Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Biomedicine & Pharmacotherapy 176 (2024) 116772

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Antioxidant effects of Paeoniflorin and relevant molecular mechanisms as


related to a variety of diseases: A review
Yansong Lu 1, Lu Yin 1, Wei Yang , Ze Wu , Jun Niu *
Department of Dermatology, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang 110016, China

A R T I C L E I N F O A B S T R A C T

Keywords: Paeoniflorin (PF), which is the main component of the Paeonia lactiflora Pall extract, is one of the traditional
Paeoniflorin Chinese medicines. The pharmacological effects associated with PF include antioxidant, immunomodulatory,
Oxidative stress anti-inflammatory, anticancer, antidepressant-like and neuroprotective effects. Our previous studies had
Antioxidant effect
revealed that PF protected melanocytes and inhibited photodamage through the suppression of oxidative stress
Clinical diseases
(OS). As OS plays a vital role in the progression of a variety of diseases, the capacity for PF to suppress OS may
exert important effects upon them. However, no review exists on these antioxidant effects of PF as related to
various diseases. Therefore, in this review we summarized studies involved with examining the antioxidant ef­
fects and molecular mechanisms of PF. Through its capacity to inhibit OS, PF has been shown to exert beneficial
effects upon several systems including nervous, cardiac/vascular, digestive, and respiratory as well as specific
diseases such as diabetes, autoimmune, pregnancy related, ocular, kidney, dermatology, along with suppression
of distal flap necrosis, postoperative adhesions, and hearing loss. Such findings provide new insights and di­
rections for future research directed at the development of PF as a natural antioxidant for the treatment of
clinical diseases.

1. Introduction a cause of several conditions including neurodegenerative diseases,


cardiovascular diseases, emphysema, cataracts, and cancer [8,9], anti­
Oxidative stress (OS), which is defined as a state of imbalance be­ oxidation represents a crucial process that can be used in the treatment
tween oxidation and antioxidation in the body with the excessive pro­ of such conditions.
duction of reactive oxygenated species (ROS), is believed to be a In recent years, natural antioxidants have been widely used in clin­
significant factor contributing to the progression of several diseases ical therapy, and traditional Chinese medicines with antioxidant effects
[1–3]. Cells are equipped with complex antioxidant defense networks, have gradually attracted increasing attention [10]. Paeoniflorin (PF) is a
which can cut ROS into less toxic substances, thus maintaining cell redox monoterpenoid glycoside extracted from Paeonia lactiflora Pall, a
homeostasis. The nuclear factor erythroid 2-related factor 2 (Nrf2) traditional Chinese herb [11]. PF exerts a variety of pharmacological
pathway, upregulating antioxidant enzymes such as superoxide dis­ effects, including antioxidant [12], immunomodulatory,
mutase (SOD), NAD(P)H: quinone oxidoreductase 1 (NQO1) and heme anti-inflammatory [13], anticancer [14], antidepressant-like [15], and
oxygenase-1 (HO-1), plays a key role in maintaining this balance of neuroprotective effects [16], etc. However, no review exists on the
redox homeostasis [4–7]. As OS-induced damage has been confirmed as antioxidant effects of PF as related to different diseases. In this report we

Abbreviations: PF, Paeoniflorin; OS, Oxidative stress; ROS, Reactive oxygenated species; Nrf2, Unclear factor erythroid 2-related factor 2; SOD, Superoxide
dismutase; P, NAD; NQO1, H:quinone oxidoreductase 1; HO-1, Heme oxygenase-1; LDH, Lactate dehydrogenase; MDA, Malondialdehyde; LD, Lactate dehydro­
genase′; SAH, Subarachnoid hemorrhage; GSH-Px, Glutathione peroxidase; CAT, Catalase activity; TLR4, Toll-like receptor 4; JAK2, Januskinase 2; STAT3, Signal
transducer and activator of transcription 3; PD, Parkinson’s disease; AD, Alzheimer’s disease; HIF-1α, Hypoxia inducible factor-1α; VEGF, Vascular endothelial
growth factor; I/R, ischemia/reperfusion; IRS, Insulin receptor substrate; GSK, Glycogen synthase kinase; SIRT1, Sirtuin-1; FOXO1a, Forkhead box O1; IBD, In­
flammatory bowel disease; AGEs, Advanced glycation end products; RPE, Retinal pigment epithelial; CKD, Chronic kidney disease; PGC-1α, Peroxisome proliferator-
activated receptor gamma coactivator-1α; UVA, Ultraviolet A; UVB, Ultraviolet B; SG, Sweat glands; NF-κB, Nuclear factor kappa-B.
* Corresponding author.
E-mail address: niujun06@126.com (J. Niu).
1
These authors contributed equally to this work and should be considered co-first authors.

https://doi.org/10.1016/j.biopha.2024.116772
Received 29 January 2024; Received in revised form 13 May 2024; Accepted 17 May 2024
Available online 28 May 2024
0753-3322/© 2024 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

review and summarize the data on the antioxidant effects (Fig. 1) and reduced [16,26].
molecular mechanisms (Table 1 and Fig. 2) of PF relating to various
diseases. Such information will be critical for the future development of
PF as a natural antioxidant in the treatment of clinical diseases. 2.2. Neural degenerative diseases

2. Inhibitory effects of PF as an antioxidant in diseases of the Different from the brain injury, aging is the major risk factor for the
nervous system neural degenerative diseases including Parkinson’s disease (PD) and
Alzheimer’s disease (AD) [27]. Currently, there is no strategy to reverse
Due to the high oxygen consumption, nervous system is particularly these diseases. However, increased expressions of OS markers were
susceptible to OS injury, which could induce irreversible damage to found in the brains of PD patients. Pretreatment of PF significantly
macromolecules [17]. As an antioxidant, PF may be a promising drug for reduced the production of ROS and lipid hydroperoxides, attenuated the
the treatment of neurological disorders. translocation of NF-κB without regulating phosphorylation of ERK1/2,
JNK, and p38, and enhanced the content of GSH in PC12 cells exposed to
the dopamine degenerating drug, 6-hydroxydopamine. In this PD model,
2.1. Brain injury PF exerted these protect effects, in part, by inhibiting the
ROS/PKCd/NF-kB pathway [28]. PF also suppressed OS in
Previous study has showed that OS is an important cause of tissue MPP+-challenged SH-SY5Y cells via ERK1/2-mediated Nrf2 activation
damage after cerebral ischemic injury, such as ischemic stroke [18–20]. [29], and suppressed H2O2 induced OS injury by activating the phos­
Pretreatment of PF significantly attenuated brain ischemic injury via phatidylinositol 3 kinase (PI3K)/protein kinase B(Akt-1) pathway in
inhibiting OS through the inhibition of Toll-like receptor 4 (TLR4)/ neural progenitor cell [30]. The phosphorylation of Akt-1 was abolished
Nuclear factor kappa-B (NF- κB) and activation of Janus kinase 2 by the inhibitor of PI3K upon OS. In an AD model, PF protected SH-SY5Y
(JAK2)/signal transducer and activator of transcription 3 (STAT3) cells from Aβ-induced cell injury via inhibiting oxidative injury and
signaling pathway [21–23]. In the injured PC12 cells, the treatment of mitochondrial dysfunction, and reversed the learning and memory
PF enhanced the levels of p‑JAK2 and p‑STAT3 in a dose-dependent deficits induced by streptozotocin [31–34]. In the C. elegans AD model,
manner and inhibited phosphorylation of p38MAPK [24]. In a rat PF alleviated the increased levels of ROS that contribute to the in vivo
model of acute cerebral infarction, PF reduced the degree of tissue toxicity of Aβ [35]. Zhai, A., et al. found that PF could reduce HT-22 cells
damage, increased SOD activities, lactate dehydrogenase (LDH) and OS induced by H2O2, which was associated with a decrease in miR-135a
Na+-K+-ATPase, while decreasing the contents of malondialdehyde via JAK2/STAT3 and ERK1/2 pathway regulation [36]. PF could sup­
(MDA) and lactate dehydrogenase (LD) [25]. This effect of PF also press ferroptosis and then reduce OS by binding to p53, that enhanced
appeared in Subarachnoid hemorrhage (SAH). OS induces the damage of the cognitive and behavioral abilities of AD mice [37].
the neurovascular unit via protein breakdown, lipid peroxidation and These studies showing an essential role of PF in the treatment of
DNA/RNA damage in early brain injury after SAH. Intraperitoneal in­ neural degenerative diseases. However, there are still limitations need to
jection of PF reduced the OS by decreasing expressions of ROS and MDA resolve in future, such as the low liposolubility of PF, making it difficult
and increasing expressions of SOD, glutathione peroxidase (GSH-Px) and to cross the blood-brain barrier. Enhancing the delivery of PF to the
catalase activity (CAT). As a result, the early brain injury after SAH were brain is the key for the future research.

Fig. 1. Antioxidant effects of PF in diseases. Through its capacity to inhibit OS, PF has been shown to exert beneficial effects upon several systems including nervous,
psychosis, cardiac/vascular, digestive, and respiratory as well as specific diseases such as diabetes, autoimmune, pregnancy related, ocular, kidney, dermatology,
along with suppression of distal flap necrosis, postoperative adhesions, and hearing loss.

2
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

Table 1
Summary of the antioxidant activities of PF. The antioxidant effects of PF in diseases, disease model, the dose of PF, the time points of evaluation, and regulation of
oxidative stress-related mechanisms were concluded.
Disease Disease model Animal and/or cell Dose of PF Time points Regulation of oxidative stress- Ref
of related mechanisms
evaluation

Nervous Ischemic stroke Oxygen‑glucose PC12 cells 25, 50, 100 and 48 h Activating the JAK2/STAT3 [23]
diseases deprivation/ 200 µM pathway
reoxygenation
Brain injuries H2O2 Schwann cells 0, 10, 20, 30, 50, 24 h Inhibiting phosphorylation of [24]
100, 200, and 300 p38MAPK
μmol⋅L− 1
Neonatal hypoxic Hypoxia/ischemia Rat pups ig. 6.25, 12.5, or 11d Inhibiting TLR4/NF-kB [21]
brain injuries 25 mg/kg
Acute Cerebral Middle cerebral artery Rats ig. 60, 120, 240 mg/ 7d Increasing the activities of [25]
Infarction occlusion kg SOD, LDH and Na+-K+-
ATPase, and decreasing the
contents of MDA and LD
Subarachnoid Endovascular perforation Rats/ Cortical i.p.5 mg/kg, twice 3d/24 h Up-regulating the Nrf2/ HO-1 [16]
hemorrhage neuron per day, /30 and pathway
100 µM
Brain injuries Lipopolysaccharide Mice BV-2 microglial 100 and 200 μM 1h Regulating NF-κB pathway [22]
cells
Nerve injuries H2O2 PC12 cells 80 µM 6h Reducing apoptosis and ROS [26]
accumulation
Parkinson’s disease MPP+/MPTP Mice/ SH-SY5Y cells ig. 50 mg/kg/3 µM 2w/24 h Through Nrf2-Dependent [29]
glutathione biosynthesis
mechanisms
Parkinson’s disease 6-hydroxydopamine PC12 cells 3, 10, or 30 µM 24 h Inhibiting ROS/PKCd/NF-kB [28]
pathway
Neural progenitor H2O2 Neural progenitor 200 μg/ml 4h Activating PI3K/Akt-1 [30]
cell injuries cells pathway
Alzheimer’s disease Streptozotocin Mice i.p.10 mg/kg 21d Attenuating mitochondrial [31]
dysfunction
Alzheimer’s disease H2O2 Hippocampal HT-22 100, 150, 200 and 24 h Down-Regulating MicroRNA- [36]
cells 300 µM 135a
Alzheimer’s disease Aβ (1–42) C. elegans strain 500 µM 36 h Regulating oxidative and heat [35]
shock stress responses
Alzheimer’s disease Aβ (25–35) SH-SY5Y cells 2, 10, 50 µM 24 h Preventing Mitochondrial [32]
Dysfunction
Alzheimer’s disease Aβ (1–42) Rats i.p.15 and 30 mg/ 20d Regulating the pathway [33]
(kg day) mediated by nerve growth
factor
Alzheimer’s disease Aβ (1–42) Rats i.p.7.5, 15 and 21d Regulating calcium [34]
30 mg/ (kg day) homeostasis
Alzheimer’s disease APP/PS1 double Mice/ PC12 cells ig.5 and 10 mg/kg/ 5w/4 min Suppressing ferroptosis and [37]
transgenic/ Erastin 2.5 µM and 5 µM then reduce OS by binding to
p53
Psychosis Schizophrenia MK-801 / H2O2 Mice /Primary ig. 10,50,100 mg/kg 6w/27 h Regulating the OS and [40]
cultured / 2 µg/ml and apoptosis pathways
hippocampal 200 ng/ml
neurons
Depression Forced swimming test Rats ig. 10 mg/kg 25 h Elevating plasma SOD and [43]
reducing MDA
Depression Corticosterone PC12 cells 1, 10 and 50 µM 48 h Up-regulating NGF expression [41]
Depression Glutamate 1, 10 and 50 µM 26 h Inhibiting Ca2+ overload [42]
Cardiac and Myocardial injury Lipopolysaccharide Mice i.p.20 mg/kg 3d Inhibiting p38 MAPK/NF-KB [48]
vascular p65
diseases
Myocardial ischemia Operation Rats ig.30 and 60 mg/kg 7d Inhibiting the expression of [49]
reperfusion injury apoptosis-related signaling
pathway
Cardiomyocyte Acrolein H9c2 100 µM 16 h Decreasing ROS production [51]
injury cardiomyocytes and down regulating caspases
cascade reaction
Cardiomyocyte Aconitine H9c2 80 mg/ml 0.5/1 h Decreasing MDA and lactate [50]
injury cardiomyocytes dehydrogenase
Cardiac hypertrophy Angiotensin II H9c2 50, 100, 200 and 24 h Increasing Nrf2/HO-1 and [52]
cardiomyocytes 400 μM PINK1/Parkin expression
Cardiomyocyte Doxorubicin H9c2 100 µmol/L 26 h Downregulating microRNA-1 [53]
injury cardiomyocytes expression
Cardiomyocyte Doxorubicin H9c2 1, 10, or 100 µmol/L 26 h Inhibiting of NOX2, NOX4 [54]
injury cardiomyocytes expression, and NOX activity
Vascular remodeling Platelet derived growth Vascular smooth 50, 100, and 200 µM 13, 25 or Regulating ROS-mediated [56]
diseases factor-BB muscle cells 49 h ERK1/2 and p38 pathways
Coronary disease Advanced oxidation Human umbilical 200 µM 25 h Blocking the ROS-HIF-1α/ [57]
protein products vein endothelial cells VEGF pathway
(continued on next page)

3
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

Table 1 (continued )
Disease Disease model Animal and/or cell Dose of PF Time points Regulation of oxidative stress- Ref
of related mechanisms
evaluation

Vascular damage H2O2 Human umbilical 10, 62.5, 250, and 26 h Scavenges Intracellular [58]
vein endothelial cells 1000 µmol/L ROS,repressing LDH
leakage, MDA production,
and restoring the activities of
total SOD and GSH-Px
Thrombosis A photo-chemical Human umbilical 10, 50, 250 mg/ml / Inhibiting of arachidonic acid [59]
reaction vein endothelial cells metabolism, the increase of t-
PA activity, and the protective
effect against free radical
Digestive Acute liver injury Lipopolysaccharide Rats i.p.60 mg/kg 7d Regulating the SIRT1/ [70]
system FOXO1a/SOD2 signaling
diseases
Hepatic fibrosis Carbon tetrachloride Mice ig. 100 mg/kg 1w Upregulating of heme [68]
oxygenase-1
Non-alcoholic fatty High-fat diet Rats ig. 20 mg/kg 4w Regulating the insulin [67]
liver disease pathway IRS/Akt/GSK3β
Hyperlipidemia High-fat diet Rats ig. 100, 12w Reducing liver MDA levels, [66]
300,500 mg/kg up-regulating SOD and Nrf2
levels
Hepatic I/Rinjury Operation Mice 5、10、20 mg/kg 24 h Decreasing the e expressions [65]
injected via the tail of apoptosis-associated
vein proteins
Liver inflammation Lipopolysaccharide Rats i.p.2.5, 5, or 10 mg/ 20d Increasing SOD, CAT and GPx [69]
kg levels as well as decreasing
GOT, GPT, LDH and MDA
levels
Cholestasis Alpha- Rats ig.0.2, 0.1 or 0.05 g/ 10d Activating Nrf2 through [72]
naphthylisothiocyanate kg PI3K/Akt-dependent pathway
Cholestasis Alpha- Rats ig.0.1 and 0.2 g/kg 10d Scavenging ROS via [73]
naphthylisothiocyanate inhibiting NADPH oxidase 4
expression
Cholestatic Hepatic Alpha- Mice/ LO2 ig.75, 150, and 10d/24 h Protecting hepatic depending [74]
Injury naphthylisothiocyanate cells 300 mg/kg/ 0, 20, Nrf2
and 100 µM
Inflammatory bowel Lipopolysaccharide Caco-2 cells 10, 50, 100 μM 24 and 48 h Inhibiting NF-κB by activating [77]
disease Nrf2/HO-1 pathway
Intestinal I/R Operation/hypoxic and IEC-6 cells ig.50 and 100 mg/ 3d/3, 6, 12, Decreasing MDA and [76]
reoxygenation kg /6.25, 12.5, 25, and 24 h increasing SOD and GSH
50, 100 and 200 μM
Respiratory Acute lung injury 5% sodium taurocholate Rats ig.40 mg/kg 24 h Activating the Nrf2/ARE [80]
diseases signaling
Asthma Ovalbumin Mice ig.10, 25 or 50 mg/ 4d Affecting glycolysis in asthma [82]
kg
Asthma Ovalbumin Mice ig.50 mg/kg 1w / [81]
Chronic obstructive Cigarette Smoke Rats 12, 24 and 48 mg/ 36w Quenching ROS and [83]
pulmonary disease kg upregulating antioxidant
enzymes via an Nrf2-
dependent mechanism
Radiation-induced γ-rays Human endothelial 50, 100 and 25 h Regulating the Nrf2/HO-1 [79]
lung injury cell lines 200 μg ml− 1 pathway
Diabetes and Diabetic cataracts High glucose Human lens 10 µM 24 h Upregulating SIRT1 [90]
complications epithelial SRA01/04
cells
Diabetic peripheral High glucose RSC96 cells 10 µM 24 h PF decreasing mitochondrial [86]
neuropathy ROS while increasing mtDNA
and mitochondrial membrane
potential
Diabetic peripheral High glucose RSC96 cells 1, 10 and 100 μM 48 h Activating Nrf2/ARE pathway [87]
neuropathy and Bcl-2-related apoptotic
pathway
Diabetic foot ulcer Streptozotocin /high Rats/ Human ig.15 and 30 mg/kg 16d/48 h Accelerating wound healing [88]
glucose immortalized /100 μM through the Nrf2 pathway
keratinocytes cells
Diabetic Streptozotocin/ high Mice/ ig. 20 and 70 mg/ 8w/- Alleviating myocardial [89]
cardiomyopathy glucose cardiomyocytes kg/- damage, resisting OS and
ferroptosis via Nrf2
Vascular Endothelial Streptozotocin /high Rats/Human ig.0.01 g/kg 6w/8d Repressing PKCβ1 protein [94]
Injuries glucose umbilical vein /100 μM level
endothelial cells
Diabetes mellitus Streptozotocin Pancreatic β-cells 20, 40, 80 μM 26 h Inhibiting the p38 MAPK and [85]
JNK signaling pathways
Diabetic osteopathy Methylglyoxal Osteoblastic MC3T3- 0.01, 0.1 and 1 μM 49 h Upregulating glyoxalase [92]
E1 cells system and mitochondrial
function
(continued on next page)

4
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

Table 1 (continued )
Disease Disease model Animal and/or cell Dose of PF Time points Regulation of oxidative stress- Ref
of related mechanisms
evaluation

Diabetic Advanced glycation end Rat HBZY-1 10− 6, 1 0− 5 and 48 h / [93]


nephropathy products mesangial cell line 10− 4 M
Autoimmune Rheumatoid arthritis Collagen-induced Rats ig.7.5 mg/kg 20d Regulating apoptosis via [96]
disease arthritis AMPK-mediated
mitochondrial fission
Rheumatoid arthritis Freund’s complete Rats i.p.5, 10 and 2 3w Inhibiting inflammation and [97]
adjuvant 0 mg/kg altering to COX-2 expression
Radiation-induced γ-rays Thymocytes 50,100,200 μg/ 24 h Scavenging ROS and [95]
thymocytes injury ml attenuating the activation of
the MAPK
Lymphocytes / Murine T-lineage 500 mg/ml 1–4 h or Reducing mitochondrial [98]
apoptosis cells and human T- 2–6 h membrane potential and
cell leukemia Jurkat increasing ROS
cells
Pregnancy Pregnancy-induced NG-nitro-L-arginine Rats/ human i.p.100 and 300 mg/ 17days/ Upregulating SIRT1 [100]
related hypertension methyl ester / H2O2 umbilical vein kg/ 50, 100 and 24 h
disease endothelial cells 200 μM
Ovarian I/R injury Operation Rats 25, 50 and 100 mg/ 30 min Increasing SOD and GSH [101]
kg levels as well as decreasing
MDA level
In vitro maturation Benzo(a)pyrene Porcine oocytes 0.1 mM / Activating the SHH signaling [102]
of porcine oocytes pathway, inhibiting ROS
production, and increasing
mitochondrial membrane
potential
Prenatal stress Glutamate Rats/ human 15, 30 and 60 mg/ 4w,24 h Regulating Nrf2/HO-1 [103]
neuroblastoma cell kg/ 10− 6, 10− 5, signaling pathway
SH-SY5Y 10− 4 M
Ocular diseases Age-related macular All-trans-retinal Human retinal 50, 100 and 200 μM 18 h Activating AMPK in a Ca2+/ [105]
degeneration pigment epithelium CaMKII-dependent manner
cells
Age-related macular H2O2 Human retinal 50,100 and 200 μg/ 24 h Decreasing ROS production [104]
degeneration pigment epithelium ml and caspase-3 activity, and
cells p38MAPK and ERK
phosphorylation
Hearing loss Cisplatin-induced Cisplatin Mice/ spiral ganglion i.p.30 mg/kg / 30 7d/50 h Decreasing ROS, increasing [107]
ototoxicity neuron μmol/L putative kinase protein 1, and
reducing apoptosis
Hearing loss Neomycin Mice/cochlear i.p.30 mg/kg / 30 7d/26 h Inhibiting ERK signaling [108]
explants μmol/L activation
Kidney diseases Chronic kidney 5/6 nephrectomized/ Rats/ mouse C2C12 ig.25 and 50 mg/ 7w/48 h Regulating AMPK/SIRT1/ [110]
disease induced TNF-α myoblasts kg/ 25 and 50 μM PGC-1α-mediated pathway
skeletal muscle
atrophy
Hypoxia/ COCL2 Human proximal 50, 100 and 200 µM 28 h Inhibiting apoptosis [111]
reoxygenation tubular epithelial and repressing oxidative
injury cells damage via Keap1/
Nrf2/HO-1 pathway
Skin diseases Photodamage Ultraviolet B Human immortalized 25、50 and 100 µM 26 h Inhibiting the ROS-p38-p53 [113]
keratinocytes cells pathway
Photodamage Ultraviolet A Human dermal 200, 400 and 24 h Regulating Nrf2/HO-1/NQ- [114]
Fibroblasts 800 μM O1 signaling pathway or
PLIN2
Vitiligo H2O2 Melanocyte 50 µM 24 h Activating JNK/Nrf2/HO-1 [115]
pathway
Bromhidrosis / Rats/primary human 2.5 mg/kg via foot 6, 24, 48 Associating with ROS [116]
sweat gland cells injection / 9.530 µM and 72 h production, apoptosis, and
/24 h autophagy.
Distal flap Skin flaps surgery Surgery Rats i.p.20 and 50 mg/kg 7d Promoting vascular [119]
necrosis hyperplasia, inhibiting
pyroptosis, and down-
regulating inflammation.
Skin flaps surgery Tert-butyl hydroperoxide Rats/ human ig. 10 mg/kg/ 0, 5, 7d/24 h Regulating Nrf2/HO-1 [118]
umbilical vein 10, and 20 μM Pathway
endothelial cells
Postoperative Postoperative Operation Rats ig.10, 20 and 7d Inhibiting inflammatory [121]
adhesions adhesions 40 mg/kg actions

3. The inhibitory effects of PF as an antioxidant in psychosis SOD and decreased MDA levels, then rescued schizophrenia -like be­
haviors [40]. Mao, Q. Q., et al. reported that PF protected PC12 cells
The psychosis is often associated with notable changes in OS markers from corticosterone-induced and glutamate-induced neurotoxicity by
[38] and the role of OS in the development of major depressive disorder inhibiting OS and Ca2+ overload, while increasing nerve growth factor
is unequivocally established [39]. In MK-801-treated mice, PF increased expression [41,42]. PF showed this anti-depressive effect by elevating

5
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

Fig. 2. The main pathway of PF as an antioxidant. The capacity to inhibit OS serves as the foundation for these beneficial effects of PF, with these effects mainly
being related to the Nrf2 pathway, including Nrf2-KEAP1, Nrf2/HO-1, Nrf2/ARE, Nrf2/HO-1/NQ-O1 and JNK/Nrf2/HO-1. However, the TLR4/NF-kB, JAK2/STAT3,
ROS/PKCd/NF-kB, PI3K/Akt-1, JAK2/STAT3, ERK1/2, NF-KB/p65, and ROS/PLIN2 pathways are also involved in the process of OS inhibition by PF.

plasma SOD and reducing MDA levels in rats subjected to a forced effects of PF on NOX activity and NOX4, NOX2 and miR-1 expressions
swimming test [43]. Through antioxidant effects, PF ameliorated the [53,54].
depressive-like behaviors in depression models.
4.2. Vascular damage
4. Inhibitory effects of PF as an antioxidant in cardiac and
vascular diseases Increases in ROS within vascular endothelial cells can be due to a
variety of factors [55]. Such increases can induce an abnormal prolif­
Substantial evidence shows that OS is a critical factor in several eration of vascular smooth muscle cells and vascular remodeling which,
cardiac and vascular diseases, such as hypertension [44], myocardial in turn, leads to atherosclerosis and stenosis. Recent evidence indicated
infarction and atherosclerosis [45]. The effects of PF as an antioxidant in that PF could serve as a possible therapy for vascular reconstruction
the treatment of several cardiac and vascular diseases by inhibiting OS diseases by inhibiting vascular smooth muscle cells proliferation
are as follows. induced by platelet derived growth factor-BB via suppression of ROS
production, an effect which was associated with the reduction of the
4.1. Myocardial injury phosphorylation of p38 and ERK1/2 [56]. PF protect human umbilical
vein endothelial cells against H2O2 and advanced oxidation protein
Myocardial oxidative injury can result from a variety of factors products as induced by OS, with the latter being associated with the
including coronary intervention, thrombolytic therapy, and environ­ down-regulation of hypoxia inducible factor-1α(HIF-1α)/vascular
mental pollutants, which induced the accumulation of ROS beyond the endothelial growth factor (VEGF) via ROS-NF-κB axis [57,58]. PF also
cellular threshold that disrupts the normal physiological function of was shown to decrease free radicals, increase t-PA activity, inhibit
cardiomyocytes [46,47]. Wang, Jia et al. reported that PF decreased arachidonic acid metabolism, and further prolong thrombosis times and
MDA, GSH and ROS in septic mice, and PF could diminish myocardial reduce vascular endothelial damage. These effects are consistent with
injury via suppressing p38 MAPK/NF-KB p65, and the phosphorylation the activation of SOD [59].
activity of p38 MAPK and p65 was inhibited [48]. PF pretreatment In conclusion, PF may present a promising alternative drug for the
protected rats from myocardial ischemia reperfusion injury by reducing management of cardiovascular-related disorders.
levels of OS through increasing SOD activity along with decreasing MDA
levels [49]. PF also reverses cardiotoxicity as induced with aconitine and 5. Inhibitory effects of PF as an antioxidant in digestive system
acrolein via decreasing MDA, LDH and ROS [50,51], and restores the diseases
expression of Nrf2/HO-1 and PINK1/Parkin to alleviate cardiac hyper­
trophy induced by AngII in H9c2 cardiomyocytes [52]. Doxorubicin, a OS is involved in the pathogenesis of several digestive system dis­
cardiotoxic anti-tumor drug, could induce cardiomyocyte apoptosis, eases, such as liver injury, cholestasis, and intestinal diseases [60]. The
which could be inhibited by PF via decreasing the reduction of ROS. effects of PF as an antioxidant in the treatment of digestive system dis­
Moreover, these reductions in ROS were closely related to the inhibitory eases by inhibiting oxidative stress are as follows.

6
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

5.1. Liver injury proteins of PF that may play an important role in OS [81]. PF inhibits OS
by modulating glycolysis, which in turn modulates autophagy to
The increase of ROS is related to the pathogenesis of hepatic fibrosis enhance immunity and inflammatory responses in asthmatic mice [82].
[61–63] and ischemia/reperfusion (I/R) injury, as well as the elevation What’s similar is that a 40 mg/kg⋅d treatment with PF was effective in
of serum free fatty acids and insulin resistance in the liver [64]. PF has attenuating OS in chronic obstructive pulmonary disease rats by inhib­
been shown to exert a protective effect upon hepatic I/R injury by iting ROS and increasing antioxidant enzymes through the Nrf2
inhibiting OS and inflammation [65]. PF can reduce MDA and pathway. In addition, the sooner PF treatment was initiated, the more
up-regulate liver SOD and Nrf2 levels which then reduces liver choles­ pronounced the protective effect of PF was [83]. Overall, PF as an
terol and steatosis as demonstrated in a rat model of hyperlipidemia antioxidant plays a key role in respiratory diseases.
[66]. It has also been reported that PF modulates the insulin receptor
substrate (IRS)-1/Akt/glycogen synthase kinase (GSK)3βpathway and 7. Inhibitory effects of PF as an antioxidant in diabetes and
ameliorates high-fat diet-induced non-alcoholic fatty liver disease by complications
inhibiting OS. PF treatment restored the decreasing phosphorylation of
Akt and GSK3β and the increasing serine phosphorylation of IRS-1 [67]. Diabetes is a metabolic disease that poses a major threat to the
The capacity for PF to alleviate liver fibrosis is related to the increased human health and well-being. The excessive production of ROS induced
expression of HO-1 and activation of hepatic stellate cells along with an by hyperglycemia is an important pathogenesis of diabetes and com­
attenuation of CCl4-induced OS [68]. In vivo, daily administrations of plications [84]. It is well known that a close relationship exists between
PF in rats significantly ameliorates the OS as induced by LPS and di­ pancreatic β-cells function and the onset of diabetes. Treatment with PF
minishes liver dysfunction [69]. Through its ability to reduce ROS and ameliorated pancreatic β-cell damage by inhibiting the JNK and
MDA levels and increase SOD activity, PF improves liver structure and p38MAPK signaling pathways, and in streptozotocin-treated INS-1 cells,
function in an acute liver injury model, an effect which was associated diminished ROS, and MDA reductions along with increasing SOD ac­
with the sirtuin-1(SIRT1)/forkhead box O1(FOXO1a)/SOD2 pathway tivity. These findings not only indicate that PF improves pancreatic
[70]. This suggests that PF protects the liver from OS damage. β-cell function by inhibiting the JNK and p38MAPK pathways but also
by inhibiting OS to maintain blood glucose levels [85]. Yang, Li et al.
5.2. Cholestasis reported that PF improved diabetic peripheral neuropathy by upregu­
lating thioredoxin based on 4D Label-free proteomic experiments in
Cholestasis is characterized by excessive ROS production and an Schwann cells mitochondria. This effect was associated with the ca­
imbalance of the redox system [71]. Chen, Ma et al. demonstrated an pacity for PF to reduce mitochondrial ROS and enhance mitochondrial
inhibitory effect of PF on OS as resulting from the capacity for PF to function in a high-glucose environment [86], along with the Nrf2/ARE
alleviate alphanaphthylisothiocyanate-induced cholestasis, a mecha­ and Bcl-2 pathways [87]. Moreover, experiments in vivo and in vitro
nism which appears to involve an activation of Nrf2 through the have demonstrated that PF inhibited OS through the Nrf2 pathway and
attenuate phosphorylations of PI3K/Akt pathway [72]. In a rat model of promoted wound healing in diabetic foot [88]. PF resisted OS and fer­
alphanaphthylisothiocyanate-induced cholestasis, PF attenuated chole­ roptosis via Nrf2 to improve cardiac dysfunction in mice with diabetic
static liver injury by scavenging ROS through suppressing the expression cardiomyopathy [89]. Whatmore, PF significantly reduced MDA and
of NADPH oxidase 4 [73], and Nrf2 is essential in this process [74]. increased SOD and GSH-Px levels in lens epithelial cells subjected to
high levels of glucose, which could be reversed by the inhibition of
5.3. Intestinal disease SIRT1. Accordingly, it appears that PF inhibited the
epithelial-mesenchymal transition of lens epithelial cells and amelio­
Excessive OS, inflammatory responses and impaired intestinal bar­ rated diabetic cataracts through a combination of OS suppression and
rier function comprise the main features of such intestinal diseases [75], SIRT1 upregulation [90]. In addition, PF also protected osteoblast
as inflammatory bowel disease (IBD) and intestinal I/R. In a MC3T3-E1 cells from methylglyoxal and antimycin-induced cytotoxic
dose-dependent manner, PF reversed the increase of MDA and decrease damage through inhibiting OS and improving mitochondrial function to
of SOD and GSH as occurs in I/R-injured rats [76]. PF also inhibited relieve diabetic osteopathy [91,92]. There is recent evidence indicating
NF-κB by activating the Nrf2/HO-1 pathway in Caco-2 cells treated with that an excessive accumulation of advanced glycation end products
LPS. This could be blocked by the RNA interference against Nrf2. As a (AGEs) leads to an imbalance of oxidant and antioxidant systems that
result, PF inhibited tight junction protein disruption and inflammatory can then damage kidney tissue and promote the development of
responses, and regulated barrier function, prevented IBD [77]. diabetic-induced nephropathy. In the experiment to evaluate the anti­
The above evidences illustrate the important role of the antioxidative oxidant capacity of PF, Zhang, Feng et al. found that PF significantly
effect of PF in the digestive system diseases. increased glutathione peroxidase and catalase activities, thereby atten­
uating AGEs-induced OS in mesangial HBZY-1 cells [93]. Through its
6. Inhibitory effects of PF as an antioxidant in respiratory ability to inhibit OS, PF has been shown to attenuate the vascular
diseases damage due to fluctuating hyperglycemia as well as diminish inflam­
matory responses and inhibit protein kinase Cβ1 protein levels [94].
Excessive ROS production, as can be induced by radiation in pul­ Based on these findings, PF may be a promising treatment strategy
monary endothelial cells, plays a key role in the pathogenesis of lung for diabetes and complications.
injury [78]. In addition, acute lung injury, as a common complication
associated with the early stages of severe acute pancreatitis, is also 8. Inhibitory effects of PF as an antioxidant in autoimmune
mainly manifested by excessive OS and inflammatory responses. Yu, J., diseases
et al. reported that PF could protect pulmonary endothelial cells from
radiation-induced oxidative damage via the Nrf2/HO-1 signaling Disruption of redox balance and mitochondrial dysfunction caused
pathway [79]. In a rat model of acute lung injury resulting from severe by excess of ROS is closely related to immune pathogenesis, PF is also an
acute pancreatitis, PF down-regulated the levels of LDH and MDA, and immunomodulator, it can weaken thymocytes injury induced by
up-regulated SOD levels in lung tissue. Simultaneously, PF increased ionizing irradiation through scavenging ROS and activating MAPK [95].
Cyt-Nrf2, HO-1 and NQ-O1 expressions, which are related to the In a rat model of collagen-induced arthritis, PF down-regulated in­
Nrf2/ARE signaling pathway [80]. In addition, transcriptional and flammatory reactions and regulated apoptosis, while simultaneously
metabolomic databases for asthma have identified some potential target down-regulating MDA and NO levels in serum and synovial tissue and

7
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

up-regulating SOD levels [96]. PF also increased the pain threshold 12. Protective effects of PF as an antioxidant in kidney diseases
within this rat model of rheumatoid arthritis and relieved arthritis
symptoms by regulating OS [97]. What’s interesting is that PF has been The imbalance between protein degradation and synthesis in chronic
shown to activate caspase with the fragmentation of DNA, and in this kidney disease (CKD), leads to skeletal muscle atrophy [109]. This pa­
way, induces human T-cell leukemia Jurkat cell and murine thology results from an excessive accumulation of ROS leading to
T-lymphocyte apoptosis. In contrast to typically observed, PF increased mitochondrial dysfunction and OS. Li, Wu et al. demonstrated that PF
ROS levels and decreased mitochondrial membrane potential in this diminished this skeletal muscle atrophy through inhibiting mitochon­
process. Therefore, it appears that PF can exert bidirectional regulatory drial dysfunction and OS in CKD, effects which were abolished by
effects upon OS [98]. Therefore, PF plays a vital role in regulating im­ knocking down peroxisome proliferator-activated receptor gamma
munity through antioxidant effects. coactivator-1α (PGC-1α) and inhibiting SIRT1and AMPK [110]. In the
model of acute kidney injury induced by H/R, PF promoted Nrf2 nuclear
9. Inhibitory effects of PF as an antioxidant in pregnancy related translocation and then increased the expression of HO-1, which could be
diseases reversed by the Nrf2 inhibitor. This study showed that PF inhibited
apoptosis and decreased OS via the Nrf2/HO-1 pathway [111]. In
OS can produce damaging effects within both the mother and fetus, summary, the utilization of PF as an antioxidant exhibits promising
such as gestational hypertension and delayed fetal brain development therapeutic potential in kidney diseases.
[99]. PF may moderate the development of gestational hypertension by
suppressing OS. In addition, the up-regulation of SIRT1 expression in 13. Inhibitory effects of PF as an antioxidant in skin diseases
gestational hypertensive rats treated with PF, diminishes vascular
endothelial cell injury/dysfunction, and reduces inflammation and OS. The skin is the largest organ of the human body. Many factors can
Within human umbilical vein endothelial cells, PF served as a protectant lead to skin diseases including inflammation, immunity, OS, etc. The
against H2O2-induced injury [100]. The Increased levels of SOD and effects of PF as an antioxidant in the treatment of dermatology diseases
GSH and decreased level of MDA were shown in the treatment of PF in by inhibiting OS are as follows.
ovarian ischemia-reperfusion injury [101]. PF enhances porcine oocyte Photodamage could be induced by ultraviolet A (UVA) or ultraviolet
maturation as demonstrated in vitro through inhibiting ROS production, B (UVB) radiation, both of which can induce an extensive generation of
increasing mitochondrial membrane potentials, and activating the SHH ROS [112]. It has been reported that PF protected human keratinocytes
pathway [102]. Similarly, in the offspring of prenatal stress rats, PF from UVB induced apoptosis through activation of the ROS-p38-p53
decreased intracellular Ca2+ concentrations and apoptosis, down­ pathway [113]. Recent results from our laboratory have indicated that
regulated ROS and NO levels and regulated mitochondrial membrane PF could suppress the OS as induced by UVA via the Nrf2/HO-1/NQ-O1
potentials. PF also has been shown to attenuate the injury of SH-SY5Y pathway or by upregulating PLIN2, thereby inhibiting photodamage
cells as induced by glutamate via regulating Nrf2/HO-1 and apoptotic [114].The increasing levels of Nrf2, HO-1, and NQ-O1 expression were
pathways, increasing the protein levels of Nrf2, HO-1, and NQO-1, observerd. What’s similar is that PF protected melanocytes from
preventing the development of fetal learning and memory lesions H2O2-induced OS, and the JNK/Nrf2/HO-1 signaling pathway played a
[103]. These results suggest that the antioxidant effects of PF exert a vital role in this process of PF protection. Accordingly, PF may serve as a
significant role in the treatment of pregnancy-related disorders. How­ potential therapy for vitiligo [115]. In primary human sweat glands (SG)
ever, it should be noted that other potential effects of PF in the fetus will cells treated with 3‑methyladenine, PF suppressed cell proliferation and
require further investigation. significantly increased autophagy and apoptosis. Interestingly, PF
increased ROS production in SG cells, suggesting that the PF’s role in the
10. Inhibitory effects of PF as an antioxidant in ocular diseases treatment of bromhidrosis involves an increase in OS within these SG
cells [116]. All in all, PF may play a major role in the treatment of
Retinal degenerative diseases are characterized by the dysfunction or dermatological diseases.
death of retinal pigment epithelial (RPE) cells. As RPE cells have a high
metabolic rate they are more susceptible to oxidative damage. In 14. Inhibitory effects of PF as an antioxidant in distal flap
response to H2O2-induced oxidative damage, PF inhibits the increase of necrosis
ROS generation and caspase-3 activity in RPE cells, while inhibiting the
phosphorylation of ERK and p38 MAPK in a dose-dependent manner The most common clinical difficulty associated with wound repair is
[104]. Similarly, in RPE cells treated with all-trans-retinal, PF pre­ distal flap necrosis. It seems likely that the OS generated in injured tissue
treatment activated AMPK in a Ca2+/CaMKII-dependent manner, can result in endothelial cell injury and decrease skin flap viability
thereby inhibiting OS and mitochondrial dysfunction [105]. These re­ [117]. Recent findings have indicated that PF inhibited endothelial cell
sults provide reliable evidence for use of PF in the treatment of ocular apoptosis through the Nrf2/HO-1 signaling pathway. The protection of
diseases. PF was partially abolished by a selective Nrf2 inhibitor [118]. Moreover,
the capacity for PF to increase SOD and decrease MDA activities served
11. Inhibitory effects of PF as an antioxidant in hearing loss to increase flap survival rates in rats. Hence, PF provides an effective
means for enhancing survival of distal flaps by inhibiting apoptosis,
Overproduction of ROS, apoptosis, and alterations in mitochondrial inflammation, and OS [119].
membrane potential lead to spiral ganglion neuronal damage, effects
which may be associated with ototoxicity from the antitumor drug, 15. Inhibitory effects of PF as an antioxidant in postoperative
cisplatin [106]. Moreover, some antibacterial drugs, such as neomycin, adhesions
can produce an excessive production of ROS leading to hear cell damage,
further instigating hearing loss. PF can significantly decrease ROS levels, There is a high incidence of adhesion formation after abdominal
increase the expression of putative kinase protein 1 and reduce surgery, which may be related to trauma, infection, or OS of the peri­
apoptosis, thereby counteracting the ototoxicity of cisplatin [107]. toneum [120]. Gao, Wei et al. found that the peritoneal adhesions in rats
Similarly, PF can exert a remarkable effect in reducing neomycin subjected to abdominal surgery could be significantly reduced by the
induced hearing loss, through its capacity to reduce OS and inhibit ERK gavage with PF. These beneficial effects of PF appear to involve a down
signaling activation [108]. These studies suggest that PF may inhibit regulation in the expressions of COX-2, TGF- β1 and IL-6, and an
hearing damage through antioxidant effects. up-regulation in the expressions of MMP-9 and SOD-2 during the

8
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

postoperative period. Accordingly, the ability for PF to diminish peri­ mechanisms and potential clinical effects of PF. As the main component
toneal adhesion involves a suppression of inflammation and OS [121]. of traditional Chinese medicine, PF will provide more and more infor­
PF may become a therapeutic agent for postoperative peritoneal mation for clinical solutions and go to the world in the new situation of
adhesions. China.

16. Discussion and conclusion CRediT authorship contribution statement

PF is a safe, effective, and nontoxic compound, whose antioxidative Jun Niu: Writing – review & editing, Validation. Yansong Lu:
effects have been extensively investigated both in vitro and in vivo as Writing – original draft, Data curation. Lu Yin: Data curation. Wei
related to several diseases. PF has been shown to exert beneficial effects Yang: Investigation. Ze Wu: Validation.
when applied to disorders involving the nervous, cardiac/vascular,
digestive, and respiratory systems, in diseases such as diabetes, auto­ Declaration of Competing Interest
immune, pregnancy related, ocular, kidney and dermatology as well as
in suppression of distal flap necrosis, postoperative adhesions and All co-authors have been seen and agreed with contents of this
hearing loss. The capacity to inhibit OS serves as the foundation for these research. The authors have declared no conflicts of interest. Neither the
beneficial effects of PF, with these effects mainly being related to the entire research nor any part of its content has been published or has been
Nrf2 pathway, including Nrf2-KEAP1, Nrf2/HO-1, Nrf2/ARE, Nrf2/HO- accepted elsewhere. It is not being submitted to any other journal.
1/NQ-O1 and JNK/Nrf2/HO-1. However, the TLR4/NF-kB, JAK2/
STAT3, ROS/PKCd/NF-kB, PI3K/Akt-1, JAK2/STAT3, ERK1/2, p38 Data Availability
MAPK/NF-KB p65, IRS/Akt/GSK3β, SIRT1/FOXO1a/SOD2, AMPK/
SIRT1/PGC-1α and ROS-p38-p53 pathways are also involved in the No data was used for the research described in the article.
process of OS inhibition by PF. The main pathway is shown in Fig. 2.
Through these pathways, PF could increase antioxidant enzymes, and Acknowledgments
decrease ROS. In addition, through its capacity to inhibit OS, PF can
mitigate the proliferation, apoptosis, ferroptosis and inflammation This work was supported by grants from the Liaoning Province
associated with several diseases. Interestingly, in the treatment of Technological Innovation Planned Project (Grant number 2022JH2/
bromhidrosis, PF appears to function by increasing OS within SG cells. 101500012), Liaoning Province Doctoral Research Initiation Fund Pro­
Accordingly, the beneficial effects of PF may not only involve an inhi­ gram Project (Grant number 2023-BS-033) and Shenyang Science and
bition of OS, but also the promotion OS, within certain diseases. In Technology Planned Project (Grant number 22–321–33–34). The lan­
addition, PF has anti-tumor effects. However, the underlying molecular guage was polished by the editorial services of ED-IT.
mechanism might be related to the inflammation and apoptosis path­
ways [13,122]. At present, the study on PF against tumors through References
regulating OS is limited. [123]. Therefore, as an antioxidant, PF’s
anti-tumor effect has a good prospect and needs further research. [1] L.J. Machlin, A. Bendich, Free radical tissue damage: protective role of
PF exhibits definite antioxidant activity similar other compounds, antioxidant nutrients, FASEB J. 1 (6) (1987) 441–445.
[2] K. Jomova, R. Raptova, S.Y. Alomar, S.H. Alwasel, E. Nepovimova, K. Kuca,
such as the antioxidant activity of quercetin in renal I/R injury [124], M. Valko, Reactive oxygen species, toxicity, oxidative stress, and antioxidants:
resveratrol in atherosclerosis [125], Curcumin in anti-aging [126], and chronic diseases and aging, Arch. Toxicol. 97 (10) (2023) 2499–2574, https://
epigallocatechin gallate in osteoporosis [127]. However, antioxidant doi.org/10.1007/s00204-023-03562-9.
[3] X. Yang, X. Liu, Y. Nie, F. Zhan, B. Zhu, Oxidative stress and ROS-mediated
activity of PF is much lower than vitamin C [128]. PF not only has cellular events in RSV infection: potential protective roles of antioxidants, Virol.
antioxidant effects, it has a variety of pharmacological activities, such as J. 20 (1) (2023) 224, https://doi.org/10.1186/s12985-023-02194-w.
immune regulation, anti-inflammatory, and anti-tumor. Thus, PF can [4] G.K. McWalter, L.G. Higgins, L.I. McLellan, C.J. Henderson, L. Song, P.
J. Thornalley, K. Itoh, M. Yamamoto, J.D. Hayes, Transcription factor Nrf2 is
improve disease from multiple ways, which is more extensive than essential for induction of NAD(P)H:quinone oxidoreductase 1, glutathione S-
vitamins. transferases, and glutamate cysteine ligase by broccoli seeds and isothiocyanates,
However, the structural modification, bioavailability, dose adjust­ J. Nutr. 134 (12 Suppl) (2004) 3499S–3506S, https://doi.org/10.1093/jn/
134.12.3499S.
ment, and toxicity of PF as an antioxidant still need to be further studied.
[5] H. Kumar, S. Koppula, I.S. Kim, S.V. More, B.W. Kim, D.K. Choi, Nuclear factor
The low bioavailability of PF due to its hydrolytic degradation in the erythroid 2-related factor 2 signaling in Parkinson disease: a promising multi
intestine and poor penetration has further limited its clinical applica­ therapeutic target against oxidative stress, neuroinflammation and cell death,
tion. Structural modification, such as paeoniflorin-phospholipid com­ CNS Neurol. Disord. Drug Targets 11 (8) (2012) 1015–1029, https://doi.org/
10.2174/1871527311211080012.
plex [129] and paeoniflorin-6′-O-benzene sulfonate, can improve the [6] J. Alam, D. Stewart, C. Touchard, S. Boinapally, A.M. Choi, J.L. Cook, Nrf2, a
bioavailability of PF [130,131]. In addition, the doses and administra­ Cap’n’Collar transcription factor, regulates induction of the heme oxygenase-1
tions of PF used in different cell or disease model were different. Gavage, gene, J. Biol. Chem. 274 (37) (1999) 26071–26078, https://doi.org/10.1074/
jbc.274.37.26071.
intraperitoneal injection and direct administration, doses were varied [7] P. Poprac, K. Jomova, M. Simunkova, V. Kollar, C.J. Rhodes, M. Valko, Targeting
from 0.1μm to 1000μm in cells and 2.5 mg/kg to 1000 mg/kg in animals free radicals in oxidative stress-related human diseases, Trends Pharmacol. Sci.
for 30 minutes to 36 weeks (see Table 1). What’s more, there has been 38 (7) (2017) 592–607, https://doi.org/10.1016/j.tips.2017.04.005.
[8] K. Xie, X. He, K. Chen, K. Sakao, D.X. Hou, Ameliorative effects and molecular
little evaluation of toxicity in PF target organs, it is encouraged to design mechanisms of vine tea on western diet-induced NAFLD, Food Funct. 11 (7)
studies with long-term application of PF to achieve the best possible (2020) 5976–5991, https://doi.org/10.1039/d0fo00795a.
level of safety [132]. Therefore, it is crucial to find the best PF con­ [9] C. Lopez-Alarcon, A. Denicola, Evaluating the antioxidant capacity of natural
products: a review on chemical and cellular-based assays, Anal. Chim. Acta 763
centration, administration, and application time to meet different (2013) 1–10, https://doi.org/10.1016/j.aca.2012.11.051.
experimental requirements. [10] H. Xu, S. Li, J. Liu, J. Cheng, L. Kang, W. Li, Y. Zhong, C. Wei, L. Fu, J. Qi,
Although a substantial amount of evidence, as presented in this re­ Y. Zhang, M. You, Z. Zhou, C. Zhang, H. Su, S. Yao, Z. Zhou, Y. Shi, R. Deng,
Q. Lv, F. Li, F. Qi, J. Chen, S. Zhang, X. Ma, Z. Xu, S. Li, Y. Xu, K. Peng, Y. Shi,
view, indicates that PF may be a promising antioxidant for the treatment
H. Jiang, G.F. Gao, L. Huang, Bioactive compounds from Huashi Baidu decoction
of a variety of diseases, there remains a significant lack of clinical possess both antiviral and anti-inflammatory effects against COVID-19, Proc.
observation and clinical research regarding the antioxidative effects of Natl. Acad. Sci. USA 120 (18) (2023) e2301775120, https://doi.org/10.1073/
PF. In this regard, to further validate the conclusions of cell and animal pnas.2301775120.
[11] X. Zhang, Y. Zhai, J. Yuan, Y. Hu, New insights into Paeoniaceae used as
experiments, researchers need to conduct large-scale, randomized, medicinal plants in China, Sci. Rep. 9 (1) (2019) 18469, https://doi.org/
controlled, double-blind clinical trials to explore the further 10.1038/s41598-019-54863-y.

9
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

[12] S.H. Kim, M.K. Lee, K.Y. Lee, S.H. Sung, J. Kim, Y.C. Kim, Chemical constituents [33] Z. Lan, L. Chen, Q. Fu, W. Ji, S. Wang, Z. Liang, R. Qu, L. Kong, S. Ma,
isolated from Paeonia lactiflora roots and their neuroprotective activity against Paeoniflorin attenuates amyloid-beta peptide-induced neurotoxicity by
oxidative stress in vitro, J. Enzym. Inhib. Med. Chem. 24 (5) (2009) 1138–1140, ameliorating oxidative stress and regulating the NGF-mediated signaling in rats,
https://doi.org/10.1080/14756360802667977. Brain Res. 1498 (2013) 9–19, https://doi.org/10.1016/j.brainres.2012.12.040.
[13] Y.X. Zhou, X.H. Gong, H. Zhang, C. Peng, A review on the pharmacokinetics of [34] S.Z. Zhong, Q.H. Ge, Q. Li, R. Qu, S.P. Ma, Peoniflorin attentuates Abeta((1-42))-
paeoniflorin and its anti-inflammatory and immunomodulatory effects, Biomed. mediated neurotoxicity by regulating calcium homeostasis and ameliorating
Pharmacother. 130 (2020) 110505, https://doi.org/10.1016/j. oxidative stress in hippocampus of rats. J. Neurol. Sci. 280 (1-2) (2009) 71–78,
biopha.2020.110505. https://doi.org/10.1016/j.jns.2009.01.027.
[14] Y. Xiang, Q. Zhang, S. Wei, C. Huang, Z. Li, Y. Gao, Paeoniflorin: a monoterpene [35] L. Ai, F. Yang, J. Song, Y. Chen, L. Xiao, Q. Wang, L. Wang, H. Li, T. Lei, Z. Huang,
glycoside from plants of Paeoniaceae family with diverse anticancer activities, Inhibition of abeta proteotoxicity by Paeoniflorin in Caenorhabditis elegans
J. Pharm. Pharmacol. 72 (4) (2020) 483–495, https://doi.org/10.1111/ through regulation of oxidative and heat shock stress responses, Rejuvenation
jphp.13204. Res. 21 (4) (2018) 304–312, https://doi.org/10.1089/rej.2017.1966.
[15] X.L. Wang, S.T. Feng, Y.T. Wang, N.H. Chen, Z.Z. Wang, Y. Zhang, Paeoniflorin: a [36] A. Zhai, Z. Zhang, X. Kong, Paeoniflorin alleviates H(2)O(2)-induced oxidative
neuroprotective monoterpenoid glycoside with promising anti-depressive injury through down-regulation of MicroRNA-135a in HT-22 cells. Neurochem
properties, Phytomedicine 90 (2021) 153669, https://doi.org/10.1016/j. Res. 44 (12) (2019) 2821–2831, https://doi.org/10.1007/s11064-019-02904-3.
phymed.2021.153669. [37] L. Zhai, H. Pei, H. Shen, Y. Yang, C. Han, Q. Guan, Paeoniflorin suppresses
[16] T. Wang, L. Xu, L. Gao, L. Zhao, X.H. Liu, Y.Y. Chang, Y.L. Liu, Paeoniflorin neuronal ferroptosis to improve the cognitive behaviors in Alzheimer’s disease
attenuates early brain injury through reducing oxidative stress and neuronal mice, Phytother. Res. 37 (10) (2023) 4791–4800, https://doi.org/10.1002/
apoptosis after subarachnoid hemorrhage in rats, Metab. Brain Dis. 35 (6) (2020) ptr.7946.
959–970, https://doi.org/10.1007/s11011-020-00571-w. [38] V. Rambaud, A. Marzo, B. Chaumette, Oxidative stress and emergence of
[17] S. Orellana-Urzua, I. Rojas, L. Libano, R. Rodrigo, Pathophysiology of Ischemic psychosis, Antioxidants 11 (10) (2022), https://doi.org/10.3390/
stroke: role of oxidative stress, Curr. Pharm. Des. 26 (34) (2020) 4246–4260, antiox11101870.
https://doi.org/10.2174/1381612826666200708133912. [39] S. Bhatt, A.N. Nagappa, C.R. Patil, Role of oxidative stress in depression, Drug
[18] C. Li, Z. Zhao, Y. Luo, T. Ning, P. Liu, Q. Chen, Y. Chu, Q. Guo, Y. Zhang, W. Zhou, Discov. Today 25 (7) (2020) 1270–1276, https://doi.org/10.1016/j.
H. Chen, Z. Zhou, Y. Wang, B. Su, H. You, T. Zhang, X. Li, H. Song, C. Li, T. Sun, drudis.2020.05.001.
C. Jiang, Macrophage-disguised manganese dioxide nanoparticles for [40] J.Q. Liang, X. Chen, Y. Cheng, Paeoniflorin rescued MK-801-induced
neuroprotection by reducing oxidative stress and modulating inflammatory Schizophrenia-like behaviors in mice via oxidative stress pathway, Front. Nutr. 9
microenvironment in acute Ischemic stroke, Adv. Sci. 8 (20) (2021) e2101526, (2022) 870032, https://doi.org/10.3389/fnut.2022.870032.
https://doi.org/10.1002/advs.202101526. [41] Q.Q. Mao, X.M. Zhong, F.M. Qiu, Z.Y. Li, Z. Huang, Protective effects of
[19] Y. Zhao, X. Zhang, X. Chen, Y. Wei, Neuronal injuries in cerebral infarction and paeoniflorin against corticosterone-induced neurotoxicity in PC12 cells,
Ischemic stroke: from mechanisms to treatment (Review), Int. J. Mol. Med. 49 (2) Phytother. Res. 26 (7) (2012) 969–973, https://doi.org/10.1002/ptr.3673.
(2022), https://doi.org/10.3892/ijmm.2021.5070. [42] Q.Q. Mao, X.M. Zhong, C.R. Feng, A.J. Pan, Z.Y. Li, Z. Huang, Protective effects of
[20] X. Hu, D. Wu, X. He, H. Zhao, Z. He, J. Lin, K. Wang, W. Wang, Z. Pan, H. Lin, paeoniflorin against glutamate-induced neurotoxicity in PC12 cells via
M. Wang, circGSK3beta promotes metastasis in esophageal squamous cell antioxidant mechanisms and Ca(2+) antagonism, Cell Mol. Neurobiol. 30 (7)
carcinoma by augmenting beta-catenin signaling, Mol. Cancer 18 (1) (2019) 160, (2010) 1059–1066, https://doi.org/10.1007/s10571-010-9537-5.
https://doi.org/10.1186/s12943-019-1095-y. [43] D.Z. Mu, M. Xue, J.J. Xu, Y. Hu, Y. Chen, P. Ren, X. Huang, Antidepression and
[21] F. Yang, Y. Li, X. Sheng, Y. Liu, Paeoniflorin treatment regulates TLR4/NF-κB prokinetic effects of Paeoniflorin on rats in the forced swimming test via
signaling, reduces cerebral oxidative stress and improves white matter integrity in polypharmacology, Evid. Based Complement Altern. Med 2020 (2020) 2153571,
neonatal hypoxic brain injury, Korean J. Physiol. Pharmacol. 25 (2) (2021) https://doi.org/10.1155/2020/2153571.
97–109, https://doi.org/10.4196/kjpp.2021.25.2.97. [44] M.D. Pokharel, D.P. Marciano, P. Fu, M.C. Franco, H. Unwalla, K. Tieu, J.
[22] Q. Chen, Y. Liu, Y. Zhang, X. Jiang, Y. Zhang, T. Asakawa, An in vitro verification R. Fineman, T. Wang, S.M. Black, Metabolic reprogramming, oxidative stress, and
of the effects of Paeoniflorin on Lipopolysaccharide-Exposed Microglia, Evid. pulmonary hypertension, Redox Biol. 64 (2023) 102797, https://doi.org/
Based Complement Altern. Med. 2020 (2020) 5801453, https://doi.org/10.1155/ 10.1016/j.redox.2023.102797.
2020/5801453. [45] X. Liu, W. Zhang, J. Luo, W. Shi, X. Zhang, Z. Li, X. Qin, B. Liu, Y. Wei, TRIM21
[23] Z. Zhang, W. Yang, Paeoniflorin protects PC12 cells from oxygen-glucose deficiency protects against atrial inflammation and remodeling post myocardial
deprivation/reoxygenation-induced injury via activating JAK2/STAT3 signaling, infarction by attenuating oxidative stress, Redox Biol. 62 (2023) 102679, https://
Exp. Ther. Med. 21 (6) (2021) 572, https://doi.org/10.3892/etm.2021.10004. doi.org/10.1016/j.redox.2023.102679.
[24] D. Zhang, B. Yang, S.Q. Chang, S.S. Ma, J.X. Sun, L. Yi, X. Li, H.M. Shi, B. Jing, Y. [46] Z.Y. Liu, K. Song, B. Tu, L.C. Lin, H. Sun, Y. Zhou, R. Li, Y. Shi, J.J. Yang,
C. Zheng, C.L. Zhang, F.G. Chen, G.P. Zhao, Protective effect of paeoniflorin on H Y. Zhang, J.Y. Zhao, H. Tao, Crosstalk between oxidative stress and epigenetic
(2)O(2) induced Schwann cells injury based on network pharmacology and marks: new roles and therapeutic implications in cardiac fibrosis, Redox Biol. 65
experimental validation. Chin. J. Nat. Med. 19 (2) (2021) 90–99, https://doi.org/ (2023) 102820, https://doi.org/10.1016/j.redox.2023.102820.
10.1016/s1875-5364(21)60010-9. [47] D. Wang, Y. Yin, S. Wang, T. Zhao, F. Gong, Y. Zhao, B. Wang, Y. Huang,
[25] W. Wu, C. Qiu, X. Feng, X. Tao, Q. Zhu, Z. Chen, X. Ma, J. Yang, X. Bao, Protective Z. Cheng, G. Zhu, Z. Wang, Y. Wang, J. Ren, G. Liang, X. Li, Z. Huang, FGF1
effect of Paeoniflorin on acute cerebral infarction in rats, Curr. Pharm. (DeltaHBS) prevents diabetic cardiomyopathy by maintaining mitochondrial
Biotechnol. 21 (8) (2020) 702–709, https://doi.org/10.2174/ homeostasis and reducing oxidative stress via AMPK/Nur77 suppression. Signal
1389201021666191224151634. Transduct. Target Ther. 6 (1) (2021) 133, https://doi.org/10.1038/s41392-021-
[26] P. Li, Z. Li, Neuroprotective effect of paeoniflorin on H(2)O(2)-induced apoptosis 00542-2.
in PC12 cells by modulation of reactive oxygen species and the inflammatory [48] S. Wang, D. Jia, H. Lu, X. Qu, Paeoniflorin improves myocardial injury via p38
response. Exp. Ther. Med. 9 (5) (2015) 1768–1772, https://doi.org/10.3892/ MAPK/NF-KB p65 inhibition in lipopolysaccharide-induced mouse, Ann. Transl.
etm.2015.2360. Med. 9 (18) (2021) 1449, https://doi.org/10.21037/atm-21-4049.
[27] K. Sun, X. Jing, J. Guo, X. Yao, F. Guo, Mitophagy in degenerative joint diseases, [49] F. Wu, B. Ye, X. Wu, X. Lin, Y. Li, Y. Wu, L. Tong, Paeoniflorin on Rat myocardial
Autophagy 17 (9) (2021) 2082–2092, https://doi.org/10.1080/ Ischemia reperfusion injury of protection and mechanism research, Pharmacology
15548627.2020.1822097. 105 (5-6) (2020) 281–288, https://doi.org/10.1159/000503583.
[28] H. Dong, R. Li, C. Yu, T. Xu, X. Zhang, M. Dong, Paeoniflorin inhibition of 6- [50] J. Li, S.H. Zhang, D. He, J.F. Wang, J.Q. Li, Paeoniflorin reduced the
hydroxydopamine-induced apoptosis in PC12 cells via suppressing reactive cardiotoxicity of aconitine in h9c2 cells, J. Biol. Regul. Homeost. Agents 33 (5)
oxygen species-mediated PKCδ/NF-κB pathway, Neuroscience 285 (2015) 70–80, (2019) 1425–1436, https://doi.org/10.23812/19-257a.
https://doi.org/10.1016/j.neuroscience.2014.11.008. [51] Q. Shao, J. Li, X. Huang, G. Zhou, Protective effects of paeoniflorin on acrolein-
[29] H. Dong, J. Zhang, H. Rong, X. Zhang, M. Dong, Paeoniflorin and Plycyrrhetinic induced apoptosis in H9c2 cardiomyocytes, Pak. J. Pharm. Sci. 33 (4) (2020)
acid synergistically alleviate MPP(+)/MPTP-induced oxidative stress through 1585–1592.
Nrf2-dependent glutathione biosynthesis mechanisms, ACS Chem. Neurosci. 12 [52] S. Ren, Y. Wang, Y. Zhang, P. Yan, D. Xiao, Y. Zhao, W. Jia, L. Ding, H. Dong,
(7) (2021) 1100–1111, https://doi.org/10.1021/acschemneuro.0c00544. C. Wei, S. Lin, Y. Lin, Paeoniflorin alleviates AngII-induced cardiac hypertrophy
[30] Y.M. Wu, R. Jin, L. Yang, J. Zhang, Q. Yang, Y.Y. Guo, X.B. Li, S.B. Liu, X.X. Luo, in H9c2 cells by regulating oxidative stress and Nrf2 signaling pathway, Biomed.
M.G. Zhao, Phosphatidylinositol 3 kinase/protein kinase B is responsible for the Pharmacother. 165 (2023) 115253, https://doi.org/10.1016/j.
protection of paeoniflorin upon H₂O₂-induced neural progenitor cell injury. biopha.2023.115253.
Neuroscience 240 (2013) 54–62, https://doi.org/10.1016/j. [53] J.Z. Li, X.N. Tang, T.T. Li, L.J. Liu, S.Y. Yu, G.Y. Zhou, Q.R. Shao, H.P. Sun, C. Wu,
neuroscience.2013.02.037. Y. Yang, Paeoniflorin inhibits doxorubicin-induced cardiomyocyte apoptosis by
[31] D. Wang, L. Liu, S. Li, C. Wang, Effects of paeoniflorin on neurobehavior, downregulating microRNA-1 expression, Exp. Ther. Med. 11 (6) (2016)
oxidative stress, brain insulin signaling, and synaptic alterations in 2407–2412, https://doi.org/10.3892/etm.2016.3182.
intracerebroventricular streptozotocin-induced cognitive impairment in mice, [54] J.Z. Li, S.Y. Yu, J.H. Wu, Q.R. Shao, X.M. Dong, Paeoniflorin protects myocardial
Physiol. Behav. 191 (2018) 12–20, https://doi.org/10.1016/j. cell from doxorubicin-induced apoptosis through inhibition of NADPH oxidase,
physbeh.2018.03.016. Can. J. Physiol. Pharmacol. 90 (12) (2012) 1569–1575, https://doi.org/10.1139/
[32] K. Wang, L. Zhu, X. Zhu, K. Zhang, B. Huang, J. Zhang, Y. Zhang, L. Zhu, B. Zhou, y2012-140.
F. Zhou, Protective effect of paeoniflorin on Aβ25-35-induced SH-SY5Y cell injury [55] Q. Zhang, J. Liu, H. Duan, R. Li, W. Peng, C. Wu, Activation of Nrf2/HO-1
by preventing mitochondrial dysfunction, Cell Mol. Neurobiol. 34 (2) (2014) signaling: an important molecular mechanism of herbal medicine in the treatment
227–234, https://doi.org/10.1007/s10571-013-0006-9. of atherosclerosis via the protection of vascular endothelial cells from oxidative
stress, J. Adv. Res. 34 (2021) 43–63, https://doi.org/10.1016/j.jare.2021.06.023.

10
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

[56] X. Fan, J. Wu, H. Yang, L. Yan, S. Wang, Paeoniflorin blocks the proliferation of colonic carcinogenesis, Pharmacol. Res 165 (2021) 105420, https://doi.org/
vascular smooth muscle cells induced by platelet‑derived growth factor‑BB 10.1016/j.phrs.2021.105420.
through ROS mediated ERK1/2 and p38 signaling pathways. Mol. Med. Rep. 17 [76] J. Wen, B. Xu, Y. Sun, M. Lian, Y. Li, Y. Lin, D. Chen, Y. Diao, M. Almoiliqy,
(1) (2018) 1676–1682, https://doi.org/10.3892/mmr.2017.8093. L. Wang, Paeoniflorin protects against intestinal ischemia/reperfusion by
[57] S. Song, X. Xiao, D. Guo, L. Mo, C. Bu, W. Ye, Q. Den, S. Liu, X. Yang, Protective activating LKB1/AMPK and promoting autophagy, Pharmacol. Res 146 (2019)
effects of Paeoniflorin against AOPP-induced oxidative injury in HUVECs by 104308, https://doi.org/10.1016/j.phrs.2019.104308.
blocking the ROS-HIF-1α/VEGF pathway, Phytomedicine 34 (2017) 115–126, [77] X.X. Wu, X.L. Huang, R.R. Chen, T. Li, H.J. Ye, W. Xie, Z.M. Huang, G.Z. Cao,
https://doi.org/10.1016/j.phymed.2017.08.010. Paeoniflorin prevents intestinal barrier disruption and inhibits lipopolysaccharide
[58] T. Chen, Z.P. Guo, X.Y. Jiao, Y.H. Zhang, J.Y. Li, H.J. Liu, Protective effects of (LPS)-induced inflammation in Caco-2 cell monolayers, Inflammation 42 (6)
peoniflorin against hydrogen peroxide-induced oxidative stress in human (2019) 2215–2225, https://doi.org/10.1007/s10753-019-01085-z.
umbilical vein endothelial cells, Can. J. Physiol. Pharmacol. 89 (6) (2011) [78] K. Shen, X. Wang, Y. Wang, Y. Jia, Y. Zhang, K. Wang, L. Luo, W. Cai, J. Li, S. Li,
445–453, https://doi.org/10.1139/y11-034. Y. Du, L. Zhang, H. Zhang, Y. Chen, C. Xu, J. Zhang, R. Wang, X. Yang, Y. Wang,
[59] J. Ye, H. Duan, X. Yang, W. Yan, X. Zheng, Anti-thrombosis effect of paeoniflorin: D. Hu, miR-125b-5p in adipose derived stem cells exosome alleviates pulmonary
evaluated in a photochemical reaction thrombosis model in vivo, Planta Med. 67 microvascular endothelial cells ferroptosis via Keap1/Nrf2/GPX4 in sepsis lung
(8) (2001) 766–767, https://doi.org/10.1055/s-2001-18364. injury, Redox Biol. 62 (2023) 102655, https://doi.org/10.1016/j.
[60] J. Duan, J.D. Matute, L.W. Unger, T. Hanley, A. Schnell, X. Lin, N. Krupka, redox.2023.102655.
P. Griebel, C. Lambden, B. Sit, J. Grootjans, M. Pyzik, F. Sommer, S. Kaiser, [79] J. Yu, X. Zhu, X. Qi, J. Che, B. Cao, Paeoniflorin protects human EA.hy926
M. Falk-Paulsen, H. Grasberger, J.Y. Kao, T. Fuhrer, H. Li, D. Paik, Y. Lee, endothelial cells against gamma-radiation induced oxidative injury by activating
S. Refetoff, J.N. Glickman, A.W. Paton, L. Bry, J.C. Paton, U. Sauer, A. the NF-E2-related factor 2/heme oxygenase-1 pathway, Toxicol. Lett. 218 (3)
J. Macpherson, P. Rosenstiel, V.K. Kuchroo, M.K. Waldor, J.R. Huh, A. Kaser, R. (2013) 224–234, https://doi.org/10.1016/j.toxlet.2013.01.028.
S. Blumberg, Endoplasmic reticulum stress in the intestinal epithelium initiates [80] Y. Hu, W. Yang, Paeoniflorin can improve acute lung injury caused by severe
purine metabolite synthesis and promotes Th17 cell differentiation in the gut, e9, acute pancreatitis through Nrf2/ARE pathway, Comput. Math. Methods Med
Immunity 56 (5) (2023) 1115–1131, https://doi.org/10.1016/j. 2022 (2022) 5712219, https://doi.org/10.1155/2022/5712219.
immuni.2023.02.018. [81] Q. Shou, L. Jin, J. Lang, Q. Shan, Z. Ni, C. Cheng, Q. Li, H. Fu, G. Cao, Integration
[61] M.J. Rodriguez, M. Sabaj, G. Tolosa, F. Herrera Vielma, M.J. Zuniga, D. of metabolomics and transcriptomics reveals the therapeutic mechanism
R. Gonzalez, J. Zuniga-Hernandez, Maresin-1 prevents liver fibrosis by targeting underlying Paeoniflorin for the treatment of allergic asthma, Front. Pharmacol. 9
Nrf2 and NF-kappaB, reducing oxidative stress and inflammation, Cells 10 (12) (2018) 1531, https://doi.org/10.3389/fphar.2018.01531.
(2021), https://doi.org/10.3390/cells10123406. [82] X. Han, S. Hu, Q. Yang, X. Sang, D. Tang, G. Cao, Paeoniflorin ameliorates airway
[62] D. Kong, Z. Zhang, L. Chen, W. Huang, F. Zhang, L. Wang, Y. Wang, P. Cao, inflammation and immune response in ovalbumin induced asthmatic mice: from
S. Zheng, Curcumin blunts epithelial-mesenchymal transition of hepatocytes to oxidative stress to autophagy, Phytomedicine 96 (2022) 153835, https://doi.org/
alleviate hepatic fibrosis through regulating oxidative stress and autophagy, 10.1016/j.phymed.2021.153835.
Redox Biol. 36 (2020) 101600, https://doi.org/10.1016/j.redox.2020.101600. [83] J. Lin, F. Xu, G. Wang, L. Kong, Q. Luo, Y. Lv, J. Liu, Y. Wei, L. Li, H. Zhang,
[63] N. Roehlen, E. Crouchet, T.F. Baumert, Liver fibrosis: mechanistic concepts and J. Dong, Paeoniflorin attenuated oxidative stress in rat COPD model induced by
therapeutic perspectives, Cells 9 (4) (2020), https://doi.org/10.3390/ cigarette smoke, Evid. Based Complement Altern. Med. 2016 (2016) 1698379,
cells9040875. https://doi.org/10.1155/2016/1698379.
[64] J. Li, T. Wang, P. Liu, F. Yang, X. Wang, W. Zheng, W. Sun, Hesperetin [84] S.K. Masenga, L.S. Kabwe, M. Chakulya, A. Kirabo, Mechanisms of oxidative
ameliorates hepatic oxidative stress and inflammation via the PI3K/AKT-Nrf2- stress in metabolic syndrome, Int. J. Mol. Sci. 24 (9) (2023), https://doi.org/
ARE pathway in oleic acid-induced HepG2 cells and a rat model of high-fat diet- 10.3390/ijms24097898.
induced NAFLD, Food Funct. 12 (9) (2021) 3898–3918, https://doi.org/10.1039/ [85] Y. Liu, J. Han, Z. Zhou, D. Li, Paeoniflorin protects pancreatic β cells from STZ-
d0fo02736g. induced damage through inhibition of the p38 MAPK and JNK signaling
[65] Y.E. Tao, Z. Wen, Y. Song, H. Wang, Paeoniflorin attenuates hepatic ischemia/ pathways, Eur. J. Pharmacol. 853 (2019) 18–24, https://doi.org/10.1016/j.
reperfusion injury via anti-oxidative, anti-inflammatory and anti-apoptotic ejphar.2019.03.025.
pathways, Exp. Ther. Med. 11 (1) (2016) 263–268, https://doi.org/10.3892/ [86] X. Yang, X. Li, Y. Zhu, Y. Gao, L. Xu, Paeoniflorin upregulates mitochondrial
etm.2015.2902. Thioredoxin of Schwann cells to improve diabetic peripheral neuropathy
[66] H. Hu, Q. Zhu, J. Su, Y. Wu, Y. Zhu, Y. Wang, H. Fang, M. Pang, B. Li, S. Chen, indicated by 4D label-free quantitative proteomics, Oxid. Med. Cell Longev. 2022
G. Lv, Effects of an enriched extract of Paeoniflorin, a monoterpene glycoside (2022) 4775645, https://doi.org/10.1155/2022/4775645.
used in chinese herbal medicine, on cholesterol metabolism in a hyperlipidemic [87] X. Yang, W. Yao, H. Shi, H. Liu, Y. Li, Y. Gao, R. Liu, L. Xu, Paeoniflorin protects
rat model, Med. Sci. Monit. 23 (2017) 3412–3427, https://doi.org/10.12659/ Schwann cells against high glucose induced oxidative injury by activating Nrf2/
msm.905544. ARE pathway and inhibiting apoptosis, J. Ethnopharmacol. 185 (2016) 361–369,
[67] Z. Ma, L. Chu, H. Liu, W. Wang, J. Li, W. Yao, J. Yi, Y. Gao, Beneficial effects of https://doi.org/10.1016/j.jep.2016.03.031.
paeoniflorin on non-alcoholic fatty liver disease induced by high-fat diet in rats, [88] X. Sun, X. Wang, Z. Zhao, J. Chen, C. Li, G. Zhao, Paeoniflorin accelerates foot
Sci. Rep. 7 (2017) 44819, https://doi.org/10.1038/srep44819. wound healing in diabetic rats though activating the Nrf2 pathway, Acta
[68] T. Wang, X. Zhou, G. Kuang, R. Jiang, X. Guo, S. Wu, J. Wan, L. Yin, Paeoniflorin Histochem. 122 (8) (2020) 151649, https://doi.org/10.1016/j.
modulates oxidative stress, inflammation and hepatic stellate cells activation to acthis.2020.151649.
alleviate CCl4-induced hepatic fibrosis by upregulation of heme oxygenase-1 in [89] H. Wu, P. Zhang, J. Zhou, S. Hu, J. Hao, Z. Zhong, H. Yu, J. Yang, J. Chi, H. Guo,
mice, J. Pharm. Pharmacol. 73 (3) (2021) 338–346, https://doi.org/10.1093/ Paeoniflorin confers ferroptosis resistance by regulating the gut microbiota and
jpp/rgaa042. its metabolites in diabetic cardiomyopathy, Am. J. Physiol. Cell Physiol. 326 (3)
[69] I.D. Kim, B.J. Ha, The effects of paeoniflorin on LPS-induced liver inflammatory (2024) C724–C741, https://doi.org/10.1152/ajpcell.00565.2023.
reactions, Arch. Pharm. Res 33 (6) (2010) 959–966, https://doi.org/10.1007/ [90] K. Zeng, W. Xi, Y. Qiao, X. Huang, X. Liu, Paeoniflorin inhibits epithelial
s12272-010-0620-8. mesenchymal transformation and oxidative damage of lens epithelial cells in
[70] L. Li, H. Wang, S. Zhao, Y. Zhao, Y. Chen, J. Zhang, C. Wang, N. Sun, H. Fan, diabetic cataract via sirtuin 1 upregulation, Bioengineered 13 (3) (2022)
Paeoniflorin ameliorates lipopolysaccharide-induced acute liver injury by 5903–5914, https://doi.org/10.1080/21655979.2021.2018534.
inhibiting oxidative stress and inflammation via SIRT1/FOXO1a/SOD2 signaling [91] E.M. Choi, Y.S. Lee, Paeoniflorin isolated from Paeonia lactiflora attenuates
in rats, Phytother. Res. (2022), https://doi.org/10.1002/ptr.7471. osteoblast cytotoxicity induced by antimycin A, Food Funct. 4 (9) (2013)
[71] M.M. Ommati, H. Niknahad, A. Najibi, A. Arjmand, S. Alidaee, S. Mazloomi, 1332–1338, https://doi.org/10.1039/c3fo60147a.
P. Ahmadi, A. Ghiasvand, M. Javadi, J. Yazdani, S. Sabouri, H. Rezaei, [92] E.M. Choi, K.S. Suh, S.Y. Rhee, Y.S. Kim, Inhibitory effect of paeoniflorin on
N. Azarpira, R. Heidari, Cholestasis-associated pulmonary inflammation, methylglyoxal-mediated oxidative stress in osteoblastic MC3T3-E1 cells,
oxidative stress, and tissue fibrosis: the protective role of the biogenic amine Phytomedicine 21 (10) (2014) 1170–1177, https://doi.org/10.1016/j.
agmatine, Pharmacology 108 (4) (2023) 379–393, https://doi.org/10.1159/ phymed.2014.05.008.
000530307. [93] M.H. Zhang, L. Feng, M.M. Zhu, J.F. Gu, C. Wu, X.B. Jia, Antioxidative and anti-
[72] Z. Chen, X. Ma, Y. Zhu, Y. Zhao, J. Wang, R. Li, C. Chen, S. Wei, W. Jiao, Y. Zhang, inflammatory activities of paeoniflorin and oxypaeoniflora on AGEs-induced
J. Li, L. Wang, R. Wang, H. Liu, H. Shen, X. Xiao, Paeoniflorin ameliorates ANIT- mesangial cell damage, Planta Med. 79 (14) (2013) 1319–1323, https://doi.org/
induced cholestasis by activating Nrf2 through an PI3K/Akt-dependent pathway 10.1055/s-0033-1350649.
in rats, Phytother. Res. 29 (11) (2015) 1768–1775, https://doi.org/10.1002/ [94] J.S. Wang, Y. Huang, S. Zhang, H.J. Yin, L. Zhang, Y.H. Zhang, Y.W. Song, D.D. Li,
ptr.5431. A Protective role of Paeoniflorin in fluctuant hyperglycemia-induced vascular
[73] Y. Zhao, G. Zhou, J. Wang, L. Jia, P. Zhang, R. Li, L. Shan, B. Liu, X. Song, S. Liu, endothelial injuries through antioxidative and anti-inflammatory effects and
X. Xiao, Paeoniflorin protects against ANIT-induced cholestasis by ameliorating reduction of PKCβ1, Oxid. Med. Cell Longev. 2019 (2019) 5647219, https://doi.
oxidative stress in rats, Food Chem. Toxicol. 58 (2013) 242–248, https://doi.org/ org/10.1155/2019/5647219.
10.1016/j.fct.2013.04.030. [95] C.R. Li, Z. Zhou, D. Zhu, Y.N. Sun, J.M. Dai, S.Q. Wang, Protective effect of
[74] L. Mao, J. Chen, K. Cheng, Z. Dou, J.D. Leavenworth, H. Yang, D. Xu, L. Luo, Nrf2- paeoniflorin on irradiation-induced cell damage involved in modulation of
dependent protective effect of paeoniflorin on alpha-naphthalene isothiocyanate- reactive oxygen species and the mitogen-activated protein kinases, Int. J.
induced hepatic injury, Am. J. Chin. Med. 50 (5) (2022) 1331–1348, https://doi. Biochem. Cell Biol. 39 (2) (2007) 426–438, https://doi.org/10.1016/j.
org/10.1142/S0192415X22500562. biocel.2006.09.011.
[75] P. Liu, Y. Wang, G. Yang, Q. Zhang, L. Meng, Y. Xin, X. Jiang, The role of short- [96] L. Huang, S. Hu, M. Shao, X. Wu, J. Zhang, G. Cao, Combined Cornus officinalis
chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and and Paeonia lactiflora pall therapy alleviates rheumatoid arthritis by regulating

11
Y. Lu et al. Biomedicine & Pharmacotherapy 176 (2024) 116772

synovial apoptosis via AMPK-mediated mitochondrial fission, Front. Pharmacol. inhibited by Paeoniflorin/Nrf2 signaling or PLIN2, Front. Pharmacol. 11 (2020)
12 (2021) 639009, https://doi.org/10.3389/fphar.2021.639009. 736, https://doi.org/10.3389/fphar.2020.00736.
[97] Z. Jia, J. He, Paeoniflorin ameliorates rheumatoid arthritis in rat models through [115] J. Yuan, Y. Lu, H. Wang, Y. Feng, S. Jiang, X.H. Gao, R. Qi, Y. Wu, H.D. Chen,
oxidative stress, inflammation and cyclooxygenase 2, Exp. Ther. Med. 11 (2) Paeoniflorin resists H(2)O(2)-induced oxidative stress in melanocytes by JNK/
(2016) 655–659, https://doi.org/10.3892/etm.2015.2908. Nrf2/HO-1 pathway. Front. Pharmacol. 11 (2020) 536, https://doi.org/10.3389/
[98] H. Tsuboi, K. Hossain, A.A. Akhand, K. Takeda, J. Du, M. Rifa’i, Y. Dai, fphar.2020.00536.
A. Hayakawa, H. Suzuki, I. Nakashima, Paeoniflorin induces apoptosis of [116] Y. Xu, H. He, P. Li, H. Liu, Paeoniflorin inhibits proliferation and promotes
lymphocytes through a redox-linked mechanism, J. Cell Biochem. 93 (1) (2004) autophagy and apoptosis of sweat gland cells, Exp. Ther. Med. 23 (1) (2022) 53,
162–172, https://doi.org/10.1002/jcb.20134. https://doi.org/10.3892/etm.2021.10975.
[99] E.H. Joo, Y.R. Kim, N. Kim, J.E. Jung, S.H. Han, H.Y. Cho, Effect of endogenic and [117] Y. Lai, N. Yang, X. Chen, X. Ma, Z. Chen, C. Dong, G. Yu, Y. Huang, D. Shi, P. Fang,
exogenic oxidative stress triggers on adverse pregnancy outcomes: Preeclampsia, K. Fu, R. Jiang, C. Mao, J. Ding, W. Gao, Dihydrocapsaicin suppresses the STING-
fetal growth restriction, gestational diabetes mellitus and preterm birth, Int. J. mediated accumulation of ROS and NLRP3 inflammasome and alleviates
Mol. Sci. 22 (18) (2021), https://doi.org/10.3390/ijms221810122. apoptosis after ischemia-reperfusion injury of perforator skin flap, Phytother. Res.
[100] J. Wu, D. Zhang, L. Hu, X. Zheng, C. Chen, Paeoniflorin alleviates NG-nitro-L- (2024), https://doi.org/10.1002/ptr.8167.
arginine methyl ester (L-NAME)-induced gestational hypertension and [118] J. Jiang, C. Dong, L. Zhai, J. Lou, J. Jin, S. Cheng, Z. Chen, X. Guo, D. Lin, J. Ding,
upregulates silent information regulator 2 related enzyme 1 (SIRT1) to reduce H W. Gao, Paeoniflorin suppresses TBHP-induced oxidative stress and apoptosis in
(2)O(2)-induced endothelial cell damage, Bioengineered 13 (2) (2022) human umbilical vein endothelial cells via the Nrf2/HO-1 signaling pathway and
2248–2258, https://doi.org/10.1080/21655979.2021.2024325. improves skin flap survival, Front. Pharmacol. 12 (2021) 735530, https://doi.
[101] P. Bayram, S.A. Karamese, H.S. Erol, B. Ozdemir, E. Toktay, C. Salum, Protective org/10.3389/fphar.2021.735530.
effects of a natural product, paeoniflorin, on ischemia reperfusion injury on rat [119] W.J. Li, Y.Y. Liu, J.B. He, X.Y. Ma, Y. Lin, P. Zheng, D.S. Lin, Effect of paeoniflorin
ovary tissue: histopathological, immunohistochemical, and biochemical study, on distal survival of random flaps, Int. Immunopharmacol. 105 (2022) 108562,
J. Histotechnol. 46 (4) (2023) 170–183, https://doi.org/10.1080/ https://doi.org/10.1016/j.intimp.2022.108562.
01478885.2023.2227409. [120] Y. Wu, X. Duan, Z. Gao, N. Yang, F. Xue, AICAR attenuates postoperative
[102] Q. Guo, S. Li, X. Wang, H.S. Han, X.J. Yin, J.C. Li, Paeoniflorin improves the in abdominal adhesion formation by inhibiting oxidative stress and promoting
vitro maturation of benzo(a)pyrene treated porcine oocytes via effects on the mesothelial cell repair, PLoS One 17 (9) (2022) e0272928, https://doi.org/
sonic hedgehog pathway, Theriogenology 180 (2022) 72–81, https://doi.org/ 10.1371/journal.pone.0272928.
10.1016/j.theriogenology.2021.12.016. [121] Q. Gao, G. Wei, Y. Wu, N. Yao, C. Zhou, K. Wang, K. Wang, X. Sun, X. Li,
[103] X. Wang, J.C. Hao, B. Shang, K.L. Yang, X.Z. He, Z.L. Wang, H.L. Jing, Y.J. Cao, Paeoniflorin prevents postoperative peritoneal adhesion formation in an
Paeoniflorin ameliorates oxidase stress in Glutamate-stimulated SY5Y and experimental rat model, Oncotarget 8 (55) (2017) 93899–93911, https://doi.org/
prenatally stressed female offspring through Nrf2/HO-1 signaling pathway, 10.18632/oncotarget.21333.
J. Affect Disord. 294 (2021) 189–199, https://doi.org/10.1016/j. [122] J. Li, C. Zhu, Z. Zhang, X. Zheng, C. Wang, H. Zhang, Paeoniflorin increases the
jad.2021.07.054. anti-tumor efficacy of sorafenib in tumor-bearing mice with liver cancer via
[104] X. Wankun, Y. Wenzhen, Z. Min, Z. Weiyan, C. Huan, D. Wei, H. Lvzhen, Y. Xu, suppressing the NF-kappab/PD-l1 axis. Heliyon 10 (2) (2024) e24461 https://doi.
L. Xiaoxin, Protective effect of paeoniflorin against oxidative stress in human org/10.1016/j.heliyon.2024.e24461.
retinal pigment epithelium in vitro, Mol. Vis. 17 (2011) 3512–3522. [123] T.W. Kim, Paeoniflorin induces ER stress-mediated apoptotic cell death by
[105] X. Zhu, K. Wang, F. Zhou, L. Zhu, Paeoniflorin attenuates atRAL-induced generating Nox4-derived ROS under radiation in Gastric cancer, Nutrients 15 (24)
oxidative stress, mitochondrial dysfunction and endoplasmic reticulum stress in (2023), https://doi.org/10.3390/nu15245092.
retinal pigment epithelial cells via triggering Ca(2+)/CaMKII-dependent [124] A. Bagheri, G. Radman, N. Aria, F. Rezaei, M. Khajenouri, S. Ghiabi, Y. Bagheri,
activation of AMPK. Arch. Pharm. Res. 41 (10) (2018) 1009–1018, https://doi. The effects of Quercetin on apoptosis and antioxidant activity in a Renal
org/10.1007/s12272-018-1059-6. Ischemia/Reperfusion injury animal model, Drug Res. 73 (5) (2023) 255–262,
[106] Y. He, Z. Zheng, C. Liu, W. Li, L. Zhao, G. Nie, H. Li, Inhibiting DNA methylation https://doi.org/10.1055/a-1999-7600.
alleviates cisplatin-induced hearing loss by decreasing oxidative stress-induced [125] L. Guo, X. Zhang, N. Lv, L. Wang, J. Gan, X. Jiang, Y. Wang, Therapeutic role and
mitochondria-dependent apoptosis via the LRP1-PI3K/AKT pathway, Acta Pharm. potential mechanism of resveratrol in atherosclerosis: TLR4/NF-kappaB/HIF-
Sin. B 12 (3) (2022) 1305–1321, https://doi.org/10.1016/j.apsb.2021.11.002. 1alpha, Mediat. Inflamm. 2023 (2023) 1097706, https://doi.org/10.1155/2023/
[107] X. Yu, R. Man, Y. Li, Q. Yang, H. Li, H. Yang, X. Bai, H. Yin, J. Li, H. Wang, 1097706.
Paeoniflorin protects spiral ganglion neurons from cisplatin-induced ototoxicity: [126] J. Xu, P. Du, X. Liu, X. Xu, Y. Ge, C. Zhang, Curcumin supplementation increases
possible relation to PINK1/BAD pathway, J. Cell Mol. Med. 23 (8) (2019) longevity and antioxidant capacity in Caenorhabditis elegans, Front. Pharmacol.
5098–5107, https://doi.org/10.1111/jcmm.14379. 14 (2023) 1195490, https://doi.org/10.3389/fphar.2023.1195490.
[108] X. Yu, Z. Fan, Y. Han, D. Zhang, L. Xu, M. Wang, Q. Yang, H. Li, M. Zhou, [127] X. Han, Y. Fu, K. Wang, S. Li, C. Jiang, S. Wang, Z. Wang, G. Liu, S. Hu,
L. Zhang, G. Sun, X. Bai, J. Li, H. Wang, Paeoniflorin reduces neomycin-induced Epigallocatechin gallate alleviates osteoporosis by regulating the gut microbiota
ototoxicity in hair cells by suppression of reactive oxygen species generation and and serum metabolites in rats, Food Funct. 14 (23) (2023) 10564–10580, https://
extracellularly regulated kinase signalization, Toxicol. Lett. 285 (2018) 9–19, doi.org/10.1039/d3fo03233g.
https://doi.org/10.1016/j.toxlet.2017.12.026. [128] Y. Wu, Y. Jiang, L. Zhang, J. Zhou, Y. Yu, Y. Zhou, T. Kang, Chemical profiling
[109] T. Higashihara, H. Nishi, K. Takemura, H. Watanabe, T. Maruyama, R. Inagi, and antioxidant evaluation of Paeonia lactiflora Pall. "Zhongjiang" by HPLC-ESI-
T. Tanaka, M. Nangaku, beta2-adrenergic receptor agonist counteracts skeletal MS combined with DPPH assay, J. Chromatogr. Sci. 59 (9) (2021) 795–805,
muscle atrophy and oxidative stress in uremic mice, Sci. Rep. 11 (1) (2021) 9130, https://doi.org/10.1093/chromsci/bmab005.
https://doi.org/10.1038/s41598-021-88438-7. [129] J. Qian, W. Cheng, C. Zhang, L. Hong, W. Chen, G. Li, Preparation,
[110] Q. Li, J. Wu, J. Huang, R. Hu, H. You, L. Liu, D. Wang, L. Wei, Paeoniflorin physicochemical characterization and pharmacokinetics of paeoniflorin-
ameliorates skeletal muscle atrophy in chronic kidney disease via AMPK/SIRT1/ phospholipid complex, Biomed. Mater. Eng. 30 (1) (2019) 11–22, https://doi.
PGC-1α-mediated oxidative stress and mitochondrial dysfunction, Front. org/10.3233/BME-181029.
Pharmacol. 13 (2022) 859723, https://doi.org/10.3389/fphar.2022.859723. [130] M. Ge, L. Wu, F. He, Y. Tai, R. Fang, D. Han, P. Guo, H. Liu, Y. Hu, S. Xu, W. Wei,
[111] D. Xing, Y. Ma, M. Lu, W. Liu, H. Zhou, Paeoniflorin alleviates hypoxia/ Q. Wang, CP-25 inhibits the hyperactivation of rheumatic synoviocytes by
reoxygenation injury in HK-2 cells by inhibiting apoptosis and repressing suppressing the switch in G(alphas)-G(alphai) coupling to the beta(2)-adrenergic
oxidative damage via Keap1/Nrf2/HO-1 pathway, BMC Nephrol. 24 (1) (2023) receptor, Cell Commun. Signal 21 (1) (2023) 346, https://doi.org/10.1186/
314, https://doi.org/10.1186/s12882-023-03366-0. s12964-023-01358-z.
[112] S. Zhu, X. Li, B. Dang, F. Wu, K. Gou, C. Wang, C. Lin, Hydrogen sulfide protects [131] J.L. Shu, X.Z. Zhang, L. Han, F. Zhang, Y.J. Wu, X.Y. Tang, C. Wang, Y. Tai, Q.
retina from blue light-induced photodamage and degeneration via inhibiting T. Wang, J.Y. Chen, Y. Chang, H.X. Wu, L.L. Zhang, W. Wei, Paeoniflorin-6’-O-
ROS-mediated ER stress-CHOP apoptosis signal, Redox Rep. 27 (1) (2022) benzene sulfonate alleviates collagen-induced arthritis in mice by downregulating
100–110, https://doi.org/10.1080/13510002.2022.2069534. BAFF-TRAF2-NF-kappaB signaling: comparison with biological agents. Acta
[113] L. Kong, S. Wang, X. Wu, F. Zuo, H. Qin, J. Wu, Paeoniflorin attenuates ultraviolet Pharmacol. Sin. 40 (6) (2019) 801–813, https://doi.org/10.1038/s41401-018-
B-induced apoptosis in human keratinocytes by inhibiting the ROS-p38-p53 0169-5.
pathway, Mol. Med. Rep. 13 (4) (2016) 3553–3558, https://doi.org/10.3892/ [132] W. Peng, Y. Chen, S. Tumilty, L. Liu, L. Luo, H. Yin, Y. Xie, Paeoniflorin is a
mmr.2016.4953. promising natural monomer for neurodegenerative diseases via modulation of Ca
[114] Y.S. Lu, Y. Jiang, J.P. Yuan, S.B. Jiang, Y. Yang, P.Y. Zhu, Y.Z. Sun, R.Q. Qi, T. Liu, (2+) and ROS homeostasis. Curr. Opin. Pharmacol. 62 (2022) 97–102, https://
H.X. Wang, Y. Wu, X.H. Gao, H.D. Chen, UVA induced oxidative stress was doi.org/10.1016/j.coph.2021.11.009.

12

You might also like