Open Access

Insight into the immunomodulatory and chemotherapeutic mechanisms of paeonol (Review)

  • Authors:
    • Precious Barnes
    • Elvis Agbo
    • Faustina Halm‑Lai
    • Kwabena Dankwa
    • Roland Osei Saahene
    • Samuel Victor Nuvor
    • Dorcas Obiri‑Yeboah
    • Ewura Seidu Yahaya
  • View Affiliations

  • Published online on: February 26, 2025     https://doi.org/10.3892/mi.2025.223
  • Article Number: 24
  • Copyright : © Barnes et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Paeonol a, pharmacologically active constituent obtained from the root bark of Paeonia suffruticosa has been extensively utilized as a traditional Chinese medicine for the treatment, prevention and control of several diseases for years. Paeonol has been reported to possess key immunomodulatory properties; however, the underlying mechanisms involved in its immunomodulatory and anticancer effects have not been extensively researched due to limitations in terms of design, conduct and interpretation. The present review focuses on both the in vitro and in vivo immunosuppressive and anticancer effects of paeonol and the underlying mechanisms of action. The present literature review aimed to include all the notable findings published on Google Scholar, PubMed, Web of Science, SciFinder and ScienceDirect. Overall, paeonol possesses multifaceted pharmacological activities with potential for use in the development of novel immunomodulator and anticancer therapeutic agents. Paeonol decreases IL‑1β expression to repress several inflammatory mediators, such as NO, iNOS, COX2 and PEG2 in the inhibition of the NLRP3 inflammasome, NF‑κB, MAPK and TLR4 pathways to provide multiple levels immunosuppression; these effects may be beneficial in immune‑related diseases. Furthermore, paeonol inhibits cancer cell growth, proliferation, invasion and metastasis by inducing cell apoptosis and the suppression of the TLR4/NF‑κB/STAT3/MAPK/PI3K/AKT/CHOP/VEGF/HIF‑1α, pathways. The present review aimed to promote further research to exploit the potential use of paeonol as a novel therapeutic agent for immunomodulation and cancer management.

1. Introduction

The immune system is comprised of white blood cells (monocytes, macrophages, dendritic cells, neutrophils, eosinophils, basophils and lymphocytes) and special immune molecules (cytokines, antibodies, complement proteins and signaling pathways) that have evolved to resist infections (1). The immune system is classified into two broad categories: The innate and adaptive immune system, which collaborate to protect the body against foreign invaders and aid in the maintenance of the normal homeostasis of the body. Innate immunity is a non-specific immune system which provides immediate initial defense against infections (2), while adaptive immunity is a specific immune system which develops more slowly and provides specific and effective defense against invading pathogens (3). Several intrinsic and extrinsic agents can affect the functioning of the immune system, rendering it either immunostimulatory or immunosuppressive (3). Immunomodulation is the alteration of the responsiveness of the body's immune system caused by a chemical substance that can suppress or activate its function in the fight against diseases (4). A malfunctioning or imbalanced immune system can result in the development of a variety of chronic diseases, such as cancer, autoimmune disorders, allergies, viral infections, inflammatory bowel diseases and numerous others (5). Immunomodulators are bioactive agents that can regulate immunological events, such as stimulating phagocytes, natural killer cell, and activating T- and B-cells (6). The organic entity of natural or artificial sources with the characteristics of inhibiting, stimulating or modulating the constituents of either non-specific or specific immunity are also referred to as immune modulators or biological response modifiers. Generally, in medical practice, these sources are grouped as immunosuppressant, immunostimulant and immunoadjuvant (7,8). Advancements in immunological research indicate that the suppression of the immune system due to stressful environmental conditions can result in the development of a variety of diseases (9). Therefore, it is evident that immunomodulators can serve as alternative therapies for the management of various diseases, such as activating immune responsiveness impairment by HIV (10,11) or suppressing it during transplantation (12). Additionally, the application of these modulators in cancer treatment is crucial due to their central role in supporting immune responses (13). As biological response modifiers, immunomodulatory agents inhibit tumor cells directly or by enhancing host defense mechanism such as induction of apoptosis against the tumor (14). The immunological effects of immunomodulators can be evaluated by their specified actions on immune cells, effector mechanisms, inhibition of nitric oxide (NO) and reactive oxygen species (ROS) production, the secretion of inflammatory cytokines, signaling pathways in macrophage cells and phagocytosis activity (15).

Cortex moutan, the root bark of Paeonia suffruticosa has been extensively utilized as a traditional Chinese medicine for the prevention and control of several diseases for years with a good safety and efficacy profile (16). Paeonol is a pharmacologically effective phenolic constituent isolated from Cortex Moutan. Paeonol has been reported to possess extensive therapeutic effects on the innate and adaptive immune system in several human diseases (17-21) and cancers (22-24), which are associated with inhibition of immune signaling pathways (25-27), NO (21) and ROS production (28), inflammatory cytokines (29,30), immune cells (20,31), phagocytes (17,32) and the induction of apoptosis (14,25,33). Paeonol has been applied together with other drugs to effectively protect against oropharyngeal candidiasis (34), hepatotoxicity (35) and cardiotoxicity (36), indicating that paeonol can potentially act as an alternative or complementary medicine to offset deficiencies and unresolved safety of current drugs for treatment of diseases. In the light of the therapeutic prospects and pharmacological activities of paeonol, the present review provides an overview of the underlying mechanisms of paeonol in immunomodulation and chemoprevention.

2. Literature search methods

To assess the existing studies on the immunomodulatory and chemotherapeutic potentials of paeonol for a wide range of human diseases, the Pubmed, Medline, Google Scholar and Web of Science, SciFinder and Science Direct databases were used to search for literature published from 2013 up to 2024. Paeonol and immunomodulation or cancer were the key words used for the search. All relevant full-text research articles with titles/abstract were incorporated without language restrictions. Citations from selected publications were tracked and potential papers added.

3. Botanical characteristics of Paeonia suffruticosa

Peonies mainly thrive well in northwest Africa, temperate Eurasia and western North America. Nevertheless, wild peony is endemic to China, where it was initially domesticated (37). It is a perennial shrub with brown-gray stems and grows to a height of 1.5 m. It has long oval leaflets, solitary, single or double flowers with an asymmetrical apex. The bark of the root is yellow to brown with lignified fleshy center and a completely developed root system (38-40). It has a tube-shaped bark, with breaks along the entire length. The bark is slightly curly inward, 3-8 cm long, 0.5-1.2 cm in diameter and 0.1-0.4 cm thick. The external surface of the bark is reddish-brown or grayish-brown. The cork drop is pink with projecting pore spaces, light brown or grayish-yellow inner surface with a fine longitudinal texture and sparkling crystals (16).

4. Paeonol: The principal active ingredient of Paeonia suffruticosa

Cortex moutan has several bioactive constituents, such as gallic acid derivatives, flavonoids, triterpenes, monoterpene glycosides, and in particular, phenolic compounds. The principal active elements of Cortex moutan comprise, paeoniflorin, gallic acid, 1,2,3,4,6-penta-O-gallic acid-β-D-glucose and the phenolic compound paeonol (16). The active ingredients have been reported to be extracted using methods, such as organic solvent extraction, ultrasonic assisted extraction, steam distillation, and CO2 supercritical fluid extraction (41-43).

5. Immunomodulatory effects of paeonol

In vitro and in vivo immunosuppressive effects of paeonol. Diverse in vitro and in vivo research exploiting paeonol has indicated its potential immunosuppressive activities. Paeonol administration was previously shown to inhibit inflammation by downregulating lipase, amylase, IL-1β and IL-6. Moreover, the drug decreased ROS, restored mitochondrial membrane potential and suppressed M1 macrophage polarization through the NOD-like receptor protein 3 (NLRP3) pathway in bone marrow-derived macrophages (21). In another study, paeonol improves fluconazole or amphotericin B application during the treatment of EC109 cells and in a mouse model of oropharyngeal candidiasis by suppressing HIF-1α, IL-17A and IL-23 expression at the mRNA and protein level (34). A previous study also demonstrates that out of seven components of Cortex moutan Radicis studied in the treatment of antibiotic-associated diarrhea in mice and RAW 264.7 cells, paeonol attenuated NO production and IL-6 expression, decreased the level of TNF-α in a concentration-dependent manner, as well as the mRNA levels of IL-6, IL-1β and TNF-α (30). Also, COX-2 and NF-κB levels decreased as confirmed by western blot analysis; however, at the concentration of 25 µM, the p-p38 MAPK level was upregulated and was downregulated at 50 µM. Furthermore, compared with the control group, the TNF-α and IL-4 levels were decreased (30). As previously reported, Miao et al (32) indicated that paeonol improved the phagocytic capacity of macrophages by restricting high-mobility group box 1 to the nucleus. Additionally, it promoted the phosphorylation of focal adhesion kinase to stimulate the development of pseudopod in enhancing phagocytosis (32). Paeonol has been documented to suppress the miR-155/JAK1-STAT1 pathway to inhibit lipopolysaccharide (LPS) and IFN-γ-induced macrophage M1 polarization via the downregulation of F4/80 and CD86, IL-6 and TNF-α in mice and RAW264.7 cells (44). A previous study demonstrated that the oral administration of paeonol suppressed the development of 1-chloro-2,4-dinitrobenzene-induced AD-like lesions in BALB/c mice by downregulating the lesion severity, epidermal thickness and infiltration of mast cells (31). Moreover, this led to decreased levels of immunoglobulin E and IL-4, histamine, IL-13, IL-31 and thymic stromal lymphopoietin, together with the modulation of the T-helper (Th1/Th2) ratio. Paeonol regulated the (Th) cell subset (Th1/Th2) ratio by downregulating immunoglobulin E, IL-4, histamine, IL-13, IL-31 and thymic stromal lymphopoietin levels. Furthermore, its application attenuated phosphorylated (p)-p38 and p-ERK. In addition, the production of TNF-α and histamine was decreased, and p38/ERK/MAPK signaling inhibited in P815 cells. The aforementioned demonstrates that the drug act on cluster of differentiation 4+ T-and mast cells to inhibit allergic inflammatory conditions (31). In another report, paeonol suppressed NF-κB, IL-6 and TNF-α through denosine 5'-monophosphate-activated protein kinase (AMPK) and the glycogen synthase kinase-3 (GSK-3) pathway to prevent inflammation (29). Liu et al (45) investigated the effects of paeonol on THP-1 cells and human umbilical vein endothelial cells (HUVECs). The authors concluded that the drug upregulated miR-223 in THP-1 extracted exosomes and in HUVECs following the intake of exosomes, but reduced STAT3 and p-STAT3 expression in HUVECs. In addition, IL-1β, IL-6, ICAM-1 and VCAM-1 levels were downregulated in HUVECs, and THP-1 cell adhesion to HUVECs was attenuated to reduce the inflammatory response (45). Another study demonstrated that paeonol, at various concentrations, exerted anti-inflammatory effects against IL-1β-induced inflammation by suppressing NO and prostaglandin E2 (PGE2) production (46). Additionally, that study revealed that inducible nitric oxide synthase (iNOS), COX-2, MMP-1, MMP-3 and MMP-13 overexpression were reversed together with inhibition of NF-κB, PI3K and AKT activated signals (46). In a study investigating the effects of paeonol in a mouse model of imiquimod-induced psoriasis-like skin lesions and murine bone marrow-derived dendritic cells stimulated by R848, it was demonstrated that the drug inhibited IL-23 and downregulated dendritic cells expressing MHCII, CD80 and CD86 in vitro (17). This indicated that paeonol suppressed the maturation and activation of dendritic cells by downregulating MyD88 and Toll-like receptor (TLR)8 proteins (17). Another study demonstrated that paeonol, in a dose-dependent manner, attenuated LPS-stimulated inflammation to protect kidney injury (18). That study employed ELISA, western blot analysis and immunohistochemistry to determine the expression of inflammatory cytokines, TLR4-NF-κB pathway, and phospho-NF-κB p65, respectively. The results revealed that paeonol repressed pro-inflammatory cytokines and upregulated anti-inflammatory cytokines. Furthermore, the p-IκBα and p-IKKβ, NF-κB p65, and NF-κB p65 DNA-binding activities were inhibited. In addition, it suppressed the TLR4-NF-κB signaling pathway (18). Paeonol has been demonstrated to suppress the PI3K/Akt/NF-κB pathway to reduce hepatotoxicity (35). As previously demonstrated, pre-treatment of BV-2 cells and mice with paeonol revealed its ability to regulate p-AMPK-α and GSK 3α/β to suppress the expression of NO, iNOS, COX-2 and ROS induced by LPS/INF-γ. Furthermore, the drug suppressed STAT3 and p38 pathways (47). In another study, the administration of paeonol suppressed IL-8 via its antioxidant activity to inhibit MAPKs/NF-κB signaling to alleviate inflammation induced by chronic cigarette smoke (26). Chen et al (48) reported that paeonol, in a concentration-dependent manner, downregulated the release of TNF-α, IL-1β, IL-6, and increased IL-10 in LPS-induced RAW 264.7 via the deactivation of IκBα, ERK1/2, JNK and p38 MAPK. The in vitro and in vivo mechanisms of action of paeonol reported above are presented in Table I.

Table I

In vitro and in vivo immunosuppressive effects of paeonol.

Table I

In vitro and in vivo immunosuppressive effects of paeonol.

Authors, year of publicationStudy typeMechanism of action(Refs.)
Yuan et al, 2022In vivo and in vitroDownregulation of IL-1β, IL-6 and ROS, and the suppression of M1 macrophage polarization and NLRP3 inflammasome(21)
Pan et al, 2022In vivo and in vitroSuppresses the HIF-1α, IL-17A and IL-23 protein level(34)
Kang et al, 2022In vivo and in vitroDownregulates NO, COX-2 and IL-6, IL-1β, TNF-α, IL-4 and NF-κB and MAPK(30)
Miao et al, 2020In vivo and in vitroRestricts HMGB1 to the nucleus and promotes focal adhesion kinase; stimulates the development of pseudopods to upregulate phagocytosis(32)
Sun et al, 2020In vivo and in vitroSuppresses miR-155/JAK1-STAT1 and downregulates F4/80 and CD86, IL-6 and TNF-α(44)
Meng et al, 2019In vivo and in vitroRegulates the Th1/Th2 ratio by downregulating IgE, IL-4, histamine, IL-13, IL-31, TNF-α and p38/ERK/MAPK(31)
Liu et al, 2018In vivo and in vitroSuppresses NF-κB, IL-6 and TNF-α through the adenosine 5'-monophosphate-activated protein kinase (AMPK) and glycogen synthase kinase-3 (GSK-3) pathways(29)
Liu et al, 2018In vivo and in vitroUpregulates miR-223, but reduces STAT3, p-STAT3, IL-1β, IL-6, ICAM-1, VCAM-1 expression(45)
Lou et al, 2017In vivo and in vitroDownregulates NO, INOS, COX-2, MMP-1, MMP-3, MMP-13, and inhibits NF-κB, PI3K and AKT(46)
Meng et al, 2017In vivo and in vitroInhibits IL-23 and downregulates dendritic cells expressing MHCII, CD80 and CD86(17)
Fan et al, 2016In vivo and in vitroRepresses pro-inflammatory cytokines and upregulates anti-inflammatory cytokines; inhibits IκBα and IKKβ, NF-κB p65, and restricts NF-κB p65 DNA-binding activity as well as TLR4-NF-κB(18)
Wu et al, 2016In vivo and in vitroSuppresses the PI3K/Akt/NF-kB pathway.(35)
Lin et al, 2015In vivo and in vitroRegulates AMPK-α and GSK 3α/β to suppress NO, iNOS, COX-2, ROS and STAT3 as well as p38 pathways(47)
Liu et al, 2014In vivo and in vitroInhibits MAPKs/NF-κB signaling pathways.(26)
Chen et al, 2014In vivo and in vitroDownregulates TNF-α, IL-1β, IL-6, upregulates IL-10 via the deactivation of IκBα, ERK1/2, JNK, and p38 MAPK(48)

[i] IL, interleukin; ROS, reactive oxygen species; NO, nitric oxide; COX-2, cyclooxygenase 2; VCAM-1, vascular cell adhesion molecule 1; ICAM-1, intercellular adhesion molecule 1; MMP, matrix metalloproteinase.

In summary, a critical analysis of both in vitro and in vivo immunomodulatory mechanisms demonstrated that paeonol suppresses the discharge of pro-inflammatory cytokines, including IL-1β, IL-6, TNF-α and IL-8, while upregulating the anti-inflammatory cytokine, IL-10, in the treatment of several inflammation in various models. However, the specific genomic and proteomic changes induced by paeonol remain underexplored. Additionally, paeonol regulates the function of macrophages in promoting tissue repair by modulating polarization to an M2 anti-inflammatory phenotype while inhibiting M1. It promotes the maturation and activation of dendritic cells to initiate T-cell adaptive immunity. The drug mitigates allergy by decreasing immunoglobulin E, histamine, hence can be used to manage conditions such as asthma, atopic dermatitis and allergic rhinitis. Moreover, paeonol protects against kidney injury and hepatotoxicity by inhibiting oxidative stress, NF-κB and MAPK pathways, and decreases the generation of ROS. Furthermore, it modulates the NF-κB, PI3K/Akt, AMPK and MAPK pathways to exert multiple level immunosuppressive effects to attenuate inflammation extensively. In addition, it exerts synergistic effects when combined with conventional antifungal agents or anti-inflammatory drugs.

In vitro immunosuppressive effects of paeonol

Recently, studies have demonstrated that paeonol promotes M2 macrophage polarization by suppressing M1 polarization (49,50) through the inhibition of the NF-κB and MAPK pathways (49). Miao et al (51) demonstrated that the promotion of nuclear p53 by paeonol served as a transcript and increased triggering receptor expressed on myeloid cells-2 to activate macrophage lipid metabolic capacity and phagocytic function. In another study, paeonol downregulated IL-1β and caspase-1 level, attenuated the monosodium urate (MSU)-induced interaction of pro-caspase-1 and apoptosis-associated speck-like protein containing caspase recruitment domain (ASC) in J774A.1 cells induced by LPS plus MSU (52). The drug decreased IL-1β, NLRP3, p-IKK, p-IκBα, and p-p65 in J774A.1 cells induced by LPS alone but did not affect the ASC level. Moreover, the IκBα content was stimulated and a greater amount of cytoplasmic p65 retained. Furthermore, paeonol decreased p65 DNA-binding action and curtailed p-JNK, p-ERK and p-p38 level. The study concluded that the inhibition of the NLRP3 inflammasome, NF-κB and MAPK pathway activation by paeonol occurred through the suppression of IL-1β (52). Another study assessed the paeonol-mediated suppression of LPS-activated inflammation through the TLR4 signaling pathway in N9 microglia cells (53). That study evaluated NO, IL-1β and PGE2 using ELISA. Additionally, COX-2, iNOS, TLR4, MyD88, IRAK4, TNFR-associated factor 6 (TRAF6), p-IkB-α, and NF-kB p65, as well as p-P38, p-JNK and p-ERK were examined using western blot analysis. The results revealed that paeonol significantly repressed the production of pro-inflammatory products, including NO, IL-1β, and PGE2 as well as the expression of iNOS and COX-2. In addition, the TLR4, MyD88, IRAK4, TRAF6, p-IkB-α and NF-κB p65, as well as p-p38, p-JNK and p-ERK expression levels were significantly reduced (53). In another study, paeonol upregulated NO, iNOS and downregulated ROS (28). Moreover, the drug attenuated TNF-α, IL-1β, IL-6 and monocyte chemoattractant protein-1 (MCP-1) with a significant decrease in RAGE and CD36, and an increase in SR-A and SR-B1 levels. The authors of that study reported that the anti-inflammatory response was likely mediated via the RAGE, CD36, SR-A and SR-B1-pathways with NADPH oxidase-dependent ROS generation (28). The aforementioned in vitro studies have revealed that paeonol decreases IL-1β to repress several inflammatory mediators, such as NO, iNOS, COX2 and PEG2 in the inhibition of the NLRP3 inflammasome, and also inhibits the NF-κB, MAPK and TLR4 pathways to provide a broad-spectrum effect on immune-related diseases, to function as an anti-inflammatory agent.

In vivo immunosuppressive effects of paeonol

In a previous study, the administration of paeonol downregulated M1 polarization markers (IL1β, iNOS, CD32 and IL6), and upregulated the M2 polarization markers (IL10, CD206 and ARG-1) by decreasing iNOS, IL1β, RhoA and Rock1 in LPS-stimulated microglia (54). Additionally, during thermal hyperalgesia, the drug downregulated IL1β and IL8 and upregulated IL4 and TGF-β levels in the serum. It was revealed that in the spinal dorsal horn the drug downregulated IBA-1, IL1β, RhoA, RhoA-GTP, COX2, Rock1, and p-p38MAPK levels to reduce neuropathic pain (54). It has also been demonstrated that paeonol acts via the dectin-1/NF-κB pathway together with TLR2 and TLR4 to relieve fungal dysbiosis-associated ulcerative colitis (27). Recently, paeonol has been reported to significantly suppress NF-κB and MCP-1 in vivo (55). In another study, paeonol was shown to inhibit the inflammatory response by suppressing the mycobiota-mediated dectin-1/IL-1β signaling pathway (56). Paeonol administration has also been shown to attenuate liver injury and fibrosis. In a previous study, paeonol downregulated ALT and AST levels, and inhibited the TGF-b/Smad3 pathway by suppressing the activation of hepatic stellate cells to downregulate IL-6 and TNF-α, and reduced oxidative stress (57). Another study indicated that paeonol modulated nuclear factor E2-related factor 2 (Nrf2)/NF-κB/NFATc1 signaling in the inhibition of osteoclastogenesis to protect against periodontitis (58). It has been well-documented that paeonol improves the asthmatic condition by downregulating IFN-γ and upregulating IL-4 via the suppression of the TLR4/NF-κB and MAPK pathway (59). In another study, in LPS/D-galactosamine-induced acute liver failure, paeonol at 100 mg/kg markedly suppressed the iNOS, NO, COX-2 and PGE2 levels (60). The phosphorylation levels of IκB kinase (IKK), IκB and NF-κB (p65), which constitute the NF-κB pathway and MAPK signaling pathway molecules, including ERK, JNK and p38 were significantly inhibited. Moreover, the decreased expression of caspase-3, -8 and -9, and Bax, with the upregulated expression of Bcl-2 observed following paeonol treatment was attributed to the inhibition of hepatocyte apoptosis (60). Another study reported that paeonol at 200 and 400 mg/kg significantly decreased IL-17 and IL-6, and upregulated TGF-β1 expression in rats with 2,4,6-trinitrobenzenesulfonic acid-induced ulcerative colitis (61). In their study, Ding et al (62) investigated the anti-oxidative stress activity of paeonol against acetaminophen (APAP)-induced hepatotoxicity and indicated that it suppressed JNK phosphorylated protein. Moreover, the drug exerted a significant suppressive effect of H2O2 or APAP-induced ROS production. Furthermore, the levels of TNF-α, MCP-1, IL-1β and IL-6 were decreased in a concentration-dependent manner, while IKKα/β, IκBα and p65 phosphorylation were significantly inhibited (62). Paeonol has been documented to exert neuroprotective effects by repressing TLR-2 and TLR-4, Iba1, NF-κB (p50) and IL-1β, as well as by suppressing apoptosis in a Sprague-Dawley rat model of cerebral ischemia-reperfusion injury (63). Lee et al (64) evaluated the renal toxic effects of paeonol and reported that it downregulated TNF-α, IL-1β and NO, as well as decreased blood urea nitrogen and serum creatine levels (64). Key signaling pathways and mediators downregulated by paeonol in vitro or in vivo are presented in Fig. 1. The in vitro or in vivo mechanisms mentioned above are presented in Table II. In summary, paeonol has the capacity to be employed for the treatment of hepatotoxicity, acute liver diseases, liver failure, gastrointestinal disorders, ulcerative colitis, renal disorders and neurological conditions. However, further transcriptomics studies are required to elucidate its molecular mechanisms and maximize its therapeutic application in medical settings.

Table II

In vitro or in vivo immunosuppressive effects of paeonol.

Table II

In vitro or in vivo immunosuppressive effects of paeonol.

Authors, year of publicationStudy typeMechanism of action(Refs.)
Niu et al, 2024In vitroRegulates M1/M2 polarization of macrophages(49)
Chen et al, 2022In vitroSuppresses IL-1β and inhibits NLRP3, NF-κB, and MAPK activated pathways(52)
He et al, 2016In vitroRepresses NO, IL-1β and PGE2, as well as the expression of iNOS and COX-2; reduces TLR4, MyD88, IRAK4, TNFR-associated factor 6 (TRAF6), p-IkB-α, and NF-κB p65, as well as p-p38, p-JNK, and p-ERK(53)
Ping et al, 2014In vitroUpregulates NO, iNOS, downregulates ROS, TNF-α, IL-1β, IL-6 and MCP-1 through the RAGE-, CD36-, SR-A- and SR-B1-mediated pathway(28)
Li et al, 2023In vivoModulating microglial M1 and M2 polarization via the RhoA/p38MAPK pathway(54)
Ge et al, 2021In vivoDecreases the dectin-1/NF-κB pathway together with TLR2 and TLR4(27)
Adki et al, 2021In vivoSuppresses NF-κB and MCP-1(55)
Wu et al, 2020In vivoSuppresses the dectin-1/IL-1β signaling pathway(56)
Wu et al, 2019In vivoInhibits TGF-β/Smad3 and downregulates IL-6 and TNF-α(57)
Li et al, 2019In vivoUpregulates Nrf2 to downregulate NF-κB/NFATc1(58)
Tang et al, 2018In vivoDownregulates IFN-γ and upregulates IL-4, suppresses TLR4/NF-κB and MAPK pathway(59)
Gong et al, 2017In vivoSuppresses iNOS, nitric oxide, COX-2, prostaglandin E2 and inhibitsNF-κB, and MAPK pathways as well as decreased hepatocyte apoptosis(60)
Zong et al, 2017In vivoDecreases IL-17 and IL-6 and upregulates TGF-β1(61)
Ding et al, 2016In vivoSuppresses c-Jun N-terminal kinase and decreases TNF-α, MCP-1, IL-1β, IL-6 and inhibits IKKα/β, IκBα and p65 phosphorylation(62)
Liao et al, 2016In vivoRepresses TLR2, TLR4, Iba1-, NF-κB- (P50-), and IL-1β and inhibits apoptosis(63)
Lee et al, 2013In vivoDownregulates TNF-α, IL-1β and nitric oxide(64)

[i] IL, interleukin; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase 2; TLR, Toll-like receptor; ROS, reactive oxygen species; PGE2, prostaglandin E2.

6. Clinical trials

A recent phase 2a randomized, placebo-controlled trial evaluated the efficacy and safety of a fixed-dose combination of apocynin (AP) and paeonol (PA) (APPA) in patients with symptomatic knee osteoarthritis (OA) (65). The study enrolled 152 participants with Kellgren-Lawrence grade 2-3 knee OA and moderate to severe pain (WOMAC pain ≥40/100). Over a period of 28 days, APPA at 800 mg twice daily demonstrated no statistically significant improvement in primary (WOMAC pain) or secondary outcomes (WOMAC function and total score) compared to the placebo. However, predefined subgroup analyses revealed a significant benefit of APPA in participants with nociplastic or neuropathic pain features. The treatment was well-tolerated with mild to moderate adverse events, primarily gastrointestinal. These findings highlight the safety profile of APPA and suggest its potential efficacy in specific OA subgroups, warranting further targeted research (65).

7. Chemopreventive potential of paeonol

The antitumor effects of paeonol on breast (14,24,36,66,67), bladder (68), colorectal (23,69), colon (70), gastric (22,71,72), lung (73-75), osteosarcoma (76), ovarian (77-81), prostate (82), chondrosarcoma (83), pancreatic (84), cervical (33,85), melanoma (25), renal (86), hepatocellular (87-89) and oral (90) cancers have been documented.

In a previous study, paeonol induced the apoptosis of breast cancer by regulating CXCL4/CXCR3-B signaling to downregulate heme oxygenase and Nrf2 to upregulate BACH1(14). In another study, paeonol and epirubicin synergistically inhibited tumor growth and promoted apoptosis via the activation of PARP, Bax and caspase-3 by suppressing p38/JNK/ERK MAPKs compared to epirubicin alone (36). Additionally, paeonol inhibited the NF-κB pathway to alleviate epirubicin-induced cardiotoxicity (36). Zhang et al (66) demonstrated that paeonol downregulated SET, protein phosphatase 2A and the PI3K/Akt pathway to prevent paclitaxel resistance in MCF-7/PTX cells. In a similar study, paeonol decreased P-glycoprotein, multidrug resistance associated protein 1, and breast cancer resistance protein to suppress transgelin 2-mediated paclitaxel resistance (67). In another study, the significant decrease in tumor weight observed by morphological analysis using hematoxylin and eosin staining, as well as TUNEL staining indicated that paeonol induced the apoptosis of tumor cells (24). Moreover, immunohistochemistry and western blot analysis demonstrated that the downregulation of Bcl-2 led to the upregulation of Bax, caspase-8 and caspase-3 to confirm the induction of apoptosis as the mechanism of inhibition employed by paeonol (24). In a recent study, the administration of paeonol exerted an inhibitory effect on cell proliferation and induced both in vitro and in vivo apoptosis by decreasing the Bcl-2/Bax ratio with the upregulation of caspase-3(68). The drug also suppressed the PI3K/AKT pathway in the inhibition of bladder cancer cell growth (68). In colorectal cancer (CRC), paeonol administration has been shown to induce apoptosis and arrest the cell cycle at the G0/G1-phase (69). Additionally, the drug downregulated β-catenin, cyclin D1, survivin and c-Myc in a concentration-dependent manner to suppress the Wnt/β-catenin signaling pathway (69). Li et al (23) investigated the effects of paeonol on CRC cells and revealed that paeonol suppressed cell proliferation by reducing COX-2 and PGE2 expression, and induced mitochondrial pathway apoptosis in a time-dependent manner. The apoptotic mechanism was associated with the upregulation of Bax and decreased Bcl-2 expression. Furthermore, the drug activated caspase-3 and caspase-9 and induced the loss of mitochondrial membrane potential (23). Flow cytometry analysis performed by Li et al (70) demonstrated that paeonol induced apoptosis and arrested the cell cycle at the G1 to S transition phase. Moreover, the treatment led to an upregulated intracellular calcium concentration and RUNX3 expression suggesting its antitumor mechanism (70). The administration of paeonol suppressed apatinib-resistant gastric cancer cell malignancy by downregulating LINC00665 and MAPK1, while upregulating miR-665(71). Paeonol has been shown to inhibit the NF-κB pathway by decreasing ERBB2 expression to suppress the proliferation of gastric cancer and induce apoptosis (72). The anticancer mechanism of paeonol has been shown to be associated with the downregulation of MMP-2 and-9 in a concentration-dependent manner to prevent invasion and migration (22). The anticancer effects of paeonol on lung cancer have been reported to be associated with the upregulation of miR-126-5p and the downregulation of zinc finger E-box-binding homeobox 2 to suppress cell viability and metastasis (73). In another study, paeonol repressed TNF-α, IL-6, IL-1β and TGF-β in the inhibition of non-small-cell lung cancer cell proliferation, invasion and migration by suppressing the STAT3/NF-κB pathway (74). Lei et al (75) reported that paeonol improved radiation-induced apoptosis via the suppression of the PI3K/AKT pathway to decrease downstream COX-2 and survivin. Recently, Zhou et al (76) investigated the effects of paeonol on osteosarcoma. They reported that the antitumor mechanisms were associated with the suppression of the invasion and migration potential, as well as the suppression of TLR4 to inhibit the activation of the MAPK/NF-κB downstream signaling pathway (76). In ovarian cancer, paeonol has been shown to suppress Akt/mTOR signaling by upregulating autophagy (77). Han et al (78) demonstrated that paeonol activated PTEN to suppress P-glycoprotein, multidrug resistant mutation 1 and metadherin to overturn resistant ovarian cancer. In another study, paeonol induced apoptosis via the upregulation of caspase-3 and caspase-9, and decreased p-Akt and p-GSK-3β signaling to inhibit ovarian cell proliferation (79). Zhou et al (80) demonstrated that paeonol increased the responsiveness of ovarian cancer cells to radiotherapy-induced apoptosis by inhibiting the PI3K/Akt/phosphatase, VEGF and HIF-1α pathways. In another study, cell viability, apoptosis, caspase-3 and survivin levels were evaluated by MTT assay, flow cytometry and Hoechst staining, and western blot analysis, respectively (81). The results revealed that cell viability was decreased, and apoptosis induced. Additionally, caspase-3 expression was upregulated and survivin protein expression was downregulated (81). In another study, paeonol administered in vivo and in vitro to human prostate cancer decreased tumor cell proliferation and suppressed tumor growth (82). In the process, the drug induced apoptosis which resulted in the upregulation of caspase-3, caspase-8, and caspase-9 levels and the downregulation of Bcl-2 with an enhanced Bax expression. Moreover, the activated PI3K/Akt pathway was inhibited. These findings suggest that the drug induces apoptosis by activating both the extrinsic and intrinsic pathways, and via the suppression PI3K/Akt pathway to inhibit tumor cell growth (82). Another study demonstrated that paeonol administration increased miR-141 by downregulating the protein kinase Cδ and c-Src pathway in the suppression of metastatic chondrosarcoma (83). Paeonol has been reported to possess anti-metastatic properties in pancreatic cancer. Researchers employed cell scratch-wound healing assay and Boyden chamber invasion assay to evaluate the migration and invasion abilities (84). In addition, the RNA and protein levels of E-cadherin, N-cadherin, vimentin and TGF-β1/Smad pathway were determined using RT-qPCR and western blot analysis. The findings revealed that paeonol suppressed epithelial-mesenchymal-transition by enhancing E-cadherin and reducing N-cadherin and vimentin. The expression levels of TGF-β1, p-Smad2/Smad2 and p-Smad3/Smad3 were also decreased to suppress the TGF-β1/Smad pathway (84). Paeonol induces apoptosis via the mitochondrial pathway and inhibits the PI3K/Akt pathway in the suppression of cervical cancer (85). It has been demonstrated that paeonol decreases 5-lipoxygenase to promote apoptosis in the inhibition of cervical cancer migration and invasion (33). It has been documented that paeonol suppresses cell proliferation, induces apoptosis and inhibits pro-inflammatory cytokine-mediated NF-κB and STAT3 signaling in the inhibition of melanoma metastasis (25). A previous study on renal cell carcinoma revealed that paeonol reduced the Bcl-2/Bax ratio in the induction of apoptosis and downregulated VEGFA to inhibit cell proliferation, invasion and metastasis (86). In another in vitro and in vivo study, paeonol downregulated miR-21-5p to upregulate KLF6 to promote apoptosis and inhibit the proliferation, migration and invasion of hepatocellular carcinoma cells (87). Li et al (88) evaluated the apoptotic activity of paeonol in hepatocellular carcinoma and reported that the drug decreased NF-κB p65/50 and protein apoptosis inhibitor-5 in the promotion of apoptosis and suppressed NF-κB signaling pathway. Fan et al (89) demonstrated the ability of paeonol to overturn endoplasmic reticulum stress-promoted resistance to doxorubicin by downregulating COX-2 to inactivate the PI3K/AKT/CHOP pathway. Paeonol has been reported to suppress tumor growth and promotes apoptosis by inhibiting mutant p53 and COX-2 and upregulating caspase-9 in 7,12-dimethylbenz(a)anthracene-induced oral carcinogenesis (90). The mechanism of paeonol in tumor immunity revealed that the drug downregulates PD1 via upregulating miR-139-5p to suppress melanoma growth (91). Research on aminothiazole-paeonol derivatives has shown that these compounds exhibit higher potency against AGS (gastric) and HT-29 (colorectal) human cancer cell lines compared to 5-fluorouracil (5-FU), a commonly used chemotherapeutic agent. Additionally, these derivatives demonstrated lower cytotoxicity towards normal cells, suggesting a potentially improved therapeutic index (92). Key signaling pathways and mediators downregulated by paeonol are illustrated in Fig. 1. The chemopreventive mechanisms mentioned above are presented in Table III.

Table III

In vitro and in vivo chemopreventive effects of paeonol.

Table III

In vitro and in vivo chemopreventive effects of paeonol.

Authors, year of publicationStudy typeMechanism of action(Refs.)
Saahene et al, 2018In vitroRegulates CXCL4/CXCR3-B signal to downregulate heme oxygenase (HO-1) and nuclear factor E2-related factor 2 (Nrf2) to upregulate BACH1 to induce apoptosis in breast cancer(14)
Wu et al, 2016In vivo and in vitroInduces apoptosis via activation of PARP, Bax, caspase 3 and suppresses MAPKs/NF-κB pathways in breast cancer(35)
Zhang et al, 2015In vitroDecreases SET, protein phosphatase 2A and phosphatidylinositol 3-kinase (PI3K)/Akt pathway in breast cancer(66)
Cai et al, 2014In vitroDecreases P-glycoprotein, multidrug resistance associated protein 1, and breast cancer resistance protein to suppress transgelin 2 resistance(67)
Qu et al, 2014In vitroDownregulates Bcl-2, upregulates Bax, caspase-8 and caspase-3 to induce breast cancer apoptosis(24)
Zhang et al, 2021In vivo and in vitroDecreases Bcl-2/Bax ratio upregulates caspase 3 and suppresses PI3K/AKT pathway in bladder cancer(68)
Liu et al, 2020In vitroInduces apoptosis, arrest the cell cycle at G0/G1-phase, downregulates β-catenin, cyclin D1, survivin and c-Myc to suppress Wnt/β-catenin pathway in colorectal cancer(69)
Li et al, 2014In vivo and in vitroDecreases cyclooxygenase-2 and prostaglandin E2 and decreased Bcl-2 and upregulates Bax, caspase 3 and caspase 9 in colorectal cancer(23)
Li et al, 2013In vitroInduces apoptosis, arrests the cell cycle at the G1 to S transition phase; upregulation of intracellular calcium concentration and RUNX3 in colon cancer(70)
Li et al, 2022In vitroDownregulation of LINC00665 and MAPK1, upregulating miR-665 in gastric cancer(71)
Fu et al, 2018In vitroInhibits NF-κB by decreasing ERBB2 and induces apoptosis gastric cancer(72)
Lyu et al, 2017In vitroDownregulation of MMP-2 and MMP-9 in gastric cancer(22)
Lv et al, 2022In vitroUpregulates miR-126-5p and downregulates ZEB2 to suppress cell viability and metastasis in lung cancer(73)
Zhang et al, 2020In vivo and in vitroInhibits TNF-α, IL-6, IL-1β, TGF-β and suppressed STAT3/NF-κB pathways in lung cancer(74)
Lei et al, 2013In vitroEnhances apoptosis, suppression of PI3K/AKT pathway and decreased cyclooxygenase-2 and survivin in lung cancer(75)
Zhou et al, 2020In vivo and in vitroSuppresses TLR4 to inhibit MAPK/NF-κB to prevent invasion and migration and induces apoptosis in osteosarcoma(76)
Gao et al, 2019In vivo and in vitroSuppresses Akt/mTOR signal by upregulating autophagy in ovarian cancer(77)
Han et al, 2018In vitroUpregulation of PTEN suppresses P-glycoprotein, multidrug resistant mutation 1 and metadherin in ovarian cancer(78)
Li et al, 2017In vitroUpregulates caspase-3 and caspase-9 to induces apoptosis and decreased phosphorylated-Akt and phosphorylated-GSK-3β in ovarian cancer(79)
Zhou et al, 2017In vitroInhibits P13K/Akt, VEGF and HIF-1α pathways via radiation induced apoptosis in ovarian cancer(80)
Yin et al, 2013In vitroUpregulates caspase-3 and downregulate of survivin to induce apoptosis in ovarian cancer(81)
Xu et al, 2017In vivo and in vitroUpregulates caspase-3, caspase-8, and caspase-9 events and downregulated Bcl-2 with an enhanced Bax; inhibits P13K/Akt pathway in prostate cancer(82)
Horng et al, 2014In vitroIncreases miR-141 and downregulated protein kinase C (PKC) δ and c-Src pathways in chondrosarcoma(83)
Cheng et al, 2020In vitroIncreases E-cadherin and reduced N-cadherin and vimentin; decreased TGF-β1, p-Smad2/Smad2 and p-Smad3/Smad3 in pancreatic cancer(84)
Du et al, 2021In vitroDownregulates Bcl-2/Bax proportion, upregulated caspase-3 by inhibiting P13K/Akt pathway in cervical cancer(85)
Sheng et al, 2021In vitroDownregulates 5-lipoxygenase to promote apoptosis to prevent invasion and migration in cervical cancer.(33)
Zhang et al, 2015In vivo and in vitroInduces apoptosis and inhibits proinflammatory cytokines mediated NF-κB and STAT3 signaling to prevent melanoma metastasis(25)
Chen et al, 2022In vitroIncreases Bax decreased Bcl-2 and downregulated VEGFA to prevent invasion and metastasis in renal cell carcinoma(86)
Cai et al, 2020In vivo and in vitroDownregulates miR-21-5p to upregulate KLF6 in the promotion of apoptosis and inhibited cell proliferation, migration and invasion of hepatocellular carcinoma.(87)
Li et al, 2019In vitroDecreases NF-κB p65/50 and protein apoptosis inhibitor-5 in the induction of apoptosis and suppressed NF-κB pathway in HCC(88)
Fan et al, 2013In vitroDownregulates COX-2 to inactivate the PI3K/AKT/CHOP pathway in hepatocellular carcinoma.(89)
Ramachandhiran et al, 2018In vivoPromotes apoptosis by inhibiting mutant p53 and COX-2 and upregulating caspase-9 in oral carcinogenesis(90)

[i] IL, interleukin; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase 2; TLR, Toll-like receptor; ROS, reactive oxygen species; PGE2, prostaglandin E2.

The chemopreventive capacity of paeonol relies on its multifaceted anticancer mechanisms by directly inhibiting cell growth and proliferation, inducing apoptosis, suppressing invasion and metastasis (MMP-2 and-9, VEGF and HIF-1α), and inhibiting the TLR4/NF-κB/STAT3/MAPK/PI3K/AKT/CHOP pathways to inhibit cancer development and progression. Moreover, the drug has demonstrated potential for combination therapy and synergy with natural compounds, by improving their potency while potentially reducing their harmful effects. Additionally, paeonol reduces drug resistance in breast cancer and ovarian cancer, by decreasing drug efflux transporters and overturning resistance-associated signaling pathways.

Whilst there are data demonstrating the anticancer potential of paeonol, a search through published literature revealed limited in vitro and in vivo information regarding the toxicity of the compound on normal cells and organs. On the contrary, there is a huge volume of data on its ability to protect against chemical induced toxicity (26,35,62). Subchronic toxicity studies in male and female laboratory rats showed no major safety concerns (64,93). This attests to the potential of paeonol as a safe alternative agent for immunomodulation.

8. Clinical relevance and therapeutic effects on the NF-κB, MAPK and PI3K/AKT pathways

The NF-κB signaling pathway is crucial in regulating immune responses and inflammation. Paeonol has been shown to inhibit this pathway, thereby reducing the expression of pro-inflammatory cytokines and promoting apoptosis in cancer cells. For instance, studies have demonstrated that paeonol can effectively decrease NF-κB activation in hepatocellular carcinoma cells, leading to enhanced apoptosis and reduced cell proliferation (88,94). The inhibition of NF-κB in preclinical models by paeonol has been associated with decreased tumor growth and improved outcomes in inflammatory conditions such as ulcerative colitis (94,95).

The MAPK pathway is essential in cellular response to stress and growth. The regulation of MAPK by paeonol has been demonstrated to be linked to suppressed cell migration and invasion in lung cancer models, indicating its significant anti-metastatic potential (74,94). The dysregulation of the MAPK cascade by paeonol, not only inhibits cancer growth, but also alleviates inflammation in patients with chronic inflammatory diseases or cancer (74,96).

The PI3K/AKT pathway modulates cell survival, proliferation and metabolism, and its hyperactivation is commonly associated with cancer progression and resistance to apoptosis. Paeonol has demonstrated the ability to inhibit PI3K/AKT signaling to induce apoptosis and suppress tumor growth (68,75,82,94,97). The PI3K/AKT signaling modulation by paeonol has been linked to an improved sensitivity to chemotherapeutic agents and diminished adverse effects from treatment, suggesting its potential as an adjunct therapy in distinct populations (66,94,97).

9. Conclusion and future perspectives

Paeonol, a bioactive component of Cortex moutan has shown promise as a naturally occurring pharmacological agent for suppressing immunological events in a variety of human diseases. The mechanism is attributed to its multifactorial activities, including immunomodulatory effects, anti-inflammatory effects and antioxidant properties. Overall, paeonol decreases IL-1β expression to repress several inflammatory mediators, such as NO, iNOS, COX2, PEG2 in the inhibition of the NLRP3 inflammasome, NF-κB, MAPK and TLR4 pathways to provide multiple levels immunosuppressive effects of immune-related diseases. Current research demonstrates the anti-inflammatory properties of paeonol through the suppression of cytokines and the modulation of immune pathways. However, further investigations are required to focus on RNA-seq analyses to identify global gene expression changes in response to paeonol. Moreover, integrating multi-omics approaches (e.g., transcriptomics, proteomics and metabolomics) to build a systems-level understanding of the actions of paeonol could ultimately lead to the development of improved therapeutic strategies for conditions characterized by inflammation. The present review suggests that paeonol may serve as an alternative immunomodulator to conventional therapeutic agents; however, its probable negative effects in clinical applications need to be assessed. The repression of the NF-κB and MAPK pathways may not only reduce pro-inflammatory determinants, but may also attenuate the induction of certain key factors in the immune response. Thus, the effects of paeonol on different cytokines, inflammatory mediators and immune responses signaling pathways need to be explored. There are limited reports available of its application in relation to the cells of the immune system, effector mechanisms and phagocytosis, and thus, further studies are warranted.

As regards cancers, it has been reported that paeonol directly suppresses the proliferation and growth of several cancer cells, arrests the cancer cell cycle, induces apoptosis, and prevents invasion and metastasis. Furthermore, the drug inhibits cancer cell growth and proliferation through the suppression of the VEGF, HIF-1α, TLR4/ NF-κB/STAT3/MAPK/PI3K/AKT/CHOP pathways. Despite promising preclinical findings, there is a notable lack of clinical trial studies evaluating the efficacy and safety of paeonol treatment for human diseases. This underscores the need to conduct large-scale, multicenter clinical trials to validate preclinical findings in conditions such as asthma, atopic dermatitis, and allergic rhinitis, kidney injury and hepatotoxicity, breast cancer, gastric cancer and ovarian cancer to assess the therapeutic efficacy and safety of this drug in diverse patient populations. Moreover, exploring the synergistic effects of paeonol in combination with paclitaxel, epirubicin, doxorubicin, and apatinib in cancer treatment is warranted.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

PB and ROS were involved in the conceptualization and design of the review, as well as in the drafting and writing of the manuscript. EA, FHL, KD, SVN, DOY and ESY were involved in the literature review, and in the drafting of the manuscript. All authors have read, edited and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Use of artificial intelligence tools

During the preparation of this work, AI tools were used to improve the readability and language of the manuscript and subsequently, the authors revised and edited the content produced by the AI tools as necessary, taking full responsibility for the ultimate content of the present manuscript.

References

1 

Jantan I, Haque MA, Ilangkovan M and Arshad L: An insight into the modulatory effects and mechanisms of action of Phyllanthus species and their bioactive metabolites on the immune system. Front Pharmacol. 10(878)2019.PubMed/NCBI View Article : Google Scholar

2 

Greenberg S and Grinstein S: Phagocytosis and innate immunity. Curr Opin Immunol. 14:136–145. 2002.PubMed/NCBI View Article : Google Scholar

3 

Janeway C, Travers P, Walport M and Shlomchik M: Immunobiology. In: TheImmune System in Health and Disease. Vol 2: Garland Science, New York, 2001.

4 

Siracusano A, Riganò R, Ortona E, Profumo E, Margutti P, Buttari B, Delunardo F and Teggi A: Immunomodulatory mechanisms during Echinococcus granulosus infection. Exp Parasitol. 119:483–489. 2008.PubMed/NCBI View Article : Google Scholar

5 

M. D. Bagatini, ‘Immune System and Chronic Diseases 2018.’.

6 

Wolska K, Górska A, Antosik K and Ługowska K: Immunomodulatory effects of propolis and its components on basic immune cell functions. Indian J Pharm Sci. 81:575–588. 2019.

7 

Ilangkovan M, Jantan I, Mesaik MA and Bukhari SNA: Immunosuppressive effects of the standardized extract of Phyllanthus amarus on cellular immune responses in Wistar-Kyoto rats. Drug Des Devel Ther. 26:4917–4930. 2015.PubMed/NCBI View Article : Google Scholar

8 

Khandelwal V, Bhati A and Anjana G: Immunomodulatory activity of hot aqueous extract of Anthocephalus cadamba leaves in albino rats. Adv Bioresearch. 7:203–206. 2016.

9 

Stromberg SP and Carlson JM: The suppression of immune system disorders by passive attrition. PLoS One. 5(e9648)2010.PubMed/NCBI View Article : Google Scholar

10 

Okoye AA and Picker LJ: CD 4+ T-cell depletion in HIV infection: Mechanisms of immunological failure. Immunol Rev. 254:54–64. 2013.PubMed/NCBI View Article : Google Scholar

11 

Wherry EJ and Kurachi M: Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 15:486–499. 2015.PubMed/NCBI View Article : Google Scholar

12 

Alpdogan O and van den Brink MR: Immune tolerance and transplantation. Semin Oncol. 39:629–642. 2012.PubMed/NCBI View Article : Google Scholar

13 

Redman JM, Gibney GT and Atkins MB: Advances in immunotherapy for melanoma. BMC Med. 14:1–11. 2016.PubMed/NCBI View Article : Google Scholar

14 

Saahene RO, Wang J, Wang ML, Agbo E and Pang D: The antitumor mechanism of paeonol on CXCL4/CXCR3-B signals in breast cancer through induction of tumor cell apoptosis. Cancer Biother Radiopharm. 33:233–240. 2018.PubMed/NCBI View Article : Google Scholar

15 

Pang KL, Vijayaraghavan K, Al Sayed B and Seyed MA: Betulinic acid-induced expression of nicotinamide adenine dinucleotide phosphate-diaphorase in the immune organs of mice: A possible role of nitric oxide in immunomodulation. Mol Med Rep. 17:3035–3041. 2018.PubMed/NCBI View Article : Google Scholar

16 

Wu M, Yu Z, Li X, Zhang X, Wang S, Yang S, Hu L and Liu L: Paeonol for the treatment of atherosclerotic cardiovascular disease: A pharmacological and mechanistic overview. Front Cardiovasc Med. 8(690116)2021.PubMed/NCBI View Article : Google Scholar

17 

Meng Y, Wang M, Xie X, Di T, Zhao J, Lin Y, Xu X, Li N, Zhai Y, Wang Y and Li P: Paeonol ameliorates imiquimod-induced psoriasis-like skin lesions in BALB/c mice by inhibiting the maturation and activation of dendritic cells. Int J Mol Med. 39:1101–1110. 2017.PubMed/NCBI View Article : Google Scholar

18 

Fan HY, Qi D, Yu C, Zhao F, Liu T, Zhang ZK, Yang MY, Zhang LM, Chen DQ and Du Y: Paeonol protects endotoxin-induced acute kidney injury: Potential mechanism of inhibiting TLR4-NF-κB signal pathway. Oncotarget. 7:39497–39510. 2016.PubMed/NCBI View Article : Google Scholar

19 

Du Q, Feng GZ, Shen L, Cui J and Cai JK: Paeonol attenuates airway inflammation and hyperresponsiveness in a murine model of ovalbumin-induced asthma. Can J Physiol Pharmacol. 88:1010–1016. 2010.PubMed/NCBI View Article : Google Scholar

20 

Kim SH, Kim SA, Park MK, Kim SH, Park YD, Na HJ, Kim HM, Shin MK and Ahn KS: Paeonol inhibits anaphylactic reaction by regulating histamine and TNF-α. Int Immunopharmacol. 4:279–287. 2004.PubMed/NCBI View Article : Google Scholar

21 

Yuan C, Xu X, Wang N, Zhu Q, Zhang J, Gong W, Ding Y, Xiao W, Chen W, Lu G, et al: Paeonol protects against acute pancreatitis by inhibiting M1 macrophage polarization via the NLRP3 inflammasomes pathway. Biochem Biophys Res Commun. 600:35–43. 2022.PubMed/NCBI View Article : Google Scholar

22 

Lyu ZK, Li CL, Jin Y, Liu YZ, Zhang X, Zhang F, Ning LN, Liang ES, Ma M, Gao W, et al: Paeonol exerts potential activities to inhibit the growth, migration and invasion of human gastric cancer BGC823 cells via downregulating MMP-2 and MMP-9. Mol Med Rep. 16:7513–7519. 2017.PubMed/NCBI View Article : Google Scholar

23 

Li M, Tan SY and Wang XF: Paeonol exerts an anticancer effect on human colorectal cancer cells through inhibition of PGE2 synthesis and COX-2 expression. Oncol Rep. 32:2845–2853. 2014.PubMed/NCBI View Article : Google Scholar

24 

Ou Y, Li Q, Wang J, Li K and Zhou S: Antitumor and apoptosis induction effects of paeonol on mice bearing EMT6 breast carcinoma. Biomol Ther (Seoul). 22:341–346. 2014.PubMed/NCBI View Article : Google Scholar

25 

Zhang L, Tao L, Shi T, Zhang F, Sheng X, Cao Y, Zheng S, Wang A, Qian W, Jiang L and Lu Y: Paeonol inhibits B 16 F 10 melanoma metastasis in vitro and in vivo via disrupting proinflammatory cytokines-mediated NF-κB and STAT 3 pathways. IUBMB life. 67:778–788. 2015.PubMed/NCBI View Article : Google Scholar

26 

Liu MH, Lin AH, Lee HF, Ko HK, Lee TS and Kou YR: Paeonol attenuates cigarette smoke-induced lung inflammation by inhibiting ROS-sensitive inflammatory signaling. Mediators Inflamm. 2014(651890)2014.PubMed/NCBI View Article : Google Scholar

27 

Ge Y, Pan M, Zhang C, Wang C, Ma K, Yan G, Wang T, Wu D and Shao J: Paeonol alleviates dextran sodium sulfate induced colitis involving Candida albicans-associated dysbiosis. Med Mycol. 59:335–344. 2021.PubMed/NCBI View Article : Google Scholar

28 

Ping M, Xiao W, Mo L, Xiao X, Song S, Tang W and Yang X: Paeonol attenuates advanced oxidation protein product-induced oxidative stress injury in THP-1 macrophages. Pharmacology. 93:286–295. 2014.PubMed/NCBI View Article : Google Scholar

29 

Liu CM, Yang HX, Ma JQ, Yang W, Feng ZJ, Sun JM, Cheng C, Li J and Jiang H: Role of AMPK pathway in lead-induced endoplasmic reticulum stress in kidney and in paeonol-induced protection in mice. Food Chem Toxicol. 122:87–94. 2018.PubMed/NCBI View Article : Google Scholar

30 

Kang B, Park DH, Lee MJ, Jeon CY, Kang KS and Choi YK: Beneficial effect of paeonol on antibiotic-associated inflammatory response in mice with diarrhea. Biomolecules. 12(1634)2022.PubMed/NCBI View Article : Google Scholar

31 

Meng Y, Liu Z, Zhai C, Di T, Zhang L, Zhang L, Zhang L, Xie X, Lin Y, Wang N, et al: Paeonol inhibits the development of 1-chloro-2, 4-dinitrobenzene-induced atopic dermatitis via mast and T cells in BALB/c mice. Mol Med Rep. 19:3217–3229. 2019.PubMed/NCBI View Article : Google Scholar

32 

Miao J, Ye P, Lan J, Ye S, Zhong J, Gresham A, Li S, You A, Chen X, Liu X and Li H: Paeonol promotes the phagocytic ability of macrophages through confining HMGB1 to the nucleus. Int Immunopharmacol. 89(107068)2020.PubMed/NCBI View Article : Google Scholar

33 

Sheng SQ, Yu LY, Zhou XW, Pan HY, Hu FY and Liu JL: Paeonol prevents migration and invasion, and promotes apoptosis of cervical cancer cells by inhibiting 5-lipoxygenase. Mol Med Rep. 23:1–8. 2021.PubMed/NCBI View Article : Google Scholar

34 

Pan M, Wang Q, Liu Y, Xiao N, Niu X, Wu D, Wang T, Yan G and Shao J: Paeonol enhances treatment of fluconazole and amphotericin B against oropharyngeal candidiasis through HIF-1α related IL-17 signaling. Med Mycol. 60(myac011)2022.PubMed/NCBI View Article : Google Scholar

35 

Wu J, Xue X, Zhang B, Jiang W, Cao H, Wang R, Sun D and Guo R: The protective effects of paeonol against epirubicin-induced hepatotoxicity in 4T1-tumor bearing mice via inhibition of the PI3K/Akt/NF-kB pathway. Chem Biol Interact. 244:1–8. 2016.PubMed/NCBI View Article : Google Scholar

36 

Wu J, Xue X, Zhang B, Cao H, Kong F, Jiang W, Li J, Sun D and Guo R: Enhanced antitumor activity and attenuated cardiotoxicity of Epirubicin combined with Paeonol against breast cancer. Tumor Biol. 37:12301–12313. 2016.PubMed/NCBI View Article : Google Scholar

37 

Zhou ZQ: Taxonomy, geographic distribution and ecological habitats of tree peonies. Genetic Resources and Crop Evolution. 53:11–22. 2006.

38 

Zhao M and Wu S: A review of the ethnobotany, phytochemistry and pharmacology of tree peony (Sect. Moutan). South African J Botany. 124:556–563. 2019.

39 

Wang X, Fan H, Li Y, Sun X, Sun X, Wang W and Zheng C: Analysis of genetic relationships in tree peony of different colors using conserved DNA-derived polymorphism markers. Scientia Horticulturae. 175:68–73. 2014.

40 

He L, Suo Z, Zhang C, Jin X, Zhao D, Zhao X, Hou B and Deng C: Classification of Chinese medicinal tree peony cultivars based on chloroplast DNA sequences. Aasri Procedia. 1:344–352. 2012.

41 

Feng Y, Yin L, Liu Y, Cao L, Zheng N, Li M and Zhan S: Quantitative determination and optimun extraction technique of nine compounds of Paeoniae Radix Alba. Zhejiang Da Xue Xue Bao Yi Xue Ban. 49:356–363. 2020.PubMed/NCBI View Article : Google Scholar : (In Chinese).

42 

Sun M, Huang L, Zhu J, Bu W, Sun J and Fang Z: Screening nephroprotective compounds from cortex Moutan by mesangial cell extraction and UPLC. Arch Pharm Res. 38:1044–1053. 2015.PubMed/NCBI View Article : Google Scholar

43 

Jiao MJ, Deng Z, Zhang J, Wang SH, Cui WJ, Zhang GY and Liu A: Preparation and quality standard of standard decoction of Chinese herbal medicine containing volatile components-case study of Moutan Cortex. Zhongguo Zhong Yao Za Zhi. 43:891–896. 2018.PubMed/NCBI View Article : Google Scholar : (In Chinese).

44 

Sun Y, Liu L, Shi XY, He H, Huang HW and Dai M: Paeonol inhibits macrophage M1 polarization by down-regulating miR-155/JAK1-STAT1 pathway. Zhongguo Zhong Yao Za Zhi. 45:2158–2164. 2020.PubMed/NCBI View Article : Google Scholar : (In Chinese).

45 

Liu Y, Li C, Wu H, Xie X, Sun Y and Dai M: Paeonol attenuated inflammatory response of endothelial cells via stimulating monocytes-derived exosomal MicroRNA-223. Front Pharmacol. 9(1105)2018.PubMed/NCBI View Article : Google Scholar

46 

Lou Y, Wang C, Tang Q, Zheng W, Feng Z, Yu X, Guo X and Wang J: Paeonol inhibits IL-1β-induced inflammation via PI3K/Akt/NF-κB pathways: In vivo and vitro studies. Inflammation. 40:1698–1706. 2017.PubMed/NCBI View Article : Google Scholar

47 

Lin C, Lin HY, Chen JH, Tseng WP, Ko PY, Liu YS, Yeh WL and Lu DY: Effects of paeonol on anti-neuroinflammatory responses in microglial cells. Int J Mol Sci. 16:8844–8860. 2015.PubMed/NCBI View Article : Google Scholar

48 

Chen N, Liu D, Soromou LW, Sun J, Zhong W, Guo W, Huo M, Li H, Guan S, Chen Z and Feng H: Paeonol suppresses lipopolysaccharide-induced inflammatory cytokines in macrophage cells and protects mice from lethal endotoxin shock. Fundam Clin Pharmacol. 28:268–276. 2014.PubMed/NCBI View Article : Google Scholar

49 

Niu Y, Jin Y, Hao Y, Liang W, Tang F, Qin Z, Liang T and Shi L: Paeonol interferes with lupus nephritis by regulating M1/M2 polarization of macrophages. Mol Immunol. 169:66–77. 2024.PubMed/NCBI View Article : Google Scholar

50 

Zhang Z, Chen T, Liu W, Xiong J, Jiang L and Liu M: Paeonol accelerates skin wound healing by regulating macrophage polarization and inflammation in diabetic rats. Korean J Physiol Pharmacol. 27:437–448. 2023.PubMed/NCBI View Article : Google Scholar

51 

Miao J, Liu X, Liao Y, Li Y, Kuang Y, Zheng J, Li Z and Lan J: Paeonol enhances macrophage phagocytic function by modulating lipid metabolism through the P53-TREM2 axis. Front Pharmacol. 14(1214756)2023.PubMed/NCBI View Article : Google Scholar

52 

Chen G, Guo T and Yang L: Paeonol reduces IL-β production by inhibiting the activation of nucleotide oligomerization domain-like receptor protein-3 inflammasome and nuclear factor-κB in macrophages. Biochem Cell Biol. 100:28–36. 2022.PubMed/NCBI View Article : Google Scholar

53 

He LX, Tong X, Zeng J, Tu Y, Wu S, Li M, Deng H, Zhu M, Li X, Nie H, et al: Paeonol suppresses neuroinflammatory responses in LPS-activated microglia cells. Inflammation. 39:1904–1917. 2016.PubMed/NCBI View Article : Google Scholar

54 

Li X, Shi H, Zhang D, Jing B, Chen Z, Zheng Y, Chang S, Gao L and Zhao G: Paeonol alleviates neuropathic pain by modulating microglial M1 and M2 polarization via the RhoA/p38MAPK signaling pathway. CNS Neurosci Ther. 29:2666–2679. 2023.PubMed/NCBI View Article : Google Scholar

55 

Adki KM and Kulkarni YA: Neuroprotective effect of paeonol in streptozotocin-induced diabetes in rats. Life Sci. 271(119202)2021.PubMed/NCBI View Article : Google Scholar

56 

Wu J, Wu D, Ma K, Wang T, Shi G, Shao J, Wang C and Yan G: Paeonol ameliorates murine alcohol liver disease via mycobiota-mediated Dectin-1/IL-1β signaling pathway. J Leucoc Biol. 108:199–214. 2020.PubMed/NCBI View Article : Google Scholar

57 

Wu S, Liu L, Yang S, Kuang G, Yin X, Wang Y, Xu F, Xiong L, Zhang M, Wan J and Gong X: Paeonol alleviates CCl4-induced liver fibrosis through suppression of hepatic stellate cells activation via inhibiting the TGF-β/Smad3 signaling. Immunopharmacol Immunotoxicol. 41:438–445. 2019.PubMed/NCBI View Article : Google Scholar

58 

Li J, Li Y, Pan S, Zhang L, He L and Niu Y: Paeonol attenuates ligation-induced periodontitis in rats by inhibiting osteoclastogenesis via regulating Nrf2/NF-κB/NFATc1 signaling pathway. Biochimie. 156:129–137. 2019.PubMed/NCBI View Article : Google Scholar

59 

Tang Y, Huang W, Song Q, Zheng X, He R and Liu J: Paeonol ameliorates ovalbumin-induced asthma through the inhibition of TLR4/NF-κB and MAPK signaling. Evid Based Complement Alternat Med. 2018(3063145)2018.PubMed/NCBI View Article : Google Scholar

60 

Gong X, Yang Y, Huang L, Zhang Q, Wan RZ, Zhang P, Wan RZ, Zhang P and Zhang B: Antioxidation, anti-inflammation and anti-apoptosis by paeonol in LPS/d-GalN-induced acute liver failure in mice. Int Immunopharmacol. 46:124–132. 2017.PubMed/NCBI View Article : Google Scholar

61 

Zong SY, Pu YQ, Xu BL, Zhang T and Wang B: Study on the physicochemical properties and anti-inflammatory effects of paeonol in rats with TNBS-induced ulcerative colitis. Int Immunopharmacol. 42:32–38. 2017.PubMed/NCBI View Article : Google Scholar

62 

Ding Y, Li Q, Xu Y, Chen Y, Deng Y, Zhi F and Qian K: Attenuating oxidative stress by paeonol protected against acetaminophen-induced hepatotoxicity in mice. PLoS One. 11(e0154375)2016.PubMed/NCBI View Article : Google Scholar

63 

Liao WY, Tsai TH, Ho TY, Lin YW, Cheng CY and Hsieh CL: Neuroprotective effect of paeonol mediates anti-inflammation via suppressing toll-like receptor 2 and toll-like receptor 4 signaling pathways in cerebral ischemia-reperfusion injured rats. Evid Based Complement Alternat Med. 2016(3704647)2016.PubMed/NCBI View Article : Google Scholar

64 

Lee H, Lee G, Kim H and Bae H: Paeonol, a major compound of moutan cortex, attenuates Cisplatin-induced nephrotoxicity in mice. Evid Based Complement Alternat Med. 2013(310989)2013.PubMed/NCBI View Article : Google Scholar

65 

Bihlet AR, Byrjalsen I, Andersen JR, Reynolds A, Larkins N, Alexandersen P, Rovsing H, Moots R and Conaghan PG: The efficacy and safety of a fixed-dose combination of apocynin and paeonol, APPA, in symptomatic knee OA: A double-blind, randomized, placebo-controlled, clinical trial. Osteoarthritis Cartilage. 32:952–962. 2024.PubMed/NCBI View Article : Google Scholar

66 

Zhang W, Cai J, Chen S, Zheng X, Hu S, Dong W, Lu J, Xing J and Dong Y: Paclitaxel resistance in MCF-7/PTX cells is reversed by paeonol through suppression of the SET/phosphatidylinositol 3-kinase/Akt pathway. Mol Med Rep. 12:1506–1514. 2015.PubMed/NCBI View Article : Google Scholar

67 

Cai J, Chen S, Zhang W, Hu S, Lu J, Xing J and Dong Y: Paeonol reverses paclitaxel resistance in human breast cancer cells by regulating the expression of transgelin 2. Phytomedicine. 21:984–991. 2014.PubMed/NCBI View Article : Google Scholar

68 

Zhang J, Cai L, Pang K, Dong Y, Zhang Z, Li B, Li R and Han CH: Paeonol inhibits proliferation and induces cell apoptosis of human T24 and 5637 bladder cancer cells in vitro and in vivo. Clin Transl Oncol. 23:601–611. 2021.PubMed/NCBI View Article : Google Scholar

69 

Liu LH, Shi RJ and Chen ZC: Paeonol exerts anti-tumor activity against colorectal cancer cells by inducing G0/G1 phase arrest and cell apoptosis via inhibiting the Wnt/β-catenin signaling pathway. Int J Mol Med. 46:675–684. 2020.PubMed/NCBI View Article : Google Scholar

70 

Li M, Tan SY, Zhang J and You HX: Effects of paeonol on intracellular calcium concentration and expression of RUNX3 in LoVo human colon cancer cells. Mol Med Rep. 7:1425–1430. 2013.PubMed/NCBI View Article : Google Scholar

71 

Li M, Cai O, Yu Y and Tan S: Paeonol inhibits the malignancy of Apatinib-resistant gastric cancer cells via LINC00665/miR-665/MAPK1 axis. Phytomedicine. 96(153903)2022.PubMed/NCBI View Article : Google Scholar

72 

Fu J, Yu L, Luo J, Huo R and Zhu B: Paeonol induces the apoptosis of the SGC-7901 gastric cancer cell line by downregulating ERBB2 and inhibiting the NF-κB signaling pathway. Int J Mol Med. 42:1473–1483. 2018.PubMed/NCBI View Article : Google Scholar

73 

Lv J, Zhu S, Chen H, Xu Y, Su Q, Yu G and Ma W: Paeonol inhibits human lung cancer cell viability and metastasis in vitro via miR-126-5p/ZEB2 axis. Drug Dev Res. 83:432–446. 2022.PubMed/NCBI View Article : Google Scholar

74 

Zhang L, Chen WX, Li LL, Cao YZ, Geng YD, Feng XJ, Wang AY, Chen ZL, Lu Y and Shen AZ: Paeonol suppresses proliferation and motility of non-small-cell lung cancer cells by disrupting STAT3/NF-κB signaling. Front Pharmacol. 11(572616)2020.PubMed/NCBI View Article : Google Scholar

75 

Lei Y, Li HX, Jin WS, Peng WR, Zhang CJ, Bu LJ, Du YY, Ma T and Sun GP: The radiosensitizing effect of Paeonol on lung adenocarcinoma by augmentation of radiation-induced apoptosis and inhibition of the PI3K/Akt pathway. Int J Radiat Biol. 89:1079–1086. 2013.PubMed/NCBI View Article : Google Scholar

76 

Zhou J, Liu Q, Qian R, Liu S, Hu W and Liu Z: Paeonol antagonizes oncogenesis of osteosarcoma by inhibiting the function of TLR4/MAPK/NF-κB pathway. Acta Histochem. 122(151455)2020.PubMed/NCBI View Article : Google Scholar

77 

Gao L, Wang Z, Lu D, Huang J, Liu J and Hong L: Paeonol induces cytoprotective autophagy via blocking the Akt/mTOR pathway in ovarian cancer cells. Cell Death Dis. 10(609)2019.PubMed/NCBI View Article : Google Scholar

78 

Han L, Guo XJ, Chen Z, Bian H, Zhang CY, Zang WH, Wang Q and Hu JL: The mechanisms of paeonol reversing multidrug resistance in ovarian cancer SKOV3/DDP cells. Acta Pharm Sin. 53:1511–1517. 2018.

79 

Li B, Yang J, Hong L, Tang J, Li Q and Fu Q: Paeonol induces apoptosis of ovarian cancer cells through the AKT/GSK-3β signaling pathway. Int J Clin Exp Med. 10:10170–10178. 2017.

80 

Zhou HM, Sun QX and Cheng Y: Paeonol enhances the sensitivity of human ovarian cancer cells to radiotherapy-induced apoptosis due to downregulation of the phosphatidylinositol-3-kinase/Akt/phosphatase and tensin homolog pathway and inhibition of vascular endothelial growth factor. Exp Ther Med. 14:3213–3220. 2017.PubMed/NCBI View Article : Google Scholar

81 

Yin J, Wu N, Zeng F, Cheng C, Kang K and Yang H: Paeonol induces apoptosis in human ovarian cancer cells. Acta Histochem. 115:835–839. 2013.PubMed/NCBI View Article : Google Scholar

82 

Xu Y, Zhu JY, Lei ZM, Wan LJ, Zhu ZW, Ye F and Tong YY: Anti-proliferative effects of paeonol on human prostate cancer cell lines DU145 and PC-3. J Physiol Biochem. 73:157–165. 2017.PubMed/NCBI View Article : Google Scholar

83 

Horng CT, Shieh PC, Tan TW, Yang WH and Tang CH: Paeonol suppresses chondrosarcoma metastasis through up-regulation of miR-141 by modulating PKCδ and c-Src signaling pathway. Int J Mol Sci. 15:11760–11772. 2014.PubMed/NCBI View Article : Google Scholar

84 

Cheng CS, Chen JX, Tang J, Geng YW, Zheng L, Lv LL, Chen LY and Chen Z: Paeonol inhibits pancreatic cancer cell migration and invasion through the inhibition of TGF-β1/smad signaling and epithelial-mesenchymal-transition. Cancer Manag Res. 29:641–651. 2020.PubMed/NCBI View Article : Google Scholar

85 

Du J, Song D, Li J, Li Y, Li B and Li L: Paeonol triggers apoptosis in HeLa cervical cancer cells: The role of mitochondria-related caspase pathway. Psychopharmacology (Berl). 239:1–10. 2021.PubMed/NCBI View Article : Google Scholar

86 

Chen Y, Jia Y, Li Y, Zheng Y, Chen G and Shi Y: Investigation on the antitumor effects of paeonol against renal cell carcinoma based on network pharmacology and experimental validation. J Ethnopharmacol. 285(114857)2022.PubMed/NCBI View Article : Google Scholar

87 

Cai M, Shao W, Yu H, Hong Y and Shi L: Paeonol inhibits cell proliferation, migration and invasion and induces apoptosis in hepatocellular carcinoma by regulating miR-21-5p/KLF6 axis. Cancer Manag Res. 17:5931–5943. 2020.PubMed/NCBI View Article : Google Scholar

88 

Li Q, Zhang Y, Sun J and Bo Q: Paeonol-mediated apoptosis of hepatocellular carcinoma cells by NF-κB pathway. Oncol Lett. 17:1761–1767. 2019.PubMed/NCBI View Article : Google Scholar

89 

Fan L, Song B, Sun G, Ma T, Zhong F and Wei W: Endoplasmic reticulum stress-induced resistance to doxorubicin is reversed by paeonol treatment in human hepatocellular carcinoma cells. PLoS One. 8(e62627)2013.PubMed/NCBI View Article : Google Scholar

90 

Ramachandhiran D, Vinothkumar V and Babukumar S: Paeonol exhibits anti-tumor effects by apoptotic and anti-inflammatory activities in 7,12-dimethylbenz (a) anthracene induced oral carcinogenesis. Biotech Histochem. 94:10–25. 2019.PubMed/NCBI View Article : Google Scholar

91 

Chen X, Xu Z, Lu M, Ding W, Zhong J, Deng S, Li S, Miao J, Liu X, Wen Q, et al: Paeonol inhibits melanoma growth by targeting PD1 through upregulation of miR-139-5p. Biochem Biophys Res Commun. 656:86–96. 2023.PubMed/NCBI View Article : Google Scholar

92 

Tsai CY, Kapoor M, Huang YP, Lin HH, Liang YC, Lin YL, Huang SC, Liao WN, Chen JK, Huang JS and Hsu MH: Synthesis and evaluation of aminothiazole-paeonol derivatives as potential anticancer agents. Molecules. 21(145)2016.PubMed/NCBI View Article : Google Scholar

93 

Adki KM, Murugesan S and Kulkarni YA: In silico and in vivo toxicological evaluation of paeonol. Chem Biodivers. 17(e2000422)2020.PubMed/NCBI View Article : Google Scholar

94 

Chang X, Feng X, Du M, Li S, Wang J, Wang Y and Liu P: Pharmacological effects and mechanisms of paeonol on antitumor and prevention of side effects of cancer therapy. Front Pharmacol. 14(1194861)2023.PubMed/NCBI View Article : Google Scholar

95 

Cheng S, Chen W, Guo Z, Ding C, Zuo R, Liao Q and Liu G: Paeonol alleviates ulcerative colitis by modulating PPAR-γ and nuclear factor-κB activation. Sci Rep. 14(18390)2024.PubMed/NCBI View Article : Google Scholar

96 

Himaya S, Ryu B, Qian ZJ and Kim SK: Paeonol from Hippocampus kuda Bleeler suppressed the neuro-inflammatory responses in vitro via NF-κB and MAPK signaling pathways. Toxicology In Vitro. 26:878–887. 2012.PubMed/NCBI View Article : Google Scholar

97 

Morsy MA, Ibrahim YF, Hafez SMN, Zenhom NM, Nair AB, Venugopala KN, Shinu P and Abdel-Gaber SA: Paeonol attenuates hepatic ischemia/reperfusion injury by modulating the Nrf2/HO-1 and TLR4/MYD88/NF-κB signaling pathways. Antioxidants (Basel). 11(1687)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-June 2025
Volume 5 Issue 3

Print ISSN: 2754-3242
Online ISSN:2754-1304

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Barnes P, Agbo E, Halm‑Lai F, Dankwa K, Saahene R, Nuvor S, Obiri‑Yeboah D and Yahaya E: Insight into the immunomodulatory and chemotherapeutic mechanisms of paeonol (Review). Med Int 5: 24, 2025.
APA
Barnes, P., Agbo, E., Halm‑Lai, F., Dankwa, K., Saahene, R., Nuvor, S. ... Yahaya, E. (2025). Insight into the immunomodulatory and chemotherapeutic mechanisms of paeonol (Review). Medicine International, 5, 24. https://doi.org/10.3892/mi.2025.223
MLA
Barnes, P., Agbo, E., Halm‑Lai, F., Dankwa, K., Saahene, R., Nuvor, S., Obiri‑Yeboah, D., Yahaya, E."Insight into the immunomodulatory and chemotherapeutic mechanisms of paeonol (Review)". Medicine International 5.3 (2025): 24.
Chicago
Barnes, P., Agbo, E., Halm‑Lai, F., Dankwa, K., Saahene, R., Nuvor, S., Obiri‑Yeboah, D., Yahaya, E."Insight into the immunomodulatory and chemotherapeutic mechanisms of paeonol (Review)". Medicine International 5, no. 3 (2025): 24. https://doi.org/10.3892/mi.2025.223