety of neurological disorders, including Alzheimer’s and Parkinson’s disease, epilepsy, multiple sclerosis, ischemic stroke, traumatic brain injury, and brain tumors. Based on the results of recent pre-clinical studies and evidence from studies in humans, this review is focused on the molecular mechanisms underlying the neuroprotective effects of chrysin in different neurological diseases. In addition, the potential challenges, and opportunities of chrysin’s inclusion in the neurotherapeutics repertoire are critically discussed.
Article
Type 2 diabetes (T2DM) is a persistent metabolic disorder rising rapidly worldwide. It is characterized by pancreatic insulin resistance and β-cell dysfunction. Hyperglycemia induced reactive oxygen species (ROS) production and oxidative stress are correlated with the pathogenesis and progression of this metabolic disease. To counteract the harmful effects of ROS, endogenous antioxidants of the body or exogenous antioxidants neutralise it and maintain bodily homeostasis. Under hyperglycemic conditions, the imbalance between the cellular antioxidant system and ROS production results in oxidative stress, which subsequently results in the development of diabetes. These ROS are produced in the endoplasmic reticulum, phagocytic cells and peroxisomes, with the mitochondrial electron transport chain (ETC) playing a pivotal role. The exacerbated ROS production can directly cause structural and functional modifications in proteins, lipids and nucleic acids. It also modulates several intracellular signaling pathways that lead to insulin resistance and impairment of β-cell function. In addition, the hyperglycemia-induced ROS production contributes to micro- and macro-vascular diabetic complications. Various in-vivo and in-vitro studies have demonstrated the anti-oxidative effects of natural products and their derived bioactive compounds. However, there is conflicting clinical evidence on the beneficial effects of these antioxidant therapies in diabetes prevention. This review article focused on the multifaceted role of oxidative stress caused by ROS overproduction in diabetes and related complications and possible antioxidative therapeutic strategies targeting ROS in this disease.
Article
Background: Neuropsychiatric symptoms are common following traumatic brain injury (TBI), but their etiological onset remains unclear. Mental health research implicates neuroinflammation in the development of psychiatric disorders. The presence of neuroinflammatory responses after TBI thus prompts an investigation of their involvement in the emergence of neuropsychiatric disorders postinjury. Objective: Review the literature surrounding the role of neuroinflammation and immune response post-TBI in the development of neuropsychiatric disorders. Methods: A search of scientific databases was conducted for original, empirical studies in human subjects. Key words such as "neuroinflammation," "TBI," and "depression" were used to identify psychopathology as an outcome TBI and the relation to neuroinflammatory response. Results: Study results provide evidence of neuroinflammation mediated post-TBI neuropsychiatric disorders including anxiety, trauma/stress, and depression. Inflammatory processes and stress response dysregulation can lead to secondary cell damage, which promote the development and maintenance of neuropsychiatric disorders postinjury. Conclusion: This review identifies both theoretical and empirical support for neuroinflammatory response as feasible mechanisms underlying neuropsychiatric disorders after TBI. Further understanding of these processes in this context has significant clinical implications for guiding the development of novel treatments to reduce psychiatric symptoms postinjury. Future directions to address current limitations in the literature are discussed.
Article
In the last decade there has been an increasing awareness that traumatic brain injury (TBI) and concussion substantially increases risk for developing psychiatric disorders. Even mild TBI increases risk for depression and anxiety disorders such as posttraumatic stress disorder (PTSD) by 2-3 fold, predisposing patients to further functional impairment. This strong epidemiological link supports examination of potential mechanisms driving neuropsychiatric symptom development after TBI. One potential mechanism for increased neuropsychiatric symptoms after TBI is via inflammatory processes, as CNS inflammation can last years after initial injury. There is emerging preliminary evidence that TBI patients with PTSD or depression exhibit increased central and peripheral inflammatory markers compared to TBI without these comorbidities. Growing evidence has demonstrated that immune signaling in animals plays an integral role in depressive- and anxiety-like behaviors after severe stress or brain injury. In this review we will (1) discuss current evidence for chronic inflammation after TBI in the development of neuropsychiatric symptoms, (2) highlight potential microglial activation and cytokine signaling contributions and (3) discuss potential promise and pitfalls for immune targeted interventions and biomarker strategies to identify and treat TBI patients with immune-related neuropsychiatric symptoms.
Article
Chronic exposure to aluminium (Al) can contribute to the progression of several neurological and neurodegenerative diseases. Al is a metal that promotes oxidative damage leading to neuronal death in different brain regions with behavior, cognition, and memory deficits. Chrysin is a flavonoid found mainly in honey, passion fruit, and propolis with antioxidant, anti-inflammatory, and cytoprotective properties. In this study, we used an integrated approach of in vitro and in vivo studies to evaluate the antioxidant and neuroprotective effects of chrysin against the neurotoxicity elicited by aluminium chloride (AlCl3). In in vitro studies, chrysin (5 µM) showed the ability to counteract the early oxidative stress elicited by tert-butyl hydroperoxide, an oxidant that mimics the lipid peroxidation and Fenton reaction in presence of AlCl3 as well as the late necrotic death triggered by AlCl3 in neuronal SH-SY5Y cells. In vivo studies in a mouse model of neurotoxicity induced by chronic exposure to AlCl3 (100 mg/kg/day) for ninety days then corroborated the antioxidant and neuroprotective effect of chrysin (10, 30, and 100 mg/kg/day) using the oral route. In particular, chrysin reduced the cognitive impairment induced by AlCl3 as well as normalized the acetylcholinesterase and butyrylcholinesterase activities in the hippocampus. In parallel, chrysin counteracted the oxidative damage, in terms of lipid peroxidation, protein carbonylation, catalase, and superoxide dismutase impairment, in the brain cortex and hippocampus. Lastly, necrotic cells frequency in the same brain regions was also decreased by chrysin. These results highlight the ability of chrysin to prevent the neurotoxic effects associated with chronic exposure to Al and suggest its potential use as a food supplement for brain health.