You are currently viewing a new version of our website. To view the old version click .
Pharmaceuticals
  • Review
  • Open Access

19 November 2025

Cardiovascular Effects, Phytochemistry, Drug Interactions, and Safety Profile of Foeniculum vulgare Mill. (Fennel): A Comprehensive Review

,
,
,
,
,
,
,
and
1
Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda 60000, Morocco
2
Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
3
Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
*
Author to whom correspondence should be addressed.
This article belongs to the Special Issue Bioactive Compounds from Plants and Foods: From Traditional Medicine to Modern Applications in Nutrition and Health

Abstract

Background/Objectives: Cardiovascular diseases remain the leading cause of mortality worldwide. According to the World Heart Federation, more than 500 million people were living with cardiovascular diseases in 2021. In this context, the use of medicinal plants has become increasingly widespread in populations as a preventive strategy against cardiovascular disorders. Foeniculum vulgare Mill., commonly known as fennel, is an aromatic and medicinal plant recognized for its beneficial properties in the treatment of various ailments, due to its richness in bioactive compounds. This review aims to summarize and analyze the cardiovascular activities of this plant, based on experimental evidence, and to provide an updated overview of its phytochemical composition and safety profile. Methods: A comprehensive literature search was conducted using databases including PubMed, Scopus, Web of Science, and Google Scholar, encompassing all publications available up to 2024. This search included research articles, reviews, mini-reviews, and clinical studies published in English. Exclusion criteria comprised publication types such as letters, conference abstracts, unpublished theses, and non-peer-reviewed reports. Studies were also excluded if they did not specifically address Foeniculum vulgare Mill. or its cardiovascular activities. All studies were screened according to predefined inclusion and exclusion criteria, and relevant data were systematically extracted and analyzed to synthesize current knowledge on the cardiovascular activities, mechanisms of action, phytochemical composition, safety, and potential drug interactions of Foeniculum vulgare Mill. Results: Numerous in vitro and in vivo studies have demonstrated that Foeniculum vulgare Mill. exhibits a wide range of activities beneficial for cardiovascular health. These include antihypertensive, cardioprotective, vasorelaxant, anti-inflammatory, antioxidant, diuretic, hypotensive, hypolipidemic, antiplatelet, and anticoagulant effects. Such pharmacological actions are largely attributed to its rich phytochemical composition, particularly its volatile oils (e.g., trans-anethole, fenchone), flavonoids (e.g., quercetin, kaempferol), and phenolic acids (e.g., p-coumaric acid, ferulic acid). Most studies report no significant signs of toxicity. Conclusions: Foeniculum vulgare Mill. emerges as a promising medicinal plant for the prevention and management of cardiovascular diseases, owing to its multifaceted beneficial effects and its favorable safety profile. However, potential interactions with cardiovascular drugs and the current limitations of existing studies highlight the need for further clinical research to fully establish its therapeutic potential.

1. Introduction

Cardiovascular disorders (CVDs) are among the leading causes of death and significantly impair the quality of life worldwide. In 2019, cardiovascular diseases were responsible for 17.9 million deaths worldwide, accounting for 32% of global deaths, according to the World Health Organization []. Among these deaths, 85% were attributed to heart attacks and strokes. Data from the World Heart Federation in 2021 indicated that over 500 million people were living with cardiovascular diseases []. Cardiomyopathy (CMD), coronary artery disease (CAD) and heart failure (HFD) are characterized by unique pathophysiological changes that drive their specific clinical outcomes []. Guidelines for prevention of CVD list hypertension, abnormal lipid levels, smoking and diabetes mellitus as the primary modifiable risk factors []. Lifestyle modifications such as limiting tobacco and alcohol use, following a balanced diet low in fat and sugar, and staying physically active to prevent obesity can reduce incidence. However, the risk of CVDs increases with age and the presence of contributing factors such as oxidative stress, chronic inflammation, and a family history of heart attacks [,]. Medicinal plants contain diverse bioactive compounds such as flavonoids, alkaloids and polyphenols which exhibit significant therapeutic and health-promoting properties []. Investigating traditionally used plants is crucial to ensure treatment effectiveness, aiming to use them for prevention and alongside conventional chemical drugs [].
Foeniculum vulgare Mill. (FV), commonly known as fennel, is a medicinal and aromatic plant from the Apiaceae (Umbelliferae) family. It has been used since ancient times, valued both as a culinary spice and for its wide range of therapeutic properties []. Numerous studies have demonstrated the efficacy of fennel in managing various health conditions, including inflammation, hyperlipidemia, hypertension, and others cardiovascular disorders [,,,]. The present review aims to compile and critically analyze the diverse biological activities of FV, with particular emphasis on its cardiovascular activities, as well as its botanical characteristics, phytochemical composition, potential drug interactions, and safety profile. By integrating current scientific findings, this review provides a comprehensive overview of the cardiovascular properties and underlying mechanisms of FV, thereby supporting its traditional use in the prevention and management of cardiovascular diseases. However, it is important to clinically validate its cardiovascular effects in order to establish its therapeutic potential.

2. Methodology of Research

To collect relevant data, a comprehensive literature search was conducted across multiple electronic databases, including PubMed, Scopus, Web of Science, and Google Scholar, covering all available publications up to 2024. The search strategy employed keywords such as Foeniculum vulgare Mill., fennel, botanical characteristics, cardiovascular pharmacology, vasorelaxation, antihypertensive, cardioprotective, diuretic, antioxidant, anti-inflammatory, antiaggregant, anticoagulant, hypolipidemic, phytochemical composition, potential drug interactions, and toxicity. The inclusion criteria covered original research articles, book chapters, reviews, mini-reviews, and clinical studies published in English that specifically focused on Foeniculum vulgare Mill. The exclusion criteria comprised letters, conference abstracts, unpublished theses, and non-peer-reviewed reports. The bibliographic data from the selected studies were extracted, analyzed, and synthesized to provide a comprehensive overview of the cardiovascular activities, underlying mechanisms, key bioactive compounds, drugs interactions, and safety profile of fennel. The chemical structures of the compounds presented in this review were drawn using ChemDraw software, version 8.

3. Botanical Description of Foeniculum vulgare Mill.

Foeniculum vulgare Mill. is one of the oldest known medicinal and aromatic plants, traditionally valued for its wide range of therapeutic properties. It comprises two main botanical varieties: Foeniculum vulgare var. dulce (sweet fennel) and Foeniculum vulgare var. vulgare (bitter fennel), both widely used in traditional and modern medicine. Historical records even cite an ancient belief attributed to Adam, suggesting that consuming fennel with sugar daily during spring could protect against all diseases, underscoring its long-standing importance in traditional health practices []. Fennel is a hardy, perennial, and aromatic herb that can reach up to 2.5 m in height. It has erect, hollow stems, and its leaves are finely dissected and feathery, reaching up to 40 cm in length, with thread-like segments approximately 0.5 mm wide. The plant produces bright yellow flowers arranged in large compound umbels at the tips of the stems []. The fruit, often referred to as a seed, is actually a dry schizocarp composed of two mericarps (also called achenes). These are elongated, oval, and ribbed, initially bluish in color and turning brownish-grey upon maturation. The typical size of the fennel fruit is 4–10 mm long and about 3 mm wide, though it may vary depending on growth stage and environmental conditions []. Fennel seeds are aromatic and flavorful, widely used as a spice and in herbal remedies. The plant is green to white in color, with bulbous leaf bases that may resemble a swollen stem, especially in cultivated varieties (Figure 1) []. It grows wild across much of temperate Europe but is generally regarded as native to the Mediterranean region. Fennel is known for producing a variety of valuable secondary metabolites, used in pharmaceuticals, agrochemicals, fragrances, dyes, biopesticides, and as food additives [].
Figure 1. Different parts of Foeniculum vulgare Mill., (A) Branched stem bearing umbels; (B) Umbel inflorescence; (C) bulbs; (D) Seeds. (Photos were taken in northeastern Morocco).

4. Phytochemistry of Foeniculum vulgare Mill.

Phytochemical investigations of FV have identified a wide array of bioactive constituents, including volatile oils, phenolic compounds, flavonoids, coumarins, alkaloids, terpenoids, glycosides, tannins, and other secondary metabolites []. The concentration and composition of these metabolites vary substantially depending on the plant’s morphological characteristics, growth stage, and geographical origin. All parts of the plant like roots, leaves, fruits, and particularly seeds are utilized for diverse therapeutic and culinary purposes []. The whole FV plant has been reported to contain approximately 6.3% moisture, 9.5% protein, 10% fatty acids, 13.4% minerals, 18.5% fiber, and 42.3% carbohydrates. Its mineral and vitamin profile includes calcium, potassium, sodium, iron, and phosphorus, as well as thiamine, riboflavin, niacin, and vitamin C. The plant’s calorific value is estimated at 370 kcal per 100 g [].

4.1. Volatile and Non-Volatile Compounds

FV is renowned for its characteristic aroma, primarily attributed to its essential oil, which is widely used as a flavoring agent in culinary and pharmaceutical preparations. The essential oil is dominated by trans-anethole (50–70%), a major phenylpropanoid compound, followed by notable amounts of estragole (10–40%), fenchone (3–10%), and limonene (2–11%). Several minor constituents also contribute to its complex chemical profile [,,,,,,,,,,,]. The relative proportions of these volatile compounds vary considerably depending on factors such as geographical origin, cultivation conditions, developmental stage, and fruit ripeness. For instance, GC-MS analyses have revealed marked regional variations: estragole predominates in Egyptian fennel seeds (51.04%), whereas trans-anethole is the major component in Chinese (54.26%) and Iraqi (43.13%) samples [,]. Furthermore, it has been found that fennel fruits contain a complex mixture of volatile constituents representing more than 98% of the total essential oil content. Among these, estragole levels may range from 34% to 89%, depending on environmental factors []. The fatty acid profile of fennel fruit oil comprises approximately 4% palmitic acid, 22% oleic acid, 14% linoleic acid, and 6% petroselinic acid. These compounds are distributed throughout the seeds, roots, stems, flowers, and fruits, with varying concentrations across the different plant tissues []. The chemical structures of the main volatile and non-volatile compounds are illustrated in Figure 2.
Figure 2. Chemical structures of the main volatile and non-volatile compounds of Foeniculum vulgare Mill. (ChemDraw 8.0).

4.2. Phenolic Compounds

FV also contains a variety of phenolic compounds, such as flavonoids, phenolic acids (hydroxycinnamic and hydroxybenzoic acids), coumarin, and tannin []. These compounds are believed to contribute to the prevention of diseases linked to oxidative stress, such as cardiovascular diseases, cancer, and inflammation [].

4.2.1. Flavonoids

Flavonoids such as eriodictyol-7-rutinoside and quercetin-3-rutinoside have been isolated from FV. In addition, its aqueous extract contains quercetin-3-O-galactoside, kaempferol-3-O-rutinoside, and kaempferol-3-O-glucoside. Further studies have identified quercetin-3-O-glucuronide, kaempferol-3-O-glucuronide, isoquercetin, and isorhamnetin-3-O-glucoside as additional flavonoid constituents of FV []. HPLC-DAD analysis of the butanolic fraction of fennel seeds also revealed the presence of epigallocatechin, vanillin (a phenolic aldehyde), rutin (a flavonoid glycoside), and naringin []. The chemical structures of the major flavonoids are illustrated in Figure 3.
Figure 3. Chemical structures of the main flavonoids of Foeniculum vulgare Mill. (ChemDraw 8.0).

4.2.2. Phenolic Acids

FV has been reported to contain a wide range of phenolic acids, including 3-O-caffeoylquinic acid, 4-O-caffeoylquinic acid, 5-O-caffeoylquinic acid, 1,3-O-dicaffeoylquinic acid, 1,4-O-dicaffeoylquinic acid, and 1,5-O-dicaffeoylquinic acid []. Moreover, HPLC-DAD analysis of the butanolic fraction of fennel seeds revealed the presence of hydroxybenzoic acids, such as syringic acid, and hydroxycinnamic acids, including chlorogenic acid, caffeic acid, p-coumaric acid, and ferulic acid []. The chemical structures of the principal phenolic acids are illustrated in Figure 4.
Figure 4. Chemical structures of the main phenolic acids of Foeniculum vulgare Mill. (ChemDraw 8.0).

4.2.3. Coumarins

Using TLC analysis, several coumarins such as imperatorin, psoralen, bergapten, xanthotoxin, and isopimpinellin were isolated from the methylene chloride extract of FV fruits []. Similarly, four coumarin compounds, namely scopoletin, 8-methoxypsoralen, bergapten, and imperatorin, were identified in the methanolic extract of fennel fruits []. The chemical structures of the major coumarins are presented in Figure 5 below.
Figure 5. Chemical structures of the main coumarin of Foeniculum vulgare Mill. (ChemDraw 8.0).

5. Cardiovascular Activities of Foeniculum vulgare Mill.

Table 1 summarizes studies investigating the cardiovascular activities of FV, including the plant parts used, tested bioactive compounds, extract types and administered doses, experimental models employed, types of tests conducted, and the cardiovascular outcomes reported in each study. Collectively, these experimental findings indicate that FV exhibits significant potential in cardiovascular protection, attributed to its hypotensive, antihypertensive, vasorelaxant, hypolipidemic, antithrombotic, anticoagulant, antioxidant, diuretic, cardioprotective, and anti-inflammatory activities. Among the various plant parts evaluated, the seeds are the most frequently used in experimental studies (43%), followed by the leaves (20.5%), aerial parts (11.4%), fruits (6.9%), stems (4.5%), bulbs (2.3%), shoots (2.3%), and inflorescences (2.3%), as illustrated in Figure 6.
Table 1. Biological and pharmacological studies on the cardiovascular activities of Foeniculum vulgare Mill.
Figure 6. Illustration showing the percentage of Foeniculum vulgare Mill. parts used in experimental studies investigating its cardiovascular-related effects.

5.1. Hypotensive Activity

Several studies have reported the hypotensive potential of FV extracts using different experimental models. The aqueous seed extract of FV (FVE) significantly reduced intraocular pressure (IOP) in normotensive rabbits, showing effects comparable to timolol []. This activity has been attributed to its anticholinesterase effect, which increases acetylcholine availability, enhances aqueous humor outflow, and consequently decreases IOP []. Similarly, the decocted aqueous extract of FV leaves (12 mg/kg/day) reduced arterial blood pressure in normotensive rats without affecting pulse or respiratory rate. This hypotensive effect appears to involve histaminergic pathways, whereas cholinergic, serotonergic, ganglionic, and adrenergic mechanisms were not implicated []. Comparable hypotensive effects were also observed for FV essential oil constituents such as estragole and trans-anethole, which induced dose-dependent decreases in mean arterial pressure and heart rate in normotensive rats []. Likewise, monoterpenes including (+)-α-pinene, (–)-β-pinene, (±)-citronellol, and (±)-linalool produced hypotension accompanied by reflex tachycardia, suggesting peripheral vasodilation and baroreflex activation []. Taken together, these findings suggest that FV exerts its hypotensive effect through multiple mechanisms, likely involving both central and peripheral pathways, partly mediated by its major volatile and non-volatile constituents.

5.2. Antihypertensive Activity

Several studies have demonstrated that FV and its bioactive compounds exert antihypertensive effects mainly through inhibition of the renin-angiotensin system, particularly the angiotensin-converting enzyme (ACE) and angiotensin II receptor pathways. In vitro investigations revealed that both the essential oil and its major constituent anethole significantly inhibited ACE activity, with IC50 values of 40.7 ± 3.5 µg/mL and 52 ± 5.8 µg/mL, respectively []. Consistently, methanolic extracts of FV leaves inhibited ACE activity by about 50% []. Similar effects were observed for phenolic acids such as gallic, caffeic, and coumaric acids, which exhibited substantial ACE inhibitory potential, with caffeic acid showing the strongest effect (IC50 = 157.3 ± 16.1 µM) []. Other polyphenols, including kaempferol and quercetin, have also been shown to inhibit ACE activity and downregulate the expression of the angiotensin II type 1 receptor (AT1R), both in vitro and in vivo [,,]. Molecular docking analyses confirmed these findings, showing a strong binding affinity of quercetin to the active site of human ACE (ΔG = −8.1 kcal/mol) []. Likewise, naringin was reported to inhibit ACE activity and suppress angiotensin II-induced gene expression []. In vivo, the aqueous extract of FV fruits (190 mg/kg/day) significantly lowered systolic blood pressure in spontaneously hypertensive rats but not in normotensive controls, suggesting a selective effect under pathological conditions []. This extract also promoted diuresis by increasing water, sodium, and potassium excretion. Similar blood pressure reductions were observed with nanoemulsions rich in FV phenolic compounds, which significantly decreased mean, systolic, and diastolic blood pressure while normalizing heart rate in salt-induced hypertensive rats []. Additionally, treatment with trans-anethole reduced AT1R expression, further supporting the role of FV constituents in modulating the renin-angiotensin system []. These results suggest that FV exerts its antihypertensive effect primarily through inhibition of ACE activity, downregulation of AT1R expression, and enhancement of renal excretory function by its bioactive constituents.

5.3. Diuretic Activity

Several studies have demonstrated that FV exhibits notable diuretic activity through multiple mechanisms involving increased urine output and modulation of electrolyte excretion. The ethanolic extract of FV fruits (500 mg/kg) significantly enhanced diuresis in rats at both 5 and 24 h post-administration, producing an effect comparable to urea (960 mg/kg) and nearly doubling urine output compared to controls, without altering sodium or potassium excretion []. Consistently, both aqueous and methanolic extracts of FV leaves (200–400 mg/kg) induced a dose-dependent increase in urine volume and significantly enhanced natriuresis, kaliuresis, and chloriuresis []. At the phytochemical level, several FV-derived compounds also contribute to its diuretic action. Phenolic acids such as chlorogenic and caffeic acids increased urine volume and sodium excretion, while rosmarinic acid displayed a potassium-sparing effect []. Similarly, the fenchone (400 mg/kg) enhanced urine output and electrolyte (Na+, K+, Ca2+) excretion in a dose-dependent manner []. Flavonoids such as isoquercitrin and (−)-epicatechin further promoted diuresis and saluresis (Na+, K+, Cl) without disturbing plasma electrolyte balance or urinary pH [,]. These findings indicate that FV promotes diuresis through synergistic actions of its phenolic, flavonoid, and monoterpene constituents, acting via osmotic and ion-transport modulation mechanisms.

5.4. Vasorelaxant Activity

Several studies have demonstrated that FV exerts significant vasorelaxant effects through multiple endothelium-dependent and -independent mechanisms involving nitric oxide (NO), calcium modulation, and potassium channel activation. In vivo, treatment with the aqueous extract of FV fruits (190 mg/kg/day for 4 days) reduced noradrenaline-induced aortic contraction by 19 ± 6.2% in SHR, but not in normotensive rats. The effect was abolished by Nω-Nitro-L-arginine methyl ester (L-NAME), indicating NO-mediated relaxation []. Similarly, aqueous and methylene chloride fractions of FV extracts induced concentration-dependent vasodilation in mesenteric arteries and isolated rat aorta, respectively, mediated via the NO/cGMP pathway and muscarinic receptor activation [,]. The butanolic fraction, rich in phenolic acids (coumaric, syringic, ferulic) and the flavonoid naringin, exhibited the strongest vasorelaxant activity in mesenteric arteries. Increased nitrite and cGMP levels following FV seed extract treatment further support enhanced endothelial NO production []. Essential oil and its major compound trans-anethole (1–300 µg/mL) also produced complete, concentration-dependent relaxation of phenylephrine- and KCl-induced contractions in isolated rat aorta, independent of the endothelium [,]. These effects may involve inhibition of store-operated and voltage-dependent Ca2+ entry, suppression of IP3-dependent Ca2+ release, and blockade of non-selective cation channels and phospholipase C activation []. In addition, FV phenolic and flavonoid constituent such as caffeic, ferulic, and coumaric acids, quercetin, kaempferol, and naringenin have demonstrated potent vasodilatory properties. These compounds act via activation of the NO/sGC/cGMP pathway, modulation of Ca2+ influx, and opening of potassium channels, with both endothelium-dependent and independent effects [,,,]. Overall, FV induces vascular relaxation via complementary mechanisms, including NO-mediated pathways, calcium channel blockade, and potassium channel activation, primarily mediated by its vasoactive compounds.

5.5. Cardioprotective Activity

Treatment with hydroalcoholic extracts of FV fruits (200–400 mg/kg, 28 days) significantly protected the myocardium against isoproterenol-induced injury in rats, as evidenced by normalized cardiac marker enzymes, improved lipid and glucose profiles, elevated glutathione levels, and histological signs of myocardial regeneration []. Similarly, preincubation of H9C2 cardiomyocytes with ethanolic extracts or anethole markedly reduced oxidative stress, DNA damage, and mitochondrial dysfunction following hypoxia/reoxygenation injury []. Several bioactive constituents of FV also demonstrated cardioprotective potential. Caffeic acid phenethyl ester derivatives (3–15 mg/kg) attenuated ischemia/reperfusion injury in rabbits by suppressing MAPK phosphorylation and proinflammatory cytokine expression (IL-1β, TNF-α) [,,,,]. Syringic acid (50 mg/kg) protected against isoproterenol-induced cardiotoxicity over 30 days in rats [], while S-limonene (1 mg/kg) prevented ECG abnormalities, infarct size, and collagen deposition, likely via the inhibition of calcium overload and oxidative stress, and the restoration of antioxidant enzyme activity []. In addition, flavonoids such as quercetin and catechin enhanced cardiac recovery after ischemia/reperfusion by reducing inflammatory cytokines (IL-1β, TNF-α, IL-6), improving mitochondrial function, and limiting apoptosis through signaling pathways involving CREB/lncRNA MIAT/Akt/GSK-3β [,]. These findings indicate that FV confers cardioprotective effects through multiple complementary pathways, involving antioxidant, anti-inflammatory, and antiapoptotic mechanisms, along with the regulation of calcium homeostasis and preservation of mitochondrial function

5.6. Anticoagulant and Antithrombotic Activities

Several studies have demonstrated that FV and its main constituents exert potent antiplatelet and anticoagulant activities through multiple mechanisms. The essential oil and its major components, particularly (+)-fenchone, estragole, and anethole, significantly inhibited platelet aggregation induced by collagen, arachidonic acid (AA), and ADP in vitro, with effects comparable to those of acetylsalicylic acid (ASA). These compounds also reduced thromboxane A2 mediated aggregation and prevented thrombin-induced clot retraction, leading to marked antithrombotic protection in vivo, where anethole and the essential oil (30 mg/kg/day) prevented 83% and 70% of paralysis events in a mouse thromboembolism model, respectively [,]. Polyphenolic compounds such as chlorogenic, p-coumaric, and syringic acids further contributed to these effects. Chlorogenic acid inhibited fibrin clot formation in a dose-dependent manner, prolonged prothrombin time (PT), and enhanced fibrinolysis []. Similarly, p-coumaric acid suppressed ADP- and AA-induced platelet aggregation in rabbits and human blood via decreased thromboxane B2 and prostaglandin E2 production []. Syringic acid also inhibited fibrin clot formation and reduced factor Xa activity, suggesting direct inhibition of procoagulant enzymes []. In addition, flavonoids such as quercetin and catechin displayed strong synergistic antiplatelet effects. Quercetin completely inhibited AA-induced aggregation and reduced thrombin-mediated platelet activation by suppressing Ca2+ mobilization and serotonin release. Together with catechin, it downregulated platelet GPIIb/IIIa expression through nitric oxide-dependent pathways [,]. In summary, these results suggest that FV exerts anticoagulant and antithrombotic effects through combined inhibition of platelet activation and aggregation, modulation of the arachidonic acid and nitric oxide pathways, and direct suppression of coagulation enzymes.

5.7. Anti-Inflammatory Activity

Numerous studies have shown that FV exhibits potent anti-inflammatory activity through the inhibition of key inflammatory mediators, signaling pathways, and enzymes. At the cellular and molecular level, the methylene chloride and ethanolic extracts of FV fruits significantly suppressed LPS-induced nitric oxide production and downregulated iNOS and COX-2 expression in RAW 264.7 macrophages. These extracts also reduced the production of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6, partly through inhibition of JNK1 and ERK1/2 phosphorylation [,]. Similarly, aqueous and hydromethanolic extracts decreased NF-κB activation, IκB-α phosphorylation, and NO release in microglial BV-2 cells, confirming their regulatory effects on NF-κB signaling and cytokine production [,]. In vivo studies further demonstrated that FV extracts alleviate acute and chronic inflammation. Methanolic and essential oil extracts markedly inhibited carrageenan- and arachidonic acid-induced paw and ear edema in rodents (up to 70% inhibition), while α-pinene and anethole exhibited comparable effects to indomethacin [,,]. Fennel essential oil (200–400 mg/kg) also protected against acetic acid-induced colitis by reducing MPO activity, TNF-α expression, and NF-κB p65 phosphorylation []. Additionally, aqueous seed extract attenuated intestinal inflammation in necrotizing enterocolitis, lowering caspase activation and inflammatory cytokine levels []. Selenium nanoparticles synthesized from fennel seeds significantly reduced arthritis severity and joint damage in arthritic mice []. Polyphenolic compounds such as quercetin, ferulic acid, chlorogenic acid derivatives, and p-coumaric acid contribute to the anti-inflammatory activity. These compounds inhibit COX-2 selectively, suppress NF-κB and NLRP3 inflammasome activation, and downregulate TNF-α, IL-6, and IL-1β expression [,,,]. Notably, quercetin glucoside, dicaffeoylquinic acid, and isorhamnetin glucuronide showed strong selective COX-2 inhibition with IC50 values between 9 and 16 µM []. Anethole and estragole also displayed anti-inflammatory potential in carrageenan models and modulated Th17/Treg immune balance [,]. These findings indicate that FV exerts its anti-inflammatory activity via several complementary mechanisms, including inhibition of NF-κB and MAPK signaling, suppression of iNOS and COX-2 expression, downregulation of pro-inflammatory cytokines, and modulation of immune cell responses.

5.8. Hypolipidemic Activity

Several experimental studies have demonstrated the hypolipidemic potential of FV through modulation of lipid metabolism, antioxidant protection, and regulation of cholesterol homeostasis. In obese male rats, oral administration of FV seed powder (300 mg/kg) for six weeks significantly reduced body weight, serum cholesterol, triglycerides, LDL, albumin, and total protein, while increasing HDL levels. Treatment also lowered liver enzyme activities (ALT, AST, ALP) and oxidative stress markers such as MDA and MPO, indicating hepatoprotective and antioxidant effects []. Similarly, intraperitoneal administration of aqueous seed extracts (50–200 mg/kg) for 14 days in BALB/c mice fed a cholesterol-enriched diet led to a significant decrease in total cholesterol levels, triglycerides, and LDL, and a dose-dependent increase in HDL levels. The extract also upregulated leptin receptor expression, suggesting improved lipid and energy metabolism regulation []. In rats fed a high-cholesterol diet, hydroalcoholic seed extract (150 mg/kg, i.p., 3 weeks) significantly reduced triglycerides, total cholesterol, LDL, LDH, ALT, and ALP, while increasing HDL levels, confirming its lipid-lowering and hepatoprotective activity []. At the molecular level, bioactive compounds from FV, including quercetin, ferulic acid, p-coumaric acid, and trans-anethole, contribute to its hypolipidemic effect through antioxidant and regulatory mechanisms. Quercetin has been shown to reduce erythrocyte cholesterol by up to 75% in hypercholesterolemic subjects and to promote LDL uptake in HepG2 cells [,]. Phenolic acids such as ferulic and p-coumaric acids inhibit lipid peroxidation and improve oxidative status, while trans-anethole decreases ROS generation and LDL oxidation, resulting in higher HDL and lower VLDL levels [,]. Limonene supplementation reduced HMG-CoA reductase activity and prevented the conversion of lb-LDL to sd-LDL, thereby decreasing atherogenic risk []. In liver cell models, quercetin, catechin, hesperidin, and isorhamnetin significantly downregulated the expression of SREBP-2 and LDLR, key regulators of cholesterol synthesis and clearance []. Overall, the evidence indicates that FV exerts hypolipidemic effects via a combination of antioxidant, hepatoprotective, and metabolic regulatory mechanisms, involving inhibition of HMG-CoA reductase, attenuation of LDL oxidation, and modulation of lipid-related gene expression.

5.9. Antioxidant Activity

Several studies have demonstrated the strong antioxidant potential of FV in both in vivo and in vitro models. Administration of the methanolic fruit extract (200 mg/kg/day) for three weeks significantly increased SOD and catalase activities compared to the control group []. Similarly, the methanolic seed extract showed strong antioxidant activity, achieving nearly complete inhibition of DPPH absorption, and provided cytoprotection against γ-irradiation by restoring MDA, catalase, and glutathione levels []. In vitro assays confirmed the antioxidant efficacy of various FV extracts (aqueous, ethanol, acetone, and methanol), with the distilled water extract exhibiting the highest lipid peroxidation inhibition in the FTC and β-carotene bleaching assays, while the acetone extract showed the greatest ABTS radical scavenging capacity []. Essential oils obtained from aerial parts also displayed potent antioxidant activity, surpassing that of α-tocopherol except at the lowest concentration tested []. Likewise, essential oil, diethyl ether, and ethyl acetate leaf extracts demonstrated significant DPPH radical scavenging effects, with IC50 values of 900, 6.2, and 1.5 µg/mL, respectively [], while the weak scavenging activity of fennel leaf oil was attributed to its anethole content []. Coumarins isolated from fennel fruits, like scopoletin, 8-methoxypsoralen, bergapten, and imperatorin exhibited strong DPPH and ABTS radical scavenging activities (50%) []. Methanolic extracts of fennel leaves, stems, shoots, and inflorescences displayed concentration-dependent increases in DPPH scavenging, reducing power, and β-carotene bleaching inhibition, with over 90% inhibition at higher doses []. Essential oils from aerial parts and fruits also showed high DPPH scavenging (>85%) and 5-lipoxygenase inhibition, though limited hydroxyl radical scavenging capacity []. Moreover, fennel leaf essential oil (EOF) reduced ABTS and H2O2 radicals by 50% and decreased ROS levels in leukocytes, accompanied by enhanced CAT, SOD, and GPx activities in a dose-dependent manner []. The hydromethanolic extract of fennel seeds increased hydroxyl radical scavenging activity up to 82.6% inhibition at 100 µg/mL []. Fennel fruit oil showed strong total antioxidant capacity (7.26 ± 0.34 mg GAE/g) and reducing power (FRAP EC50 = 63.44 ± 2.29 mg/mL) while reducing MDA and enhancing SOD and CAT activities in bleomycin-induced pulmonary fibrosis []. The aqueous and butanolic fractions of FV seeds also demonstrated potent, concentration-dependent antioxidant activities in DPPH and β-carotene assays, with butanolic fraction showing higher efficacy []. Other studies highlighted the contribution of individual compounds. α-Pinene and trans-anethole increased SOD, POX, CAT, and GST activities in insect models []. In colitis models, (–)-fenchone (150 mg/kg) elevated GSH and SOD while reducing MDA and MPO levels []. Flavonoids such as quercetin and kaempferol enhanced Nrf2 and antioxidant gene expression (SOD1, GPX3) and restored glutathione under oxidative stress [,]. Likewise, epicatechin and rutin exhibited potent radical scavenging abilities, while p-coumaric, chlorogenic, ferulic, and syringic acids provided additional antioxidant protection through ROS scavenging, metal chelation, and upregulation of endogenous defenses [,,,,]. These findings suggest that the antioxidant potential of FV may result from the combined actions of its phenolic, flavonoid, and terpenoid constituents, which contribute to radical scavenging, metal chelation, and reinforcement of antioxidant defense systems. Figure 7 bellow presents the cardiovascular and biological activities of FV, highlighting the principal underlying mechanisms involved.
Figure 7. Comprehensive summary of the mechanisms underlying the cardiovascular activities of Foeniculum vulgare Mill. ⊘ Inhibition; Increase; Decrease.
FV exerts its cardiovascular activities through multiple and complementary pathways, as summarized in Table 2. The table also highlights the bioactive compounds associated with these mechanisms.
Table 2. The summary of mechanisms and major bioactive compounds involved in cardiovascular activities.

6. Safety Profile of Foeniculum vulgare Mill.

Several studies have confirmed the non-toxic and safe nature of FV across different experimental models. In acute toxicity tests, ethanolic extracts administered to mice at doses up to 3 g/kg showed no mortality or major behavioral or physiological alterations, even at the highest concentration []. Only slight and transient reductions in locomotor activity and mild piloerection were observed []. Other solvent extracts (n-hexane, methylene chloride, ethyl acetate, methanol) were also found to be non-toxic up to 5.5 g/kg in mice [], and repeated oral administration up to 2 g/kg for 10 days caused no neurological or systemic toxicity []. Similarly, the butanolic fraction (2 g/kg) produced no adverse effects or mortality over a 14-day follow-up []. The lethal dose 50 of anethole in rats is estimated at 2090 mg/kg, and that of fennel essential oil at 1326 mg/kg. Subchronic exposure to one-third of this dose (695 mg/kg) induced only mild hepatic alterations, while long-term dietary administration caused no liver damage, confirming its low hepatotoxic potential []. However, some caution is warranted regarding estragole, a minor constituent of fennel oil, which has shown dose-dependent genotoxic and carcinogenic potential in certain models [,,,]. Due to its estrogenic activity, fennel preparations used for dysmenorrhea have also raised questions about possible teratogenic risks, though studies report no teratogenic effects up to 9.3 mg/mL []. Interestingly, anethole exhibits anti-genotoxic properties against several mutagens []. Finally, a 90-day subchronic study in rats receiving daily doses up to 1.56 g/kg confirmed the absence of chronic toxicity, supporting the plant’s long-term safety []. According to the U.S. Food and Drug Administration (FDA), FV is classified as a Generally Recognized As Safe (GRAS) substance under Title 21 CFR §182.10, as listed in the “Substances Added to Food” database (formerly EAFUS). This recognition supports the safety of FV for food and medicinal applications [].

7. Interactions Between Foeniculum vulgare Mill. and Cardiovascular Drugs

It has been demonstrated that FV inhibits cytochrome P450 3A4 (CYP3A4). Consequently, fennel may reduce the metabolism of several cardiovascular drugs that are primarily metabolized by this enzyme, which could alter their bioavailability and therapeutic efficacy. Among these drugs are diuretics (e.g., eplerenone, spironolactone, indapamide), whose reduced metabolism may increase plasma concentrations, thereby enhancing the diuretic effect and potentially leading to electrolyte imbalances such as hypokalemia or hyponatremia [,]. For calcium channel blockers (e.g., amlodipine, diltiazem, felodipine, isradipine, nifedipine, nimodipine, nitrendipine, nisoldipine, verapamil), inhibition of CYP3A4 by fennel may cause elevated plasma levels, resulting in excessive vasodilation, hypotension, or bradycardia [,,]. Similarly, β blockers (e.g., betaxolol, bisoprolol, carvedilol) may exhibit increased plasma concentrations due to decreased metabolic clearance, which could enhance their pharmacological effects and increase the risk of fatigue, bradycardia, or excessive hypotension [,,].

8. Limitations and Future Perspectives

Despite the promising findings highlighting the cardiovascular potential of FV, most of the available data are derived from in vitro and in vivo studies. However, these experimental results remain largely unconfirmed by clinical investigations in humans. Moreover, the pharmacokinetic and pharmacodynamic profiles of the plant’s bioactive constituents have not been fully elucidated, limiting our understanding of their absorption, metabolism, and mechanism of action. Comprehensive clinical trials and detailed pharmacological evaluations are therefore essential to validate these preclinical results and ensure the safety and efficacy of FV in cardiovascular therapy.

9. Conclusions

Experimental studies demonstrate that FV exhibits a wide spectrum of pharmacological and biological activities beneficial for cardiovascular health. These include hypotensive, diuretic, antihypertensive, cardioprotective, vasorelaxant, anti-inflammatory, antioxidant, hypolipidemic, and antiaggregant effects, all contributing to the maintenance of vascular and cardiac function. These effects are mainly attributed to its rich phytochemical composition, particularly flavonoids, phenolic acids, and volatile constituents, which act synergistically through multiple molecular pathways. Toxicological investigations indicate that FV extracts are generally safe and well tolerated at experimental doses, although caution should be exercised regarding the toxicity of estragole under conditions of chronic or high-dose exposure. Furthermore, possible pharmacological interactions between FV and cardiovascular drugs deserve further investigation to ensure safe therapeutic use. Overall, this review provides an integrated overview of the cardiovascular activities, phytochemical composition, potential drug interactions, and safety profile of FV. These findings scientifically support its traditional use and highlight its potential as a promising natural source for cardiovascular protection.

Author Contributions

Conceptualization, A.Z. (Amal Zahi) and A.L.; Methodology, A.Z. (Amal Zahi), A.L., A.Z. (Abderrahim Ziyyat), H.M. and B.K.; Validation, A.Z. (Amal Zahi), A.L., and B.K.; Writing—original draft, A.Z. (Amal Zahi) and A.L.; Writing—review and editing, A.Z. (Amal Zahi), A.L., A.R., N.A., M.R., M.N.P. and B.K.; Visualization, A.Z. (Amal Zahi) and A.L.; Supervision, A.L., A.Z. (Abderrahim Ziyyat), and B.K.; Funding acquisition, B.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2020R1I1A2066868), the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. RS-2020-NR049559), and the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT)(RS-2024-00350362).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ACEAngiotensin-Converting Enzyme
AKTProtein Kinase B
ALPAlkaline Phosphatase
ALTAlanine Aminotransferase
ASTAspartate Aminotransferase
CATCatalase
COX1/2Cyclooxygenase-1/2
CPKCreatine Phosphokinase
CREBcAMP Response Element-Binding Protein
cGMPCyclic Guanosine Monophosphate
CRPC-Reactive Protein
ERK1/2Extracellular Signal-Regulated Kinases 1 and 2
FVFoeniculum vulagre Mill.
GC-MSGas Chromatography-Mass Spectrometry
GPxGlutathione Peroxidase
GSK-3βGlycogen Synthase Kinase-3 Beta
GSTGlutathione S-Transferase
HepG2Human Liver Cancer Cell Line G2
HMG-CoA3-Hydroxy-3-Methylglutaryl-Coenzyme A
HPLC-DADHigh-Performance Liquid Chromatography with Diode Array Detection
IFN-γInterferon-Gamma
IKB-αInhibitor of Nuclear Factor Kappa-B Alpha
IL-1βInterleukin-1 Beta
IL-4Interleukin-4
IncRNA MIATLong Non-Coding RNA Myocardial Infarction Associated Transcript
iNOSInducible Nitric Oxide Synthase
JNK1c-Jun N-terminal Kinase 1
LDHLactate Dehydrogenase
L-NAMEN-G-Nitro-L-Arginine Methyl Ester
MAPKMitogen-Activated Protein Kinase
MPOMyeloperoxidase
NF-κBNuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells
NLRP3NOD-Like Receptor Pyrin Domain Containing 3
Nrf2Nuclear Factor Erythroid 2-Related Factor 2
PMAPhorbol 12-Myristate 13-Acetate
SODSuperoxide Dismutase
SREBP-2Sterol Regulatory Element-Binding Protein 2
TNF-αTumor Necrosis Factor Alpha

References

  1. WHO. Cardiovascular Diseases. 2021. Available online: https://www.who.int/health-topics/cardiovascular-diseases/#tab=tab_1 (accessed on 3 June 2024).
  2. WHR. Confronting the World’s Number One Killer; World Heart Federation. 2023. Available online: https://world-heart-federation.org/wp-content/uploads/World-Heart-Report-2023.pdf (accessed on 4 June 2024).
  3. Moon, K.Z.; Rahman, M.H.; Alam, M.J.; Hossain, M.A.; Hwang, S.; Kang, S.; Moon, S.; Park, M.N.; Ahn, C.-H.; Kim, B. Unraveling the interplay between cardiovascular diseases and alcohol use disorder: A bioinformatics and network-based exploration of shared molecular pathways and key biomarkers validation via western blot analysis. Comput. Biol. Chem. 2025, 115, 108338. [Google Scholar] [CrossRef]
  4. Covani, M.; Niccoli, G.; Fujimoto, D.; Scalamera, R.; Vergallo, R.; Porto, I.; McNulty, I.; Lee, H.; Kakuta, T.; Jang, I.-K. Plaque vulnerability and cardiovascular risk factor burden in acute coronary syndrome: An optical coherence tomography analysis. J. Am. Coll. Cardiol. 2025, 86, 77–89. [Google Scholar] [CrossRef] [PubMed]
  5. Naveed, M.; Majeed, F.; Taleb, A.; Zubair, H.M.; Shumzaid, M.; Farooq, M.A.; Baig, M.M.F.A.; Abbas, M.; Saeed, M.; Changxing, L. A Review of Medicinal Plants in Cardiovascular Disorders: Benefits and Risks. Am. J. Chin. Med. 2020, 48, 259–286. [Google Scholar] [CrossRef] [PubMed]
  6. Mota, A.H. A review of medicinal plants used in therapy of cardiovascular diseases. Int. J. Pharmacogn. Phytochem. Res. 2016, 8, 572–591. [Google Scholar]
  7. Kheirkhah, A.H.; Kavianpour, M.; Ababzadeh, S. Herbal medicine: A potent booster for stem cell therapy in cardiovascular diseases. Adv. Tradit. Med. 2025, 25, 395–410. [Google Scholar] [CrossRef]
  8. Celiński, R.; Krzemińska, B.; Grzywa-Celińska, A.; Szewczyk, G.; Szewczyk, K.D.S. A Review on the Potential Use of Medicinal Plants from the Apiaceae and the Rosaceae Families in Cardiovascular Diseases—Experimental Evidence and Traditional Applications. Appl. Sci. 2024, 14, 3728. [Google Scholar] [CrossRef]
  9. Muckensturm, B.; Foechterlen, D.; Reduron, J.-P.; Danton, P.; Hildenbrand, M. Phytochemical and chemotaxonomic studies of Foeniculum vulgare. Biochem. Syst. Ecol. 1997, 25, 353–358. [Google Scholar] [CrossRef]
  10. Choi, E.-M.; Hwang, J.-K. Antiinflammatory, analgesic and antioxidant activities of the fruit of Foeniculum vulgare. Fitoterapia 2004, 75, 557–565. [Google Scholar] [CrossRef]
  11. Zakernezhad, F.; Barati, M.; Sanadgol, N.; Movahedi, M.; Majd, A.; Golab, F. Assessing the Association Between Fennel (Foeniculum vulgare) Extract and Serum Lipid Profile and Leptin Receptor Expression. Basic Clin. Neurosci. J. 2021; in press. [Google Scholar]
  12. Zahi, A.; Driouech, M.; Hakkou, Z.; Mansouri, F.; El Hajji, F.; Ziyyat, A.; Mekhfi, H.; Bnouham, M.; Legssyer, A. Vasorelaxant effect of fennel seeds (Foeniculum vulgare Mill.) extracts on rat mesenteric arteries: Assessment of phytochemical profiling and antioxidant potential. Fitoterapia 2025, 181, 106359. [Google Scholar] [CrossRef]
  13. Natarajan, P.; Grace, J. Effect of Mill. on Isoproterenol induced myocardial infarction in Foeniculum vulgare albino rats. Asian J. Pharm. Pharmacol. 2019, 5, 259–265. [Google Scholar]
  14. Rahimi, R.; Ardekani, M.R.S. Medicinal properties of Foeniculum vulgare Mill. in traditional Iranian medicine and modern phytotherapy. Chin. J. Integr. Med. 2013, 19, 73–79. [Google Scholar] [CrossRef]
  15. Badgujar, S.B.; Patel, V.V.; Bandivdekar, A.H. Foeniculum vulgare Mill: A review of its botany, phytochemistry, pharmacology, contemporary application, and toxicology. BioMed Res. Int. 2014, 2014, 842674. [Google Scholar] [CrossRef]
  16. Septina, E.; Yetti, R.D.; Rivai, H. Overview of Traditional Use, Phytochemical, and Pharmacological Activities of Chinese Petai (Leucaena leucocephala). Int. J. Pharm. Sci. Med. 2020, 5, 1–10. [Google Scholar] [CrossRef]
  17. Rizvi, M.A.; Ali, S.A. Medicinal flowers of Pakistan. Int. J. Adv. Res 2016, 4, 1313–1341. [Google Scholar]
  18. Al-Snafi, A.E. The chemical constituents and pharmacological effects of Foeniculum vulgare-A review. IOSR J. Pharm. 2018, 8, 81–96. [Google Scholar]
  19. Khandelwal, K.R. Practical Pharmacognosy; Pragati Books Pvt. Ltd.: Pune, India, 2008. [Google Scholar]
  20. Díaz-Maroto, M.C.; Pérez-Coello, M.S.; Esteban, J.; Sanz, J. Comparison of the volatile composition of wild fennel samples (Foeniculum vulgare Mill.) from Central Spain. J. Agric. Food Chem. 2006, 54, 6814–6818. [Google Scholar] [CrossRef] [PubMed]
  21. Anubhuti, P.; Rahul, S.; Kant, K.C. Standardization of fennel (Foeniculum vulgare), its oleoresin and marketed ayurvedic dosage forms. Int. J. Pharm Sci Drug Res 2011, 3, 265–269. [Google Scholar]
  22. Telci, I.; Demirtas, I.; Sahin, A. Variation in plant properties and essential oil composition of sweet fennel (Foeniculum vulgare Mill.) fruits during stages of maturity. Ind. Crops Prod. 2009, 30, 126–130. [Google Scholar] [CrossRef]
  23. Diao, W.-R.; Hu, Q.-P.; Zhang, H.; Xu, J.-G. Chemical composition, antibacterial activity and mechanism of action of essential oil from seeds of fennel (Foeniculum vulgare Mill.). Food Control 2014, 35, 109–116. [Google Scholar] [CrossRef]
  24. Viuda-Martos, M.; Mohamady, M.A.; Fernández-López, J.; Abd ElRazik, K.A.; Omer, E.A.; Pérez-Alvarez, J.A.; Sendra, E. In vitro antioxidant and antibacterial activities of essentials oils obtained from Egyptian aromatic plants. Food Control 2011, 22, 1715–1722. [Google Scholar] [CrossRef]
  25. Damayanti, A.; Setyawan, E. Essential oil extraction of fennel seed (Foeniculum vulgare) using steam distillation. Int. J. Sci. Eng. 2012, 3, 12–14. [Google Scholar] [CrossRef]
  26. Ahmad, B.S.; Talou, T.; Saad, Z.; Hijazi, A.; Cerny, M.; Kanaan, H.; Chokr, A.; Merah, O. Fennel oil and by-products seed characterization and their potential applications. Ind. Crops Prod. 2018, 111, 92–98. [Google Scholar] [CrossRef]
  27. Ahmed, A.F.; Shi, M.; Liu, C.; Kang, W. Comparative analysis of antioxidant activities of essential oils and extracts of fennel (Foeniculum vulgare Mill.) seeds from Egypt and China. Food Sci. Hum. Wellness 2019, 8, 67–72. [Google Scholar] [CrossRef]
  28. Abdullah, B.H.; Abbas, I.S.; Jasiem, T.M. Phytochemical study and evaluation of Iraqi fennel seed oil as antibacterial of urinary tract infection. Indian J. Forensic Med. Toxicol. 2020, 14, 842–846. [Google Scholar]
  29. Díaz-Maroto, M.C.; Díaz-Maroto Hidalgo, I.J.; Sánchez-Palomo, E.; Pérez-Coello, M.S. Volatile components and key odorants of Fennel (Foeniculum vulgare Mill.) and Thyme (Thymus vulgaris L.) oil extracts obtained by simultaneous distillation− extraction and supercritical fluid extraction. J. Agric. Food Chem. 2005, 53, 5385–5389. [Google Scholar] [CrossRef]
  30. Mimica-Dukić, N.; Kujundžić, S.; Soković, M.; Couladis, M. Essential oil composition and antifungal activity of Foeniculum vulgare Mill. obtained by different distillation conditions. Phytother. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2003, 17, 368–371. [Google Scholar] [CrossRef]
  31. Mehra, N.; Tamta, G.; Nand, V. Phytochemical screening and in vitro antioxidant assays in Foeniculum vulgare Mill.(Fennel) seeds collected from Tarai region in the Uttarakhand. Indian J. Nat. Prod. Resour. IJNPR Former. Nat. Prod. Radiance NPR 2022, 13, 213–222. [Google Scholar]
  32. Napoli, E.M.; Curcuruto, G.; Ruberto, G. Screening the essential oil composition of wild Sicilian fennel. Biochem. Syst. Ecol. 2010, 38, 213–223. [Google Scholar] [CrossRef]
  33. Barakat, H.; Alkabeer, I.A.; Aljutaily, T.; Almujaydil, M.S.; Algheshairy, R.M.; Alhomaid, R.M.; Almutairi, A.S.; Mohamed, A. Phenolics and Volatile Compounds of Fennel (Foeniculum vulgare) Seeds and Their Sprouts Prevent Oxidative DNA Damage and Ameliorates CCl4-Induced Hepatotoxicity and Oxidative Stress in Rats. Antioxidants 2022, 11, 2318. [Google Scholar] [CrossRef]
  34. Parejo, I.; Viladomat, F.; Bastida, J.; Schmeda-Hirschmann, G.; Burillo, J.; Codina, C. Bioguided isolation and identification of the nonvolatile antioxidant compounds from fennel (Foeniculum vulgare Mill.) waste. J. Agric. Food Chem. 2004, 52, 1890–1897. [Google Scholar] [CrossRef]
  35. Faudale, M.; Viladomat, F.; Bastida, J.; Poli, F.; Codina, C. Antioxidant activity and phenolic composition of wild, edible, and medicinal fennel from different Mediterranean countries. J. Agric. Food Chem. 2008, 56, 1912–1920. [Google Scholar] [CrossRef] [PubMed]
  36. Nassar, M.I.; Aboutabl, E.-s.A.; Makled, Y.A.; El-Khrisy, E.-D.; Osman, A.F. Secondary metabolites and pharmacology of Foeniculum vulgare Mill. Subsp. Piperitum. Rev. Latinoam. Química 2010, 38, 103–112. [Google Scholar]
  37. Yang, I.J.; Lee, D.U.; Shin, H.M. Anti-inflammatory and antioxidant effects of coumarins isolated from Foeniculum vulgare in lipopolysaccharide-stimulated macrophages and 12-O-tetradecanoylphorbol-13-acetate-stimulated mice. Immunopharmacol. Immunotoxicol. 2015, 37, 308–317. [Google Scholar] [CrossRef]
  38. Agarwal, R.; Gupta, S.K.; Agrawal, S.S.; Srivastava, S.; Saxena, R. Oculohypotensive effects of Foeniculum vulgare in experimental models of glaucoma. Indian J. Physiol. Pharmacol. 2008, 52, 77–83. [Google Scholar]
  39. Abdul-Ghani, A.-S.; Amin, R. The vascular action of aqueous extracts of Foeniculum vulgare leaves. J. Ethnopharmacol. 1988, 24, 213–218. [Google Scholar] [CrossRef]
  40. Bardai, S.E.; Lyoussi, B.; Wibo, M.; Morel, N. Pharmacological evidence of hypotensive activity of Marrubium vulgare and Foeniculum vulgare in spontaneously hypertensive rat. Clin. Exp. Hypertens. 2001, 23, 329–343. [Google Scholar] [CrossRef]
  41. Tognolini, M.; Ballabeni, V.; Bertoni, S.; Bruni, R.; Impicciatore, M.; Barocelli, E. Protective effect of Foeniculum vulgare essential oil and anethole in an experimental model of thrombosis. Pharmacol. Res. 2007, 56, 254–260. [Google Scholar] [CrossRef]
  42. Swaminathan, A.; Sridhara, S.R.C.; Sinha, S.; Nagarajan, S.; Balaguru, U.M.; Siamwala, J.H.; Rajendran, S.; Saran, U.; Chatterjee, S. Nitrites Derived From Foneiculum Vulgare (Fennel) Seeds Promotes Vascular Functions. J. Food Sci. 2012, 77, H273–H279. [Google Scholar] [CrossRef]
  43. Tettey, C.; Yang, I.; Ocloo, A.; Shin, H. Vasorelaxant and Anti-Inflammatory Activities of the Methylene Chloride Fraction of F oeniculum vulgare Fruit Extract. J. Food Biochem. 2015, 39, 55–63. [Google Scholar] [CrossRef]
  44. Chaudhary, S.K.; Maity, N.; Nema, N.K.; Bhadra, S.; Saha, B.P.; Mukherjee, P.K. Angiotensin converting enzyme inhibition activity of fennel and coriander oils from India. Nat. Prod. Commun. 2013, 8, 1934578X1300800531. [Google Scholar] [CrossRef]
  45. Abu-Zaiton, A.; Alu’datt, M.; Wafa, M. Evaluating the effect of Foeniculum vulgare extract on enzymes related with blood pressure and diabetes (in vitro study). Int. J. Adv. Chem. Eng. Biol. Sci. 2015, 2, 77–80. [Google Scholar]
  46. Han, A.Y.; Lee, H.S.; Seol, G.H. Foeniculum vulgare Mill. increases cytosolic Ca2+ concentration and inhibits store-operated Ca2+ entry in vascular endothelial cells. Biomed. Pharmacother. 2016, 84, 800–805. [Google Scholar] [CrossRef] [PubMed]
  47. Das, S.; Rani, S. Formulation and Evaluation of Nanoemulsion of the Phenolic Content of Foeniculum vulgare for Antidepressant and Antihypertensive Potentiality. Indian J. Pharm. Educ. Res. 2023, 57, s660–s666. [Google Scholar] [CrossRef]
  48. Seo, J.W.; Habiba, S.U.; Munni, Y.A.; Choi, H.J.; Aktar, A.; Mazumder, K.; Nah, D.-Y.; Yang, I.-J.; Moon, I.S. Protective Effects of Anethole in Foeniculum vulgare Mill. Seed Ethanol Extract on Hypoxia/Reoxygenation Injury in H9C2 Heart Myoblast Cells. Antioxidants 2024, 13, 1161. [Google Scholar] [CrossRef]
  49. Tanira, M.; Shah, A.; Mohsin, A.; Ageel, A.; Qureshi, S. Pharmacological and toxicological investigations on Foeniculum vulgare dried fruit extract in experimental animals. Phytother. Res. 1996, 10, 33–36. [Google Scholar] [CrossRef]
  50. Jemal, A. Evaluation of the Diuretic Activity of Aqueous and 80% Methanol Extracts of Foeniculum vulgare Mill (Apiaceae) Leaf in Rats. Masters’s Thesis, Addis Ababa University, Addis Ababa, Ethiopia, 2015. [Google Scholar]
  51. Mansouri, E.; Kooti, W.; Bazvand, M.; Boroon, M.G.; Amirzargar, A.; Afrisham, R.; Afzalzadeh, M.R.; Ashtary-Larky, D.; Jalali, N. The effect of hydro-alcoholic extract of Foeniculum vulgare Mill on leukocytes and hematological tests in male rats. Jundishapur J. Nat. Pharm. Prod. 2015, 10, e18396. [Google Scholar] [CrossRef]
  52. Lutviani, S.; Anisa, I.N.; Soemardji, A.A. Anti-Inflammatory and Mucolytic Activity Test of Ethanol Extract Fennel Leaf (Foeniculum vulgare Mill.). In Proceedings of the Conference on Natural Resources and Life Sciences 2022 (NRLS-BIO 2022), Online, 24–25 August 2022; Atlantis Press: Dordrecht, The Netherlands, 2023; pp. 133–137. [Google Scholar]
  53. Tanveer, M.; Shehzad, A.; Butt, M.; Shahid, M. Pharmacodynamics of Foeniculum vulgare against ulcer, inflammation, hepatotoxicity and nephrotoxicity. J. Anim. Plant Sci. 2021, 31, 841–853. [Google Scholar] [CrossRef]
  54. Panthong, S.; Itharat, A.; Aukkanibut, S.; Intarawan, T. Anti-inflammatory effect and total flavonoid content of the ethanolic seed extracts of three umbelliferae species. Sci. Technol. Asia 2020, 25, 174–181. [Google Scholar]
  55. Arif, A.; Bhatti, A.; John, P. Therapeutic potential of Foeniculum vulgare mill. Derived selenium nanoparticles in arthritic Balb/c mice. Int. J. Nanomed. 2019, 14, 8561–8572. [Google Scholar] [CrossRef]
  56. Özbek, H. The Anti-inflammatory Activity of the Foeniculum vulgare L. Essential Oil and Investigation of its Median Lrthal Dose in Rats and Mice. Int. J. Pharmacol. 2005, 50, 329–331. [Google Scholar]
  57. Özbek, H.; Sever Yilmaz, B. Anti-inflammatory and hypoglycemic activities of alpha-pinene. Acta Pharm. Sci. 2017, 55, 7. [Google Scholar] [CrossRef]
  58. Rezayat, S.M.; Dehpour, A.-R.; Motamed, S.M.; Yazdanparast, M.; Chamanara, M.; Sahebgharani, M.; Rashidian, A. Foeniculum vulgare essential oil ameliorates acetic acid-induced colitis in rats through the inhibition of NF-kB pathway. Inflammopharmacology 2018, 26, 851–859. [Google Scholar] [CrossRef] [PubMed]
  59. Crescenzi, M.A.; D’Urso, G.; Piacente, S.; Montoro, P. UPLC-ESI-QTRAP-MS/MS analysis to quantify bioactive compounds in fennel (foeniculum vulgare mill.) waste with potential anti-inflammatory activity. Metabolites 2022, 12, 701. [Google Scholar] [CrossRef] [PubMed]
  60. Yakut, H.I.; Koyuncu, E.; Cakir, U.; Tayman, C.; Koyuncu, İ.; Taskin Turkmenoglu, T.; Cakir, E.; Ozyazici, A.; Aydogan, S.; Zenciroglu, A. Preventative and therapeutic effects of fennel (Foeniculum vulgare) seed extracts against necrotizing enterocolitis. J. Food Biochem. 2020, 44, e13284. [Google Scholar] [CrossRef]
  61. Koppula, S.; Alluri, R.; Kopalli, S.R. Foeniculum vulgare Mill. inhibits lipopolysaccharide-induced microglia activation and ameliorates neuroinflammation-mediated behavioral deficits in mice. Asian Pac. J. Trop. Biomed. 2024, 14, 28–39. [Google Scholar] [CrossRef]
  62. Cherbal, A.; Bouabdallah, M.; Benhalla, M.; Hireche, S.; Desdous, R. Phytochemical screening, phenolic content, and anti-inflammatory effect of Foeniculum vulgare seed extract. Prev. Nutr. Food Sci. 2023, 28, 141. [Google Scholar] [CrossRef]
  63. Darzi, S.E.; Khazraei, S.P.; Amirghofran, Z. The immunoinhibitory and apoptosis-inducing activities of Foeniculum vulgare on human peripheral blood lymphocytes. Res. Pharm. Sci. 2018, 13, 103–110. [Google Scholar] [CrossRef]
  64. Lee, H.-S. Anticoagulant properties of compounds derived from Fennel (Foeniculum vulgare Gaertner) fruits. Food Sci. Biotechnol. 2006, 15, 763–767. [Google Scholar]
  65. Elghazaly, N.A.; Radwan, E.H.; Zaatout, H.H.; Elghazaly, M.M.; El din Allam, N. Beneficial effects of fennel (Foeniculum vulgare) in treating obesity in rats. J. Obes. Manag. 2019, 1, 16–33. [Google Scholar] [CrossRef]
  66. Naderi, G.A.; Roghani, M.; Esmaeil Jamaat, E.; Zahedi, E.; Sanaeirad, A. The effect of Foeniculum vulgare (Fennel) hydroalcoholic extract on serum lipid profiles and liver enzymes in male rats fed a high cholesterol regimen. J. Basic Clin. Pathophysiol. 2019, 7, 20–27. [Google Scholar]
  67. Mohamad, R.H.; El-Bastawesy, A.M.; Abdel-Monem, M.G.; Noor, A.M.; Al-Mehdar, H.A.R.; Sharawy, S.M.; El-Merzabani, M.M. Antioxidant and anticarcinogenic effects of methanolic extract and volatile oil of fennel seeds (Foeniculum vulgare). J. Med. Food 2011, 14, 986–1001. [Google Scholar] [CrossRef] [PubMed]
  68. Noreen, S.; Tufail, T.; Ain, H.; Khalid, W.; Hanif, A.; Ali, B.; Khan, M.; Iqbal, R.; Alwahibi, M.; ERCİŞLİ, S. Assessment of antioxidant activities of flaxseed (Linum usitatisimum L.) and fennel seed (Foeniculum vulgare Mill.) extracts. Pol. J. Environ. Stud. 2024, 33, 3359–3366. [Google Scholar] [CrossRef] [PubMed]
  69. Ruberto, G.; Baratta, M.T.; Deans, S.G.; Dorman, H.D. Antioxidant and antimicrobial activity of Foeniculum vulgare and Crithmum maritimum essential oils. Planta Medica 2000, 66, 687–693. [Google Scholar] [CrossRef] [PubMed]
  70. Ouariachi, E.; Lahhit, N.; Bouyanzer, A.; Hammouti, B.; Paolini, J.; Majidi, L.; Desjobert, J.M.; Costa, J. Chemical composition and antioxidant activity of essential oils and solvent extracts of Foeniculum vulgare Mill. from Morocco. J. Chem. Pharm. Res. 2014, 6, 743–748. [Google Scholar]
  71. Senatore, F.; Oliviero, F.; Scandolera, E.; Taglialatela-Scafati, O.; Roscigno, G.; Zaccardelli, M.; De Falco, E. Chemical composition, antimicrobial and antioxidant activities of anethole-rich oil from leaves of selected varieties of fennel [Foeniculum vulgare Mill. ssp. vulgare var. azoricum (Mill.) Thell]. Fitoterapia 2013, 90, 214–219. [Google Scholar] [CrossRef]
  72. Barros, L.; Heleno, S.A.; Carvalho, A.M.; Ferreira, I.C. Systematic evaluation of the antioxidant potential of different parts of Foeniculum vulgare Mill. from Portugal. Food Chem. Toxicol. 2009, 47, 2458–2464. [Google Scholar] [CrossRef]
  73. Miguel, M.G.; Cruz, C.; Faleiro, L.; Simões, M.T.; Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G. Foeniculum vulgare essential oils: Chemical composition, antioxidant and antimicrobial activities. Nat. Prod. Commun. 2010, 5, 1934578X1000500231. [Google Scholar] [CrossRef]
  74. Di Napoli, M.; Castagliuolo, G.; Badalamenti, N.; Maresca, V.; Basile, A.; Bruno, M.; Varcamonti, M.; Zanfardino, A. Antimicrobial, antibiofilm, and antioxidant properties of essential oil of Foeniculum vulgare Mill. leaves. Plants 2022, 11, 3573. [Google Scholar] [CrossRef]
  75. Singh, A.; Raja, W. Assessment of antioxidant activity of Foeniculum vulgare seed extract using fenton reaction. Res. J. Med. Plants Ayurveda 2020, 1, 1–7. [Google Scholar]
  76. Khammassi, M.; Abidi, A.; Ochi, N.; Ayadi, A.; Mabrouk, Y.; Amri, I.; De Feo, V.; Sebai, H.; Polito, F. Protective effect of essential oil of Foeniculum vulgare Mill. against bleomycin-induced pulmonary fibrosis and oxidative stress in rat. Phytomed. Plus 2024, 4, 100660. [Google Scholar] [CrossRef]
  77. Joshi, H.; Parle, M. Cholinergic basis of memory-strengthening effect of Foeniculum vulgare Linn. J. Med. Food 2006, 9, 413–417. [Google Scholar] [CrossRef] [PubMed]
  78. de Siqueira, R.J.; Magalhães, P.J.; Leal-Cardoso, J.H.; Duarte, G.P.; Lahlou, S. Cardiovascular effects of the essential oil of Croton zehntneri leaves and its main constituents, anethole and estragole, in normotensive conscious rats. Life Sci. 2006, 78, 2365–2372. [Google Scholar] [CrossRef] [PubMed]
  79. Menezes, I.A.; Barreto, C.M.; Antoniolli, A.R.; Santos, M.R.; de Sousa, D.P. Hypotensive activity of terpenes found in essential oils. Z. Naturforsch. C J. Biosci. 2010, 65, 562–566. [Google Scholar] [CrossRef] [PubMed]
  80. Hidalgo, M.; Martin-Santamaria, S.; Recio, I.; Sanchez-Moreno, C.; de Pascual-Teresa, B.; Rimbach, G.; de Pascual-Teresa, S. Potential anti-inflammatory, anti-adhesive, anti/estrogenic, and angiotensin-converting enzyme inhibitory activities of anthocyanins and their gut metabolites. Genes Nutr. 2012, 7, 295–306. [Google Scholar] [CrossRef]
  81. Olszanecki, R.; Bujak-Gizycka, B.; Madej, J.; Suski, M.; Wołkow, P.; Jawień, J.; Korbut, R. Kaempferol, but not resveratrol inhibits angiotensin converting enzyme. J. Physiol. Pharmacol. 2008, 59, 387–392. [Google Scholar]
  82. Maneesai, P.; Iampanichakul, M.; Potue, P.; Khamseekaew, J.; Tong-Un, T.; Prachaney, P.; Settheetham-Ishida, W.; Pakdeechote, P. Kaempferol Ameliorates Renal Remodelling by Inhibiting the Renin-Angiotensin System Cascade in Hypertensive Rats. J. Food Biochem. 2024, 2024, 8810152. [Google Scholar] [CrossRef]
  83. Huang, W.Y.; Fu, L.; Li, C.Y.; Xu, L.P.; Zhang, L.X.; Zhang, W.M. Quercetin, hyperin, and chlorogenic acid improve endothelial function by antioxidant, antiinflammatory, and ACE inhibitory effects. J. Food Sci. 2017, 82, 1239–1246. [Google Scholar] [CrossRef]
  84. Amal, Z.; Mounime, K.; Mounia, D.; Abderrahim, Z.; Hassane, M.; Mohamed, B.; Abdelkhaleq, L. The Contribution of the Rat Mesenteric Vascular Bed Model to Phytopharmacology with Computational Studies of the Main Vasorelaxant Phytochemicals. Lett. Drug Des. Discov. 2024, 21, 4184–4204. [Google Scholar] [CrossRef]
  85. Mahadevaswamy, M.; Suchitha, G.; Pavan, S.; Vivek, H.; Nithya, S.; Chandan, S.; Prasad, S.K.; Keshava Prasad, T.; Ahmad, S.F.; Attia, S.M. Naringin attenuates angiotensin II induced cardiac hypertrophy by inhibiting carbonic anhydrase II. Sci. Rep. 2025, 15, 11789. [Google Scholar]
  86. Samadi-Noshahr, Z.; Ebrahimzadeh-Bideskan, A.; Hadjzadeh, M.A.; Shafei, M.N.; Salmani, H.; Hosseinian, S.; Khajavi-Rad, A. trans-Anethole attenuated renal injury and reduced expressions of angiotensin II receptor (AT1R) and TGF-β in streptozotocin-induced diabetic rats. Biochimie 2021, 185, 117–127. [Google Scholar] [CrossRef]
  87. Moser, J.C.; Cechinel-Zanchett, C.C.; Mariano, L.N.B.; Boeing, T.; da Silva, L.M.; de Souza, P. Diuretic, Natriuretic and Ca2+-Sparing Effects Induced by Rosmarinic and Caffeic Acids in Rats. Rev. Bras. Farmacogn. 2020, 30, 588–592. [Google Scholar] [CrossRef]
  88. Bashir, A.; Mushtaq, M.N.; Younis, W.; Anjum, I. Fenchone, a monoterpene: Toxicity and diuretic profiling in rats. Front. Pharmacol. 2023, 14, 1119360. [Google Scholar] [CrossRef] [PubMed]
  89. Junior, A.G.; Gasparotto, F.M.; Boffo, M.A.; Lourenço, E.L.B.; Stefanello, M.É.A.; Salvador, M.J.; da Silva-Santos, J.E.; Marques, M.C.A.; Kassuya, C.A.L. Diuretic and potassium-sparing effect of isoquercitrin—An active flavonoid of Tropaeolum majus L. J. Ethnopharmacol. 2011, 134, 210–215. [Google Scholar] [CrossRef]
  90. Mariano, L.N.B.; Boeing, T.; da Silva, R.d.C.M.V.d.A.F.; Cechinel-Filho, V.; Niero, R.; da Silva, L.M.; de Souza, P.; Andrade, S.F.d. Preclinical evaluation of the diuretic and saluretic effects of (-)-epicatechin and the result of its combination with standard diuretics. Biomed. Pharmacother. 2018, 107, 520–525. [Google Scholar] [CrossRef] [PubMed]
  91. Mudnic, I.; Modun, D.; Rastija, V.; Vukovic, J.; Brizic, I.; Katalinic, V.; Kozina, B.; Medic-Saric, M.; Boban, M. Antioxidative and vasodilatory effects of phenolic acids in wine. Food Chem. 2010, 119, 1205–1210. [Google Scholar] [CrossRef]
  92. Yuan, T.-Y.; Zi-Ran, N.; Di, C.; Yu-Cai, C.; Hui-Fang, Z.; Lian-Hua, F.; Du, G.-H. Vasorelaxant effect of quercetin on cerebral basilar artery in vitro and the underlying mechanisms study. J. Asian Nat. Prod. Res. 2018, 20, 477–487. [Google Scholar] [CrossRef]
  93. Mahobiya, A.; Singh, T.U.; Rungsung, S.; Kumar, T.; Chandrasekaran, G.; Parida, S.; Kumar, D. Kaempferol-induces vasorelaxation via endothelium-independent pathways in rat isolated pulmonary artery. Pharmacol. Rep. 2018, 70, 863–874. [Google Scholar] [CrossRef]
  94. Tan, C.S.; Tew, W.Y.; Jingying, C.; Yam, M.F. Vasorelaxant effect of 5,7,4′-Trihydroxyflavanone (Naringenin) via endothelium dependent, potassium and calcium channels in Sprague Dawley rats: Aortic ring model. Chem. Biol. Interact. 2021, 348, 109620. [Google Scholar] [CrossRef]
  95. Opie, L.H.; Commerford, P.J.; Gersh, B.J.; Pfeffer, M.A. Controversies in ventricular remodelling. Lancet 2006, 367, 356–367. [Google Scholar] [CrossRef]
  96. Tan, J.; Ma, Z.; Han, L.; Du, R.; Zhao, L.; Wei, X.; Hou, D.; Johnstone, B.H.; Farlow, M.R.; Du, Y. Caffeic acid phenethyl ester possesses potent cardioprotective effects in a rabbit model of acute myocardial ischemia-reperfusion injury. Am. J. Physiol. -Heart Circ. Physiol. 2005, 289, H2265–H2271. [Google Scholar] [CrossRef]
  97. Aswar, U.; Mahajan, U.; Kandhare, A.; Aswar, M. Ferulic acid ameliorates doxorubicin-induced cardiac toxicity in rats. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 659–668. [Google Scholar] [CrossRef]
  98. Ding, S.K.; Wang, L.X.; Guo, L.S.; Luo, P.; Du, J.J.; Zhao, Z.L.; Wang, G.G. Syringic acid inhibits apoptosis pathways via downregulation of p38MAPK and JNK signaling pathways in H9c2 cardiomyocytes following hypoxia/reoxygenation injury. Mol. Med. Rep. 2017, 16, 2290–2294. [Google Scholar] [CrossRef]
  99. Shaik, A.H.; Al Omar, S.Y.; Mohammad, A.; Kodidhela, L.D. Combined cardio-protective ability of syringic acid and resveratrol against isoproterenol induced cardio-toxicity in rats via attenuating NF-kB and TNF-α pathways. Sci. Rep. 2020, 10, 1–13. [Google Scholar]
  100. Rhana, P.; Barros, G.M.; Santos, V.C.d.O.; Costa, A.D.; Santos, D.M.d.; Fernandes-Braga, W.; Durço, A.O.; Santos, M.R.V.; Roman-Campos, D.; Vasconcelos, C.M.L.d.; et al. S-limonene protects the heart in an experimental model of myocardial infarction induced by isoproterenol: Possible involvement of mitochondrial reactive oxygen species. Eur. J. Pharmacol. 2022, 930, 175134. [Google Scholar] [CrossRef] [PubMed]
  101. Liu, Y.; Song, Y.; Li, S.; Mo, L. Cardioprotective effect of quercetin against ischemia/reperfusion injury is mediated through NO system and mitochondrial K-ATP channels. Cell J. (Yakhteh) 2021, 23, 184. [Google Scholar]
  102. Cong, L.; Su, Y.; Wei, D.; Qian, L.; Xing, D.; Pan, J.; Chen, Y.; Huang, M. Catechin relieves hypoxia/reoxygenation-induced myocardial cell apoptosis via down-regulating lncRNA MIAT. J. Cell. Mol. Med. 2020, 24, 2356–2368. [Google Scholar] [CrossRef]
  103. Choi, J.H.; Kim, S. Investigation of the anticoagulant and antithrombotic effects of chlorogenic acid. J. Biochem. Mol. Toxicol. 2017, 31, e21865. [Google Scholar] [CrossRef]
  104. Luceri, C.; Giannini, L.; Lodovici, M.; Antonucci, E.; Abbate, R.; Masini, E.; Dolara, P. p-Coumaric acid, a common dietary phenol, inhibits platelet activity in vitro and in vivo. Br. J. Nutr. 2007, 97, 458–463. [Google Scholar] [CrossRef]
  105. Choi, J.-H.; Kim, S. Mechanisms of attenuation of clot formation and acute thromboembolism by syringic acid in mice. J. Funct. Foods 2018, 43, 112–122. [Google Scholar] [CrossRef]
  106. Pignatelli, P.; Pulcinelli, F.M.; Celestini, A.; Lenti, L.; Ghiselli, A.; Gazzaniga, P.P.; Violi, F. The flavonoids quercetin and catechin synergistically inhibit platelet function by antagonizing the intracellular production of hydrogen peroxide. Am. J. Clin. Nutr. 2000, 72, 1150–1155. [Google Scholar] [CrossRef]
  107. Navarro-Núñez, L.; Rivera, J.; Guerrero, J.A.; Martínez, C.; Vicente, V.; Lozano, M. Differential effects of quercetin, apigenin and genistein on signalling pathways of protease-activated receptors PAR1 and PAR4 in platelets. Br. J. Pharmacol. 2009, 158, 1548–1556. [Google Scholar] [CrossRef]
  108. Zhu, H.; Liang, Q.-h.; Xiong, X.-g.; Wang, Y.; Zhang, Z.-h.; Sun, M.-j.; Lu, X.; Wu, D. Anti-inflammatory effects of p-coumaric acid, a natural compound of Oldenlandia diffusa, on arthritis model rats. Evid. Based Complement. Altern. Med. 2018, 2018, 5198594. [Google Scholar] [CrossRef]
  109. Liu, Y.; Shi, L.; Qiu, W.; Shi, Y. Ferulic acid exhibits anti-inflammatory effects by inducing autophagy and blocking NLRP3 inflammasome activation. Mol. Cell. Toxicol. 2022, 18, 509–519. [Google Scholar] [CrossRef]
  110. Zhang, L.; Fan, Y.; Su, H.; Wu, L.; Huang, Y.; Zhao, L.; Han, B.; Shu, G.; Xiang, M.; Yang, J.-M. Chlorogenic acid methyl ester exerts strong anti-inflammatory effects via inhibiting the COX-2/NLRP3/NF-κB pathway. Food Funct. 2018, 9, 6155–6164. [Google Scholar] [CrossRef]
  111. Ponte, E.L.; Sousa, P.L.; Rocha, M.V.; Soares, P.M.; Coelho-de-Souza, A.N.; Leal-Cardoso, J.H.; Assreuy, A. Comparative study of the anti-edematogenic effects of anethole and estragole. Pharmacol. Rep. 2012, 64, 984–990. [Google Scholar] [CrossRef]
  112. Zhang, S.; Chen, X.; Devshilt, I.; Yun, Q.; Huang, C.; An, L.; Dorjbat, S.; He, X. Fennel main constituent, trans-anethole treatment against LPS-induced acute lung injury by regulation of Th17/Treg function. Mol. Med. Rep. 2018, 18, 1369–1376. [Google Scholar] [CrossRef]
  113. Duchnowicz, P.; Broncel, M.; Podsędek, A.; Koter-Michalak, M. Hypolipidemic and antioxidant effects of hydroxycinnamic acids, quercetin, and cyanidin 3-glucoside in hypercholesterolemic erythrocytes (in vitro study). Eur. J. Nutr. 2012, 51, 435–443. [Google Scholar] [CrossRef]
  114. Ahmad, S.; Beg, Z.H. Hypolipidemic and antioxidant activities of thymoquinone and limonene in atherogenic suspension fed rats. Food Chem. 2013, 138, 1116–1124. [Google Scholar] [CrossRef] [PubMed]
  115. Rajeshwari, U.; Shobha, I.; Andallu, B. Comparison of aniseeds and coriander seeds for antidiabetic, hypolipidemic and antioxidant activities. Spatula DD 2011, 1, 9–16. [Google Scholar] [CrossRef]
  116. Dhivya, K.; Vengateswari, G.; Arunthirumeni, M.; Karthi, S.; Senthil-Nathan, S.; Shivakumar, M.S. Bioprospecting of Prosopis juliflora (Sw.) DC seed pod extract effect on antioxidant and immune system of Spodoptera litura (Lepidoptera: Noctuidae). Physiol. Mol. Plant Pathol. 2018, 101, 45–53. [Google Scholar] [CrossRef]
  117. Araruna, M.E.C.; Alves Júnior, E.B.; de Lima Serafim, C.A.; Pessoa, M.M.B.; de Souza Pessôa, M.L.; Alves, V.P.; Sobral, M.V.; da Silva, M.S.; Alves, A.F.; de Paiva Sousa, M.C. (−)-Fenchone Ameliorates TNBS-Induced Colitis in Rats via Antioxidant, Immunomodulatory, and Cytoprotective Mechanisms. Pharmaceuticals 2024, 18, 18. [Google Scholar] [CrossRef]
  118. Prasad, J.; Baitharu, I.; Sharma, A.K.; Dutta, R.; Prasad, D.; Singh, S.B. Quercetin Reverses Hypobaric Hypoxia-Induced Hippocampal Neurodegeneration and Improves Memory Function in the Rat. High Alt. Med. Biol. 2013, 14, 383–394. [Google Scholar] [CrossRef]
  119. Hsu, J.-Y.; Lin, H.-H.; Chyau, C.-C.; Wang, Z.-H.; Chen, J.-H. Aqueous Extract of Pepino Leaves Ameliorates Palmitic Acid-Induced Hepatocellular Lipotoxicity via Inhibition of Endoplasmic Reticulum Stress and Apoptosis. Antioxidants 2021, 10, 903. [Google Scholar] [CrossRef]
  120. Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef]
  121. Lee, S.-J.; Mun, G.-I.; An, S.-M.; Boo, Y.-C. Evidence for the association of peroxidases with the antioxidant effect of p-coumaric acid in endothelial cells exposed to high glucose plus arachidonic acid. BMB Rep. 2009, 42, 561–567. [Google Scholar] [CrossRef]
  122. Peng, J.; Zheng, T.-t.; Liang, Y.; Duan, L.-f.; Zhang, Y.-d.; Wang, L.-J.; He, G.-m.; Xiao, H.-t. p-Coumaric Acid Protects Human Lens Epithelial Cells against Oxidative Stress-Induced Apoptosis by MAPK Signaling. Oxidative Med. Cell. Longev. 2018, 2018, 8549052. [Google Scholar] [CrossRef]
  123. Bian, Y.-Y.; Guo, J.; Majeed, H.; Zhu, K.-X.; Guo, X.-N.; Peng, W.; Zhou, H.-M. Ferulic acid renders protection to HEK293 cells against oxidative damage and apoptosis induced by hydrogen peroxide. Vitr. Cell. Dev. Biol. Anim. 2015, 51, 722–729. [Google Scholar] [CrossRef]
  124. Li, Y.; Zhang, L.; Wang, X.; Wu, W.; Qin, R. Effect of Syringic acid on antioxidant biomarkers and associated inflammatory markers in mice model of asthma. Drug Dev. Res. 2019, 80, 253–261. [Google Scholar] [CrossRef]
  125. Shah, A.; Qureshi, S.; Ageel, A. Toxicity studies in mice of ethanol extracts of Foeniculum vulgare fruit and Ruta chalepensis aerial parts. J. Ethnopharmacol. 1991, 34, 167–172. [Google Scholar] [CrossRef]
  126. Kishore, R.N.; Anjaneyulu, N.; Ganesh, M.N.; Sravya, N. Evaluation of anxiolytic activity of ethanolic extract of Foeniculum vulgare in mice model. Int. J. Pharm. Pharm. Sci. 2012, 4, 584–586. [Google Scholar]
  127. Ostad, S.; Soodi, M.; Shariffzadeh, M.; Khorshidi, N.; Marzban, H. The effect of fennel essential oil on uterine contraction as a model for dysmenorrhea, pharmacology and toxicology study. J. Ethnopharmacol. 2001, 76, 299–304. [Google Scholar] [CrossRef] [PubMed]
  128. Drinkwater, N.R.; Miller, E.C.; Miller, J.A.; Pitot, H.C. Hepatocarcinogenicity of estragole (1-allyl-4-methoxybenzene) and 1′-hydroxyestragole in the mouse and mutagenicity of 1′-acetoxyestragole in bacteria. J. Natl. Cancer Inst. 1976, 57, 1323–1331. [Google Scholar] [CrossRef] [PubMed]
  129. Miller, J.A.; Miller, E.C. The metabolic activation and nucleic acid adducts of naturally-occurring carcinogens: Recent results with ethyl carbamate and the spice flavors safrole and estragole. Br. J. Cancer 1983, 48, 1–15. [Google Scholar] [CrossRef]
  130. Miller, E.C.; Swanson, A.B.; Phillips, D.H.; Fletcher, L.; Liem, A.; Miller, J.A. Structure-activity studies of the carcinogenicities in the mouse and rat of some naturally occurring and synthetic alkenylbenzene derivatives related to safrole and estragole. Cancer Res. 1983, 43, 1124–1134. [Google Scholar] [PubMed]
  131. Paini, A.; Punt, A.; Viton, F.; Scholz, G.; Delatour, T.; Marin-Kuan, M.; Schilter, B.; van Bladeren, P.J.; Rietjens, I.M. A physiologically based biodynamic (PBBD) model for estragole DNA binding in rat liver based on in vitro kinetic data and estragole DNA adduct formation in primary hepatocytes. Toxicol. Appl. Pharmacol. 2010, 245, 57–66. [Google Scholar] [CrossRef]
  132. Ostad, S.; Khakinegad, B.; Sabzevari, O. Evaluation of the teratogenicity of fennel essential oil (FEO) on the rat embryo limb buds culture. Toxicol. Vitr. 2004, 18, 623–627. [Google Scholar] [CrossRef]
  133. Abraham, S. Anti-genotoxicity of trans-anethole and eugenol in mice. Food Chem. Toxicol. 2001, 39, 493–498. [Google Scholar] [CrossRef]
  134. Dai, G.; Wang, C.; Tang, W.; Liu, J.; Xue, B. A 90-Day Oral Toxicity Study of the Ethanol Extract from Eupatorium japonicum Thunb and Foeniculum vulgare in Rats. BioMed Res. Int. 2020, 2020, 6859374. [Google Scholar] [CrossRef]
  135. FDA. U.S. Food and Drug Administration. Fennel, Sweet (Foeniculum vulgare mill. var. Dulce); FDA: Silver Spring, MD, USA, 2025.
  136. Zisaki, A.; Miskovic, L.; Hatzimanikatis, V. Antihypertensive drugs metabolism: An update to pharmacokinetic profiles and computational approaches. Curr. Pharm. Des. 2015, 21, 806–822. [Google Scholar] [CrossRef]
  137. Danjuma, M.I.; Mukherjee, I.; Makaronidis, J.; Osula, S. Converging indications of aldosterone antagonists (spironolactone and eplerenone): A narrative review of safety profiles. Curr. Hypertens. Rep. 2014, 16, 414. [Google Scholar] [CrossRef]
  138. Flockhart, D.A.; Thacker, D.; McDonald, C.; Desta, Z. The Flockhart Cytochrome P450 Drug-Drug Interaction Table. 2021. Available online: https://drug-interactions.medicine.iu.edu/ (accessed on 21 October 2025).
  139. Akbar, M.; Henderson, C.; Helen Grant, M. Inhibition of cell growth and glutathione depletion by calcium channel blockers. Toxicology 2008, 253, 14–15. [Google Scholar] [CrossRef]
  140. Mazzari, A.L.; Prieto, J.M. Herbal medicines in Brazil: Pharmacokinetic profile and potential herb-drug interactions. Front. Pharmacol. 2014, 5, 162. [Google Scholar] [CrossRef]
  141. Wessler, J.D.; Grip, L.T.; Mendell, J.; Giugliano, R.P. The P-glycoprotein transport system and cardiovascular drugs. J. Am. Coll. Cardiol. 2013, 61, 2495–2502. [Google Scholar] [CrossRef]
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.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.