Next Article in Journal
Periplaneta americana (L.) Extract PAS840 Promotes Ischemic Stroke Recovery by Inhibiting Inflammasome Activation
Previous Article in Journal
Role of ACE1, ACE2, and CCR5-Δ32 Polymorphisms in the Transmission of SARS-CoV-2 to Intimate Contacts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Osthole: A Coumarin with Dual Roles in Biology and Chemistry

1
School of Marine Sciences, Ningbo University, Ningbo 315832, China
2
College of Plant Protection, Northwest A&F University, Xianyang 712100, China
*
Author to whom correspondence should be addressed.
Biology 2025, 14(6), 588; https://doi.org/10.3390/biology14060588
Submission received: 20 March 2025 / Revised: 10 May 2025 / Accepted: 13 May 2025 / Published: 22 May 2025

Simple Summary

Osthole is a natural coumarin-like compound isolated from various plant species. Osthole and its derivatives have received much attention due to their lots of interesting biological activities. In order to timely summarize their recent advances on biology and chemistry from 2018 to early 2025, here the biological activities, mechanisms of action, total synthesis, structural modifications, structureactivity relationships (SARs) of osthole and its derivatives are presented. Hopefully, this review can be used as the useful reference for researchers interested in the biology and chemistry of osthole derivatives for further in-depth research.

Abstract

Osthole is a natural coumarin-like compound isolated from the Fructus cnidii. In the last few years, this plant-derived product and its derivatives have aroused much attention for their interesting biological activities, including anticancer, anti-inflammatory, neuroprotective, and insecticidal effects. This review summarizes the recent progress on the biological activities of osthole and its derivatives from 2018 to early 2025, with a focus on their total synthesis, structural modifications, and mechanisms of action. Additionally, structureactivity relationships (SARs) of osthole derivatives are presented. This review aims to serve as a comprehensive reference for future research on osthole and its derivatives in both medicinal and agricultural applications.
Key Contribution: This review presents an overview on the biological activities; mechanisms of action; structural modifications; and structure–activity relationships of osthole and its derivatives.

1. Introduction

Coumarin (Figure 1), which contains a 2 H-benzopyran-2-one core structure, is a naturally occurring compound with an aromatic odor that is widely distributed in various plant species. Vogel first isolated it from tonka beans (Dipteryx odoranta Wild; Fabaceae family) in 1820. Since then, thousands of natural coumarins have been isolated, described, and synthesized, and their biological activities have been studied from a variety of sources, including plants, bacteria, fungi, and chemical synthesis [1]. Osthole (1, Figure 1), often referred to as osthol (7-methoxy-8-(3-methyl-2-butenyl) coumarin), is a coumarin analog that was initially isolated from the Cnidium plant [2]. Osthole has shown many biological properties, including anticancer [3], anti-inflammatory [4], osteogenic [5], and neuroprotective activities, without significant toxic side effects [6]. It also has insecticidal activity in agriculture [7].
In this paper, recent advances on biological activities, total synthesis, and structural modifications of osthole and its derivatives from 2018 to early 2025 were reviewed. In addition, their structureactivity relationships (SARs) and mechanisms of action were presented for further in-depth research.

2. Bioactivities of Osthole and Its Derivatives

The biological activities of osthole and its derivatives are outlined in Figure 2.

2.1. Anticancer Activity

Osthole has broad-spectrum anticancer activities against a range of tumor cells (Table 1), including endometrial carcinoma (EC) cells [8], breast cancer cells [9,10,11,12,13], ovarian carcinoma (OC) cells [14,15], colorectal (CRC) cancer cells [16,17,18,19,20], hepatocellular carcinoma (HCC) cells [21,22,23,24], bladder cancer cells [25], retinoblastoma (RB) cells [26], cervical cancer cells [27,28,29], head and neck squamous cell carcinoma (HNSCC) cells [30], glioma cells [31,32,33], gallbladder cancer (GBC) cells [34], lung cancer cells [35], human gastric cancer cells [36,37], esophageal squamous cell carcinoma (ESCC) cells [38], rhabdomyosarcoma (RMS) cells [39], pancreatic cancer cells [40], and nasopharyngeal cancer (NPC) cells [41]. Osthole can inhibit the PI3K/Akt signaling pathway in various cancer cells, such as in endometrial carcinoma, bladder cancer, retinoblastoma, cervical cancer, head and neck squamous cell carcinoma, and gastric cancer, which causes cancer cell proliferation, migration, invasion, and death [25,26,27,29,30,36]. Osthole has inhibited 4T1 cell proliferation through mTOR/SREBP1/FASN signaling, blocked cell entry into s phase, and induced apoptosis in breast cancer cells [11]. It can decrease the ASC/caspase-1/IL-1β inflammasome pathway and control colorectal cancer progression by regulating autophagy and mitochondrial signaling [17]. Osthole has inhibited glycolysis regulated by the GSK-3β/AMPK/mTOR pathway, increasing radiosensitivity of subcutaneously transplanted hepatocellular carcinoma in nude mice [21].

2.2. Immunomodulatory and Anti-Inflammatory Activities

Natural products offer several benefits, including the regulation of immune cell differentiation, mitigation of oxidative stress, and a reduction in inflammation. Osthole has acted on DSS-induced UC and LPS-induced ALI in mice by regulating p38 and JNK phosphorylation in the MAPK pathway [42]. Osthole has the potential to treat polycystic ovarian syndrome in mice by activating the Nrf2-Foxo1-GSH-NF-κB pathway [43]. Osthole can effectively prevent DSS-induced ulcerative colitis (UC) and LPS-induced acute lung injury (ALI) [42,44] and inhibit oleic acid/lipopolysaccharide-induced lipid accumulation and inflammatory response in hepatocytes via activating the PPARα signaling pathway [45,46]. Oshtole can treat rheumatoid arthritis (RA) and suppress knee osteoarthritis (KOA) by enhancing autophagy activated by the AMPK/ULK1 pathway [47,48,49,50]. Osthole significantly reduced skin damage and chronic itching associated with atopic dermatitis (AD) by increasing ZO-3 expression and blocking Akt phosphorylation in an AD mouse model [51,52,53,54], as well as having a protective effect against lipopolysaccharide-induced inflammation in BV2 cells [55,56]. Oshthole can ameliorate neurogenic and inflammatory nociceptive sensitization through the modulation of iNOS, COX-2, and inflammatory cytokines, and reduce the role of COX-2 in inflammation and ASD development [57,58].

2.3. Neuroprotective Effects

Osthole has had a neuroprotective impact on Alzheimer’s disease. Intranasal administration of osthole/borneols thermosensitive gel enhanced intracerebral bioavailability and reduced cognitive impairment in APP/PS1 transgenic mice [59,60,61,62]. Osthole can effectively reduce Parkinson’s disease symptoms by inhibiting JAK/STAT and MAPK pathways through attenuating 6-OHDA cytotoxicity in SH-SY5Y cells [63,64,65]. Osthole may delay neural stem cell (NSC) senescence through the p16-pRb signaling pathway [66]. Osthole provided protection against mechanical brain injury, effectively promoted cognitive dysfunction and neuronal recovery, and improved neurological function [67]. Osthole showed a substantial ability to lessen seizures [68]. Osthole reduced pain depression by inhibiting inflammation and oxidative stress-mediated neurotransmitter dysregulation [69].

2.4. Osteogenic Effects and Cardiovascular Benefits

Osthole may be used therapeutically to prevent osteoporosis [70]. Osthole has promoted osteogenic differentiation and inhibited osteoclastogenesis and osteoblast activity [71,72,73]. Osthole has been shown to have cardioprotective properties [74,75], and it can promote autophagy and protect rats from cardiac ischemia/reperfusion injury [76,77].

2.5. Hepatoprotective Effects and Therapeutic Potential in Metabolic Disease

Osthole has protected against D-Gal-induced liver injury through TLR4/MAPK/NF-κB pathways, and also prevented tamoxifen- and acetaminophen-induced liver injury [78,79,80]. Osthole has inhibited renal fibrosis through the IL-11/ERK1/2 signaling pathway, which is independent of TGF-β1 signaling [81,82]. Osthole has had the potential to help prevent and treat diabetic kidney disease (DKD) [83].

2.6. Antimicrobial and Antiparasitic Activities

Osthole has demonstrated inhibitory effects against a wide range of viruses, bacteria, and parasites. Natural products represent valuable sources of novel antimicrobial agents. Against Listeria monocytogenes, osthole strongly decreased Na+-K+-ATPase and Ca2+-Mg2+-ATPase activities, increased the production of reactive oxygen species (ROS), and disrupted the integrity and permeability of cell membranes [84,85,86]. Osthole has been successfully developed for its significant inhibitory effect on phytopathogenic fungi (Table 2) [87,88,89,90]. Osthole can effectively inhibit SARS-CoV-2 as well as tobacco mosaic virus (TMV) infection [91,92]. Furthermore, osthole demonstrated a high activity against Rhipicephalus sanguineus [93].

2.7. Insecticidal Activity

Osthole and its derivatives have shown pesticidal effects on Mythimna separata, Myzus persicae, Tetranychus cinnabarinus, and Plutella xylostella Linnaeus [94,95,96]. It can also be a strong attractant for oviposition on Bactrocera dorsalis [97]. Osthole had a significant lethal effect on the second instar larvae of Spodoptera frugiperda [98]. Tetranychus urticae Koch can be controlled by combining matrine and osthole in a certain ratio [99]; osthole had good contact and stomach poisoning effects on Coptotermes formosanus, Sitophilus zeamais Motschusky, and Plutella xylostella Linnaeus [100,101,102]. After treatment of osthole at 125 × 10−3 mg/mL for 24 h, the glutathione-S-transferase (GST) and acetylcholinesterase (AChE) activities in the P. xylostella larvae were significantly reduced. Furthermore, osthole showed larvicidal activity against mosquitoes [7,103].

2.8. Other Activities

Osthole can exert antipruritic effects by inhibiting or desensitizing cation channels TRPV1 and TRPV3 [104,105]. Osthole inhibited phosphodiesterase 4D activity, making it a promising bronchodilator for asthma treatment without targeting β2-adrenoceptors [106,107]. Osthole can also dramatically lower blood pressure by inhibiting the vascular constriction mediated by angiotensin II and CaCl2 [108].

3. Total Synthesis of Osthole and Its Derivatives

As described in Scheme 1, Liu et al. reported the synthesis of osthole (1) by 8-allyl-7-methoxycoumarin olefin complexation using 2,4 dihydroxybenzaldehyde (2) as a raw material [109]. Compound 3 was formed by reacting compound 2 with ethyl 2-chloroacetate. Subsequently, in the presence of potassium carbonate and potassium iodide, compound 3 was reacted with 3-chloropropene to afford compound 4, which was then C-8 Claisen rearranged to give compound 5. Compound 5 was methylated with dimethyl sulfate in acetone to give compound 6. Ultimately, compound 1 was created with a 77.6% yield by reacting compound 6 with 3-chloro-3-methyl-1-butylene. Meanwhile, Yin et al. developed a palladium-catalyzed coupling reaction of tributyl (3-methylbut-2-en-1-yl) stannane with 7-methoxy-8-iodocoumarin (8), which was from 7-hydroxy-8-iodocoumarin (7), to synthesize compound 1 [110].
The use of a microwave-promoted tandem Claisen rearrangement/olefin/cyclization method was an efficient approach for the synthesis of compound 1. As shown in Scheme 2, Konrádová et al. prepared compound 1 from an aldehyde synthon (9) by substitution and reduction reactions at a 78% yield [111]. Schmidt et al. obtained compound 11 straight from compound 9 by substitution reaction, skipping the reduction step from compound 10 to compound 11, and the yield of compound 1 was 59% [112]. Bromination or iodination of 2-hydroxy-4-methoxybenzaldehyde (9) with Br2 or NIS in the presence of aluminum chloride produced compounds 12a and 12b. These compounds were then reacted with the Wittig reagent to provide compounds 13a and 13b. The C-H activation/cyclization of compounds 13a and 13b was catalyzed by rhodium (III) to give coumarins 14a and 14b, respectively. Finally, compound 1 was synthesized from compound 14a by Stille cross-coupling reaction [113]. Compared to palladium and nickel reagents, Grignard reagents align better with sustainable development principles. They can be modified into highly active derivatives by incorporating inexpensive and environmentally friendly transition metals, such as iron, zinc, and copper. Liu et al. therefore employed a non-precious metal-assisted Grignard reagent (using CuI and LiCl as promoters) to synthesize compound 1 from compound 14b at an 80% yield. [7].
As shown in Scheme 3, a series of oxime derivatives of (E)-4-chloro-2-oxo-2H-chromene-3-carbaldehyde (19a19z and 19a’19d’) were prepared through the successive three-step reactions from 2-hydroxyacetophenones (15a15e) [114]. Among them, target compounds 19a (EC50: 8.06 µg/mL), 19g (EC50: 4.75 µg/mL), and 19n (EC50: 5.43 µg/mL) showed a slightly lower inhibitory effect against Botrytis cinerea than boscalid (EC50: 0.88 µg/mL). In addition, the majority of compounds exhibited similar inhibitory effectiveness to boscalid against Alternaria solani; in particular, compounds 19c (EC50: 5.39 µg/mL) and 19g (EC50: 5.80 µg/mL) showed greater inhibitory effects than boscalid (EC50: 13.60 µg/mL). Against Alternaria alternata, compound 19g (EC50: 5.74 µg/mL) displayed comparable inhibitory activity to boscalid (EC50: 2.55 µg/mL).
As shown in Scheme 4, Ma et al. developed a series of coumarin derivatives using ring-merging and ring-opening strategies [115]. The intermediates 2022 were synthesized in multistep reactions using 2,4-dihydroxybenzaldehyde (2) as the raw material. The treatment of compound 20 with the corresponding bromides provided compounds 23a23c. Acylation of 2022 gave derivatives 24a24d, 25a25c, and 26a26e. Compound 24a (EC50: 1.56 µg/mL) showed high inhibitory activity against Pyricularia grisea, which was more than 3 times more potent than chlorothalonil (EC50: 5.81 µg/mL). Compound 24a showed good lethal activity against Ostrinia nubilalis, and its fatality rate was 67%, which was similar to that of matrine.
As shown in Scheme 5, a highly effective microwave-promoted technique was created for the synthesis of a number of pyrano [3,2-c]chromene-2,5-diones (28a28o) and furo [3,2-c]coumarins (29(ad)–33(ad)) [116,117]. Among them, compounds 30c, 32b, 32c, and 33b showed high inhibitory activity against B. cinerea, Colletotrichum capsica, and Rhizoctorzia solani. In particular, compound 30c was more effective than azoxystrobin (EC50: 0.158 µM) against B. cinerea (EC50: 0.110 µM) and C. capsica (EC50: 0.040 µM), making it the most promising choice. Additionally, compound 33b (EC50: 0.051 µM) was equally as effective as the control azoxystrobin (EC50: 0.053 µM) against R. solani. As shown in Scheme 6, two series of 7-pyrazolecoumarins (36a36p) and 7-pyrrolecoumarins (38a38h) were synthesized from m-aminophenol (34) via the intermediates 35a35h and 37 [118]. In particular, compounds 36g, 38ad, and 38h showed good antifungal efficacy against Cucumber anthrax and Alternaria leaf spot, which was superior to the control osthole. Compared to osthole (EC50: 67.2 µg/mL), compound 36g exhibited a greater inhibitory effect against R. solani (EC50: 15.4 µg/mL).

4. Structural Modifications of Osthole and Its Derivatives

Structural modifications of osthole at different positions are outlined in Figure 3.

4.1. Structural Modifications on the Lactone Ring of Osthole

One of the key methods for finding novel insecticides is the structural modification of natural products (NPs). By opening the lactone of osthole (1), Xu et al. produced two series of amide/ester derivatives of osthole [95]. As seen in Scheme 7, compounds 41b, 41l, 42l, 42m, 44h, 44l, and 44m showed more pronounced growth inhibition against M. separata than the precursor osthole, with corrected final mortality rates (CFMRs) of 62.0–68.9% at 1 mg/mL. The acaricidal activity of compounds 41b (LC50: 0.238 mg/mL), 41i (LC50: 0.226 mg/mL), and 42m (LC50: 0.205 mg/mL) against T. cinnabarinus was 5.7–6.6 times higher than that of osthole (LC50: 1.359 mg/mL); thus, compounds 41b and 42m have potential for further development as pesticides.
As shown in Scheme 8, a series of novel osthole-type ester derivatives (47a47z) was prepared by modifying the osthole lactone ring [119]. Among them, compounds 47c (LC50: 0.31 mg/mL), 47d (LC50: 0.24 mg/mL), and 47e (LC50: 0.28 mg/mL) showed strong acaricidal activity against T. cinnabarinus, as well as high control efficiency. In particular, compound 47d had more than five times the acaricidal activity of precursor 1 (LC50: 1.22 mg/mL), and compounds 47i, 47n, and 47o had 1.6–1.8 times the insecticidal activity against M. separata compared to 1. Scheme 9 illustrates how N-hydroxyamide 51 was made by hydrolyzing 1 [120]. Compound 51 inhibited angiogenesis and suppressed tumor cell proliferation by targeting VEGFR2 signaling. In addition, HDAC6-induced Hsp90 hyperacetylation, and HDAC6-enhanced cyclin D1 and CDK4 degradation may be mediated by N-hydroxyamide 52-induced G1 growth arrest [121].

4.2. Structural Modifications at the C-7 Position of Osthole

As depicted in Scheme 10, Zhou et al. prepared a series of osthole derivatives and tested their anti-inflammatory properties [42]. The majority of them were able to successfully suppress the release of inflammatory cytokines, including IL-6 in mouse macrophages J774A activated by LPS. Compound 56m (IC50: 4.57 μM) showed the strongest in vitro anti-inflammatory activity, which was 32 times higher than that of osthole. Compound 56m significantly reduced DSS-induced ulcerative colitis and LPS-induced acute lung damage, and it is a potential anti-inflammatory candidate. As described in Scheme 11, Zhang et al. used an MTT assay to evaluate the cytoprotective ability of osthole-based compounds [122]. At 10 μM, compound 59c demonstrated significant neuroprotective efficacy against H2O2 (45.7 ± 5.5%), oxygen glucose deprivation (64.6 ± 4.8%), and Aβ42 (61.4 ± 5.2%).
The ability of compound 60c (50.4 ± 7.1%) to suppress NO has outperformed that of the positive medication indomethacin. The neuroprotective effect of osthole may be improved by the addition of piperazine, tetrahydropyrrole, and aromatic amine groups.

4.3. Structural Modifications at the C-8 Position of Osthole

The carboxylic acid ester scaffold, as a potent pharmacophore, is widely utilized in structural modification strategies [123]. As depicted in Scheme 12, Hao et al. designed and synthesized a series of osthole ester derivatives (66a66y) and evaluated their agricultural bioactivities [124]. Compound 66n exhibited significant insecticidal activity against M. separata as compared to that of precursor 1. Compounds 66i and 66j demonstrated 2.6 and 3.3 times the acaricidal activity of 1, and good control effects in the glasshouse. As shown in Scheme 13, Hao et al. prepared two series of novel ester and amide derivatives modifying the C-4′ position of osthole (68a68w and 69a69j) [125]. Compounds 68b (LC50: 0.492 mg/mL) and 68e (LC50: 0.509 mg/mL) showed 3.2 and 3.1 times more acaricidal activity than 1 (LC50: 1.578 mg/mL) against T. cinnabarinus, and the aphidicidal activities of compounds 68w (LC50: 0.039 mg/mL), 69f (LC50: 0.034 mg/mL), and 69g (LC50: 0.038 mg/mL) against Aphis citricola were 1.9–2.1 times that of 1 (LC50: 0.073 mg/mL).
As shown in Scheme 14, Ren et al. regioselectively semi-prepared series of novel derivatives (70a70s) containing hydrazone/acylhydrazone/sulfonylhydrazone skeletons at the C-8 position of osthole [126]. Benzoylhydrazone 70b demonstrated 1.6 times the insecticidal activity of 1 against M. separata. The insecticidal activity can be enhanced by adding acylhydrazones to the 3′-methyl-2′-butylenyl fragment at the C-8 position of the osthole. As shown in Scheme 15, a number of osthole-based N-hydroxycinnamates (7179) were prepared and examined for their ability to inhibit enzymes [127]. Comparable to that of suberoylanilide hydroxamic acid (SAHA; IC50: 24.5 nM), compounds 74d (IC50: 24.5 nM), 74e (IC50: 20.0 nM), and 74g (IC50: 19.6 nM) showed good inhibitory effects against nuclear HDACs in HeLa cells. Furthermore, three different kinds of osthole derivatives were combined with SAHA-like hydroxamates. Compounds 78c, 78d, and 78c had greater anti-HDAC-8 efficacy in addition to SAHA-like activity against HDAC-1, HDAC-4, and HDAC-6 [128].
A series of oxime ester derivatives of osthole (81a81z, Scheme 16) were synthesized by selectively oxidizing 1 in the presence of selenium dioxide [129]. The acaricidal activity of compound 81c (LC50: 0.316 mg/mL) against T. cinnabarinus was more than three times that of its precursor 1 (LC50: 1.011 mg/mL), and it was particularly effective in the greenhouse. Among them, compounds 81c and 81f had the strongest acaricidal activity against M. separata, with final mortality rates of 70.4% and 70.4%, respectively. Scheme 17 illustrated the construction of novel osthole esters (83a83s) containing the isoxazoline fragment [130]. Compounds 82 (LC50: 0.75 mg/mL) and 83h (LC50: 0.76 mg/mL) showed more than 1.5 times the acaricidal activity against T. cinnabarinus than 1 (LC50: 1.14 mg/mL), and also had good control in the greenhouse. At 1 mg/mL, compounds 83a (FMR: 55.1%), 83c (FMR: 62.0%), and 83o (FMR: 55.1%) showed 1.5–1.6 times the growth inhibition against M. separata compared to 1. The addition of the acryloyloxy group to the C-4′ position of osthole was critical for insecticidal and acaricidal properties. As shown in Scheme 18, the new dihydropyrimidinone analog of osthole (84) was synthesized by the Biginelli reaction [131]. Compound 84 exhibited potent inhibitory effects on colon (Colo-205), prostate (PC-3), leukemia (THP-1), and lung (A549) cancer cell lines, as well as on normal cell line fR-2, with IC50 values of 37.6, 40.8, 65.8, 67.8, and 68.8 µM, respectively.

4.4. Structural Modifications at the C-7 and C-8 Positions and on the Lactone Ring of Osthole

As described in Scheme 19, a series of new 2-isopropanol-4-methoxy-7-alkyl/aryloxycarbonyl-(E)-vinyl-2,3-dihydrobenzofuran derivatives (87a87z and 87a’87h’) were designed and synthesized. Among all derivatives, compound 87b’ exhibited promising insecticidal activity against P. xylostella with an LC50 value of 0.759 mg/mL, which was higher than that of 1 (LC50: 1.415 mg/mL). Additionally, compound 87e’ displayed 3.3 times higher acaricidal activity and good control effects compared to those of 1 [94]. As shown in Scheme 20, Ren et al. used osthole as raw material to prepare a series of new acrylate derivatives (90a90z and 90a’90m’) of isopropene-2,3-dihydrobenzofuran via consecutive bromination, rearrangement, and esterization processes [96]. Among them, the insecticidal activity of compound 90d’ (LC50: 0.368 mg/mL) against P. xylostella was 3.5 times greater than that of 1 (LC50: 1.281 mg/mL). The acaricidal activity of compound 90l’ (LC50: 0.165 mg/mL) was 8.3 times higher than that of 1 (LC50: 1.367 mg/mL) against T. cinnabarinus, and it could be further developed as an acaricide. As illustrated in Scheme 21, You et al. synthesized a series of 3-aryl-substituted derivatives (92a92j) by structural modification at the C-7 and C-8 positions and on the lactone ring of osthole [132]. Some derivatives showed good inhibitory effects, among which compound 92e inhibited MCF-7 and MDA-MB-231, with IC50 values of 0.24 and 0.31 µM, respectively, surpassing the parent compound osthole by over 100 times. The 7-methoxy and 3-aryl ring were critical for preserving cytotoxicity potency.

5. StructureActivity Relationships of Osthole and Its Derivatives

As described in Figure 4, the structureactivity relationships (SARs) of osthole and its derivatives can be generally summarized as follows: (i) the introduction of an aromatic ring at the C-3 position of osthole can improve anticancer activity [120,121]. Incorporating heterocyclic fused fragments at the C-3 and C-4 positions resulted in derivatives showing good antifungal activity [116,117]. Opening the lactone ring and introducing aliphatic chain ester or amide groups were necessary for the insecticidal activity [94,95,119]. Additionally, the introduction of an N-hydroxylamide group can also augment anticancer activity [120,121]. (ii) The introduction of the pyrrole/pyrazole group at the C-7 position was necessary for the antifungal activity [118]. Adding the N-hydroxycinnamate group can also improve the anticancer activity [127]. Moreover, the introduction of AMP mimics, flexible carbon chains, and amide groups were beneficial for anti-inflammatory activity [42]. (iii) The isopentenyl fragment underwent oxidation and was subsequently converted into ester derivatives that exhibited inhibitory activity against acetylcholinesterase [62,124]. In particular, the introduction of N-benzoylthioureas/isoxazolines, hydrazones/acylhydrazones/sulfonylhydrazones, oxime esters, amides, and carboxylate esters groups were important for the pesticidal activity [125,126,131].

6. Conclusions

Recent advances in the biological activities, total synthesis, and structural modification of osthole and its derivatives were reviewed in the present work. Furthermore, the mechanisms of action and structureactivity relationships (SARs) of osthole and its derivatives were summarized. To find more pronounced compounds, extensive structural modification of osthole and its derivatives should be performed, and the scope of bioactivities can be broadened. With ongoing in-depth research on osthole and its derivatives, it is anticipated that the development of more osthole-type products will promote their practical applications in medicinal and agricultural fields, thereby contributing to human health and sustainable agricultural development.

Author Contributions

Conceptualization, M.L., H.D. and H.X.; methodology, M.L. and H.D.; software, H.D.; validation, M.L., H.D. and H.X.; formal analysis, M.L. and H.D.; resources, M.L., H.D. and H.X.; data curation, H.D.; writing—original draft preparation, H.D.; writing—review and editing, M.L., H.D. and H.X.; supervision, M.L. and H.X.; project administration, M.L.; funding acquisition, M.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research supported by the Scientific Research Fund of Ningbo University (project No. ZX2025000463).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AktProtein kinase B
AMPAdenosine 5′-monophosphate
AMPKAdenosine 5′-monophosphate-activated protein kinase
ASDAutistic spectrum disorders
COX-2Cyclooxygenase 2
CREBcAMP-response element binding protein
ERK1/2Phosphor-extracellular signalregulated kinase 1/2
FASNFatty acid synthase
HDACHistone deacetylase
HER2Human epidermal growth factor receptor 2
MAPKMitogen-activated protein kinase
NOSNitric oxide synthase
JAKJanus kinase
NF-κBNuclear factor kappa-B
PI3KPhosphatidylinositol 3 kinase
PTENPhosphate and tension homology deleted on chromosome ten
ROSReactive oxygen species
STAT3Signal transducer and activator of transcription 3

References

  1. Annunziata, F.; Pinna, C.; Dallavalle, S.; Tamborini, L.; Pinto, A. An overview of coumarin as a versatile and readily accessible scaffold with broad-ranging biological activities. Int. J. Mol. Sci. 2020, 21, 4618. [Google Scholar] [CrossRef] [PubMed]
  2. Stefanachi, A.; Leonetti, F.; Pisani, L.; Catto, M.; Carotti, A. Coumarin: A natural, privileged and versatile scaffold for bioactive compounds. Molecules 2018, 23, 250. [Google Scholar] [CrossRef] [PubMed]
  3. Yang, S.J.; Dai, W.L.; Wang, J.N.; Zhang, X.L.; Zheng, Y.T.; Bi, S.Y.; Pang, L.W.; Ren, T.Q.; Yang, Y.; Sun, Y.; et al. Osthole: An up-to-date review of its anticancer potential and mechanisms of action. Front. Pharmacol. 2022, 13, 945627. [Google Scholar] [CrossRef]
  4. Li, S.H.; Li, M.Y.; Yuan, T.T.; Wang, G.W.; Zeng, J.B.; Shi, Z.M.; Liu, J.H.; Su, J.C. Osthole activates the cholinergic anti-inflammatory pathway via α7nAChR upregulation to alleviate inflammatory responses. Chem. Biodivers. 2024, 21, e202400290. [Google Scholar] [CrossRef]
  5. Hu, H.Y.; Chen, M.; Dai, G.Z.; Du, G.Q.; Wang, X.Z.; He, J.; Zhao, Y.F.; Han, D.P.; Cao, Y.L.; Zheng, Y.X.; et al. An inhibitory role of osthole in rat MSCs osteogenic differentiation and proliferation via Wnt/β-catenin and Erk1/2-MAPK pathways. Cell. Physiol. Biochem. 2016, 38, 2375–2388. [Google Scholar] [CrossRef]
  6. Li, S.H.; Yan, Y.H.; Jiao, Y.A.; Gao, Z.; Xia, Y.; Kong, L.; Yao, Y.J.; Tao, Z.Y.; Song, J.; Yan, Y.P.; et al. Neuroprotective effect of osthole on neuron synapses in an Alzheimer’s disease cell model via upregulation of microRNA-9. J. Mol. Neurosci. 2016, 60, 71–81. [Google Scholar] [CrossRef] [PubMed]
  7. Liu, M.; Liu, Y.; Hua, X.W.; Wu, C.C.; Zhou, S.; Wang, B.L.; Li, Z.M. Synthesis of osthole derivatives with grignard reagents and their larvicidal activities on mosquitoes. Chin. J. Chem. 2015, 33, 1353–1358. [Google Scholar] [CrossRef]
  8. Liang, L.; Yang, B.; Wu, Y.Y.; Sun, L. Osthole suppresses the proliferation and induces apoptosis via inhibiting the PI3K/AKT signaling pathway of endometrial cancer JEC cells. Exp. Ther. Med. 2021, 22, 10605. [Google Scholar] [CrossRef]
  9. Chen, Y.Q.; Song, H.Y.; Zhou, Z.Y.; Ma, J.; Luo, Z.Y.; Zhou, Y.; Wang, J.Y.; Liu, S.; Han, X.H. Osthole inhibits the migration and invasion of highly metastatic breast cancer cells by suppressing ITGα3/ITGβ5 signaling. Acta Pharmacol. Sin. 2022, 43, 1544–1555. [Google Scholar] [CrossRef]
  10. Dai, X.X.; Yin, C.T.; Zhang, Y.; Guo, G.L.; Zhao, C.G.; Wang, O.C.; Xiang, Y.Q.; Zhang, X.H.; Liang, G. Osthole inhibits triple negative breast cancer cells by suppressing STAT3. J. Exp. Clin. Cancer Res. 2018, 37, 322. [Google Scholar] [CrossRef]
  11. Li, X.Y.; Zhang, C.L.; Wu, E.H.; Han, L.; Deng, X.L.; Shi, Z.F. UPLC-Q-TOF/MS-based metabolomics approach reveals osthole intervention in breast cancer 4T1 cells. Int. J. Mol. Sci. 2023, 24, 1168. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, N.; Tian, H.; Zhang, G.D.; Sun, N.; Wang, S.M. Effect of combined treatment with lobaplatin and osthole on inducing apoptosis and inhibiting proliferation in human breast cancer MDA-MB-231 cells. Med. Oncol. 2022, 39, 103579. [Google Scholar] [CrossRef]
  13. Park, W.; Park, S.; Song, G.; Lim, W. Inhibitory effects of osthole on human breast cancer cell progression via induction of cell cycle arrest, mitochondrial dysfunction, and ER stress. Nutrients 2019, 11, 2777. [Google Scholar] [CrossRef] [PubMed]
  14. Bae, H.; Lee, J.Y.; Song, J.; Song, G.; Lim, W. Osthole interacts with an ER-mitochondria axis and facilitates tumor suppression in ovarian cancer. J. Cell. Physiol. 2021, 236, 1025–1042. [Google Scholar] [CrossRef] [PubMed]
  15. Liang, J.; Zhou, J.L.; Xu, Y.Q.; Huang, X.F.; Wang, X.F.; Huang, W.H.; Li, H. Osthole inhibits ovarian carcinoma cells through LC3-mediated autophagy and GSDME-dependent pyroptosis except for apoptosis. Eur. J. Pharmacol. 2020, 874, 172990. [Google Scholar] [CrossRef]
  16. Arslan, A.K.K.; Uzunhisarcikli, E.; Ökçesiz, A.; Eken, A.; Yerer, M.B. Anticaner effects of coumarin compounds osthole and imperatorin, alone and in combination with 5-fluorouacil in colon carcinoma cells. Acta Pol. Pharm. 2021, 78, 243–251. [Google Scholar]
  17. Song, J.; Ham, J.; Park, W.; Song, G.; Lim, W. Osthole impairs mitochondrial metabolism and the autophagic flux in colorectal cancer. Phytomedicine 2024, 125, 115383. [Google Scholar] [CrossRef]
  18. Torshizi, G.H.; Tabrizi, M.H.; Karimi, E.; Younesi, A.; Larian, Z. Designing nanostructured lipid carriers modified with folate-conjugated chitosan for targeted delivery of osthole to HT-29 colon cancer cells: Investigation of anticancer, antioxidant, and antibacterial activities. Cancer Nanotechnol. 2024, 15, 7. [Google Scholar] [CrossRef]
  19. Zhou, X.H.; Kang, J.; Zhang, L.L.; Cheng, Y. Osthole inhibits malignant phenotypes and induces ferroptosis in KRAS-mutant colorectal cancer cells via suppressing AMPK/Akt signaling. Cancer Chemother. Pharmacol. 2023, 92, 119–134. [Google Scholar] [CrossRef]
  20. Zhou, X.H.; Kang, J.; Zhong, Z.D.; Cheng, Y. Osthole induces apoptosis of the HT-29 cells via endoplasmic reticulum stress and autophagy. Oncol. Lett. 2021, 22, 12987. [Google Scholar] [CrossRef]
  21. Huang, H.; Xue, J.; Xie, M.L.; Xie, T. Osthole inhibits GSK-3β/AMPK/mTOR pathway-controlled glycolysis and increases radiosensitivity of subcutaneous transplanted hepatocellular carcinoma in nude mice. Strahlenther. Onkol. 2024, 200, 444–452. [Google Scholar] [CrossRef] [PubMed]
  22. Mo, Y.S.; Wu, Y.; Li, X.; Rao, H.; Tian, X.X.; Wu, D.N.; Qiu, Z.P.; Zheng, G.H.; Hu, J.J. Osthole delays hepatocarcinogenesis in mice by suppressing AKT/FASN axis and ERK phosphorylation. Eur. J. Pharmacol. 2020, 867, 172788. [Google Scholar] [CrossRef]
  23. Yao, F.; Zhang, L.R.; Jiang, G.R.; Liu, M.; Liang, G.Q.; Yuan, Q. Osthole attenuates angiogenesis in an orthotopic mouse model of hepatocellular carcinoma via the downregulation of nuclear factor-κB and vascular endothelial growth factor. Oncol. Lett. 2018, 16, 4471–4479. [Google Scholar] [CrossRef] [PubMed]
  24. Ye, J.F.; Sun, D.; Yu, Y.; Yu, J.H. Osthole resensitizes CD133+ hepatocellular carcinoma cells to cisplatin treatment via PTEN/AKT pathway. Aging 2020, 12, 14406–14417. [Google Scholar] [CrossRef]
  25. Jiang, Y.Z.; Zhang, M.Z.; Wang, L.; Zhang, L.; Ma, M.H.; Jing, M.X.; Li, J.P.; Song, R.D.; Zhang, Y.Q.; Yang, Z.Z.; et al. Potential mechanisms of osthole against bladder cancer cells based on network pharmacology, molecular docking, and experimental validation. BMC Complement. Med. Ther. 2023, 23, 122. [Google Scholar] [CrossRef]
  26. Lv, X.F.; Yang, H.J.; Zhong, H.; He, L.; Wang, L. Osthole exhibits an antitumor effect in retinoblastoma through inhibiting the PI3K/AKT/mTOR pathway via regulating the hsa_circ_0007534/miR-214-3p axis. Pharm. Biol. 2022, 60, 417–426. [Google Scholar] [CrossRef] [PubMed]
  27. Che, Y.L.; Li, J.; Li, Z.J.; Li, J.; Wang, S.; Yan, Y.; Zou, K.; Zou, L.J. Osthole enhances antitumor activity and irradiation sensitivity of cervical cancer cells by suppressing ATM/NF-κB signaling. Oncol. Rep. 2018, 40, 737–747. [Google Scholar] [CrossRef]
  28. Jakubowicz-Gil, J.; Paduch, R.; Skalicka-Wozniak, K.; Sumorek-Wiadro, J.; Zajac, A.; Gawron, A. Hsps responsible for apoptosis induction failure in cervical cancer cells upon osthole and tamoxifen treatment. Postępy Hig. Med. Doświadczalnej 2019, 73, 563–571. [Google Scholar] [CrossRef]
  29. Su, J.; Zhang, F.; Li, X.; Liu, Z. Osthole promotes the suppressive effects of cisplatin on NRF2 expression to prevent drug-resistant cervical cancer progression. Biochem. Biophys. Res. Commun. 2019, 514, 510–517. [Google Scholar] [CrossRef]
  30. Yang, J.; Zhu, X.J.; Jin, M.Z.; Cao, Z.W.; Ren, Y.Y.; Gu, Z.W. Osthole induces cell cycle arrest and apoptosis in head and neck squamous cell carcinoma by suppressing the PI3K/AKT signaling pathway. Chem.-Biol. Interact. 2020, 316, 108934. [Google Scholar] [CrossRef]
  31. Huangfu, M.J.; Wei, R.M.; Wang, J.; Qin, J.L.; Yu, D.; Guan, X.; Li, X.M.; Fu, M.L.; Liu, H.P.; Chen, X. Osthole induces necroptosis via ROS overproduction in glioma cells. FEBS Open Bio 2021, 11, 456–467. [Google Scholar] [CrossRef] [PubMed]
  32. Sumorek-Wiadro, J.; Zajac, A.; Badziul, D.; Langner, E.; Skalicka-Wozniak, K.; Maciejczyk, A.; Wertel, I.; Rzeski, W.; Jakubowicz-Gil, J. Coumarins modulate the anti-glioma properties of temozolomide. Eur. J. Pharmacol. 2020, 881, 173207. [Google Scholar] [CrossRef] [PubMed]
  33. Sumorek-Wiadro, J.; Zajac, A.; Langner, E.; Skalicka-Wozniak, K.; Maciejczyk, A.; Rzeski, W.; Jakubowicz-Gil, J. Antiglioma potential of coumarins combined with sorafenib. Molecules 2020, 25, 5192. [Google Scholar] [CrossRef]
  34. Zou, T.L.; Wang, H.F.; Ren, T.; Shao, Z.Y.; Yuan, R.Y.; Gao, Y.; Zhang, Y.J.; Wang, X.A.; Liu, Y.B. Osthole inhibits the progression of human gallbladder cancer cells through JAK/STAT3 signal pathway both in vitro and in vivo. Anti-Cancer Drugs 2019, 30, 1022–1030. [Google Scholar] [CrossRef] [PubMed]
  35. Abosharaf, H.A.; Diab, T.; Atlam, F.M.; Mohamed, T.M. Osthole extracted from a citrus fruit that affects apoptosis on A549 cell line by histone deacetylasese inhibition (HDACs). Biotechnol. Rep. 2020, 28, e00531. [Google Scholar] [CrossRef]
  36. Xu, X.J.; Liu, X.Y.; Zhang, Y. Osthole inhibits gastric cancer cell proliferation through regulation of PI3K/AKT. PLoS ONE 2018, 13, e0193449. [Google Scholar] [CrossRef]
  37. Yang, Y.; Ren, F.; Tian, Z.Y.; Song, W.; Cheng, B.F.; Feng, Z.W. Osthole synergizes with HER2 inhibitor, trastuzumab in HER2-overexpressed N87 gastric cancer by inducing apoptosis and inhibition of AKT-MAPK pathway. Front. Pharmacol. 2018, 9, 1392. [Google Scholar] [CrossRef]
  38. Zhu, X.B.; Li, Z.Z.; Li, T.T.; Long, F.; Lv, Y.S.; Liu, L.; Liu, X.F.; Zhan, Q.M. Osthole inhibits the PI3K/AKT signaling pathway via activation of PTEN and induces cell cycle arrest and apoptosis in esophageal squamous cell carcinoma. Biomed. Pharmacother. 2018, 102, 502–509. [Google Scholar] [CrossRef]
  39. Jarzab, A.; Luszczki, J.; Guz, M.; Skalicka-Wozniak, K.; Halasa, M.; Smok-Kalwat, J.; Polberg, K.; Stepulak, A. Combination of osthole and cisplatin against rhabdomyosarcoma TE671 cells yielded additive pharmacologic interaction by means of isobolographic analysis. Anticancer Res. 2018, 38, 205–210. [Google Scholar]
  40. Wang, B.T.; Zheng, X.; Liu, J.; Zhang, Z.; Qiu, C.Y.; Yang, L.; Zhang, L.Q.; Zhang, Q.; Gao, H.W.; Wang, X.M. Osthole inhibits pancreatic cancer progression by directly exerting negative effects on cancer cells and attenuating tumor-infiltrating M2 macrophages. J. Pharmacol. Sci. 2018, 137, 290–298. [Google Scholar] [CrossRef]
  41. Liu, P.Y.; Chang, D.C.; Lo, Y.S.; Hsi, Y.T.; Lin, C.C.; Chuang, Y.C.; Lin, S.H.; Hsieh, M.J.; Chen, M.K. Osthole induces human nasopharyngeal cancer cells apoptosis through Fas-Fas ligand and mitochondrial pathway. Environ. Toxicol. 2018, 33, 446–453. [Google Scholar] [CrossRef]
  42. Zhou, Y.; Du, Z.T.; Wu, Q.Q.; Guo, M.; Chen, Z.C.; Sun, C.H.; Li, X.B.; Zou, Y.; Zheng, Z.W.; Chen, P.; et al. Discovery of novel osthole derivatives exerting anti-inflammatory effect on DSS-induced ulcerative colitis and LPS-induced acute lung injury in mice. Eur. J. Med. Chem. 2024, 268, 152864. [Google Scholar] [CrossRef] [PubMed]
  43. Jin, S.; Wang, Y.S.; Huang, J.C.; Wang, T.T.; Li, B.Y.; Guo, B.; Yue, Z.P. Osthole exhibits the remedial potential for polycystic ovary syndrome mice through Nrf2-Foxo1-GSH-NF-κB pathway. Cell Biol. Int. 2024, 48, 1111–1123. [Google Scholar] [CrossRef] [PubMed]
  44. Fan, H.Y.; Gan, Z.F.; Ji, K.; Li, X.; Wu, J.B.; Liu, Y.; Wang, X.K.; Liang, H.Y.; Liu, Y.N.; Li, X.T.; et al. The in vitro and in vivo anti-inflammatory effect of osthole, the major natural coumarin from Cnidium monnieri (L.) Cuss, via the blocking of the activation of the NF-κB and MAPK/p38 pathways. Phytomedicine 2019, 58, 116252. [Google Scholar] [CrossRef] [PubMed]
  45. Zhao, X.; Wang, F.; Zhou, R.J.; Zhu, Z.Y.; Xie, M.L. PPARα/γ antagonists reverse the ameliorative effects of osthole on hepatic lipid metabolism and inflammatory response in steatohepatitic rats. Inflammopharmacology 2018, 26, 425–433. [Google Scholar] [CrossRef]
  46. Zhao, X.; Xue, J.; Xie, M.L. Osthole inhibits oleic acid/lipopolysaccharide-induced lipid accumulation and inflammatory response through activating PPARα signaling pathway in cultured hepatocytes. Exp. Gerontol. 2019, 119, 7–13. [Google Scholar] [CrossRef]
  47. Jiang, X.L.; Lu, Z.J.; Zhang, Q.; Yu, J.L.; Han, D.; Liu, J.H.; Li, P.; Li, F. Osthole: A potential AMPK agonist that inhibits NLRP3 inflammasome activation by regulating mitochondrial homeostasis for combating rheumatoid arthritis. Phytomedicine 2023, 110, 154640. [Google Scholar] [CrossRef]
  48. Lin, X.; Chen, J.; Tao, C.; Luo, L.; He, J.; Wang, Q. Osthole regulates N6-methyladenosine-modified TGM2 to inhibit the progression of rheumatoid arthritis and associated interstitial lung disease. MedComm 2023, 4, e219. [Google Scholar] [CrossRef]
  49. Zhang, W.L.; Li, Z.C.; Dai, Z.W.; Chen, S.Y.; Guo, W.X.; Wang, Z.W.; Wei, J.S. GelMA hydrogel as a promising delivery system for osthole in the treatment of rheumatoid arthritis: Targeting the miR-1224-3p/AGO1 axis. Int. J. Mol. Sci. 2023, 24, 13210. [Google Scholar] [CrossRef]
  50. Ma, T.; Wang, X.P.; Qu, W.J.; Yang, L.S.; Jing, C.; Zhu, B.R.; Zhang, Y.K.; Xie, W.P. Osthole suppresses knee osteoarthritis development by enhancing autophagy activated via the AMPK/ULK1 pathway. Molecules 2022, 27, 8624. [Google Scholar] [CrossRef]
  51. Fu, X.P.; Hong, C.H. Osthole attenuates mouse atopic dermatitis by inhibiting thymic stromal lymphopoietin production from keratinocytes. Exp. Dermatol. 2019, 28, 561–567. [Google Scholar] [CrossRef] [PubMed]
  52. Kordulewska, N.K.; Król-Grzymala, A. The effect of osthole on transient receptor potential channels: A possible alternative therapy for atopic dermatitis. J. Inflamm. Res. 2024, 17, 881–898. [Google Scholar] [CrossRef] [PubMed]
  53. Kordulewska, N.K.; Topa, J.; Stryinski, R.; Jarmolowska, B. Osthole inhibits expression of genes associated with toll-like receptor 2 signaling pathway in an organotypic 3D skin model of human epidermis with atopic dermatitis. Cells 2022, 11, 88. [Google Scholar] [CrossRef]
  54. Zhou, Y.; Shi, J.X.; Qi, M.X.; Li, X.; Yang, Y.; Zhu, C.; Wang, C.M.; Tang, Z.X.; Ma, Y.X.; Yu, G. Osthole relieves skin damage and inhibits chronic itch through modulation of Akt/ZO-3 pathway in atopic dermatitis. Eur. J. Pharmacol. 2023, 947, 175649. [Google Scholar]
  55. Bao, Y.X.; Meng, X.L.; Liu, F.N.; Wang, F.; Yang, J.H.; Wang, H.Y.; Xie, G.H. Protective effects of osthole against inflammation induced by lipopolysaccharide in BV2 cells. Mol. Med. Rep. 2018, 17, 4561–4566. [Google Scholar] [CrossRef]
  56. Liu, C.H.; Chen, M.Y.; Kuo, Y.H.; Cheng, J.C.; Chang, L.Z.; Chang, M.S.; Chuang, T.N.; Hsieh, W.T.; Xiao, Y.R.; Wu, B.T.; et al. Osthole antagonizes microglial activation in an NRF2-dependent manner. Molecules 2023, 28, 28020507. [Google Scholar] [CrossRef]
  57. Singh, G.; Bhatti, R.; Mannan, R.; Singh, D.; Kesavan, A.; Singh, P. Osthole ameliorates neurogenic and inflammatory hyperalgesia by modulation of iNOS, COX-2, and inflammatory cytokines in mice. Inflammopharmacology 2019, 27, 949–960. [Google Scholar] [CrossRef]
  58. Kordulewska, N.K.; Kostyra, E.; Chwala, B.; Moszynska, M.; Cieslinska, A.; Fiedorowicz, E.; Jarmolowska, B. A novel concept of immunological and allergy interactions in autism spectrum disorders: Molecular, anti-inflammatory effect of osthole. Int. Immunopharmacol. 2019, 72, 1–11. [Google Scholar] [CrossRef]
  59. Chu, Q.B.; Zhu, Y.F.; Cao, T.J.; Zhang, Y.; Chang, Z.C.; Liu, Y.; Lu, J.H.; Zhang, Y.Z. Studies on the neuroprotection of osthole on glutamate-induced apoptotic cells and an Alzheimer’s disease mouse model via modulation oxidative stress. Appl. Biochem. Biotechnol. 2020, 190, 634–644. [Google Scholar] [CrossRef]
  60. Liu, J.M.; Wu, Q.H.; Wu, Q.Q.; Zhong, G.C.; Liang, Y.; Gu, Y.; Hu, Y.H.; Wang, W.J.; Hao, N.; Fang, S.H.; et al. Modulating endoplasmic reticulum stress in APP/PS1 mice by Gomisin B and osthole in Bushen-Yizhi formula: Synergistic effects and therapeutic implications for Alzheimer’s disease. Phytomedicine 2023, 119, 155023. [Google Scholar] [CrossRef]
  61. Wang, X.; Fu, X.M.; Luo, X.R.; Lai, Y.Y.; Cai, C.P.; Liao, Y.F.; Dai, Z.; Fang, S.H.; Fang, J.S. Network proximity analysis deciphers the pharmacological mechanism of osthole against D-galactose induced cognitive disorder in rats. Molecules 2024, 29, 21. [Google Scholar] [CrossRef] [PubMed]
  62. Yu, X.; Zhang, Y.; Zhang, M.J.; Chen, Y.F.; Yang, W.D. Natural products as sources of acetylcholinesterase inhibitors: Synthesis, biological activities, and molecular docking studies of osthole-based ester derivatives. Front. Plant Sci. 2022, 13, 1054650. [Google Scholar] [CrossRef] [PubMed]
  63. Barangi, S.; Hosseinzadeh, P.; Karimi, G.; Tayarani-Najaran, Z.; Mehri, S. Osthole attenuated cytotoxicity induced by 6-OHDA in SH-SY5Y cells through inhibition of JAK/STAT and MAPK pathways. Iran. J. Basic Med. Sci. 2023, 26, 953–959. [Google Scholar]
  64. Wang, S.G.; Ding, S.L.; Wang, Y.L.; He, C.Y.; Yao, R.P.; Jin, H. Osthole exerts neuroprotection in a mouse model of Parkinson’s disease. Curr. Top. Nutraceutical Res. 2023, 21, 10–17. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, Y.; Zhou, Y.; Wang, X.; Zhen, F.; Chen, R.; Geng, D.Q.; Yao, R.Q. Osthole alleviates MPTP-induced Parkinson’s disease mice by suppressing Notch signaling pathway. Int. J. Neurosci. 2019, 129, 833–841. [Google Scholar] [CrossRef]
  66. Yao, Y.J.; Liang, X.C.; Shi, Y.; Lin, Y.; Yang, J.X. Osthole delays tert-butyl hydroperoxide-induced premature senescence in neural stem cells. Cell. Reprogramm. 2018, 20, 268–274. [Google Scholar] [CrossRef]
  67. Yan, Y.H.; Kong, L.; Xia, Y.; Liang, W.B.; Wang, L.T.; Song, J.; Yao, Y.J.; Lin, Y.; Yang, J.X. Osthole promotes endogenous neural stem cell proliferation and improved neurological function through Notch signaling pathway in mice acute mechanical brain injury. Brain Behav. Immun. 2018, 67, 118–129. [Google Scholar] [CrossRef]
  68. Zhao, C.; Rollo, B.; Shahid Javaid, M.; Huang, Z.; He, W.; Xu, H.; Kwan, P.; Zhang, C. An integrated in vitro human iPSCs-derived neuron and in vivo animal approach for preclinical screening of anti-seizure compounds. J. Adv. Res. 2024, 64, 249–262. [Google Scholar] [CrossRef]
  69. Singh, L.; Kaur, A.; Garg, S.; Bhatti, R. Skimmetin/osthole mitigates pain-depression dyad via inhibiting inflammatory and oxidative stress-mediated neurotransmitter dysregulation. Metab. Brain Dis. 2021, 36, 111–121. [Google Scholar] [CrossRef]
  70. Jin, Z.X.; Liao, X.Y.; Da, W.W.; Zhao, Y.J.; Li, X.F.; Tang, D.Z. Osthole enhances the bone mass of senile osteoporosis and stimulates the expression of osteoprotegerin by activating β-catenin signaling. Stem Cell Res. Ther. 2021, 12, 154. [Google Scholar] [CrossRef]
  71. Chen, J.C.; Liao, X.F.; Gan, J.W. Review on the protective activity of osthole against the pathogenesis of osteoporosis. Front. Pharmacol. 2023, 14, 1236892. [Google Scholar] [CrossRef] [PubMed]
  72. Ma, Y.; Wang, L.N.; Zheng, S.Y.; Xu, J.K.; Pan, Y.L.; Tu, P.C.; Sun, J.; Guo, Y. Osthole inhibits osteoclasts formation and bone resorption by regulating NF-κB signaling and NFATc1 activations stimulated by RANKL. J. Cell. Biochem. 2019, 120, 16052–16061. [Google Scholar] [CrossRef] [PubMed]
  73. Xu, T.S.; Yin, J.Y.; Dai, X.; Liu, T.Y.; Shi, H.F.; Zhang, Y.Y.; Wang, S.; Yue, G.Y.; Zhang, Y.F.; Zhao, D.D.; et al. Cnidii Fructus: A traditional Chinese medicine herb and source of antiosteoporotic drugs. Phytomedicine 2024, 128, 155375. [Google Scholar] [CrossRef] [PubMed]
  74. García-Arroyo, F.E.; Gonzaga-Sánchez, G.; Silva-Palacios, A.; Roldán, F.J.; Loredo-Mendoza, M.L.; Alvarez-Alvarez, Y.Q.; Coyotl, J.A.D.; Orozco, K.A.V.; Tapia, E.; Osorio-Alonso, H.; et al. Osthole prevents heart damage induced by diet-induced metabolic syndrome: Role of fructokinase (KHK). Antioxidants 2023, 12, 1023. [Google Scholar] [CrossRef]
  75. Li, Y.L.; Li, Y.Q.; Shi, F.G.; Wang, L.N.; Li, L.S.; Yang, D.L. Osthole attenuates right ventricular remodeling via decreased myocardial apoptosis and inflammation in monocrotaline-induced rats. Eur. J. Pharmacol. 2018, 818, 525–533. [Google Scholar] [CrossRef]
  76. Hou, H.; Yang, Y.P.; Chen, R.; Guo, Z.P. Osthole protects H9c2 cardiomyocytes against trastuzumab-induced damage by enhancing autophagy through the p38MAPK/mTOR signaling pathway. Toxicol. In Vitro 2023, 93, 105704. [Google Scholar] [CrossRef]
  77. Lu, W.F.; He, Y.; Yu, T.; Huang, K.L.; Liu, S.Z. Osthole down-regulates miR-30a and promotes autophagy to protect rats against myocardial ischemia/reperfusion injury. Turk Gogus Kalp Damar. 2019, 27, 178–184. [Google Scholar] [CrossRef]
  78. Cai, Y.; Sun, W.; Zhang, X.X.; Lin, Y.D.; Chen, H.; Li, H. Osthole prevents acetaminophen-induced liver injury in mice. Acta Pharmacol. Sin. 2018, 39, 74–84. [Google Scholar] [CrossRef]
  79. Ma, Z.; Peng, L.; Chu, W.H.; Wang, P.; Fu, Y.Q. Osthole alleviates D-galactose-induced liver injury in vivo via the TLR4/MAPK/NF-κB pathways. Molecules 2023, 28, 443. [Google Scholar] [CrossRef]
  80. Zhou, W.B.; Zhang, X.X.; Cai, Y.; Sun, W.; Li, H. Osthole prevents tamoxifen-induced liver injury in mice. Acta Pharmacol. Sin. 2019, 40, 608–619. [Google Scholar] [CrossRef]
  81. Wu, F.; Zhao, Y.; Shao, Q.Q.; Fang, K.; Dong, R.L.; Jiang, S.J.; Lu, F.E.; Luo, J.L.; Chen, G. Ameliorative effects of osthole on experimental renal fibrosis in vivo and in vitro by inhibiting IL-11/ERK1/2 signaling. Front. Pharmacol. 2021, 12, 646331. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, S.P.; Huang, Q.; Cai, X.X.; Jiang, S.; Xu, N.; Zhou, Q.; Cao, X.Y.; Hultström, M.; Tian, J.; Lai, E.Y. Osthole ameliorates renal fibrosis in mice by suppressing fibroblast activation and epithelial-mesenchymal transition. Front. Physiol. 2018, 9, 1650. [Google Scholar] [CrossRef] [PubMed]
  83. Li, Q.S.; Wang, Y.F.; Yan, J.; Yuan, R.Y.; Zhang, J.M.; Guo, X.H.; Zhao, M.M.; Li, F.F.; Li, X.T. Osthole ameliorates early diabetic kidney damage by suppressing oxidative stress, inflammation and inhibiting TGF-81/Smads signaling pathway. Int. Immunopharmacol. 2024, 133, 112131. [Google Scholar] [CrossRef] [PubMed]
  84. Guo, Y.Y.; Chen, J.B.; Ren, D.; Du, B.; Wu, L.; Zhang, Y.Y.; Wang, Z.Y.; Qian, S. Synthesis of osthol-based botanical fungicides and their antifungal application in crop protection. Bioorg. Med. Chem. 2021, 40, 116184. [Google Scholar] [CrossRef]
  85. Kong, Y.; Yan, H.; Hu, J.J.; Dang, Y.X.; Han, Z.H.; Tian, B.; Wang, P.X. Antibacterial activity and mechanism of action of osthole against Listeria monocytogenes. J. Agric. Food Chem. 2024, 72, 10853–10861. [Google Scholar] [CrossRef]
  86. Zheng, H.L.; Chen, Y.H.; Guo, Q.L.; Wei, H.; Yue, J.Y.; Zhou, H.Y.; Zhao, M.M. Inhibitory effect of osthole from Cnidium monnieri (L.) Cusson on Fusarium oxysporum, a common fungal pathogen of potato. Molecules 2021, 26, 3818. [Google Scholar] [CrossRef]
  87. Hu, K.; Li, R.Y.; Mo, F.X.; Ding, Y.; Zhou, A.A.; Guo, X.; Li, R.T.; Li, M.; Ou, M.G.; Li, M. Natural product osthole can significantly disrupt cell wall integrity and dynamic balance of Fusarium oxysporum. Pestic. Biochem. Physiol. 2023, 196, 105623. [Google Scholar] [CrossRef]
  88. Hu, X.F.; Wang, J.; Zhang, Y.B.; Li, R.Y.; Li, M. Molecular mechanism of osthole against chitin synthesis of Ustilaginoidea virens based on combined transcriptome and metabolome analyses. Pestic. Biochem. Physiol. 2023, 196, 105612. [Google Scholar] [CrossRef]
  89. Lai, D.; Wang, D.L.; Shao, X.H.; Qin, J.; Zhuang, Q.L.; Xu, H.H.; Xiao, W.Q. Comparative physiological and transcriptome analysis provide insights into the inhibitory effect of osthole on Penicillium choerospondiatis. Pestic. Biochem. Physiol. 2024, 198, 105749. [Google Scholar] [CrossRef]
  90. Sun, Y.B.; Chen, Y.; Liu, T.; Wang, Y.Y.; Wang, Y.; Han, L.R.; Ma, Z.Q.; Feng, J.T. Evaluating the efficacy of osthole and matrine for control of Sorghum purple spot. J. Plant Dis. Prot. 2021, 128, 1263–1268. [Google Scholar] [CrossRef]
  91. Yu, C.Q.; Xie, T.T.; Liu, S.H.; Bai, L.G. Fabrication of a biochar-doped monolithic adsorbent and its application for the extraction and determination of coumarins from Angelicae Pubescentis Radix. J. Chromatogr. A 2024, 1714, 115940. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, Y.H.; Guo, D.S.; Lu, M.H.; Yue, J.Y.; Liu, Y.; Shang, C.M.; An, D.R.; Zhao, M.M. Inhibitory effect of osthole from Cnidium monnieri on tobacco mosaic virus (TMV) infection in Nicotiana glutinosa. Molecules 2020, 25, 65. [Google Scholar] [CrossRef] [PubMed]
  93. Ni, J.; Ren, Q.Y.; Luo, J.; Chen, Z.; Xu, X.F.; Guo, J.H.; Tan, Y.C.; Liu, W.G.; Qu, Z.Q.; Wu, Z.G.; et al. Ultrasound-assisted extraction extracts from Stemona japonica (Blume) Miq. and Cnidium monnieri (L.) Cuss. could be used as potential Rhipicephalus sanguineus control agents. Exp. Parasitol. 2020, 217, 107955. [Google Scholar] [CrossRef]
  94. Ren, Z.L.; Lv, M.; Yang, Y.L.; Gu, S.Y.; Li, L.L.; Liu, H.Q.; Xu, H. Structural optimization of natural plant products: Construction, pesticidal activities, and toxicology study of new 2-isopropanol-4-methoxy-7-alkyl/aryloxycarbonyl-(E)-vinyl-2,3-dihydrobenzofurans. J. Agric. Food Chem. 2025, 73, 1804–1812. [Google Scholar] [CrossRef]
  95. Xu, J.W.; Lv, M.; Li, T.Z.; Wen, H.P.; Xu, H. Optimization of osthole in the lactone ring as an agrochemical candidate: Synthesis, characterization, and pesticidal activities of osthole amide/ester derivatives and their effects on morphological changes of mite epidermis. J. Agric. Food Chem. 2023, 71, 6570–6583. [Google Scholar] [CrossRef] [PubMed]
  96. Ren, Z.L.; Lv, M.; Liu, H.Q.; Wen, H.P.; Zhang, Y.L.; Xu, H. Optimization of osthole as a pesticide candidate: Synthesis, crystal structures, and agrochemical properties of acrylate derivatives of isopropenyl 2,3-dihydrobenzofurans. J. Agric. Food Chem. 2023, 71, 18301–18311. [Google Scholar] [CrossRef]
  97. Dong, F.; Chen, X.; Men, X.Y.; Li, Z.; Kong, Y.J.; Yuan, Y.Y.; Ge, F. Contact toxicity, antifeedant activity, and oviposition preference of osthole against agricultural pests. Insects 2023, 14, 725. [Google Scholar] [CrossRef]
  98. Idrees, A.; Afzal, A.; Chohan, T.A.; Hayat, S.; Qadir, Z.A.; Gaafar, A.R.Z.; Zuan, A.T.K.; Li, J. Laboratory evaluation of selected botanicals and insecticides against invasive Spodoptera frugiperda (Lepidoptera: Noctuidae). J. King Saud Univ. Sci. 2023, 35, 102811. [Google Scholar] [CrossRef]
  99. Zhang, Z.B.; Hu, Q.; Du, X.G. Acaricidal of activity of matrine and osthole mixture against Tetranychus urticae Koch on Strawberry. Chin. J. Biol. Control 2022, 38, 1037–1042. [Google Scholar]
  100. Zeng, X.H.; Chen, S.H.; Wang, S.Z.; Zhang, L.; Xu, P.; Guo, Y.H. Study on the contact toxicity and stomach poisoning of four botanical pesticide against Coptotermes formosanus. Chin. J. Hyg. Insectic. Equip. 2019, 25, 370–373. [Google Scholar]
  101. Xia, C.X.; Li, S.Q.; Cai, W.L.; Yan, X.; Zhang, H.Y. Insecticidal activity of osthole powder and its effect on enzyme activity of Sitophilus zeamais. Chin. Bull. Entomol. 2009, 46, 740–744. [Google Scholar]
  102. Meng, L.L.; Wang, J.Z.; Zhang, Z.Y.; Sun, S.L.; Zhang, M.Z.; Zhang, P.F.; Ding, B.L.; Pu, Y.Y. Stomach action of osthole and its effect on two enzyme activities of Plutella xylostella larvae. J. Beijing Univ. Agric. 2010, 25, 24–27. [Google Scholar]
  103. Wang, Z.Q.; Kim, J.R.; Wang, M.; Shu, S.H.; Ahn, Y.J. Larvicidal activity of Cnidium monnieri fruit coumarins and structurally related compounds against insecticide-susceptible and insecticide-resistant Culex pipiens pallens and Aedes aegypti. Pest Manag. Sci. 2012, 68, 1041–1047. [Google Scholar] [CrossRef]
  104. Sun, X.Y.; Sun, L.L.; Qi, H.; Gao, Q.; Wang, G.X.; Wei, N.N.; Wang, K. Antipruritic effect of natural coumarin osthole through selective inhibition of thermosensitive TRPV3 channel in the skin. Mol. Pharmacol. 2018, 94, 1164–1173. [Google Scholar] [CrossRef]
  105. Torres, K.V.; Pantke, S.; Rudolf, D.; Eberhardt, M.M.; Leffler, A. The coumarin osthole is a non-electrophilic agonist of TRPA1. Neurosci. Lett. 2022, 789, 136878. [Google Scholar] [CrossRef]
  106. Wang, P.P.; Fan, Z.J.; Wei, W.P.; Yang, C.S.; Wang, Y.; Shen, X.; Yan, X.; Zhou, Z.H. Biosynthesis of the plant coumarin osthole by engineered Saccharomyces cerevisiae. ACS Synth. Biol. 2023, 12, 2455–2462. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, S.; Xie, Y.; Huo, Y.W.; Li, Y.; Abel, P.W.; Jiang, H.H.; Zou, X.H.; Jiao, H.Z.; Kuang, X.L.; Wolff, D.W.; et al. Airway relaxation mechanisms and structural basis of osthole for improving lung function in asthma. Sci. Signal. 2020, 13, eaax0273. [Google Scholar] [CrossRef]
  108. Park, J.; Shin, S.; Bu, Y.; Choi, H.Y.; Lee, K. Vasorelaxant and blood pressure-lowering effects of Cnidium monnieri fruit ethanol extract in sprague dawley and spontaneously hypertensive rats. Int. J. Mol. Sci. 2024, 25, 4223. [Google Scholar] [CrossRef]
  109. Liu, G.L.; Hao, B.; Liu, S.P.; Wang, G.X. Synthesis and anthelmintic activity of osthol analogs against Dactylogyrus intermedius in goldfish. Eur. J. Med. Chem. 2012, 54, 582–590. [Google Scholar] [CrossRef]
  110. Yin, Q.; Yan, H.; Zhang, Y.Q.; Wang, Y.G.; Zhang, G.J.; He, Y.X.; Zhang, W.H. Palladium-catalyzed synthesis of 8-allyl or 8-prenylcoumarins by using organotin reagents as multicoupling nucleophiles. Appl. Organomet. Chem. 2013, 27, 85–88. [Google Scholar] [CrossRef]
  111. Konrádová, D.; Kozubíková, H.; Dolezal, K.; Pospísil, J. Microwave-assisted synthesis of phenylpropanoids and coumarins: Total synthesis of osthol. Eur. J. Org. Chem. 2017, 2017, 5204–5213. [Google Scholar] [CrossRef]
  112. Schmidt, B.; Riemer, M. Synthesis of allyl- and prenylcoumarins via microwave-promoted tandem claisen rearrangement/wittig olefination. Synthesis 2016, 48, 141–149. [Google Scholar] [CrossRef]
  113. Gulías, M.; Marcos-Atanes, D.; Mascareñas, J.L.; Font, M. Practical, large-scale preparation of benzoxepines and coumarins through Rhodium(III)-catalyzed C-H activation/annulation reactions. Org. Process Res. Dev. 2019, 23, 1669–1673. [Google Scholar] [CrossRef]
  114. Dai, P.; Wang, Q.Q.; Teng, P.; Jiao, J.; Li, Y.F.; Xia, Q.; Zhang, W.H. Design, synthesis, antifungal activity, and 3D-QASR of novel oxime ether-containing coumarin derivatives as potential fungicides. J. Agric. Food Chem. 2024, 72, 5983–5992. [Google Scholar] [CrossRef] [PubMed]
  115. Ma, H.N.; Wang, K.H.; Wang, B.B.; Wang, Z.W.; Liu, Y.X.; Wang, Q.M. Design, synthesis, and biological activities of novel coumarin derivatives as pesticide candidates. J. Agric. Food Chem. 2024, 72, 4658–4668. [Google Scholar] [CrossRef]
  116. Zhang, M.Z.; Zhang, R.R.; Wang, J.Q.; Yu, X.; Zhang, Y.L.; Wang, Q.Q.; Zhang, W.H. Microwave-promoted synthesis of novel fused osthole analogues. Chin. J. Chem. 2016, 34, 1344–1352. [Google Scholar] [CrossRef]
  117. Zhang, M.Z.; Zhang, R.R.; Wang, J.Q.; Yu, X.; Zhang, Y.L.; Wang, Q.Q.; Zhang, W.H. Microwave-assisted synthesis and antifungal activity of novel fused Osthole derivatives. Eur. J. Med. Chem. 2016, 124, 10–16. [Google Scholar] [CrossRef]
  118. Zhang, S.G.; Liang, C.G.; Sun, Y.Q.; Teng, P.; Wang, J.Q.; Zhang, W.H. Design, synthesis and antifungal activities of novel pyrrole- and pyrazole-substituted coumarin derivatives. Mol. Divers. 2019, 23, 915–925. [Google Scholar] [CrossRef]
  119. Li, S.C.; Lv, M.; Sun, Z.Q.; Hao, M.; Xu, H. Optimization of osthole in the lactone ring: Structural elucidation, pesticidal activities, and control efficiency of osthole ester derivatives. J. Agric. Food Chem. 2021, 69, 6465–6474. [Google Scholar] [CrossRef]
  120. Yang, H.Y.; Hsu, Y.F.; Chiu, P.T.; Ho, S.J.; Wang, C.H.; Chi, C.C.; Huang, Y.H.; Lee, C.F.; Li, Y.S.; Ou, G.; et al. Anti-cancer activity of an osthole derivative, NBM-T-BMX-OS01: Targeting vascular endothelial growth factor receptor signaling and angiogenesis. PLoS ONE 2013, 8, e81592. [Google Scholar] [CrossRef]
  121. Pai, J.T.; Hsu, C.Y.; Hua, K.T.; Yu, S.Y.; Huang, C.Y.; Chen, C.N.; Liao, C.H.; Weng, M.S. NBM-T-BBX-OS01, semisynthesized from osthole, induced G1 growth arrest through HDAC6 inhibition in lung cancer cells. Molecules 2015, 20, 8000–8019. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, L.; Wu, Y.H.; Yang, G.X.; Gan, H.X.; Sang, D.Y.; Zhou, J.Y.; Su, L.; Wang, R.; Ma, L. Design, synthesis and biological evaluation of novel osthole-based derivatives as potential neuroprotective agents. Bioorg. Med. Chem. Lett. 2020, 30, 127633. [Google Scholar] [CrossRef] [PubMed]
  123. Ren, Z.L.; Lv, M.; Xu, H. Osthole: Synthesis, structural modifications, and biological properties. Mini-Rev. Med. Chem. 2022, 22, 2124–2137. [Google Scholar]
  124. Hao, M.; Jiang, L.L.; Lv, M.; Ding, H.X.; Zhou, Y.M.; Xu, H. Plant natural product-based pesticides in crop protection: Semi-synthesis, mono-crystal structures and agrochemical activities of osthole ester derivatives, and study of their toxicology against Tetranychus cinnabarinus (Boisduval). Pest Manag. Sci. 2024, 80, 6356–6365. [Google Scholar] [CrossRef]
  125. Hao, M.; Lv, M.; Zhou, L.; Li, H.J.; Xu, J.W.; Xu, H. Construction, pesticidal activities, control effects, and detoxification enzyme activities of osthole ester/amide derivatives. J. Agric. Food Chem. 2022, 70, 9337–9345. [Google Scholar] [CrossRef] [PubMed]
  126. Ren, Z.L.; Lv, M.; Sun, Z.Q.; Li, T.Z.; Zhang, S.Y.; Xu, H. Regioselective hemisynthesis and insecticidal activity of C8-hydrazones/acylhydrazones/sulfonylhydrazones coumarin-type derivatives of osthole. Bioorg. Med. Chem. Lett. 2021, 40, 127962. [Google Scholar] [CrossRef]
  127. Huang, W.J.; Chen, C.C.; Chao, S.W.; Lee, S.S.; Hsu, F.L.; Lu, Y.L.; Hung, M.F.; Chang, C.I. Synthesis of N-hydroxycinnamides capped with a naturally occurring moiety as inhibitors of histone deacetylase. Chemmedchem 2010, 5, 598–607. [Google Scholar] [CrossRef]
  128. Huang, W.J.; Chen, C.C.; Chao, S.W.; Yu, C.C.; Yang, C.Y.; Guh, J.H.; Lin, Y.C.; Kuo, C.I.; Yang, P.; Chang, C.I. Synthesis and evaluation of aliphatic-chain hydroxamates capped with osthole derivatives as histone deacetylase inhibitors. Eur. J. Med. Chem. 2011, 46, 4042–4049. [Google Scholar] [CrossRef]
  129. Ren, Z.L.; Lv, M.; Li, T.Z.; Hao, M.; Li, S.C.; Xu, H. Construction of oxime ester derivatives of osthole from Cnidium monnieri, and evaluation of their agricultural activities and control efficiency. Pest Manag. Sci. 2020, 76, 3560–3567. [Google Scholar] [CrossRef]
  130. Shan, X.J.; Lv, M.; Wang, J.R.; Qin, Y.J.; Xu, H. Acaricidal and insecticidal efficacy of new esters derivatives of a natural coumarin osthole. Ind. Crops Prod. 2022, 182, 114855. [Google Scholar] [CrossRef]
  131. Farooq, S.; Alharthi, F.A.; Alsalme, A.; Hussain, A.; Dar, B.A.; Hamid, A.; Koul, S. Dihydropyrimidinones: Efficient one-pot green synthesis using Montmorillonite-KSF and evaluation of their cytotoxic activity. RSC Adv. 2020, 10, 42221. [Google Scholar] [CrossRef] [PubMed]
  132. You, L.S.; An, R.; Wang, X.H.; Li, Y.M. Discovery of novel osthole derivatives as potential anti-breast cancer treatment. Bioorg. Med. Chem. Lett. 2010, 20, 7426–7428. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The chemical structures of coumarin and osthole (1).
Figure 1. The chemical structures of coumarin and osthole (1).
Biology 14 00588 g001
Figure 2. The biological activities of osthole and its derivatives.
Figure 2. The biological activities of osthole and its derivatives.
Biology 14 00588 g002
Scheme 1. Synthesis of osthole (1) by Liu et al. and Yin et al. [109,110].
Scheme 1. Synthesis of osthole (1) by Liu et al. and Yin et al. [109,110].
Biology 14 00588 sch001
Scheme 2. Synthesis of osthole (1) by Konrádová et al., Schmidt et al., Gulías et al., and Liu et al. [7,111,112,113].
Scheme 2. Synthesis of osthole (1) by Konrádová et al., Schmidt et al., Gulías et al., and Liu et al. [7,111,112,113].
Biology 14 00588 sch002
Scheme 3. Synthesis of osthole derivatives 19a19z and 19a’19d’.
Scheme 3. Synthesis of osthole derivatives 19a19z and 19a’19d’.
Biology 14 00588 sch003
Scheme 4. Synthesis of osthole derivatives 23a23c, 24a24d, 25a25c, and 26a26e.
Scheme 4. Synthesis of osthole derivatives 23a23c, 24a24d, 25a25c, and 26a26e.
Biology 14 00588 sch004
Scheme 5. Synthesis of osthole derivatives 2833.
Scheme 5. Synthesis of osthole derivatives 2833.
Biology 14 00588 sch005
Scheme 6. Synthesis of osthole derivatives 36a36p and 38a38h.
Scheme 6. Synthesis of osthole derivatives 36a36p and 38a38h.
Biology 14 00588 sch006
Figure 3. Structural modifications of osthole at different positions.
Figure 3. Structural modifications of osthole at different positions.
Biology 14 00588 g003
Scheme 7. Synthesis of osthole derivatives 4144.
Scheme 7. Synthesis of osthole derivatives 4144.
Biology 14 00588 sch007
Scheme 8. Synthesis of osthole derivatives 47a47z.
Scheme 8. Synthesis of osthole derivatives 47a47z.
Biology 14 00588 sch008
Scheme 9. Synthesis of osthole derivatives 4852.
Scheme 9. Synthesis of osthole derivatives 4852.
Biology 14 00588 sch009
Scheme 10. Synthesis of osthole derivatives 5357.
Scheme 10. Synthesis of osthole derivatives 5357.
Biology 14 00588 sch010
Scheme 11. Synthesis of osthole derivatives 5863.
Scheme 11. Synthesis of osthole derivatives 5863.
Biology 14 00588 sch011
Scheme 12. Synthesis of osthole-type ester derivatives 66a66y.
Scheme 12. Synthesis of osthole-type ester derivatives 66a66y.
Biology 14 00588 sch012
Scheme 13. Synthesis of osthole derivatives 68a68w and 69a69j.
Scheme 13. Synthesis of osthole derivatives 68a68w and 69a69j.
Biology 14 00588 sch013
Scheme 14. Synthesis of osthole derivatives 70a70s.
Scheme 14. Synthesis of osthole derivatives 70a70s.
Biology 14 00588 sch014
Scheme 15. Synthesis of osthole derivatives 7179.
Scheme 15. Synthesis of osthole derivatives 7179.
Biology 14 00588 sch015
Scheme 16. Synthesis of osthole oxime ester derivatives 81a81z.
Scheme 16. Synthesis of osthole oxime ester derivatives 81a81z.
Biology 14 00588 sch016
Scheme 17. Synthesis of osthole derivatives 83a83s.
Scheme 17. Synthesis of osthole derivatives 83a83s.
Biology 14 00588 sch017
Scheme 18. Synthesis of osthole derivative 84.
Scheme 18. Synthesis of osthole derivative 84.
Biology 14 00588 sch018
Scheme 19. Synthesis of osthole derivatives 87a87z and 87a’87h’.
Scheme 19. Synthesis of osthole derivatives 87a87z and 87a’87h’.
Biology 14 00588 sch019
Scheme 20. Synthesis of osthole derivatives 90a90z and 90a’90m’.
Scheme 20. Synthesis of osthole derivatives 90a90z and 90a’90m’.
Biology 14 00588 sch020
Scheme 21. Synthesis of osthole derivatives 92a92j.
Scheme 21. Synthesis of osthole derivatives 92a92j.
Biology 14 00588 sch021
Figure 4. The structureactivity relationships of osthole and its derivatives.
Figure 4. The structureactivity relationships of osthole and its derivatives.
Biology 14 00588 g004
Table 1. The anticancer activity of osthole.
Table 1. The anticancer activity of osthole.
TumorCell Lines/ModelInvolved MechanismPeriod (h)IC50 (µM)
Endometrial
carcinoma
JEC, Ishikawa and KLE cellsSuppressing the proliferation and inducing apoptosis via inhibiting the PI3K/AKT signaling pathway\\ [8]
Breast cancer4T1 cellsInhibiting cell proliferation, blocking the cells from remaining in S-phase, and inducing apoptosis48178.40 [11]
MDA-MB-231 cellsInducing apoptosis arrest by elevation in the translation level of p53/Bax/caspase-3 p17 and downregulating of the Bcl-2 protein\\ [12]
MDA-MB-231 cells
BT-549 cells
MDA-MB-468 cells
MCF-7 cells
Inducing apoptosis and cell cycle arrest in TNBC cells through inhibition of STAT3 phosphorylation and nuclear translocation24129.40 [10]
106.20 [10]
105.40 [10]
168.00 [10]
MDA-MB-231
MDA-MB-231BO
MCF-7 cells
Inhibiting cell migration and invasion via suppressing of ITGα3 and ITGβ5 signaling2429.8 [9]
111.4 [9]
260.6 [9]
BT-474 and MCF-7 cellsInducing cell cycle arrest, mitochondrial dysfunction, and ER stress\\ [13]
Ovarian carcinomaA2780 cell
OVCAR3 cell
Inducing LC3-mediated autophagy and GSDME-dependent pyroptosis2475.24 [15]
73.58 [15]
ES2 and OV90 cellsTargeting PI3K/MAPK signaling pathway\\ [14]
Colorectal cancerHCT116 and HT29 cellsRegulating autophagy- and mitochondria-mediated signal transduction.\\ [17]
HT-29 cellsInducing apoptosis via cycle arrest and activating the intrinsic apoptotic pathway24115.08 [18]
HCT116 and SW480 cellsInhibits malignant phenotypes and inducing ferroptosis in KRAS mutant cells via suppressing AMPK/Akt signaling\\ [19]
CCL-222 cellsReducing the Akt mRNA expression and the activity p38 MAPK-α2423.00 [16]
Colorectal cancerHT-29 cellsInducing apoptosis of the cells via endoplasmic reticulum stress and autophagy\\ [20]
Hepatocellular carcinomaHCC-LM3 cellsInhibiting GSK-3β/AMPK/mTOR pathway-controlled glycolysis\[21]
AKT/c-Met-driven HCC mouse modelDelaying hepatocarcinogenesis by suppressing AKT/FASN axis and ERK phosphorylation\[22]
Huh7 and HepG2 cellsResensitizing CD133+ hepatocellular carcinoma cells to cisplatin treatment via PTEN/AKT pathway\[24]
Hepa1–6 cellsInhibiting angiogenesis in an orthotopic mouse model of HCC via the NF-κB/VEGF signaling pathway.\[23]
Bladder cancer5637 cells
253 J cells
Inhibiting invasion, migration, and epithelial–mesenchymal transition by inhibiting PI3K-AKT and JAK/STAT3 pathways24146.40 [25]
160.80 [25]
RetinoblastomaY-79 cellsInhibiting the PI3K/AKT/mTOR pathway via regulating the hsa_circ_0007534/miR-214-3p axis24200.00 [26]
Cervical cancerHela B cellsTargeting DCLK1 mechanistically via interacting with Val468.\\ [28]
CDDP-resistant cervical cancer cellsReversing chemoresistance of CDDP-resistant cervical cancer to CDDP through repressing NRF2 expression partly associated with PI3K/AKT blockage\\ [29]
HeLa, SiHa, C-33A and CaSki cellsSuppressing ATM/NF-κB signaling\\ [27]
Head and neck squamous cell carcinomaFaDu cells
Cal27 cells
SCC25 cells
HN4 cells
Inducing cell cycle arrest and apoptosis via suppressing the PI3K/AKT signaling pathway24122.35 [30]
183.32 [30]
189.26 [30]
237.42 [30]
GliomaU87 and C6 cellsTriggering glioma cell necroptosis via ROS overproduction\\ [31]
MOGGCCM and T98G cellsInhibiting of anti-apoptotic Bcl-2 expression, decreasing mitochondrial membrane potential, and activation of caspase 3 via the mitochondrial (internal) pathway\\ [32]
AA and GBM cellsIncreasing the level of apoptosis via blocking of the expression of Bcl-2 and PI3K \\ [33]
Gallbladder cancerNOZ and SGC-996 cellsInhibiting the progression of human gallbladder cancer cells via the JAK/STAT3 signal pathway\\ [34]
Lung cancerA549 cellsInducing the apoptosis in A549 cells by inhibiting the histone deacetylase24188.50 [35]
Gastric cancerHGC-27 and SGC-7901 cellsInducting cell cycle arrest at G2/M phase by the regulation of PI3K/AKT\\ [36]
N87 and SK-BR-3 cellsInducing apoptosis and inhibiting AKT/MAPK pathway\\ [37]
Esophageal squamous cell carcinomaKYSE150 cells
KYSE410 cells
Inhibiting the PI3K/AKT signaling pathway via activation of PTEN24198.45 [38]
235.67 [38]
RhabdomyosarcomaTE671 cellsInhibiting of cell cycle progression and inducing of apoptosis via the AKT and ERK kinase signaling pathways964.05 * [39]
Pancreatic cancerPanc 02 cells and RAW 264.7 cellsAttenuating tumor-infiltrating M2 macrophages\\ [40]
Nasopharyngeal cancerNPC-039 and NPC-BM cellsInducing apoptosis mediated by the Fas-Fas ligand and mitochondrial pathway\\ [41]
* µg/mL.
Table 2. Antibacterial and antifungal activities of osthole.
Table 2. Antibacterial and antifungal activities of osthole.
ObjectPeriod (h)Values (µg/mL)
Listeria monocytogenes ATCC191156MIC = 62.5 [85]
Staphylococcus aureus ATCC2592320–24MIC = 128 [84]
Escherichia coli ATCC25922MIC = 128 [84]
Staphylococcus aureus (MRSA) ATCC33591MIC = 128 [84]
Staphylococcus aureus (Methicillin-resistant isolate) ATCC43300MIC = 128 [84]
Erwinia carotorora4898.00% 1 [84]
Ralstonia solanacearum25.00% 1 [84]
Pseudomonas syringae pv. actinidiae68.00% 1 [84]
Fusarium oxysporum7257.40% 2 [86]
Fusarium moniliforme J. Sheld70.66% 2 [86]
Thanatephorus cucumeris Donk75.90% 2 [86]
Penicillium choerospondiatis144EC50 = 9.86 [89]
Ustilaginoidea virens240EC50 = 5.04 [88]
Fusarium oxysporum168EC50 = 6.40 [87]
Cercospora sorghi72EC50 = 1.46 [90]
1 At 0.1 mg/mL; 2 At 5 mg/mL.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lv, M.; Ding, H.; Xu, H. Osthole: A Coumarin with Dual Roles in Biology and Chemistry. Biology 2025, 14, 588. https://doi.org/10.3390/biology14060588

AMA Style

Lv M, Ding H, Xu H. Osthole: A Coumarin with Dual Roles in Biology and Chemistry. Biology. 2025; 14(6):588. https://doi.org/10.3390/biology14060588

Chicago/Turabian Style

Lv, Min, Haixia Ding, and Hui Xu. 2025. "Osthole: A Coumarin with Dual Roles in Biology and Chemistry" Biology 14, no. 6: 588. https://doi.org/10.3390/biology14060588

APA Style

Lv, M., Ding, H., & Xu, H. (2025). Osthole: A Coumarin with Dual Roles in Biology and Chemistry. Biology, 14(6), 588. https://doi.org/10.3390/biology14060588

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop