Next Article in Journal
Screening of Antifungal Lactic Acid Bacteria and Their Impact on the Quality and Shelf Life of Rye Bran Sourdough Bread
Previous Article in Journal
Obtention and Characterisation of Antioxidant-Rich Peptides from Defatted Grape Seed Meal Using Different Enzymes
Previous Article in Special Issue
Nutritional Enhancement of Rice Noodles with Watermeal (Wolffia globosa)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Comprehensive Review of Perilla frutescens: Chemical Composition, Pharmacological Mechanisms, and Industrial Applications in Food and Health Products

College of Biological and Food Engineering, Hubei Minzu University, Enshi 445000, China
*
Author to whom correspondence should be addressed.
Foods 2025, 14(7), 1252; https://doi.org/10.3390/foods14071252
Submission received: 8 March 2025 / Revised: 27 March 2025 / Accepted: 1 April 2025 / Published: 3 April 2025

Abstract

:
Perilla frutescens (L.) Britt., a multifunctional herbaceous plant, is widely used in traditional medicine and cuisine due to its rich array of bioactive compounds. To date, many key phytochemicals in P. frutescens have been identified, including volatile terpenoids (perillaldehyde, limonene,), flavonoids (luteolin, apigenin), and phenolic acids (rosmarinic acid derivatives), which exhibit significant antioxidant, anti-inflammatory, antiviral, anticancer, antibacterial, and blood sugar-lowering effects. Studies have shown that volatile oils, flavonoids, and phenolic acids in P. frutescens exert their effects in various experimental models. In food and industrial applications, P. frutescens shows innovative potential in functional foods, natural preservatives, and novel food additives, effectively extending food shelf life and providing antimicrobial protection. Moreover, research on the biology and genetic improvement of P. frutescens has provided new approaches to enhance its yield and bioactive content. Finally, this paper also discusses the safety and standardization issues of the plant, providing theoretical support for its widespread application.

1. Introduction

Perilla frutescens (L.) Britt. is an annual, erect herbaceous plant of the genus Perilla in the Lamiaceae family, widely distributed across Asia. It has been traditionally employed in medicinal and culinary practices for centuries across China, Japan, Korea, and other Asian countries [1]. Leaf color is a critical attribute for consumer preference in P. frutescens [2]. The plant species P. frutescens exhibits three distinct chemotypes: green, bicolored (green/red), and red (purple), characterized by differential pigmentation patterns during leaf and stem development [3,4]. Red perilla manifests uniform anthocyanin expression across both adaxial (upper) and abaxial (lower) leaf surfaces, presenting as dark erythroid pigmentation, while the bicolored phenotype displays this chromatic characteristic exclusively on the abaxial surface (Figure 1). The purple and green/purple P. frutescens leaves are considered medicinal, while the green leaves are considered edible [5]. As both a medicinal plant and an industrial crop, P. frutescens is extensively employed in the food, pharmaceutical, and cosmetic industries, largely due to its distinctive aromatic compounds [6].
P. frutescens is a versatile medicinal and edible plant with extensive utilization potential, as its leaves, stems, and seeds possess distinct bioactive properties [6]. Tender leaves are eaten fresh in salads, brewed into herbal tea, or added to soups, while the stems and leaves are commonly used for pickling. In traditional Chinese cuisine, P. frutescens leaves are frequently paired with seafood to alleviate gastrointestinal discomfort. Notably, P. frutescens leaf decoctions are traditionally consumed in summer to reduce heat stress and stimulate appetite [7]. In Japanese and Korean cuisines, fresh P. frutescens leaves are commonly used as a raw vegetable or garnish.
Comprehensive phytochemical studies have identified key bioactive constituents of P. frutescens, including volatile oils (perillaldehyde, limonene), flavonoids (apigenin, luteolin), phenolic acids (rosmarinic acid, caffeic acid), and triterpenoids. These bioactive compounds contribute to its diverse pharmacological activities, including antimicrobial, antiviral, anticancer, anti-inflammatory, antioxidant, hypoglycemic, and neuroprotective effects [8,9,10,11]. A variety of P. frutescens-derived products, including P. frutescens seed oil (with an omega-3 content exceeding 60%), functional teas, and alcoholic beverages, have gained prominence in niche markets, benefiting from the plant’s GRAS (Generally Recognized As Safe) status.
This review aims to integrate interdisciplinary research progress on P. frutescens, with a focus on its chemical composition, pharmacological mechanisms, applications in the food industry, and advances in synthetic biology. By addressing current research gaps and outlining future industrial development pathways, we propose a comprehensive “Resource Development-Mechanistic Investigation-Technological Transformation-Standard Regulation” framework. This framework offers a structured and scientific roadmap for the comprehensive exploration of P. frutescens’s potential, ensuring its safety, regulatory compliance, and long-term sustainability. This proposed framework not only optimizes the multifaceted value of P. frutescens but also sets a benchmark for the innovation and advancement of natural food additives. With its implementation, P. frutescens is expected to evolve from a traditional herb into a globally competitive health food ingredient, contributing to the green transformation of the food industry. The research direction of P. frutescens in the latest five years are shown in Table 1.

2. Chemical Composition

2.1. Volatile Oils

The essential oils of P. frutescens, predominantly composed of monoterpenes and sesquiterpenes, possess distinctive aromatic profiles and significant bioactivities, rendering them highly versatile in various applications [12,13]. Comprehensive phytochemical analyses have identified key constituents such as perillaldehyde, perilla ketone, limonene, shisofuran, farnesenes (Z, E, α), and trans-shisool [14]. GC-MS analysis identified perilla ketone as the dominant volatile constituent, constituting 80.76% of the essential oil profile. Other components included β-caryophyllene (1.65%), linalool (1.15%), caryophyllene oxide (1.12%), and apiol (1.19%) [8]. Among these compounds, perillaldehyde is the most characteristic component of P. frutescens leaf essential oil, distinguished by its strong herbal aroma and potent antimicrobial and anti-inflammatory properties [15]. Limonene, another key monoterpene, possesses diverse biological activities, including anticancer, antioxidant, and antiviral effects [16,17]. Additionally, perilla alcohol and β-caryophyllene have been validated for their pharmacological potential in preclinical models [1]. The composition of P. frutescens essential oils (PEOs) is subject to various environmental factors, including humidity, soil conditions, temperature, seasonal variations, harvest timing, sowing period, geographical distribution, and phenological stages [18]. Notably, PEOs modulate key targets such as SLC6A4 (serotonin transporter) and SLC6A3 (dopamine transporter), regulating serotonergic and dopaminergic synaptic pathways and thereby alleviating menopause-associated depression [19]. Additionally, PEOs exhibit potent larvicidal activity against Aedes aegypti mosquitoes, leading to a 68% reduction in dengue fever transmission rates in endemic regions [20]. PEOs have been formulated into bioinsecticides, exhibiting effectiveness against Tribolium castaneum and Lasioderma serricorne [21].
The biosynthesis of PEOs is predominantly driven by terpenoid metabolism, wherein monoterpenoids (such as perillaldehyde and limonene) are synthesized through the coordinated action of the mevalonate (MVA) and methylerythritol phosphate (MEP) pathways (Figure 2A). Typically, the MVA pathway operates in the cytoplasm of eukaryotic cells, whereas the MEP pathway functions within the plastids of plants and prokaryotes. In this process, glucose enters the cell and undergoes glycolysis in the cytoplasm, yielding pyruvate, which is subsequently converted into acetyl-CoA within the mitochondria. Acetyl-CoA then enters the MVA pathway to generate isopentenyl pyrophosphate (IPP), while its isomer, dimethylallyl pyrophosphate (DMAPP), is produced via the MEP pathway. These two intermediates serve as precursors for the synthesis of geranyl pyrophosphate (GPP), the fundamental building block for monoterpenoid biosynthesis. Under the catalysis of specific enzymes, GPP gives rise to various constituents of PEOs, leading to the classification of different chemical types (Figure 2B,C).

2.2. Flavonoids

P. frutescens is acknowledged as a valuable source of structurally diverse flavonoids, which have been extensively studied for their pharmacological properties. Wang et al. [22] identified three primary flavonoid aglycones (luteolin, apigenin, and 5,7,2′,5′-tetrahydroxyflavone), five flavonoid glycosides (luteolin-7-O-diglucuronide, apigenin-7-O-diglucuronide, luteolin-7-O-glucuronide, apigenin-7-O-glucuronide, and quercetin-3-O-β-D-glucuronide), and two anthocyanins (shisonin and malonylshisonin) in P. frutescens. Among these, luteolin (3′,4′,5,7-tetrahydroxyflavone) and apigenin (4′,5,7-trihydroxyflavone) are the principal bioactive flavonoids, demonstrating potent anticancer and anti-inflammatory activities [23,24,25]. Luteolin and apigenin are structurally homologous flavonoids, both possessing a common flavone backbone, while the presence of a 3′-hydroxyl group in luteolin’s B-ring confers greater lipophilicity relative to apigenin [26]. The flavonoid content in leaves, stems, and seeds varies considerably, and fluctuations in these levels can influence the overall antioxidant potential of P. frutescens [27]. P. frutescens leaves contain significantly higher levels of flavonoids compared to stems, with the flavonoid content in leaves exceeding twice that in stems. Furthermore, the total flavonoid content in leaves is approximately five times greater than in stems.

2.3. Phenolic Acids

P. frutescens is rich in diverse phenolic acids, including rosmarinic acid, caffeic acid, and their derivatives, such as 3′-dehydroxyrosmarinic acid-3-O-glucoside and rosmarinic acid-3-O-glucoside. Additional identified compounds include methyl rosmarinate, caffeic acid-3-O-glucoside, various caffeic acid esters, vanillic acid, protocatechuic acid, and chlorogenic acid [28,29]. Among these, P. frutescens leaves exhibit the highest concentration of phenolic acids, significantly surpassing those found in stems and seeds. P. frutescens floral tissues harvested at 20 days post anthesis demonstrate maximal phenolic compound yields, with rosmarinic acid constituting the predominant phytochemical component during this developmental phase [30]. Phytochemical profiling revealed significant interspecific and organ-specific variations, with rosmarinic acid identified as the predominant phenolic constituent in seed extracts across analyzed taxa (4606.4 μg/g). Strikingly, Perilla frutescens seeds exhibited exceptional phenolic accumulation, achieving the maximal total phenolic content of 4856.8 μg/g DW. Compared to other plant parts, such as the main stem, lateral stem, and petiole, both perilla leaves and roots also exhibit high phenolic content, ranging from 272.4 to 886.9 μg/g and 354.7 to 993.3 μg/g, respectively [10].
Studies have shown that P. frutescens polyphenol extracts enhance the freshness, herbal aroma, and overall sensory acceptability of grass carp (Ctenopharyngodon idella) by mitigating fishy odors resulting from lipid oxidation during cooking [31]. Both caffeic acid and rosmarinic acid enhance the hepatic synthesis of endogenous antioxidant enzymes and glutathione (GSH); however, rosmarinic acid demonstrates a superior ability to scavenge DPPH free radicals compared to caffeic acid [32,33]. Proton ionic liquids, in combination with microwave-assisted techniques, have been employed to extract rosmarinic acid with a purity reaching 90.4% [34]. Moreover, rosmarinic acid has been found to inhibit inflammatory responses and disrupt the metabolic and glycolytic pathways of Trichophyton mentagrophytes [35]. The synthesis pathway of P. frutescens flavonoids and phenolic acids is shown in Figure 3.

2.4. Anthocyanins

Anthocyanins exhibit a wide range of biological activities in traditional Chinese medicine, including antioxidative, antihypertensive, anti-inflammatory, anti-atherosclerotic, and anticancer effects. In P. frutescens, the anthocyanin profile predominantly consists of cyanidin-type derivatives, with malonylshisonin being the most abundant component followed by shisonin. The pigmentation of P. frutescens leaves is primarily attributed to anthocyanins; however, these compounds are highly susceptible to degradation under high pH, light exposure, heat, and ionizing substances [36]. Light conditions play a crucial role in anthocyanin biosynthesis, with red and blue light treatments enhancing anthocyanin accumulation by 4.3-fold compared to green light exposure [37]. In the red (purple) perilla, anthocyanins are responsible for the deep red and purple hues of the leaves, while in green perilla, anthocyanins are either absent or present in very low concentrations, resulting in entirely green leaves. The green/red perilla variety exhibits a unique combination of green leaves with purple undersides, where anthocyanins accumulate predominantly on the leaf backs. These differences in anthocyanin content and distribution are crucial for distinguishing and classifying the various P. frutescens varieties. Moreover, anthocyanins have been reported to regulate glucose metabolism, enhance insulin sensitivity, protect against ultraviolet-induced damage, and facilitate collagen synthesis [38].

2.5. Fatty Acids

P. frutescens seeds exhibit high concentrations of fatty acids, predominantly composed of palmitic acid (PA, C16:0), stearic acid (SA, C18:0), oleic acid (OA, C18:1), linoleic acid (LA, C18:2), and α-linolenic acid (ALA, C18:3). Notably, the seed oil of P. frutescens serves as an exceptional botanical source of polyunsaturated fatty acids (PUFA), distinguished by its uniquely high α-linolenic acid content. Accounting for 54–64% of total fatty acids, the ALA proportion in P. frutescens seed oil significantly surpasses that of most other plant-derived oils, establishing it as a preeminent natural reservoir of omega-3 fatty acids [6]. Fatty acid desaturase 3 (FAD3) serves as a critical enzymatic mediator in PUFA biosynthesis, specifically catalyzing the desaturation of linoleic acid into α-linolenic acid. Notably, genetic variations in FAD3, particularly single nucleotide polymorphisms (SNPs), have been strongly associated with quantitative alterations in fatty acid profiles, demonstrating significant correlations with LA and ALA content modulation across diverse biological systems [39]. Currently, P. frutescens seed oil has gained prominence as a natural functional edible oil, with its development and utilization expanding rapidly in both nutraceutical and culinary applications.

2.6. Polysaccharides

A novel polysaccharide (PFP) isolated from Perilla frutescens exhibits a unique compositional profile, comprising 89.73% neutral sugars, 8.93% uronic acid, and 1.56% protein, with a molecular weight of 1.18 × 106 Da. Integrated spectroscopic (FT-IR, NMR) and ion chromatographic analyses confirmed the pyranose configuration of PFP, revealing a monosaccharide composition of rhamnose, arabinose, galactose, glucose, xylose, and galacturonic acid in molar ratios of 0.13:0.55:1.40:1.00:0.13:0.22. Structural modeling suggests a backbone comprising α-L-Araf-(1→, →6)-β-D-Galp-(1→, →4)-α-D-Glcp-(1→, →1,4)-β-D-Xylp-(1→, →4)-α-GalpA-(1→, →2,4)-α-L-Rhap-(1→. Functionally, PFP demonstrates dual therapeutic potential by significantly inhibiting the proliferation of H22 hepatoma cells while concurrently mitigating chemotherapy-induced immunosuppressive effects on immune organs, highlighting its promise as a multifunctional adjuvant in oncological interventions [40].

3. Functional Activity

3.1. Antioxidant

The antioxidant capacity of P. frutescens is primarily assessed using in vitro assays that measure its free radical scavenging ability. A comparative analysis of 12 Perilla cultivars demonstrated substantial variability in their antioxidant activity [41]. The potent antioxidant properties of P. frutescens are attributed to its abundance of flavonoids, phenolic acids, and essential oils. Moreover, metabolite composition is a key determinant of P. frutescens leaf antioxidant activity, with higher metabolite levels correlating with enhanced antioxidant capacity [42].
Among the various P. frutescens varieties, purple-leaved P. frutescens demonstrates the highest antioxidant activity, primarily attributed to its elevated rosmarinic acid content (314.3 mg/g) [43]. Cold-pressed P. frutescens oil (CPPO) demonstrates significant anti-photoaging efficacy, effectively attenuating ultraviolet-induced wrinkle formation and melanin deposition in hairless murine models. Mechanistic studies revealed its potent antioxidant capacity through dual modulation of oxidative stress markers: suppressing reactive oxygen species (ROS) generation while downregulating superoxide dismutase (SOD) activity in normal human dermal fibroblasts (NHDFs). These dose-dependent effects (0.625–0.25%) confirm CPPO’s multimodal antioxidant-photo-protective properties [44].
Perillaldehyde, a key constituent of PEO, inhibits ROS production and exerts antioxidant effects by activating the nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) pathways in human keratinocytes [45]. Caffeic acid and rosmarinic acid enhance hepatic antioxidant enzyme activity, thereby exerting protective effects [32,46]. Luteolin has been reported to attenuate ROS generation and promote neuronal survival [47].

3.2. Anti-Inflammatory

P. frutescens demonstrates strong anti-inflammatory properties against multiple inflammatory disorders, including otitis, hepatitis, colitis, and airway inflammation [48]. The anti-inflammatory potential of P. frutescens has been predominantly investigated in animal models. P. frutescens extract (containing caffeic acid, rosmarinic acid, luteolin, and apigenin) from the whole plant markedly inhibits key inflammatory transcriptional regulators, such as nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3), thereby mitigating colitis symptoms in mice [49]. Additionally, it reduces the phosphorylation of Bruton’s tyrosine kinase (Tyr223 and Tyr174) in fMLF-activated human neutrophils while lowering intracellular Ca2+ levels [50].
P. frutescens seed oil exhibits inhibitory effects on ethyl phenylpropiolate (EPP)-induced auricular edema in rats, demonstrating anti-inflammatory activity. However, due to the required pharmacokinetic processes including absorption, metabolism, and systemic distribution to target cells, the inhibition rates of P. frutescens seed oil reached only 59.3% (0.1 mL/ear) and 65.7% (1 mL/ear) at the 15 min time point [51]. Moreover, perillaldehyde, perilla ketone, isoperilla ketone, ursolic acid, rosmarinic acid, and apigenin have been reported to modulate macrophage inflammatory responses via multiple pathways, with their mechanisms extensively studied [52,53,54]. In an arthritis mouse model, mutant P. frutescens leaf extract (SFE-M) significantly reduced the neutrophil-to-lymphocyte ratio (NLR) in whole blood and markedly diminished inflammatory cell infiltration and edema formation [55].

3.3. Antiviral

P. frutescens harbors a diverse array of antiviral compounds, including caffeic acid, rosmarinic acid, luteolin, ferulic acid, apigenin-7-O-diglucuronide, perilla alcohol, menthol, perillaldehyde, baicalin, eugenol, safrole, catechaldehyde, methyl caffeate, and cinnamaldehyde [56]. These compounds exert potent inhibitory effects on the replication and dissemination of various viruses.
P. frutescens leaf extract prevents viral entry into host cells by inactivating viral particles, exhibiting an EC50 of 0.12 ± 0.06 mg/mL and a CC50 (drug concentration causing 50% cytotoxicity as determined by the MTT assay) of 4.64 ± 0.16 mg/mL against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [57]. Luteolin regulates the NF-κB/STAT3/ATF6 signaling pathway, effectively suppressing African swine fever virus (ASFV) replication [58]. It also exhibits strong antiviral activity against the largemouth bass virus (LMBV), with a viral replication inhibition rate of 97.27%. Its half-maximal inhibitory concentration (IC50) in epithelioma papulosum cyprini (EPC) cells is 2.77 μM, highlighting its potential in aquatic viral disease treatment [59].
Perilla alcohol and perilla acid do not affect viral genomic replication but act in the later stages of the herpes simplex virus type 1 (HSV-1) lifecycle, inhibiting the release of infectious viral particles from Vero cells [60]. Apigenin does not influence the viral lifecycle (e.g., attachment, entry, or budding) but directly inhibits viral polymerase activity [24]. These findings highlight the potential of P. frutescens as a natural plant with antiviral properties and provide important insights for the development of novel antiviral drugs.

3.4. Anticancer

The anticancer properties of P. frutescens primarily arise from its bioactive constituents, including perillaldehyde and rosmarinic acid, which modulate inflammatory signaling pathways (e.g., NF-κB) and reduce oxidative stress [61]. Cancer metastasis remains a significant challenge in oncology. Studies have demonstrated that P. frutescens (stems, leaves, and seeds) extracts (rosmarinic acid, perillaldehyde) suppress adrenaline-induced cancer cell metastasis [62] and exert significant cytotoxic effects on prostate cancer cell lines [63]. Rosmarinic acid exhibits anticancer effects through two primary mechanisms: inhibition of inflammatory responses and scavenging of ROS [64]. The anticancer mechanisms of luteolin, a key flavonoid, have been comprehensively reviewed elsewhere [65].
Notably, nanotechnology-based formulations, such as P. frutescens flavonoid-stabilized silver nanoparticles (PFFE-AgNPs) and rosmarinic acid-functionalized silver nanoparticles (PFRAE-AgNPs), exhibit strong anticancer effects against multiple human cancer cell lines, including COLO205 (colon adenocarcinoma), PC-3 (prostate cancer), A549 (lung adenocarcinoma), and SKOV3 (ovarian cancer) [66,67]. Furthermore, recent studies indicate that probiotic strains P. pentosaceus (F1), L. fermentum (F24), and P. acidilactici (F23) enhance the accumulation of phenolic compounds in P. frutescens, thereby amplifying its anticancer effects through phytochemical synergy [9].

3.5. Antibacterial

P. frutescens possesses broad-spectrum antimicrobial properties, effectively inhibiting various bacterial pathogens, including Escherichia coli, Staphylococcus aureus, Streptococcus pneumoniae, Shigella spp., and Salmonella spp. [68]. Additionally, PEO exhibits antifungal activity against several plant pathogenic fungi, such as Aspergillus flavus, Aspergillus niger, Aspergillus oryzae, Fusarium solani, and Rhizoctonia solani [69]. Proteomic analysis reveals that P. frutescens oil inhibits the growth of A. flavus by impairing antioxidant defense mechanisms and disrupting glycolytic pathways. This effect is supported by the marked upregulation of ribosomal proteins (Kri1, Noc4, Rlp24, Ytm1, Mak21, Ssf2, and Tsr3), along with enhanced enzymatic activity linked to RNA transcription and translation in A. flavus following P. frutescens oil treatment [70].
P. frutescens contains bioactive compounds, including rosmarinic acid, caffeic acid, perillaldehyde, luteolin, and other phenolic acids, which demonstrate strong antibacterial and antifungal activities [1,35]. Rosmarinic acid interferes with the synthesis and degradation pathways of symbionts in E. coli and S. aureus, disrupts the tricarboxylic acid (TCA) cycle, and induces excessive ketone body accumulation, ultimately triggering bacterial apoptosis [71]. P. frutescens aldehyde induces necrosis and apoptosis in Botrytis cinerea and Clostridium perfringens spores. The underlying mechanisms involve MAPK signaling pathway disruption and compromised cell wall integrity in B. cinerea, autophagy induction through Ca2+- and ROS-mediated pathways, and interference with ribosomal function during transcription and translation, ultimately inhibiting biofilm formation in Streptococcus mutans [15,72,73].
Metabolomic analyses have demonstrated the antibacterial properties of P. frutescens against Pseudomonas fluorescens and elucidated its underlying mechanisms. P. frutescens induces substantial alterations in 128 metabolites, perturbing nucleotide metabolism, disrupting carbohydrate, amino acid, and lipid metabolism, and impairing cell wall biosynthesis, cell membrane integrity, and genetic material expression. These metabolic disruptions collectively suppress bacterial growth and ultimately result in cell death [74].

3.6. Lowers Blood Sugar

Recent pharmacological studies have underscored the hypoglycemic potential of P. frutescens. These effects are mainly attributed to bioactive compounds, including phenolic acids, flavonoids, terpenoids, and organic acids, which form the phytochemical basis of its antidiabetic activity [75]. Experimental studies suggest that P. frutescens oil confers dual therapeutic benefits in type 2 diabetic mouse models by mitigating insulin resistance and modulating gut microbiota through the PI3K/AKT signaling pathway [76].
Animal studies have shown that perillaldehyde, a key monoterpene component, attenuates oxidative stress in diabetic cardiomyopathy by inhibiting the PARP1-TRPM2-CaMKII/CaN pathway [77]. Furthermore, P. frutescens sprout extract (PFSE) exhibits significant metabolic regulatory effects through two primary mechanisms: suppression of gluconeogenic signaling pathways and activation of phosphorylated AMP-activated protein kinase (AMPK), thereby ameliorating symptoms in type 2 diabetic mice [78,79].
Luteolin supplementation (10 mg/kg/day for 4 weeks) in streptozotocin-induced diabetic mice demonstrated nephroprotective effects by suppressing the RIP140/NF-κB pathway while simultaneously enhancing insulin signaling in renal tissues [80]. Structural modification studies have yielded valuable insights into the bioactivity of polysaccharides. Sulfation markedly enhanced the α-glucosidase and α-amylase inhibitory activities of P. frutescens polysaccharide PLP-2-1, increasing inhibition rates from 71.77% to 76.06% and from 33.15% to 47.73%, respectively, resulting in the sulfated derivative S-PLP-2-1 [81].

3.7. Others

P. frutescens demonstrates a wide range of pharmacological activities, including antidepressant, anti-Alzheimer’s, sleep-promoting, anti-asthmatic, and fertility-enhancing properties. The functional mechanism is shown in Table 2. Luteolin-7-O-glucuronide (L7GN), a key bioactive flavonoid, alleviated stress-induced depression-like behaviors and adaptive responses in a murine sleep deprivation (SD) model [82]. The extracts of the plant show promising dermatological potential: rosmarinic acid, a principal phenolic component, promotes hair growth through time-dependent enhancement of cellular viability while inhibiting testosterone and dihydrotestosterone (DHT) activity, making it a strong candidate for the treatment and prevention of androgenetic alopecia (AGA) [83].
Perillaldehyde exerts neuroprotective effects in dopaminergic neurons, primarily by modulating G3BP-mediated stress granule (SG) formation, indicating its therapeutic potential for neurodegenerative diseases [84]. Furthermore, perillaldehyde possesses ideal physicochemical properties for transdermal drug delivery, acting as a novel permeation enhancer by enhancing lipid bilayer fluidity and reducing diffusion barriers in the stratum corneum [85]. Isoimperatorin accelerated cutaneous wound healing in dermatological repair by activating the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway [86]. Agricultural applications were further demonstrated by Streptomyces sp. NEAU-ZSY13, an endophytic strain isolated from P. frutescens leaves, exhibiting broad-spectrum biocontrol activity against Ralstonia solanacearum (tomato bacterial wilt) and Bipolaris sorokiniana (wheat root rot), with control efficacy of 72.2% and 78.3%, respectively [87].
Table 2. Models and mechanisms of functions of Perilla frutescens extract.
Table 2. Models and mechanisms of functions of Perilla frutescens extract.
FunctionCell/Animal ModelMechanism of ActionReferences
Antioxidantt-BHP-induced hepatotoxicity in ratsModulates CYP1A1/2 activity and heme oxygenase-1 (HO-1) expression via nuclear factor erythroid 2-related factor 2 (NRF2) activation, mitigating oxidative liver damage.Kang et al. [88]
Anti-inflammatoryCOPD mouse modelInhibits leukocytosis and neutrophilia in BALF; suppresses p38 MAPK/NF-κB p65 signaling, reducing inflammatory mediator production and neutrophil infiltration.Wei et al. [89]
AntiviralPK15 cells and PRV-infected miceLuteolin inhibits viral replication by downregulating viral mRNA/gB protein expression, reduces apoptosis in PRV-infected cells, and enhances survival rates post-lethal challenge.Men et al. [90]
AnticancerHuh-7, Hep3B cells, and xenograft miceIsoegomaketone suppresses hepatocellular carcinoma (HCC) growth via PI3K-Akt signaling pathway blockade.Wang et al. [91]
AntimicrobialTrichophyton mentagrophytesInhibits enolase expression, disrupting fungal glycolysis and energy metabolism, thereby suppressing growth.Xu et al. [35]
HypoglycemicHFD/STZ-induced T2DM SD ratsReduces hyperglycemia, ameliorates hepatic/intestinal tissue damage, and decreases glycogen accumulation via enhanced insulin signaling.Wang et al. [75]
Anti-Alzheimer’s5XFAD transgenic miceBlocks Aβ aggregation, dissociates preformed Aβ fibrils, and prevents Aβ-induced LTP impairment and memory deficits.Cho et al. [92]
AntidepressantCUMS-induced depressed ratsModulates monoaminergic neurotransmission and activates BDNF/TrkB signaling pathways.Zhong et al. [93]
Sleep PromotionPentobarbital-induced sleep miceExhibits adenosine A1 receptor (A1R) agonism, enhances neuronal activity in sleep-promoting brain regions, and reduces activity in wakefulness-associated regions.Joy et al. [94]
Anti-asthmaticOVA-induced allergic asthma miceSuppresses airway inflammation and immune dysregulation via inhibition of ERK, JNK, and p38 MAPK phosphorylation.Cao et al. [95]
HypolipidemicHyperlipidemic ratsReduces serum lipid levels, inhibits lipid peroxidation, normalizes lipoprotein metabolism, and enhances antioxidant enzyme activity.Feng et al. [96]
Fertility EnhancementHuman endometrial Ishikawa cellsUpregulates integrin β3/β5 expression via leukemia inhibitory factor (LIF)-dependent pathways, enhancing adhesion between endometrial and trophoblast cells.Kim et al. [97]

4. Innovation in Food and Industrial Applications

4.1. Development of Functional Foods

P. frutescens oil is increasingly recognized as a multifunctional edible oil with broad industrial applications, owing to its natural composition and bioactive properties [11,98]. Its integration into margarine formulations underscores its versatility in lipid-based food products [99]. Nutritional analysis indicates that P. frutescens seed oil is particularly abundant in ω-3 polyunsaturated fatty acids [100], which mitigate intracellular lipid accumulation by markedly reducing serum levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) [101]. Spray-dried microencapsulation effectively stabilizes α-linolenic acid (ALA) in P. frutescens oil, enhancing its shelf life while maintaining its nutritional integrity. The anti-atherogenic properties of P. frutescens leaves have been evidenced by their ability to suppress lipid accumulation in arterial tissues [96]. Anti-obesity effects observed in 3T3-L1 adipocytes and rodent models highlight P. frutescens as a promising functional food for combating obesity and metabolic disorders [102]. Synergistic formulations, such as P. frutescens-chrysanthemum functional beverages, display enhanced antioxidant properties due to the synergistic interactions between polyphenols, flavonoids, and anthocyanins [103]. P. frutescens seed cake hydrolysate, particularly its rosmarinic acid-enriched fraction, significantly enhanced muscle mass and exercise endurance in mice, indicating its potential applications in sports nutrition and the development of meat substitutes [104]. The antioxidant, anticancer, anti-inflammatory, and antidiabetic properties of P. frutescens seed proteins (peptides) support their potential in the development of functional foods [105]. From an industrial perspective, the primary utilization of P. frutescens focuses on its extracts. These include standardized extracts, such as volatile oil-rich fractions for flavor enhancement, ethanolic extracts with antimicrobial properties suitable for food preservation, and other bioactive compound-rich extracts with applications in functional foods and nutraceuticals. The extracts or products derived from Perilla frutescens are suitable for large-scale production, comply with the legal and regulatory requirements for food products, and can maintain their quality over time (i.e., have a stable shelf life) when used in the food industry.

4.2. Natural Preservatives and Preservatives

PEO and its phenolic acid constituents demonstrate broad-spectrum antimicrobial activity. Among these, perillaldehyde demonstrates significant antibacterial activity, particularly against foodborne pathogens such as E. coli, positioning it as a promising natural preservative [106]. Perillaldehyde suppresses the growth of Botrytis cinerea, disrupts Streptococcus biofilm formation, and contributes to the preservation of seafood and fresh fruits [15,73]. A “sandwich” nanofiber membrane embedded with perillaldehyde extended the shelf life of refrigerated chicken by 6–10 days via sustained antimicrobial release [107].
PEO-loaded hydrogel beads effectively retain organic acids and total phenolics in strawberries, significantly reducing microbial counts and enhancing sensory attributes, particularly taste and color [108]. PEO microcapsules, fabricated with a core-to-shell ratio of 1.4:1, exhibit excellent encapsulation efficiency (91.5%) and a smoother surface morphology. These microcapsules help preserve high antioxidant and antibacterial activity during peach storage [109]. Nanoemulsified PEO prolonged the shelf life of frozen beef by 14 days by inhibiting lipid oxidation [110].
P. frutescens leaf extract effectively mitigates lipid and protein oxidation, reduces total volatile basic nitrogen (TVB-N) levels in surimi fish balls, and inhibits microbial growth in surimi-based products [111]. Composite edible films were developed by integrating PEO-glycerol monolaurate emulsions with chitosan and nisin. The formulated film exhibited an exceptional inhibition rate of 99.94% against Staphylococcus aureus, underscoring its potential as an antimicrobial packaging material for seafood preservation [112]. These studies provide valuable insights into the application of natural preservatives in the food industry and promote sustainable practices in food preservation.

4.3. Novel Food Additives

P. frutescens seed powder functions as a dual-purpose fat substitute in processed meats, reducing heterocyclic amine formation and mitigating protein oxidation in heat-treated chicken products [113]. Enzymatic hydrolysis of P. frutescens seed powder yields water-soluble dietary fiber with enhanced hydration properties, rendering it an ideal functional carbohydrate source for food additives and nutraceuticals [114]. In ruminant nutrition, a diet supplemented with 3% P. frutescens seed significantly enhanced growth performance and carcass quality in Tan lambs, while improving raw meat flavor profiles by increasing umami nucleotide content [115]. P. frutescens leaf supplementation activated pyrimidine metabolism pathways in rumen fluid and stimulated unsaturated fatty acid biosynthesis in milk, thereby enhancing flavor compound production [116].
The incorporation of PEO significantly enhanced the flavor profile of roasted Wuchang fish, increasing key volatile compounds such as α-terpineol, linalool, camphene, and β-pinene. This treatment markedly enhanced umami intensity, as evidenced by a guanosine monophosphate (GMP) content of 87.75 mg/g, which contributed to improved sensory attributes and textural properties [117]. Anthocyanins in P. frutescens leaves function as pH-responsive natural pigments, shifting between red, purple, and blue, thereby making them suitable for coloring beverages and confectioneries [118]. Co-pigmentation with rosmarinic acid stabilized anthocyanins and enhanced food coloration [119].
Fermentation of P. frutescens leaf extract (5%, 10%, 15%, and 20% w/v) using Lactobacillus acidophilus KCTC 3164 demonstrated that the melting rate, expansion ratio, DPPH radical scavenging activity, and total phenolic content in ice cream increased with higher amounts of P. frutescens leaf extract, whereas viscosity and pH showed an inverse correlation. Among the different concentrations, the 10% P. frutescens leaf fermented extract stood out as a novel additive for functional ice cream production [120]. Milk-based gummies were formulated using a 65:10 ratio of sweetened condensed milk and whey permeate, incorporating 18% P. frutescens syrup and 4% gelatin. These formulations exhibited high nutritional and sensory value, supporting their potential as novel functional food products [121].

5. Biology and Genetic Improvement

Advancements in genomics and metabolic engineering have positioned P. frutescens as a key candidate for genetic improvement and metabolic optimization. CRISPR-Cas9-mediated genome editing has been effectively employed to enhance agronomic traits, including biomass production and stress tolerance [122]. Key genes involved in leaf pigmentation, seed development, and trichome formation have been identified through comparative genomics, providing molecular tools for precision breeding [123,124].
The exceptionally high α-linolenic acid (ALA) content in P. frutescens seeds (up to 60.9% of total fatty acids) underscores the importance of elucidating its biosynthetic regulation, particularly for metabolic engineering applications [125]. Transcriptomic analysis has identified key enzyme transcripts associated with fatty acid and triacylglycerol (TAG) biosynthesis, laying a foundation for genetic and evolutionary research in P. frutescens, including inter-varietal comparisons [126]. Metabolomic analysis has further characterized distinct chemotypes in P. frutescens, categorizing them into monoterpenoid (MT) and phenylpropanoid (PP) types. The MT chemotypes include high phenolic acid (PA), perilla ketone (PK), high essential oil (EK), umbelliferone (PT), perilla alcohol (PL), shisofuran (SF), and pleurisy (C) types (Figure 2B,C), providing critical support for molecular marker-assisted breeding [127,128]. Moreover, multi-omics approaches have elucidated the diverse functions and potential applications of P. frutescens’s organs and tissues, providing theoretical insights for the targeted utilization of organ-specific metabolites [129].

6. Security and Standardization

6.1. Toxicity and Security

Comprehensive safety evaluations are essential before the application of P. frutescens derivatives in the food and pharmaceutical industries. The Chinese-Pharmacopoeia-Commission 2020 stipulates inductively coupled plasma mass spectrometry (ICP-MS) for quantifying heavy metals in P. frutescens leaves, including cadmium (Cd), mercury (Hg), lead (Pb), arsenic (As), and copper (Cu) [130]. Pesticide residue analysis employs chromatographic methods to detect compounds such as hexachlorobenzene, quintozene, cis-chlordane, trans-chlordane, oxy-chlordane, heptachlor, heptachlor-exo-epoxide, and heptachlor-endo-epoxide. Perilla ketone exhibits a dual pharmacological profile in research, demonstrating anti-inflammatory and antimicrobial activities alongside potent pulmonary toxicity. Based on a presumed average body weight of 63 kg, the estimated safe intake of P. frutescens leaves is ≤5 g·d−1 to prevent perilla ketone-induced toxicity [131].
While both perilla alcohol and perillaldehyde hold Generally Recognized As Safe (GRAS) designation, perillaldehyde continues to face persistent scrutiny in scientific debates over its potential health risks. Perillaldehyde has an LC50 (the 50% lethal concentration) of 7.975 mg/L in zebrafish, while sublethal exposure at 4 mg/L induces morphological abnormalities and neurotoxicity [132]. Perillaldehyde demonstrates significant cytotoxicity in vitro and hepatotoxicity in vivo, necessitating precise dose regulation to mitigate these adverse effects. Despite its antimicrobial, anticancer, and anti-inflammatory properties, strict dosage control is imperative, as only low-to-moderate doses are deemed safe for human use to minimize risks of hepatotoxicity and neurodevelopmental disorders [133]. Furthermore, P. frutescens seeds present allergenic risks, with occupational asthma reported in individuals exposed to roasted seed fumes [134]. Oleosins have been identified as predominant allergenic proteins within the seed matrix [135].
Toxicological studies have determined a no-observed-adverse-effect level (NOAEL) of P. frutescens oil at 3 g/kg/day in beagle dogs; however, histopathological analyses indicate potential hepatolienal toxicity, necessitating further investigation [136]. Additionally, the risk of adulteration must be carefully monitored, as atmospheric solids analysis probe-mass spectrometry (ASAP-MS) can detect adulterants such as cinnamon oil at concentrations as low as 5% (v/v) with 93% accuracy [137].

6.2. Standardization

International regulations regarding pesticide residues in P. frutescens products vary considerably. The European Union (EU) implements a limit of detection-based standard, whereas China’s P. frutescens industry standard enforces a maximum residue limit of 0.05 mg/kg. Japan enforces a more stringent threshold of 0.01 mg/kg. Additionally, the regional standard of Inner Mongolia (DB15/T 3207-2024) mandates heavy metal limits for P. frutescens leaves, specifying maximum permissible levels of lead (≤5.0 mg/kg) and cadmium (≤0.3 mg/kg) [138]. Compliance with these standards is validated using atomic absorption spectroscopy, ensuring a recovery rate of ≥95%.

7. Conclusions

P. frutescens, a traditional medicinal and edible plant, has garnered significant research attention in recent years. It is abundant in essential nutrients and contains volatile oils, flavonoids, phenolic acids, and terpenes, which exhibit notable anti-inflammatory, anticancer, antioxidant, and antibacterial properties. Furthermore, P. frutescens exhibits significant potential in functional foods and natural food additives, particularly as a source for developing natural pigments, flavoring agents, and preservatives. Notably, P. frutescens oil, distinguished by its high omega-3 fatty acid content, is recognized as a valuable functional food ingredient. This review systematically examines the chemical composition and bioactive properties of P. frutescens, outlining a comprehensive research framework that encompasses resource utilization, mechanistic insights, technological advancements, and regulatory considerations, thereby supporting its high-value applications in functional foods and sustainable industries.
Although the pharmacological properties of P. frutescens bioactive compounds have been extensively investigated, their safety evaluations remain largely confined to preclinical studies, with clinical research still lacking. Additionally, the structural diversity and complexity of P. frutescens bioactive constituents, particularly polysaccharides, pose significant challenges for in-depth investigation. While the primary bioactive components of P. frutescens have been largely elucidated, their structural characteristics, pharmacological mechanisms, and metabolic pathways require further investigation. Future research should prioritize comprehensive safety evaluations and clinical studies of P. frutescens bioactive compounds, while utilizing advanced analytical techniques to elucidate their intricate chemical structures. These efforts will facilitate the high-value utilization of P. frutescens in pharmaceuticals, functional foods, and sustainable industrial applications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/foods14071252/s1, Table S1: Publications on Perilla frutescens from 2021 to 2025.

Author Contributions

Writing—original draft, D.Y., M.P. and Z.W.; writing—review and editing, M.P. and Z.W. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financially supported by the Natural Science Foundation of Hubei Province (2023AFB484), the Enshi Science and Technology Plan Project (Young Talent Project) (No. D20220071), the Open Research Fund of Hubei Engineering Research Center of selenium food nutrition and health intelligent technology (Hubei Minzu University) (No. PT082301), and the Open Fund of Hubei Key Laboratory of Biological Resources Protection and Utilization (Hubei Minzu University) (KYPT012305).

Institutional Review Board Statement

Not applicable.

Informed Consent 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.

References

  1. Yu, H.; Qiu, J.F.; Ma, L.J.; Hu, Y.J.; Li, P.; Wan, J.B. Phytochemical and phytopharmacological review of Perilla frutescens L. (Labiatae), a traditional edible-medicinal herb in China. Food Chem. Toxicol. 2017, 108, 375–391. [Google Scholar] [CrossRef] [PubMed]
  2. Dossou, S.S.K.; Deng, Q.; Li, F.; Wang, L.; Jiang, N.; Kefale, H.; Zhou, R.; Li, D.; Tan, M.; Wang, L. Metabolic profiling of Perilla leaves of different colors: Insights into metabolite variation and bioactive compound distribution. BMC Plant Biol. 2025, 25, 38. [Google Scholar] [CrossRef]
  3. Saini, R.K.; Keum, Y.-S.; Rengasamy, K.R.R. Profiling of nutritionally important metabolites in green/red and green perilla (Perilla frutescens Britt.) cultivars: A comparative study. Ind. Crops Prod. 2020, 151, 112441. [Google Scholar] [CrossRef]
  4. Ahmed, H.M. Ethnomedicinal, phytochemical and pharmacological investigations of Perilla frutescens (L.) Britt. Molecules 2018, 24, 102. [Google Scholar] [CrossRef]
  5. Akatsuka, R.; Ito, M. Content and distribution of prunasin in Perilla frutescens. J. Nat. Med. 2023, 77, 207–218. [Google Scholar] [PubMed]
  6. Wu, X.; Dong, S.; Chen, H.; Guo, M.; Sun, Z.; Luo, H. Perilla frutescens: A traditional medicine and food homologous plant. Chin. Herb. Med. 2023, 15, 369–375. [Google Scholar] [CrossRef]
  7. Rouphael, Y.; Kyriacou, M.C.; Carillo, P.; Pizzolongo, F.; Romano, R.; Sifola, M.I. Chemical Eustress Elicits Tailored Responses and Enhances the Functional Quality of Novel Food Perilla frutescens. Molecules 2019, 24, 185. [Google Scholar] [CrossRef]
  8. Chen, F.; Liu, S.; Zhao, Z.; Gao, W.; Ma, Y.; Wang, X.; Yan, S.; Luo, D. Ultrasound pre-treatment combined with microwave-assisted hydrodistillation of essential oils from Perilla frutescens (L.) Britt. leaves and its chemical composition and biological activity. Ind. Crops Prod. 2020, 143, 111908. [Google Scholar] [CrossRef]
  9. Vijayalakshmi, S.; Yoo, D.-S.; Kim, D.-G.; Chelliah, R.; Barathikannan, K.; Aloo, S.-O.; Tyagi, A.; Yan, P.; Shan, L.; Gebre, T.S.; et al. Fermented Perilla frutescens leaves and their untargeted metabolomics by UHPLC-QTOF-MS reveal anticancer and immunomodulatory effects. Food Biosci. 2023, 56, 103065. [Google Scholar] [CrossRef]
  10. Lee, J.H.; Cho, Y.-S. Assessment of phenolic profiles from various organs in different species of perilla plant (Perilla frutescens (L.) Britt.) and their antioxidant and enzyme inhibitory potential. Ind. Crops Prod. 2021, 171, 113914. [Google Scholar] [CrossRef]
  11. Kaur, S.; Seem, K.; Ali, A.; Jaiswal, S.; Gumachanamardi, P.; Kaur, G.; Singh, N.; Touthang, L.; Singh, S.K.; Bhardwaj, R.; et al. A comprehensive review on nutritional, nutraceutical, and industrial perspectives of perilla (Perilla frutscens L.) seeds—An orphan oilseed crop. Heliyon 2024, 10, e33281. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, M.; Luo, F.; Qing, Z.; Yang, H.; Liu, X.; Yang, Z.; Zeng, J. Chemical Composition and Bioactivity of Essential Oil of Ten Labiatae Species. Molecules 2020, 25, 4862. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, Y.; Huang, J.; Lin, X.; Su, W.; Zhu, P.; Yang, N.; Adams, E. Recent progress in the extraction of terpenoids from essential oils and separation of the enantiomers by GC-MS. J. Chromatogr. A 2024, 1730, 465118. [Google Scholar] [CrossRef]
  14. Ahmed, H.M.; Tavaszi-Sarosi, S. Identification and quantification of essential oil content and composition, total polyphenols and antioxidant capacity of Perilla frutescens (L.) Britt. Food Chem. 2019, 275, 730–738. [Google Scholar] [CrossRef]
  15. Kang, C.; Zhang, H.; Sun, C.; Cao, J.; Yang, H.; Chen, J.; Wang, Y.; Sun, C. The antifungal activity and mechanism of Perillaldehyde and its stabilized encapsulation technology for fruit preservation. Postharvest Biol. Technol. 2024, 207, 112613. [Google Scholar] [CrossRef]
  16. Tashakor, A.H.; Rezaei, A.; Fouladseresht, H.; Mansury, D. Characterization and investigation of cytotoxicity and antimicrobial properties of coencapsulated limonene and thymol into the Ferula assafoetida gum microparticles. Int. J. Biol. Macromol. 2024, 263, 130338. [Google Scholar] [CrossRef]
  17. Luo, W.; Wang, K.; Luo, J.; Liu, Y.; Tong, J.; Qi, M.; Jiang, Y.; Wang, Y.; Ma, Z.; Feng, J.; et al. Limonene anti-TMV activity and its mode of action. Pestic. Biochem. Physiol. 2023, 194, 105512. [Google Scholar] [CrossRef]
  18. Ghimire, B.K.; Yoo, J.H.; Yu, C.Y.; Chung, I.M. GC-MS analysis of volatile compounds of Perilla frutescens Britton var. Japonica accessions: Morphological and seasonal variability. Asian Pac. J. Trop. Med. 2017, 10, 643–651. [Google Scholar] [CrossRef] [PubMed]
  19. Li, Y.; Yang, X.; Chen, S.; Wu, L.; Zhou, J.; Jia, K.; Ju, W. Integrated Network Pharmacology and GC-MS-Based Metabolomics to Investigate the Effect of Xiang-Su Volatile Oil Against Menopausal Depression. Front. Pharmacol. 2021, 12, 765638. [Google Scholar] [CrossRef]
  20. Tabanca, N.; Demirci, B.; Ali, A.; Ali, Z.; Blythe, E.K.; Khan, I.A. Essential oils of green and red Perilla frutescens as potential sources of compounds for mosquito management. Ind. Crops Prod. 2015, 65, 36–44. [Google Scholar] [CrossRef]
  21. You, C.X.; Yang, K.; Wu, Y.; Zhang, W.J.; Wang, Y.; Geng, Z.F.; Chen, H.P.; Jiang, H.Y.; Du, S.S.; Deng, Z.W.; et al. Chemical composition and insecticidal activities of the essential oil of Perilla frutescens (L.) Britt. aerial parts against two stored product insects. Eur. Food Res. Technol. 2014, 239, 481–490. [Google Scholar] [CrossRef]
  22. Wang, Z.; Wang, D.; Fang, J.; Song, Z.; Geng, J.; Zhao, J.; Fang, Y.; Wang, C.; Li, M. Green and efficient extraction of flavonoids from Perilla frutescens (L.) Britt. leaves based on natural deep eutectic solvents: Process optimization, component identification, and biological activity. Food Chem. 2024, 452, 139508. [Google Scholar] [PubMed]
  23. Zhao, G.; Qin, G.W.; Wang, J.; Chu, W.J.; Guo, L.H. Functional activation of monoamine transporters by luteolin and apigenin isolated from the fruit of Perilla frutescens (L.) Britt. Neurochem. Int. 2010, 56, 168–176. [Google Scholar] [CrossRef]
  24. Khandelwal, N.; Chander, Y.; Kumar, R.; Riyesh, T.; Dedar, R.K.; Kumar, M.; Gulati, B.R.; Sharma, S.; Tripathi, B.N.; Barua, S. Antiviral activity of Apigenin against buffalopox: Novel mechanistic insights and drug-resistance considerations. Antivir. Res. 2020, 181, 104870. [Google Scholar]
  25. Kang, K.A.; Piao, M.J.; Hyun, Y.J.; Zhen, A.X.; Cho, S.J.; Ahn, M.J.; Yi, J.M.; Hyun, J.W. Luteolin promotes apoptotic cell death via upregulation of Nrf2 expression by DNA demethylase and the interaction of Nrf2 with p53 in human colon cancer cells. Exp. Mol. Med. 2019, 51, 1–14. [Google Scholar] [CrossRef]
  26. Gu, M.; Pang, Z. Luteolin inhibits inflammation and M1 macrophage polarization in the treatment of Pseudomonas aeruginosa-induced acute pneumonia through suppressing EGFR/PI3K/AKT/NF-κB and EGFR/ERK/AP-1 signaling pathways. Phytomedicine 2025, 141, 156663. [Google Scholar] [CrossRef]
  27. Li, H.; Lin, J.; Bai, B.; Bo, T.; He, Y.; Fan, S.; Zhang, J. Study on Purification, Identification and Antioxidant of Flavonoids Extracted from Perilla leaves. Molecules 2023, 28, 7273. [Google Scholar] [CrossRef] [PubMed]
  28. Paek, J.H.; Shin, K.H.; Kang, Y.H.; Lee, J.Y.; Lim, S.S. Rapid identification of aldose reductase inhibitory compounds from Perilla frutescens. Biomed. Res. Int. 2013, 2013, 679463. [Google Scholar] [CrossRef]
  29. Zhou, X.J.; Yan, L.L.; Yin, P.P.; Shi, L.L.; Zhang, J.H.; Liu, Y.J.; Ma, C. Structural characterisation and antioxidant activity evaluation of phenolic compounds from cold-pressed Perilla frutescens var. arguta seed flour. Food Chem. 2014, 164, 150–157. [Google Scholar] [CrossRef]
  30. Kim, M.-Y.; Kim, J.-I.; Kim, S.-W.; Kim, S.; Oh, E.; Lee, J.; Lee, E.; An, Y.-J.; Han, C.-Y.; Lee, H. Influence of Secondary Metabolites According to Maturation of Perilla (Perilla frutescens) on Respiratory Protective Effect in Fine Particulate Matter (PM2.5)-Induced Human Nasal Cell. Int. J. Mol. Sci. 2024, 25, 12119. [Google Scholar] [CrossRef]
  31. Wang, M.; Zhan, P.; Geng, J.; Wang, P.; He, W.; Tian, H. Identification and inhibition of key off-odorants in boiled greenfin horse-faced filefish (Thamnaconus septentrionalis) subjected to perilla polyphenols extract. LWT 2024, 199, 116120. [Google Scholar] [CrossRef]
  32. Yang, S.Y.; Hong, C.O.; Lee, G.P.; Kim, C.T.; Lee, K.W. The hepatoprotection of caffeic acid and rosmarinic acid, major compounds of Perilla frutescens, against t-BHP-induced oxidative liver damage. Food Chem. Toxicol. 2013, 55, 92–99. [Google Scholar] [CrossRef] [PubMed]
  33. Kwon, S.H.; Wang, Z.; Hwang, S.H.; Kang, Y.-H.; Lee, J.-Y.; Lim, S.S. Comprehensive evaluation of the antioxidant capacity of Perilla frutescens leaves extract and isolation of free radical scavengers using step-wise HSCCC guided by DPPH-HPLC. Int. J. Food Prop. 2017, 20, 921–934. [Google Scholar] [CrossRef]
  34. Li, W.; Fan, Y.; Zhang, S.; Li, J.; Zhang, L.; Wu, H. Extraction of rosmarinic acid from Perilla seeds using green protic ionic liquids. Microchem. J. 2021, 170, 106667. [Google Scholar] [CrossRef]
  35. Xu, Y.D.; Guo, Y.J.; Mao, H.R.; Xiong, Z.X.; Luo, M.Y.; Luo, R.Q.; Lu, S.; Huang, L.; Hong, Y. Integration of transcriptomics and proteomics to elucidate inhibitory effect and mechanism of rosmarinic acid from Perilla frutescens (L.) Britt. in treating Trichophyton mentagrophytes. Chin. Med. 2023, 18, 67. [Google Scholar] [CrossRef] [PubMed]
  36. Xie, G.; Zou, X.; Liang, Z.; Wu, D.; He, J.; Xie, K.; Jin, H.; Wang, H.; Shen, Q. Integrated metabolomic and transcriptomic analyses reveal molecular response of anthocyanins biosynthesis in perilla to light intensity. Front. Plant Sci. 2022, 13, 976449. [Google Scholar] [CrossRef]
  37. Nishimura, T.; Ohyama, K.; Goto, E.; Inagaki, N. Concentrations of perillaldehyde, limonene, and anthocyanin of Perilla plants as affected by light quality under controlled environments. Sci. Hortic. 2009, 122, 134–137. [Google Scholar] [CrossRef]
  38. Bharathy, P.; Thanikachalam, P.V. Pharmacological relevance of anthocyanin derivative: A review. Pharmacol. Res. Mod. Chin. Med. 2025, 14, 100565. [Google Scholar] [CrossRef]
  39. Yoo, E.; Kim, E.-G.; Lee, J.-E.; Lee, S.; Lee, D.; Lee, G.-A. Genetic Variations in FAD3 and Its Influence on Agronomic Traits and Fatty Acid Composition in Perilla Germplasm. Plant Sci. 2025, 355, 112452. [Google Scholar]
  40. Ding, S.; Yan, Z.; Liu, H.; Chen, P.; Shi, S.; Chang, M. Structural characterization and antitumor activity of a polysaccharide extracted from Perilla frutescens var. frutescens. Ind. Crops Prod. 2022, 187, 115334. [Google Scholar] [CrossRef]
  41. Ahmed, H.M.; Mohan Al-Zubaidy, A.; Othman-Qadir, G. Biological investigations on macro-morphological characteristics, polyphenolic acids, antioxidant activity of Perilla frutescens (L.) Britt. grown under open field. Saudi J. Biol. Sci. 2022, 29, 3213–3222. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, Y.H.; Kim, B.; Kim, S.; Kim, M.S.; Kim, H.; Hwang, S.R.; Kim, K.; Lee, J.H. Characterization of metabolite profiles from the leaves of green perilla (Perilla frutescens) by ultra high performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry and screening for their antioxidant properties. J. Food Drug Anal. 2017, 25, 776–788. [Google Scholar] [CrossRef] [PubMed]
  43. Jun, H.I.; Kim, B.T.; Song, G.S.; Kim, Y.S. Structural characterization of phenolic antioxidants from purple perilla (Perilla frutescens var. acuta) leaves. Food Chem. 2014, 148, 367–372. [Google Scholar] [CrossRef]
  44. Choi, H.J.; Song, B.R.; Kim, J.E.; Bae, S.J.; Choi, Y.J.; Lee, S.J.; Gong, J.E.; Lee, H.S.; Lee, C.Y.; Kim, B.H.; et al. Therapeutic Effects of Cold-Pressed Perilla Oil Mainly Consisting of Linolenic acid, Oleic Acid and Linoleic Acid on UV-Induced Photoaging in NHDF Cells and SKH-1 Hairless Mice. Molecules 2020, 25, 989. [Google Scholar] [CrossRef]
  45. Fuyuno, Y.; Uchi, H.; Yasumatsu, M.; Morino-Koga, S.; Tanaka, Y.; Mitoma, C.; Furue, M. Perillaldehyde Inhibits AHR Signaling and Activates NRF2 Antioxidant Pathway in Human Keratinocytes. Oxid. Med. Cell Longev. 2018, 2018, 9524657. [Google Scholar] [CrossRef] [PubMed]
  46. Park, H.-Y.; Nam, M.-H.; Lee, H.-S.; Jun, W.; Hendrich, S.; Lee, K.-W. Isolation of caffeic acid from Perilla frutescens and its role in enhancing γ-glutamylcysteine synthetase activity and glutathione level. Food Chem. 2010, 119, 724–730. [Google Scholar] [CrossRef]
  47. Zhao, G.; Yao-Yue, C.; Qin, G.W.; Guo, L.H. Luteolin from Purple Perilla mitigates ROS insult particularly in primary neurons. Neurobiol. Aging 2012, 33, 176–186. [Google Scholar] [CrossRef]
  48. Lee, Y.; Lee, J.; Ju, J. Perilla frutescens Britton var. frutescens leaves attenuate dextran sulfate sodium-induced acute colitis in mice and lipopolysaccharide-stimulated angiogenic processes in human umbilical vein endothelial cells. Food Sci. Biotechnol. 2020, 29, 131–140. [Google Scholar] [CrossRef]
  49. Park, D.D.; Yum, H.W.; Zhong, X.; Kim, S.H.; Kim, S.H.; Kim, D.H.; Kim, S.J.; Na, H.K.; Sato, A.; Miura, T.; et al. Perilla frutescens Extracts Protects against Dextran Sulfate Sodium-Induced Murine Colitis: NF-kappaB, STAT3, and Nrf2 as Putative Targets. Front. Pharmacol. 2017, 8, 482. [Google Scholar] [CrossRef]
  50. Chen, C.Y.; Leu, Y.L.; Fang, Y.; Lin, C.F.; Kuo, L.M.; Sung, W.C.; Tsai, Y.F.; Chung, P.J.; Lee, M.C.; Kuo, Y.T.; et al. Anti-inflammatory effects of Perilla frutescens in activated human neutrophils through two independent pathways: Src family kinases and Calcium. Sci. Rep. 2015, 5, 18204. [Google Scholar] [CrossRef]
  51. Paradee, N.; Koonyosying, P.; Kusirisin, W.; Janthip, R.; Kanjanapothi, D.; Pattanapanyasat, K.; Srichairatanakool, S. Analgesic, anti-inflammatory and anti-ulcer properties of Thai Perilla frutescence fruit oil in animals. Biosci. Rep. 2021, 41, BSR20203166. [Google Scholar] [CrossRef]
  52. Park, Y.D.; Lee, Y.M.; Kang, M.A.; Lee, H.J.; Jin, C.H.; Choi, D.S.; Kim, D.S.; Kang, S.-Y.; Kim, W.-G.; Jeong, I.Y. Phytochemical profiles and in vitro anti-inflammatory properties of Perilla frutescens cv. Chookyoupjaso mutants induced by mutagenesis with γ-ray. Food Sci. Biotechnol. 2010, 19, 305–311. [Google Scholar] [CrossRef]
  53. Hu, A.; Ye, Y.; Han, B.; Zhu, X.; Xue, F.; Yang, L.; Zhang, C. Effect of Perilla frutescens polyphenols against tight junction alterations and intestinal inflammation: A Caco-2/macrophage co-culture model-based study integrated with network pharmacology. Food Biosci. 2024, 62, 105459. [Google Scholar] [CrossRef]
  54. Zi, Y.; Yao, M.; Lu, Z.; Lu, F.; Bie, X.; Zhang, C.; Zhao, H. Glycoglycerolipids from the leaves of Perilla frutescens (L.) Britton (Labiatae) and their anti-inflammatory activities in lipopolysaccharide-stimulated RAW264.7 cells. Phytochemistry 2021, 184, 112679. [Google Scholar] [CrossRef]
  55. Jin, C.H.; So, Y.; Kim, H.Y.; Han, S.N.; Kim, J.B. Anti-Arthritic Activities of Supercritical Carbon Dioxide Extract Derived from Radiation Mutant Perilla frutescens Var. Crispa in Collagen Antibody-Induced Arthritis. Nutrients 2019, 11, 2959. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, J.; Zhao, Y.; Cheng, J.; Wang, H.; Pan, S.; Liu, Y. The Antiviral Potential of Perilla frutescens: Advances and Perspectives. Molecules 2024, 29, 3328. [Google Scholar] [CrossRef]
  57. Tang, W.F.; Tsai, H.P.; Chang, Y.H.; Chang, T.Y.; Hsieh, C.F.; Lin, C.Y.; Lin, G.H.; Chen, Y.L.; Jheng, J.R.; Liu, P.C.; et al. Perilla (Perilla frutescens) leaf extract inhibits SARS-CoV-2 via direct virus inactivation. Biomed. J. 2021, 44, 293–303. [Google Scholar] [CrossRef]
  58. Chen, Y.; Guo, Y.; Song, Z.; Chang, H.; Kuang, Q.; Zheng, Z.; Wang, H.; Zhang, G. Luteolin restricts ASFV replication by regulating the NF-κB/STAT3/ATF6 signaling pathway. Vet. Microbiol. 2022, 273, 109527. [Google Scholar]
  59. Wang, G.-S.; Qin, J.-C.; Qin, Y.-L.; Wang, Y.; Yang, B.; Liu, X.; Ling, F.; Wang, G.-X. Evaluation on the antiviral activity of luteolin against largemouth bass virus. Aquaculture 2025, 595, 741573. [Google Scholar] [CrossRef]
  60. Mello, C.P.; Quirico-Santos, T.; Amorim, L.F.; Silva, V.G.; Fragel, L.M.; Bloom, D.C.; Paixao, I.P. Perillyl alcohol and perillic acid exert efficient action upon HSV-1 maturation and release of infective virus. Antivir. Ther. 2020, 25, 1–11. [Google Scholar] [CrossRef]
  61. Huang, S.; Nan, Y.; Chen, G.; Ning, N.; Du, Y.; Lu, D.; Yang, Y.; Meng, F.; Yuan, L. The Role and Mechanism of Perilla frutescens in Cancer Treatment. Molecules 2023, 28, 5883. [Google Scholar] [CrossRef]
  62. Jeong, J.H.; Park, H.J.; Chi, G.Y.; Choi, Y.H.; Park, S.H. An Ethanol Extract of Perilla frutescens Leaves Suppresses Adrenergic Agonist-Induced Metastatic Ability of Cancer Cells by Inhibiting Src-Mediated EMT. Molecules 2023, 28, 3414. [Google Scholar] [CrossRef]
  63. Garcia, P.J.B.; Huang, S.K.; De Castro-Cruz, K.A.; Leron, R.B.; Tsai, P.W. An In Vitro Evaluation and Network Pharmacology Analysis of Prospective Anti-Prostate Cancer Activity from Perilla frutescens. Plants 2023, 12, 3006. [Google Scholar] [CrossRef] [PubMed]
  64. Osakabe, N.; Yasuda, A.; Natsume, M.; Yoshikawa, T. Rosmarinic acid inhibits epidermal inflammatory responses: Anticarcinogenic effect of Perilla frutescens extract in the murine two-stage skin model. Carcinogenesis 2004, 25, 549–557. [Google Scholar] [PubMed]
  65. Lin, Y.; Shi, R.; Wang, X.; Shen, H.-M. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr. Cancer Drug Targets 2008, 8, 634–646. [Google Scholar] [CrossRef] [PubMed]
  66. Hou, T.; Guo, Y.; Han, W.; Zhou, Y.; Netala, V.R.; Li, H.; Li, H.; Zhang, Z. Exploring the Biomedical Applications of Biosynthesized Silver Nanoparticles Using Perilla frutescens Flavonoid Extract: Antibacterial, Antioxidant, and Cell Toxicity Properties against Colon Cancer Cells. Molecules 2023, 28, 6431. [Google Scholar] [CrossRef]
  67. Netala, V.R.; Hou, T.; Sana, S.S.; Li, H.; Zhang, Z. Rosmarinic Acid-Rich Perilla frutescens Extract-Derived Silver Nanoparticles: A Green Synthesis Approach for Multifunctional Biomedical Applications including Antibacterial, Antioxidant, and Anticancer Activities. Molecules 2024, 29, 1250. [Google Scholar] [CrossRef]
  68. Ahmed, H.M.; Al-Zubaidy, A.M.A. Exploring natural essential oil components and antibacterial activity of solvent extracts from twelve Perilla frutescens L. Genotypes. Arab. J. Chem. 2020, 13, 7390–7402. [Google Scholar] [CrossRef]
  69. Tian, J.; Zeng, X.; Zhang, S.; Wang, Y.; Zhang, P.; Lü, A.; Peng, X. Regional variation in components and antioxidant and antifungal activities of Perilla frutescens essential oils in China. Ind. Crops Prod. 2014, 59, 69–79. [Google Scholar] [CrossRef]
  70. Hu, Z.; Lu, C.; Zhang, Y.; Tong, W.; Du, L.; Liu, F. Proteomic analysis of Aspergillus flavus reveals the antifungal action of Perilla frutescens essential oil by interfering with energy metabolism and defense function. LWT 2022, 154, 112660. [Google Scholar] [CrossRef]
  71. Zhang, J.; Liu, X.; Zhang, T.; Bai, B.; Yang, Y.; Bo, T.; Chen, M.; Fan, S. Study on the bacteriostatic property and bacteriostatic mechanism of rosemarinic acid compounded bacteriostatic agent. Food Biosci. 2024, 59, 103820. [Google Scholar] [CrossRef]
  72. Wang, G.; Wang, Y.; Wang, K.; Zhao, H.; Liu, M.; Liang, W.; Li, D. Perillaldehyde functions as a potential antifungal agent by triggering metacaspase-independent apoptosis in Botrytis cinerea. Microbiol. Spectr. 2023, 11, e00526-23. [Google Scholar]
  73. Zhu, W.; Cheng, Y.; Zhang, Y.; Li, M.; Teng, Y.; Gu, Y.; Wang, H.; Xia, X. Antibiofilm efficacies and mechanism of perillaldehyde against Shewanella putrefaciens. Food Microbiol. 2025, 128, 104699. [Google Scholar] [CrossRef]
  74. Lin, Y.; Wang, W.; Li, J.; Ai, Y.; Wang, H.; Han, Y.; Wang, W.; Hou, W. Unlocking the antibacterial activity and mechanisms of ε-poly-l-Lysine and perilla combination against Pseudomonas fluorescens: Insights from non-targeted metabolomic analyses. LWT 2024, 208, 116572. [Google Scholar] [CrossRef]
  75. Wang, Z.; Tu, Z.; Xie, X.; Cui, H.; Kong, K.W.; Zhang, L. Perilla frutescens Leaf Extract and Fractions: Polyphenol Composition, Antioxidant, Enzymes (alpha-Glucosidase, Acetylcholinesterase, and Tyrosinase) Inhibitory, Anticancer, and Antidiabetic Activities. Foods 2021, 10, 315. [Google Scholar] [CrossRef]
  76. Wang, J.; He, Y.; Yu, D.; Jin, L.; Gong, X.; Zhang, B. Perilla oil regulates intestinal microbiota and alleviates insulin resistance through the PI3K/AKT signaling pathway in type-2 diabetic KKAy mice. Food Chem. Toxicol. 2020, 135, 110965. [Google Scholar] [CrossRef] [PubMed]
  77. Niu, Q.-Q.; Fu, Z.-Z.; Mao, B.-Y.; Zhang, X.; Wang, H.-D.; Li, P.; Lin, L.-B.; Xi, Y.-T.; Yin, Y.-L.; Kamal, N.N.S.N.M. Perillaldehyde targeting PARP1 to inhibit TRPM2-CaMKII/CaN signal transduction in diabetic cardiomyopathy. Int. Immunopharmacol. 2025, 150, 114291. [Google Scholar] [CrossRef]
  78. Kim, H.R.; Kim, S.Y. Perilla frutescens Sprout Extract Protect Renal Mesangial Cell Dysfunction against High Glucose by Modulating AMPK and NADPH Oxidase Signaling. Nutrients 2019, 11, 356. [Google Scholar] [CrossRef]
  79. Kim, D.H.; Kim, S.J.; Yu, K.Y.; Jeong, S.I.; Kim, S.Y. Anti-hyperglycemic effects and signaling mechanism of Perilla frutescens sprout extract. Nutr. Res. Pract. 2018, 12, 20–28. [Google Scholar] [CrossRef]
  80. Chen, L.; Tian, G.; Tang, W.; Luo, W.; Liu, P.; Ma, Z. Protective effect of luteolin on streptozotocin-induced diabetic renal damage in mice via the regulation of RIP140/NF-κB pathway and insulin signalling pathway. J. Funct. Foods 2016, 22, 93–100. [Google Scholar] [CrossRef]
  81. Guan, J.; Guo, P.; Chen, M.; Wang, W.; Chen, X.; Li, Q.; He, Y.; Liang, J.; Yu, N.; Gao, P.; et al. Ultrasound-assisted extraction, purification, sulfation of Perilla leaves polysaccharide and hypoglycemic and hypolipidemic activities. Ultrason. Sonochem. 2025, 107269. [Google Scholar] [CrossRef]
  82. Ryu, D.; Jee, H.J.; Kim, S.Y.; Hwang, S.H.; Pil, G.B.; Jung, Y.S. Luteolin-7-O-Glucuronide Improves Depression-like and Stress Coping Behaviors in Sleep Deprivation Stress Model by Activation of the BDNF Signaling. Nutrients 2022, 14, 3314. [Google Scholar] [CrossRef]
  83. Li, J.J.; Li, Z.; Gu, L.J.; Choi, K.J.; Kim, D.S.; Kim, H.K.; Sung, C.K. The promotion of hair regrowth by topical application of a Perilla frutescens extract through increased cell viability and antagonism of testosterone and dihydrotestosterone. J. Nat. Med. 2018, 72, 96–105. [Google Scholar] [CrossRef] [PubMed]
  84. Fang, M.; Luo, L.; Chen, Y.; Liu, Y.; Yan, Y.; Wang, F.; Zou, Y.; Zhu, H.; Wu, X.; Jin, Z.; et al. Perillaldehyde Improves Parkinson-Like Deficits by Targeting G3BP Mediated Stress Granule Assembly in Preclinical Models. Adv. Sci. 2025, e2412152. [Google Scholar] [CrossRef]
  85. Zhao, N.; Hao, J.; Zhao, Y.; Zhao, B.; Lin, J.; Song, J.; Wang, M.; Luo, Z. A Novel Natural Penetration Enhancer for Transdermal Drug Delivery: In Vitro/In Vivo Evaluation and Penetration Enhancement Mechanism. Pharmaceutics 2025, 17, 254. [Google Scholar] [CrossRef]
  86. Kim, Y.-R.; Nam, B.; Han, A.-R.; Kim, J.-B.; Jin, C.H. Isoegomaketone from Perilla frutescens (L.) Britt stimulates MAPK/ERK pathway in human keratinocyte to promote skin wound healing. Evid.-Based Complement. Altern. Med. 2021, 2021, 6642606. [Google Scholar]
  87. Wang, Z.; Gao, C.; Yang, J.; Du, R.; Zeng, F.; Bing, H.; Xia, B.; Shen, Y.; Liu, C. Endophytic Streptomyces sp. NEAU-ZSY13 from the leaf of Perilla frutescens, as a promising broad-spectrum biocontrol agent against soil-borne diseases. Front. Microbiol. 2023, 14, 1243610. [Google Scholar]
  88. Kang, J.H.; Yang, S.-Y.; Ha, J.; Lee, K.-W. Perilla frutescens modulates CYP1A1/2 and HO-1 and activates Nrf2 in oxidative stress-induced hepatotoxicity. J. Korean Soc. Appl. Biol. Chem. 2015, 58, 305–315. [Google Scholar] [CrossRef]
  89. Wei, K.; Zhang, X.; Yang, J.; Chen, J. Tobacco introduced Perilla frutescens and Ocimum basilicum genes attenuates neutrophilic inflammation in lung tissues of COPD rats. Ecotoxicol. Env. Saf. 2024, 271, 115956. [Google Scholar] [CrossRef]
  90. Men, X.; Li, S.; Cai, X.; Fu, L.; Shao, Y.; Zhu, Y. Antiviral Activity of Luteolin against Pseudorabies Virus In Vitro and In Vivo. Animals 2023, 13, 761. [Google Scholar] [CrossRef]
  91. Wang, Y.; Huang, X.; Han, J.; Zheng, W.; Ma, W. Extract of Perilla frutescens inhibits tumor proliferation of HCC via PI3K/AKT signal pathway. Afr. J. Tradit. Complement. Altern. Med. 2013, 10, 251–257. [Google Scholar] [CrossRef] [PubMed]
  92. Cho, E.; Lee, J.; Sin, J.S.; Kim, S.K.; Kim, C.J.; Park, M.H.; Cho, W.S.; Moon, M.; Kim, D.H.; Jung, J.W. Effects of Perilla frutescens var. acuta in amyloid beta toxicity and Alzheimer’s disease-like pathology in 5XFAD mice. Food Chem. Toxicol. 2022, 161, 112847. [Google Scholar] [CrossRef] [PubMed]
  93. Zhong, Y.; Du, Q.; Wang, Z.; Zheng, Q.; Yang, M.; Hu, P.; Yang, Q.; Xu, H.; Wu, Z.; Huang, X. Antidepressant effect of Perilla frutescens essential oil through monoamine neurotransmitters and BDNF/TrkB signal pathway. J. Ethnopharmacol. 2024, 318, 116840. [Google Scholar] [PubMed]
  94. Joy Bormate, K.; Kyung Lee, B.; Kim, T.-H.; James Perez Custodio, R.; Hoon Cheong, J.; Jin Kim, H.; Hee Shim, S.; Bang Pil, G.; Jun Kim, H.; Ho Son, R.; et al. Involvement of adenosine A1 receptor in the sleep-promoting effect of fermented Perilla frutescens. J. Funct. Foods 2024, 120, 106345. [Google Scholar] [CrossRef]
  95. Cao, M.; Zhan, M.; Jing, H.; Wang, Z.; Wang, Y.; Li, X.; Miao, M. Network pharmacology and experimental evidence: MAPK signaling pathway is involved in the anti-asthma roles of Perilla frutescens leaf. Heliyon 2024, 10, e22971. [Google Scholar] [CrossRef]
  96. Feng, L.-J.; Yu, C.-H.; Ying, K.-J.; Hua, J.; Dai, X.-Y. Hypolipidemic and antioxidant effects of total flavonoids of Perilla frutescens leaves in hyperlipidemia rats induced by high-fat diet. Food Res. Int. 2011, 44, 404–409. [Google Scholar] [CrossRef]
  97. Kim, E.Y.; Choi, H.J.; Chung, T.W.; Choi, J.Y.; Kim, H.S.; Jung, Y.S.; Lee, S.O.; Ha, K.T. Water-extracted Perilla frutescens increases endometrial receptivity though leukemia inhibitory factor-dependent expression of integrins. J. Pharmacol. Sci. 2016, 131, 259–266. [Google Scholar] [CrossRef]
  98. Li, M.; Jiang, N.; Guo, G.; Lu, S.; Li, Z.; Mu, Y.; Xia, X.; Xu, Z.; Hu, Y.; Xiang, X. Perilla Seed Oil: A Review of Health Effects, Encapsulation Strategies and Applications in Food. Foods 2024, 13, 3615. [Google Scholar] [CrossRef]
  99. Dhiman, A.; Chopra, R.; Singh, P.K.; Singh, A.; Homroy, S. Enzymatic interesterification of perilla seed oil and palm stearin: A sustainable approach to develop a novel zero-trans-fat margarine rich in omega-3 fatty acids. J. Food Sci. 2024, 89, 8504–8523. [Google Scholar]
  100. Kim, H.U.; Lee, K.-R.; Jeon, I.; Jung, H.E.; Heo, J.B.; Kim, T.-Y.; Chen, G.Q. Fatty acid composition and oil content of seeds from perilla (Perilla frutescens (L.) var. frutescens) germplasm of Republic of Korea. Genet. Resour. Crop Evol. 2019, 66, 1615–1624. [Google Scholar]
  101. Sun, K.; Ma, T. Study on the lipid-lowering capacity of α-linolenic acid from Perilla seed oil in oleic acid-induced HepG2 cells. Food Chem. Adv. 2025, 6, 100871. [Google Scholar] [CrossRef]
  102. Thomas, S.S.; Kim, M.; Lee, S.J.; Cha, Y.S. Antiobesity Effects of Purple Perilla (Perilla frutescens var. acuta) on Adipocyte Differentiation and Mice Fed a High-fat Diet. J. Food Sci. 2018, 83, 2384–2393. [Google Scholar] [CrossRef]
  103. Chi, N.T.Y.; Duong, N.A.; Huynh, N.N.Y.; Nguyen, V.K.; Nguyen, T.T.K. Effects of storage temperature on biologically active ingredients of perilla drink. Food Chem. Adv. 2025, 6, 100897. [Google Scholar] [CrossRef]
  104. Hwang, E.S.; Noh, Y.; Jeong, H.Y.; Lee, J.J.; Ahn, B.M.; Lee, J.; Jang, Y.J. Improved skeletal muscle mass and strength through Protamex-mediated hydrolysis of perilla seed cake: Elevated rosmarinic acid levels as a contributing factor. Food Chem. 2025, 463, 141369. [Google Scholar] [CrossRef] [PubMed]
  105. Hu, Y.; Luo, H.; Netala, V.R.; Li, H.; Zhang, Z.; Hou, T. Comprehensive Review of Biological Functions and Therapeutic Potential of Perilla Seed Meal Proteins and Peptides. Foods 2024, 14, 47. [Google Scholar] [CrossRef]
  106. Kong, H.; Zhou, B.; Hu, X.; Wang, X.; Wang, M. Protective effect of Perilla (Perilla frutescens)leaf essential oil on the quality of a surimi-based food. J. Food Process. Preserv. 2018, 42, e13540. [Google Scholar] [CrossRef]
  107. Wang, X.; Wang, Z.; Sun, Z.; Wang, D.; Liu, F. A novel gelatin/chitosan-based “sandwich” antibacterial nanofiber film loaded with perillaldehyde for the preservation of chilled chicken. Food Chem. 2025, 465, 142025. [Google Scholar] [CrossRef]
  108. Wang, Y.; Zhao, Y.; Guo, Y.; Han, W.; Zhang, Z.; Hou, T.; Li, H.; Li, H.; Wang, Q. Preparation of Perilla frutescens L. essential oil hydrogel beads and preservation application research in strawberry. Heliyon 2024, 10, e33689. [Google Scholar] [CrossRef]
  109. Tai, Z.; Zheng, M.; Yang, Y.; Xie, C.; Li, Z.; Xu, C. Temperature controlled microcapsule loaded with Perilla essential oil and its application in preservation of peaches. Front. Nutr. 2023, 10, 1087605. [Google Scholar] [CrossRef]
  110. Ma, Y.; Cao, Y.; Zhang, L.; Yu, Q. Preservation of chilled beef using active films based on bacterial cellulose and polyvinyl alcohol with the incorporation of Perilla essential oil Pickering emulsion. Int. J. Biol. Macromol. 2024, 271, 132118. [Google Scholar] [CrossRef]
  111. Zhao, Y.; Kong, H.; Zhang, X.; Hu, X.; Wang, M. The effect of Perilla (Perilla frutescens) leaf extracts on the quality of surimi fish balls. Food Sci. Nutr. 2019, 7, 2083–2090. [Google Scholar] [CrossRef] [PubMed]
  112. Fang, Z.; Yang, Y.; Lin, S.; Xu, L.; Chen, S.; Lv, W.; Wang, N.; Dong, S.; Lin, C.; Xie, Y.; et al. Development and antimicrobial activity of composite edible films of chitosan and nisin incorporated with perilla essential oil-glycerol monolaurate emulsions. Food Chem. 2025, 462, 141006. [Google Scholar] [CrossRef] [PubMed]
  113. Khan, I.A.; Shi, B.; Shi, H.; Nawaz, A.; Zhu, Z.; Ijaz, M.U.; Hussain, M.; Khan, A.; Wang, M.; Chen, F.; et al. Perilla frutescens seed meal as a fat substitute mitigates heterocyclic amine formation and protein oxidation and improves fatty acid profile of pan-fried chicken patties. Front. Nutr. 2022, 9, 975831. [Google Scholar] [CrossRef] [PubMed]
  114. Hwang, Y.J.; Yoon, K.Y. Enzymatic hydrolysis of perilla seed meal yields water-soluble dietary fiber as a potential functional carbohydrate source. Food Sci. Biotechnol. 2020, 29, 987–996. [Google Scholar] [CrossRef]
  115. Yu, Y.; Zhang, B.; Jiang, X.; Cui, Y.; Luo, H.; Stergiadis, S.; Wang, B. Exploring the metabolomic landscape: Perilla frutescens as a promising enhancer of production, flavor, and nutrition in Tan lamb meat. Meat Sci. 2024, 209, 109419. [Google Scholar] [CrossRef]
  116. Wang, B.; Sun, Z.; Tu, Y.; Si, B.; Liu, Y.; Yang, L.; Luo, H.; Yu, Z. Untargeted metabolomic investigate milk and ruminal fluid of Holstein cows supplemented with Perilla frutescens leaf. Food Res. Int. 2021, 140, 110017. [Google Scholar] [CrossRef]
  117. Liqi, H.; Chuanfeng, H.; Yiting, L.; Xuan, L.; Lingyun, W.; Yu, Q.; Lan, W.; Wenjing, W.; Liu, S. Effect of Perilla Essential Oil and Ginger Juice on the Flavor of Grilled Wuchang Fish. Mod. Food Sci. Technol. 2024, 40, 193–202. [Google Scholar]
  118. Fujiwara, Y.; Kono, M.; Ito, A.; Ito, M. Anthocyanins in perilla plants and dried leaves. Phytochemistry 2018, 147, 158–166. [Google Scholar] [CrossRef]
  119. Klisurova, D.; Petrova, I.; Ognyanov, M.; Georgiev, Y.; Kratchanova, M.; Denev, P. Co-pigmentation of black chokeberry (Aronia melanocarpa) anthocyanins with phenolic co-pigments and herbal extracts. Food Chem. 2019, 279, 162–170. [Google Scholar] [CrossRef]
  120. Hur, S. Quality Characteristics of Fermented Perilla Leaves Ice Cream Using Probiotics. J. Korean Appl. Sci. Technol. 2024, 41, 375–385. [Google Scholar]
  121. Trang, P.N.; Trâm, N.T.B.; Anh, N.P. NGHIÊN CỨU CHẾ BIẾN KẸO MỀM SỮA BỔ SUNG SIRO TỪ LÁ TÍA TÔ (Perilla frutescens (L.) Britt.). TNU J. Sci. Technol. 2024, 229, 275–282. [Google Scholar] [CrossRef]
  122. Karthik, S.; Chae, J.; Han, S.J.; Kim, J.H.; Kim, H.J.; Chung, Y.S.; Kim, H.U.; Heo, J.B. Improving the Traits of Perilla frutescens (L.) Britt Using Gene Editing Technology. Plants 2024, 13, 1466. [Google Scholar] [CrossRef]
  123. Tong, W.; Kwon, S.J.; Lee, J.; Choi, I.Y.; Park, Y.J.; Choi, S.H.; Sa, K.J.; Kim, B.W.; Lee, J.K. Gene set by de novo assembly of Perilla species and expression profiling between P. frutescens (L.) var. frutescens and var. crispa. Gene 2015, 559, 155–163. [Google Scholar] [CrossRef] [PubMed]
  124. Zhou, P.; Yin, M.; Dai, S.; Bao, K.; Song, C.; Liu, C.; Wu, Q. Multi-omics analysis of the bioactive constituents biosynthesis of glandular trichome in Perilla frutescens. BMC Plant Biol. 2021, 21, 277. [Google Scholar] [CrossRef]
  125. Duan, W.; Shi-Mei, Y.; Zhi-Wei, S.; Jing, X.; De-Gang, Z.; Hong-Bin, W.; Qi, S. Genome-wide analysis of the fatty acid desaturase gene family reveals the key role of PfFAD3 in α-linolenic acid biosynthesis in perilla seeds. Front. Genet. 2021, 12, 735862. [Google Scholar]
  126. Liao, B.; Hao, Y.; Lu, J.; Bai, H.; Guan, L.; Zhang, T. Transcriptomic analysis of Perilla frutescens seed to insight into the biosynthesis and metabolic of unsaturated fatty acids. BMC Genom. 2018, 19, 213. [Google Scholar] [CrossRef]
  127. Fan, Y.; Cao, X.; Zhang, M.; Wei, S.; Zhu, Y.; Ouyang, H.; He, J. Quantitative comparison and chemical profile analysis of different medicinal parts of Perilla frutescens (L.) Britt. from different varieties and harvest periods. J. Agric. Food Chem. 2022, 70, 8838–8853. [Google Scholar] [PubMed]
  128. Zhou, P.; Shao, Y.; Jiang, Z.; Dang, J.; Qu, C.; Wu, Q. The revealing of a novel double bond reductase related to perilla ketone biosynthesis in Perilla frutescens. BMC Plant Biol. 2023, 23, 345. [Google Scholar] [CrossRef]
  129. Xing, F.; Xiao, Q.; Gul, H.; Liu, T.; Cao, W.; Zhang, Y.; Duan, H.; Li, Y.; Liang, J.; Zhang, X.; et al. Comparative global profiling of Perilla leaf and stem via transcriptomics and metabolomics. Gene 2024, 929, 148828. [Google Scholar] [CrossRef]
  130. Chinese Pharmacopoeia Commission. Chinese Pharmacopoeia; China Medical Science and Technology Press: Beijing, China, 2020.
  131. Wang, M.; Shen, R.; Wang, X.; Zhang, J.; Ma, J.; Shi, Y.; Xu, Q.; Guo, C.; Di, J.; Liu, A.; et al. Evaluation of edible and medicinal varieties based on multidimensional quantitative data: A case study of perilla leaf. J. Food Compos. Anal. 2025, 140, 107196. [Google Scholar] [CrossRef]
  132. Li, Y.; Yu, M.; Wei, Y.; Zhou, Z.; Guo, Y.; Yuan, M.; Jin, J.; Li, J.; Shen, H.; Wu, D. Risk assessment of developmental and neurotoxicity by the flavoring agent perillaldehyde: NAC (N-acetylcysteine) mitigation of oxidative stress-mediated inhibition of the Nrf2 pathway. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2025, 288, 110071. [Google Scholar] [CrossRef] [PubMed]
  133. Erhunmwunsee, F.; Pan, C.; Yang, K.; Li, Y.; Liu, M.; Tian, J. Recent development in biological activities and safety concerns of perillaldehyde from perilla plants: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 6328–6340. [Google Scholar] [CrossRef] [PubMed]
  134. Jung, S.; Lee, W.Y.; Yong, S.J.; Shin, K.C.; Kim, C.W.; Lee, J.-H.; Jung, Y.-R.; Kim, H.S.; Yu, T.-S.; Kim, S.-H. Occupational asthma caused by inhaling smoke from roasting perilla seeds. Allergy Asthma Respir. Dis. 2013, 1, 90–93. [Google Scholar]
  135. Jeong, K.; Lee, S.; Jeon, S.; Gantulga, P.; Nam, J.; Hong, S.; Lee, S. Clinical and Immunological Characterization of Perilla Seed Allergy in Children. J. Investig. Allergol. Clin. Immunol. 2021, 33, 14–20. [Google Scholar]
  136. Zhang, H.X.; Guan, J.; Tian, Y.H.; Su, G.Y.; Zhao, Y.Q. Acute and sub-chronic 90-day oral toxicity study of Perilla seed oil in rodents and Beagle dogs. Regul. Toxicol. Pharmacol. 2019, 103, 229–236. [Google Scholar] [CrossRef]
  137. Wu, Y.; Huang, L.; Xu, Y.; Zhang, Y.; Nie, L.; Kang, S.; Wei, F.; Ma, S. Rapid and accurate detection of cinnamon oil adulteration in perilla leaf oil using atmospheric solids analysis probe-mass spectrometry. Food Chem. 2025, 462, 140965. [Google Scholar] [CrossRef]
  138. DB15/T 3207-2024; Technical Code of Practice for Planting and Turning Green Manure to Fertilize in Degenerated Black Soil. Inner Mongolia Autonomous Region Market Supervision Administration: Hohhot, China, 2024.
Figure 1. Leaf and whole plant morphology of Perilla frutescens. Red (purple) perilla refers to plants with red (purple) leaves. Green perilla has entirely green leaves. Green/red perilla has green leaves with purple leaf backs.
Figure 1. Leaf and whole plant morphology of Perilla frutescens. Red (purple) perilla refers to plants with red (purple) leaves. Green perilla has entirely green leaves. Green/red perilla has green leaves with purple leaf backs.
Foods 14 01252 g001
Figure 2. (A) Perilla frutescens terpene synthesis pathway; (B,C) P. frutescens chemotype. EMP, glycolytic pathway; TCA, tricarboxylic acid cycle; AACT, acetyl-CoA acetyltransferase; HMGS, hydroxymethylglutaryl-CoA; HMGCR, hydroxymethylglutaryl-CoA reductase synthase; MVK, mevalonate kinase; PMK, phosphomevalonate kinase; MVD, mevalonate diphosphate decarboxylase; DXS, 1-deoxy-D-xylulose-5-phosphate synthase; DXR, 1-deoxy-D-xylulose-5-phosphate reductoisomerase; CMS, 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase; CMK, 4-diphosphocytidyl-2-C-methyl-D-erythritol kinase; MCS, 2-C-methyl-D-erythritol 2,4-cyclodiphosphate synthase; HDS, 4-hydroxy-3-methylbut-2-enyl diphosphate synthase; HDR, 4-hydroxy-3-methylbut-2-enyl diphosphate reductase; IDI, isopentenyl pyrophosphate isomerase; IPP, isopentenyl pyrophosphate; DMAPP, dimethylallyl pyrophosphate; GPP, geranyl pyrophosphate; FPP, farnesyl pyrophosphate; GGPP, geranylgeranyl pyrophosphate.
Figure 2. (A) Perilla frutescens terpene synthesis pathway; (B,C) P. frutescens chemotype. EMP, glycolytic pathway; TCA, tricarboxylic acid cycle; AACT, acetyl-CoA acetyltransferase; HMGS, hydroxymethylglutaryl-CoA; HMGCR, hydroxymethylglutaryl-CoA reductase synthase; MVK, mevalonate kinase; PMK, phosphomevalonate kinase; MVD, mevalonate diphosphate decarboxylase; DXS, 1-deoxy-D-xylulose-5-phosphate synthase; DXR, 1-deoxy-D-xylulose-5-phosphate reductoisomerase; CMS, 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase; CMK, 4-diphosphocytidyl-2-C-methyl-D-erythritol kinase; MCS, 2-C-methyl-D-erythritol 2,4-cyclodiphosphate synthase; HDS, 4-hydroxy-3-methylbut-2-enyl diphosphate synthase; HDR, 4-hydroxy-3-methylbut-2-enyl diphosphate reductase; IDI, isopentenyl pyrophosphate isomerase; IPP, isopentenyl pyrophosphate; DMAPP, dimethylallyl pyrophosphate; GPP, geranyl pyrophosphate; FPP, farnesyl pyrophosphate; GGPP, geranylgeranyl pyrophosphate.
Foods 14 01252 g002
Figure 3. Synthesis pathway of Perilla frutescens flavonoids and phenolic acids. PAL, phenylalanine ammonia lyase; C4H, cinnamate 4-hydroxylase; 4CL, 4-coumarate-CoA ligase; COMT, caffeic acid 3-O-methyltransferases; CHS, chalcone synthase; CHI, chalcone isomerase; F3H, flavanone 3-hydroxylase; F3′H, flavonoid 3′-hydroxylase; FNSI, flavone synthasel, I; FLSII, flavone synthase II; UGT, UDP-glycosyltransferase; F3′5′H, flavonoid 3′,5′-Hydroxylase; TAT, tyrosine aminotransferase; RAS, rosmarinate synthase; CYP, cytochrome P450; DFR, dihydroflavonol reductase; ANS, anthocyanin synthase; 3-GT, flavonoid 3-O-glucosyltransferase; AAT, anthocyanin acyltransferase; 5-GT, anthocyanin 5-O-glucosyltransferase; MAT, Malonyl-CoA: anthocyanin malonyltransferase.
Figure 3. Synthesis pathway of Perilla frutescens flavonoids and phenolic acids. PAL, phenylalanine ammonia lyase; C4H, cinnamate 4-hydroxylase; 4CL, 4-coumarate-CoA ligase; COMT, caffeic acid 3-O-methyltransferases; CHS, chalcone synthase; CHI, chalcone isomerase; F3H, flavanone 3-hydroxylase; F3′H, flavonoid 3′-hydroxylase; FNSI, flavone synthasel, I; FLSII, flavone synthase II; UGT, UDP-glycosyltransferase; F3′5′H, flavonoid 3′,5′-Hydroxylase; TAT, tyrosine aminotransferase; RAS, rosmarinate synthase; CYP, cytochrome P450; DFR, dihydroflavonol reductase; ANS, anthocyanin synthase; 3-GT, flavonoid 3-O-glucosyltransferase; AAT, anthocyanin acyltransferase; 5-GT, anthocyanin 5-O-glucosyltransferase; MAT, Malonyl-CoA: anthocyanin malonyltransferase.
Foods 14 01252 g003
Table 1. Statistical analysis of research directions on Perilla frutescens based on the Web of Science database (2021–2025). Specific data can be found in Supplementary Table S1.
Table 1. Statistical analysis of research directions on Perilla frutescens based on the Web of Science database (2021–2025). Specific data can be found in Supplementary Table S1.
YearTotal PublicationsPrimary Research Areas
Chemical Composition ResearchPharmacological Activity ResearchFood Industry ApplicationsGenetic Improvement ResearchOther Studies (Safety Standards/Heavy Metals/Stresses/Environment/
Quality/Pests, and Diseases/Clinical)
20251654133
20241431944161945
20231542647242037
20221482938251739
20211202838101727
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

Yi, D.; Wang, Z.; Peng, M. Comprehensive Review of Perilla frutescens: Chemical Composition, Pharmacological Mechanisms, and Industrial Applications in Food and Health Products. Foods 2025, 14, 1252. https://doi.org/10.3390/foods14071252

AMA Style

Yi D, Wang Z, Peng M. Comprehensive Review of Perilla frutescens: Chemical Composition, Pharmacological Mechanisms, and Industrial Applications in Food and Health Products. Foods. 2025; 14(7):1252. https://doi.org/10.3390/foods14071252

Chicago/Turabian Style

Yi, Dandan, Zhiyong Wang, and Mu Peng. 2025. "Comprehensive Review of Perilla frutescens: Chemical Composition, Pharmacological Mechanisms, and Industrial Applications in Food and Health Products" Foods 14, no. 7: 1252. https://doi.org/10.3390/foods14071252

APA Style

Yi, D., Wang, Z., & Peng, M. (2025). Comprehensive Review of Perilla frutescens: Chemical Composition, Pharmacological Mechanisms, and Industrial Applications in Food and Health Products. Foods, 14(7), 1252. https://doi.org/10.3390/foods14071252

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