Next Article in Journal
Comparative Metabolomic Analysis of Three Medicinal Paphiopedilum Species Reveals Divergence in Antioxidant Capacity and Functional Compound Profiles
Next Article in Special Issue
Death of Leukemia Cells and Platelets Induced by 3,3′-Dihydroxy-4,5-Dimethoxybibenzyl Is Mediated by p38 Mitogen-Activated Protein Kinase Pathway
Previous Article in Journal
Bioactive Cyclopeptide Alkaloids and Ceanothane Triterpenoids from Ziziphus mauritiana Roots: Antiplasmodial Activity, UHPLC-MS/MS Molecular Networking, ADMET Profiling, and Target Prediction
Previous Article in Special Issue
A Review of Botany, Phytochemistry, and Biological Activities of Eight Salvia Species Widespread in Kazakhstan
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparative Phytochemical Analysis and Antimicrobial Properties of Ethanol and Macerated Extracts from Aerial and Root Parts of Achillea nobilis

by
Aiman Berdgaleeva
1,*,
Zere Zhalimova
1,
Akzharkyn Saginbazarova
1,
Gulbanu Tulegenova
1,
Dana Zharylkassynova
1,
Aliya Bazargaliyeva
2,
Zhaidargul Kuanbay
2,
Svetlana Sakhanova
3,
Akmaral Ramazanova
4,
Akzhamal Bilkenova
5 and
Aigul Sartayeva
6
1
Department of Pharmaceutical Disciplines, Marat Ospanov West Kazakhstan Medical University, 66 Maresyev Street, Aktobe 030019, Kazakhstan
2
Department of Biology, Natural Sciences Faculty, K.Zhubanov Aktobe Regional University, 34 Moldagulova Avenue, Aktobe 030012, Kazakhstan
3
Scientific and Practical Center, Marat Ospanov West Kazakhstan Medical University, 74 Maresyev Street, Aktobe 030019, Kazakhstan
4
Department of Neurology with the Course of Psychiatry and Narcology, Marat Ospanov West Kazakhstan Medical University, 66 Maresyev Street, Aktobe 030019, Kazakhstan
5
Department of Natural Sciences, Marat Ospanov West Kazakhstan Medical University, 66 Maresyev Street, Aktobe 030019, Kazakhstan
6
Department of General Medical Practice No. 2, Marat Ospanov West Kazakhstan Medical University, 66 Maresyev Street, Aktobe 030019, Kazakhstan
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(14), 2957; https://doi.org/10.3390/molecules30142957
Submission received: 9 June 2025 / Revised: 6 July 2025 / Accepted: 7 July 2025 / Published: 14 July 2025
(This article belongs to the Special Issue Advances in Natural Products and Their Biological Activities)

Abstract

Achillea nobilis represents a species of considerable medicinal importance within the Asteraceae family, historically employed in Central Asia and various Eurasian territories for the management of inflammatory, microbial, and gastrointestinal ailments. Notwithstanding its extensive ethnopharmacological significance, the phytochemical profile and pharmacological attributes of its various anatomical components have not been comprehensively investigated. This research endeavor sought to delineate the phytochemical constituents and evaluate the antimicrobial efficacy of ethanol extracts derived from both the aerial and root segments of A. nobilis. Qualitative phytochemical analysis and GC–MS characterization unveiled a diverse array of bioactive compounds, encompassing flavonoids, phenolic compounds, organic acids, lactones, alcohols, and heterocyclic derivatives. In particular, the aerial oil extract exhibited the presence of terpenoids, fatty acids and their esters, sterols, hydrocarbons, and minor organosilicon and cyclobutanone derivatives, with notable compounds such as linoleic acid (8.08%), 6-tetradecyne (14.99%), isopropyl linoleate (14.64%), and E,Z-1,3,12-nonadecatriene (22.25%). In vitro antimicrobial activity was assessed against eight clinically relevant microbial strains employing the broth microdilution technique. The aerial ethanol extract exhibited pronounced antimicrobial properties, particularly against MRSA and C. albicans, with MICs ranging from 0.5 to 2 mg/mL, whereas the root ethanol extract displayed MICs of 1 to 3 mg/mL. Additionally, the aerial oil extract showed moderate inhibitory activity, with MIC values ranging from 1.5 to 3 mg/mL, demonstrating effectiveness particularly against C. albicans, C. neoformans, and MRSA. These findings underscore the therapeutic potential of A. nobilis, particularly its aerial component, as a viable natural source of antimicrobial agents.

1. Introduction

The focused inquiry into flora possessing ethnopharmacological significance persists as an essential avenue for the identification of novel bioactive compounds exhibiting therapeutic potential [1]. Achillea nobilis, in conjunction with its subspecies (A. nobilis subsp. sipylea, A. nobilis subsp. neilreichii, and A. nobilis subsp. nobilis), forms a taxonomic assemblage of aromatic medicinal plants that have been historically acknowledged for their varied healing properties throughout Eurasian territories [2,3,4]. Members of this assemblage, classified within the Asteraceae family, have been deeply entrenched in numerous traditional medicinal practices, utilized in the form of decoctions, infusions, essential oils, and topical applications. These formulations have been primarily applied to address a broad range of ailments, encompassing inflammatory conditions, pain syndromes, gastrointestinal disturbances, respiratory infections, and dermatological issues [5,6,7,8,9,10]. The persistent application of A. nobilis in ethnomedicine is particularly notable within the healing customs of nations such as Bosnia and Herzegovina, Turkey, Iran, China, and Kazakhstan, where the plant is imbued with both cultural and pharmacological importance (Figure 1) [1,2,11].
A. nobilis, or “noble yarrow,” is a perennial herbaceous species of the Asteraceae family, typically growing 15–70 cm tall. It features hairy, longitudinally ridged stems and woolly-pubescent, pinnately divided leaves that vary between basal and upper parts. The plant forms dense, corymbose inflorescences with numerous small capitula containing pale yellow and white florets. Flowering occurs from June to August, and the species is well-adapted to temperate Eurasian habitats [1,2,3].
Between the years 1978 and 2011, phytochemical investigations concerning A. nobilis predominantly concentrated on the aerial segments of the organism, with insufficient focus directed towards its subterranean roots and lipid-soluble constituents [12]. The most commonly employed methodology for extraction was hydrodistillation, particularly for the purpose of isolating volatile compounds from the flowering aerial tissues [13,14,15,16,17,18,19]. Initial studies employed chromatographic techniques in conjunction with ultraviolet and infrared spectroscopy to identify flavonoid glycosides such as apigenin-7-glycoside (cosmosiin) and luteolin-7-glucoside (cynaroside) [17,18]. Subsequent investigations integrated advanced methodologies, including infrared spectroscopy, nuclear magnetic resonance, thin-layer chromatography, ultraviolet spectroscopy, gas chromatography–mass spectrometry, electrospray ionization mass spectrometry, and high-performance liquid chromatography, to further elucidate the plant’s secondary metabolites [16,17,18]. Through the application of these methodologies, a diverse array of chemical classes were discerned, encompassing sesquiterpene lactones, methoxylated flavones, dihydroxyflavones, and peroxide derivatives. Compounds such as anobin, estafiatin, hanphyllin, tanaparthin-β-peroxide, and chrysartemin A were successfully isolated from chloroform and ethanol extracts, while 1,8-cineole, camphor, α-thujone, β-caryophyllene, and artemisia ketone were identified in hexane and dichloromethane fractions utilizing gas chromatography–mass spectrometry [20,21,22,23]. Despite some investigations also employing liquid–liquid partitioning and solid-phase microextraction techniques, the chemical profiling of non-polar fractions and subterranean components has remained largely unexplored. The persistent emphasis on aerial parts and polar solvent systems underscores a significant gap in the phytochemical characterization of A. nobilis, thereby accentuating the imperative for future research endeavors aimed at investigating root-derived and lipid-soluble constituents to comprehensively assess the plant’s medicinal potential.
Pharmacological research on A. nobilis has shown a variety of bioactivities that back its traditional applications and healing potential. Importantly, ethanolic extracts of A. nobilis showed considerable anticonvulsant effects in animal seizure models, such as maximal electroshock (MES), pentylenetetrazole (PTZ), and strychnine nitrate (STN)-induced seizures. The effects were dependent on the dosage, with 60% protection noted at 300 mg/kg in the STN model, indicating a role in glycine-mediated neurotransmission [7,24]. Additionally, A. nobilis subsp. neilreichii showed antinociceptive and anti-inflammatory properties in rodent studies, particularly alleviating inflammatory pain in formalin and carrageenan-induced paw edema tests, although it was less efficient in thermal nociception trials [8,25]. A. nobilis subsp. sipylea demonstrated antispasmodic effects in isolated rat duodenum samples, functioning through calcium channel modulation instead of muscarinic antagonism, suggesting smooth muscle relaxation capabilities [7,8,22,23]. Research on antioxidants showed that A. nobilis infusions increased catalase and glutathione peroxidase activities in erythrocytes and leucocytes, offering protection against oxidative damage caused by hydrogen peroxide [7,23,24].
The antimicrobial efficacy of A. nobilis and its various subspecies has been corroborated by numerous investigations, which illustrate inhibitory effects against a wide array of Gram-positive and Gram-negative microorganisms. In a comparative analysis, essential oils derived from A. nobilis subsp. sipylea and A. nobilis subsp. neilreichii displayed significant activity against pathogens such as Escherichia coli, Staphylococcus aureus, Salmonella typhimurium, and Candida albicans, with inhibition zones ranging from 9 to 41 mm contingent upon the specific bacterial strain and the provenance of the plant material [9]. Furthermore, when assessed alongside conventional antibiotics such as amoxicillin and sulbactam/ampicillin, the essential oils demonstrated comparable effectiveness in certain instances, particularly against S. aureus and P. vulgaris [9]. Additional investigations revealed that A. nobilis essential oil accomplished inhibition zones of 25 mm against E. coli, Klebsiella pneumoniae, and S. aureus, and 23 mm against Pseudomonas aeruginosa, thereby surpassing A. crithmifolia and even thymol in specific scenarios [15]. A comprehensive screening employing the disc diffusion methodology indicated that A. nobilis subsp. neilreichii essential oil inhibited various strains, including Bacillus megaterium, Listeria monocytogenes, and K. pneumoniae, with inhibition zones ranging from 3 to 13 mm [21]. These outcomes bolster the proposition that the antimicrobial effects are contingent upon species and strain, likely influenced by the phytochemical constituents of the essential oil. Moreover, the choice of solvent and extraction method significantly influences the antimicrobial potency, as illustrated in antioxidant studies which demonstrated that ethanol and ethanol–water extracts exhibited superior radical scavenging and reducing capabilities, potentially correlating with antimicrobial efficacy [20].
Our research aims to provide a comprehensive analysis of the phytochemical composition of A. nobilis by examining both the aerial and root parts using two complementary extraction approaches: ethanol extraction aided by vortexing and maceration-based oil extraction. Subsequently, GC–MS profiling was performed to identify a broad spectrum of bioactive compounds potentially responsible for the plant’s traditional medicinal applications. Previous studies have largely focused on hydrodistilled or aqueous–alcoholic extracts from the aerial parts, often neglecting the chemical diversity of underground organs and lipid-soluble fractions. To overcome this limitation, we implemented vortex-assisted ethanol extraction, which enhances solvent penetration and facilitates efficient recovery of both polar and moderately non-polar constituents. In parallel, macerated oil extraction of the aerial part was conducted to selectively isolate lipophilic compounds, such as fatty acids, terpenoids, sterols, and hydrocarbons, which are not readily extracted with polar solvents. The inclusion of oil-based extraction was further motivated by its practical relevance for topical formulations. Given the traditional external use of A. nobilis in folk medicine, the oil extract serves as a prototype for the development of therapeutic ointments and dermal applications, where lipid-soluble phytoconstituents can exert antimicrobial, anti-inflammatory, or antioxidant effects directly at the site of application. This dual-extraction strategy not only expands the phytochemical profile of the species but also provides a pharmacologically meaningful foundation for the development of evidence-based phytotherapeutic products targeting both systemic and topical uses.

2. Results

2.1. Screening for Bioactive Phytochemical Classes

Phytochemicals consist of a wide variety of naturally found plant substances, numerous of which are linked to medicinal attributes. A qualitative phytochemical analysis was performed to acquire an initial insight into the medicinal potential of A. nobilis. As outlined in Table 1, extracts from both the aerial and root sections of the plant, as well as the aerial oil extract, demonstrated the existence of several important phytochemical categories. The screening validated the presence of biologically active components including alcohols, aldehydes, amines, amides, alkaloids, triterpenoids, tannins, flavonoids, and glycosides.
In the aerial oil extract, alcohols, aldehydes, and triterpenoids were prominently detected, while flavonoids and glycosides were also present in moderate amounts. In contrast, amines, amides, tannins, and alkaloids were not detected in the oil-based extract.

2.2. Secondary Metabolite Analysis (GC–MS)

An extensive GC–MS evaluation of the aerial ethanol extract, root ethanol extract, and aerial oil extract of A. nobilis showed a wide variety of phytochemical compounds. Ethanol was chosen as the extraction solvent because of its wide-ranging ability to dissolve various polar to moderately non-polar bioactive substances and its appropriateness for subsequent biological and pharmacological assessments. A total of 38 compounds were recognized in the aerial ethanol extract (Table 2, Figure 2), while 58 compounds were found in the root ethanol extract (Table 3, Figure 3). Of these, 22 compounds were found in both plant sections, indicating a level of phytochemical similarity. These common constituents were additionally categorized according to their relative concentrations into three specific groups: minor compounds (present at concentrations <1%), common compounds (n = 22), and major compounds (concentrations ≥1%).
Furthermore, 25 compounds were identified in the aerial oil extract (Table 4, Figure 4), of which 9 were minor compounds (present at concentrations <1%) and 16 were major compounds (≥1%). These compounds belonged to diverse phytochemical classes including terpenoids, heterocyclic compounds, fatty acids, fatty acid esters, hydrocarbons, amides, vitamins, cyclobutanone derivatives, organosilicon compounds, and sterols. Compared to the aerial ethanol extract and root ethanol extract, the aerial oil extract exhibited a more selective enrichment of lipophilic bioactives, especially terpenoid and fatty acid derivatives, consistent with the solvent’s affinity for non-polar constituents.
In the aerial ethanol extract, the compounds identified at concentrations under 1% included R-(–)-1,2-propanediol (0.89%), γ-butyrolactone (0.95%), N-nitrosohexamethyleneimine (0.19%), 1,2-cyclopentanedione (0.54%), and 5-tert-butylpyrogallol (0.60%). In comparison, the root ethanol extract displayed a broader spectrum of minor elements, including but not restricted to 4-cyclopentene-1,3-dione (0.81%), 2(5H)-furanone, 3-methyl- (0.39%), 2(5H)-furanone (0.97%), 1,2-cyclopentanedione, 3-methyl- (0.69%), and 2-cyclopenten-1-one, 3-ethyl-2-hydroxy- (0.54%). Other minor components included ethanone, 1-(1H-pyrrol-2-yl)- (0.45%), 2(3H)-furanone, 5-heptyldihydro- (0.70%), allyl acetate (0.85%), 1,2,3-propanetriol, 1-acetate (0.42%), acetaminophen (0.09%), and uric acid (0.70%). Trace amounts of various nitrogen-containing heterocycles and phenolic derivatives were also detected, including 2-aminopyrimidine-1-oxide (0.53%), 3-pyridinol, 6-methyl- (0.32%), and uracil (0.56%).
The aerial ethanol extract comprised a collection of significant compounds, such as 2,3-butanediol (6.08%), butanoic acid (1.89%), (L)-α-terpineol (1.53%), methyl N-hydroxybenzenecarboximidate (2.69%), and ethanol, 2,2’-oxybis- (1.82%). Other notable components included phenol, 4-ethyl-2-methoxy- (1.08%), 1,3-propanediol (2.20%), eugenol (1.49%), and dianhydromannitol (1.07%). Significantly, 3-isobutylhexahydropyrrolo [1,2-a]pyrazine-1,4-dione (1.27%) and octadecanoic acid (1.42%) also played a major role in shaping the extract’s chemical profile. In contrast, the root ethanol extract demonstrated a wider range of chemical variety among its primary components. Significant compounds included 2-propenoic acid (1.16%), 2-furanmethanol (1.14%), 2,4-dimethyl-2-oxazoline-4-methanol (3.01%), and 1,2-cyclopentanedione (2.55%). The compound present in the highest quantity was benzaldehyde, 3-hydroxy-, oxime (6.65%). Other notable compounds included cyclopropyl carbinol (1.06%), 1,3-dioxol-2-one, 4,5-dimethyl- (1.39%), α-hydroxy-γ-butyrolactone (1.26%), and 4H-pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6-methyl- (9.39%). The latter showed the greatest relative abundance among all recognized components. In addition, 3-pyridyl ester of benzoic acid (1.88%) and butyl 9-decenoate (1.58%) were also common.
The aerial oil extract contained a distinct composition with 25 identified compounds. Among these, 9 were minor compounds (present at <1%): camphor (1.41%), 2,4-decadienal (0.46%), 2-furanacetaldehyde, α-propyl- (0.69%), 3-(hydroxymethylene)indolin-2-one (0.66%), hedycaryol (0.70%), 6-methyl-2,4(1H,3H)-pteridinedione (0.57%), tridecanoic acid, methyl ester (0.92%), 11,14-octadecadienoic acid, methyl ester (0.68%), 4-methyl-3-pentenal (0.60%), and 1-(trimethylsilyl)-1-propyne (0.49%). The major compounds (≥1%) in the aerial oil extract were hexadecanoic acid, ethyl ester (1.15%), linoleic acid (8.08%), linoleic acid ethyl ester (1.75%), oleic acid ethyl ester (2.28%), palmitoyl chloride (2.50%), α-tocopherol (8.36%), E,Z-1,3,12-nonadecatriene (22.25%), oleic acid, 3-hydroxypropyl ester (6.92%), 2-dodecylcyclobutanone (1.24%), isopropyl linoleate (14.64%), cis-13,16-docasadienoic acid (1.42%), chondrillasterol (1.54%), and squalene (2.35%).
The GC–MS analysis showed a wide range of chemical classes present in the aerial ethanol extract, root ethanol extract, and aerial oil extract of A. nobilis (Table 5). Both ethanol-based extracts contained high levels of alcohols, carboxylic acids, ketones, phenols, and heterocyclic compounds. Alcohols constituted an important category, including various diols and polyether diols found in both sections of the plant. Carboxylic acids, especially saturated fatty acids and short-chain fatty acids, were prevalent in both extracts, highlighting the metabolic diversity of the plant.
Significantly, the root ethanol extract exhibited a greater presence of distinct chemical classes like carbonate derivatives, sugar lactones, nitrosoamines, and oxazoline derivatives, which were not found in the aerial ethanol extract. In contrast, dioxolones and imidates were only found in the aerial ethanol extract. Additionally, phenolic compounds, often linked to antioxidant and antimicrobial benefits, were found in both extracts, although certain phenol derivatives were exclusive to one section. Overall, the root ethanol extract demonstrated the greatest variety of chemical classes, while the aerial oil extract exhibited selective enrichment in lipid-soluble phytochemicals, contributing additional pharmacologically relevant classes not seen in polar extracts.
The categorization of the detected compounds revealed clear differences in distribution patterns between the aerial and root extracts. The aerial extract of the alcohols mainly included diols and monoterpene alcohols, whereas the root extract presented other subclasses like cyclopropyl alcohols and polyether diols. Among the carboxylic acids, both extracts included short-chain fatty acids and γ-hydroxy acids; however, the root ethanol extract specifically had α,β-unsaturated acids and hydroxybutyrolactones.
The aerial oil extract contained significant amounts of long-chain unsaturated fatty acids, such as linoleic and oleic acid derivatives, along with their esters. This extract also included tocopherols, triterpenoids, and cyclobutanone derivatives, representing lipid-soluble classes rarely detected in ethanol-based profiles.
Phenolic subclasses also exhibited variation, with methoxyphenols, dimethoxyphenols, and hydroquinone derivatives found in both parts, whereas alkylated methoxyphenols and trihydroxybenzene derivatives were observed solely in the aerial ethanol extract. The root ethanol extract exhibited significant diversity in the heterocyclic class, featuring oxazoline, pyrimidine, and pyridinol derivatives, while the aerial extract contained various pyrrole and imidate derivatives. Furthermore, subclasses like sugar alcohols, dihydroxybenzenes, and pyranones distinguished the two extracts further.
Of the 22 shared phytochemical components found in both aerial ethanol and root ethanol extracts of A. nobilis, several were detected at levels under 1% in both sections of the plant (Figure 5, Table 6). These comprised propanoic acid (0.92% in aerial ethanol extract, 0.96% in root ethanol extract), benzofuran, 2,3-dihydro- (0.60%, 0.41%), succinimide (0.48%, 0.34%), and 3-methyl-4-phenyl-1H-pyrrole (0.61%, 0.48%). Moreover, β-D-glucopyranose, 1,6-anhydro- (1.39%, 0.33%) and 1,4:3,6-dianhydro-α-D-glucopyranose (1.76%, 0.4%) demonstrated asymmetric occurrence, surpassing 1% solely in the aerial ethanol extract. These minor components, despite being found in comparatively small amounts, can have considerable pharmacological impacts via synergistic processes or act as indicators for particular therapeutic functions.
In the aerial oil extract, additional minor compounds identified at levels under 1% included 2,4-decadienal (0.46%), 2-furanacetaldehyde, α-propyl- (0.69%), 3-(hydroxymethylene)indolin-2-one (0.66%), hedycaryol (0.70%), 6-methyl-2,4(1H,3H)-pteridinedione (0.57%), 4-methyl-3-pentenal (0.60%), 11,14-octadecadienoic acid, methyl ester (0.68%), and 1-(trimethylsilyl)-1-propyne (0.49%).
Multiple compounds were consistently found at elevated levels (>1%) in both aerial ethanol and root ethanol extracts, suggesting their prevalence and possible functional significance in A. nobilis. 4-hydroxybutanoic acid (7.76% in aerial ethanol extract, 2.34% in root ethanol extract) and glycerin (15.97%, 6.73%) were some of the main components. 2-methoxy-4-vinylphenol, a recognized phenolic substance, was significantly found (3.26%, 1.10%), in addition to phenol, 2,6-dimethoxy- (2.85%, 1.57%). Other frequently occurring shared compounds included triethylene glycol (2.80%, 1.05%), tetraethylene glycol (3.22%, 1.95%), hydroquinone (1.74%, 1.35%), and pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)- (2.19%, 1.55%). These findings emphasize that both parts of the plant hold a fundamental set of key bioactive compounds that could greatly enhance the overall medicinal properties of A. nobilis. In the aerial oil extract, compounds detected at levels >1% included camphor (1.41%), hexadecanoic acid, ethyl ester (1.15%), linoleic acid ethyl ester (1.75%), oleic acid ethyl ester (2.28%), 2,2,2-trifluoro-N-(hydroxymethyl)acetamide (3.35%), palmitoyl chloride (2.50%), α-tocopherol (8.36%), E,Z-1,3,12-nonadecatriene (22.25%), oleic acid, 3-hydroxypropyl ester (6.92%), 2-dodecylcyclobutanone (1.24%), isopropyl linoleate (14.64%), cis-13,16-docasadienoic acid (1.42%), chondrillasterol (1.54%), and squalene (2.35%).

2.3. Determination of Antimicrobial Potency

The expected minimum inhibitory concentrations (MICs) of ethanol extracts from the aerial and root sections of A. nobilis against a selection of clinically significant microbial strains are presented in the related data (Table 7). The aerial extract showed enhanced antimicrobial effectiveness overall, exhibiting lower MIC values specifically against C. albicans (0.75 mg/mL), MRSA (0.50 mg/mL), and C. neoformans (0.85 mg/mL), in contrast to the root extract. Additionally, the aerial oil extract demonstrated considerable antimicrobial effects, with MIC values of 0.9 mg/mL against MRSA, 1.2 mg/mL against C. albicans, and 1.4 mg/mL against C. neoformans.
Despite the root extract demonstrating less potent inhibitory effects, it still displayed moderate activity against all tested strains, indicating that both parts of the plant have broad-spectrum antimicrobial potential. The aerial oil extract also showed MIC values of 1.5–3.2 mg/mL against Gram-negative bacteria, reinforcing its comparable effectiveness. Importantly, the extracts showed efficacy against Gram-positive and Gram-negative bacteria, along with fungal pathogens, reinforcing the traditional application of A. nobilis for treating infections and emphasizing its potential as a source of natural antimicrobial compounds, which may contribute to alternative therapeutic strategies amid rising antibiotic resistance and the need for plant-derived agents with multifunctional bioactivity.

3. Discussion

The increasing worldwide fascination with bioactive natural products highlights the essential requirement for thorough scientific research on medicinal plant species. Plants historically utilized in folk medicine for various health issues are now gaining heightened interest from researchers in various fields because of their possible pharmacological properties and enduring ethnomedicinal significance. A. nobilis is one such plant, historically esteemed in Central Asia for its medicinal qualities, akin to other species in the Achillea genus [7,8,17]. Even with its recorded traditional uses, a considerable gap persists in focused phytochemical studies that specifically target this species. The lack of thorough chemical and pharmacological investigations not only restricts our comprehension of its possible therapeutic advantages but also hinders the identification of new compounds that might aid in creating evidence-based therapies. As a result, an important source of natural therapeutic compounds might be neglected. Thus, prioritizing the phytochemical identification and pharmacological assessment of important regional plants such as A. nobilis is crucial to reveal their complete medicinal capabilities and facilitate their incorporation into contemporary healthcare practices.
The preliminary phytochemical screening of A. nobilis revealed the presence of various major classes of bioactive compounds. In the aerial ethanol extract, alcohols, aldehydes, amines, flavonoids, triterpenoids, and glycosides were detected. The aerial oil extract tested positive for alcohols, aldehydes, alkaloids, and triterpenoids. Meanwhile, the root ethanol extract contained flavonoids, alkaloids, and glycosides. These findings are consistent with previous studies on various members of the Achillea genus, where flavonoids, terpenoids, and phenolic compounds have been widely reported as dominant constituents contributing to their anti-inflammatory, antimicrobial, and antioxidant properties [61,62,63,64]. Compared to species such as A. millefolium and A. wilhelmsii, which are well-characterized for their high flavonoid and sesquiterpene lactone contents, A. nobilis shows a similar phytochemical pattern but demonstrates a broader distribution of classes across both plant organs [64]. Moreover, the presence of glycosides and triterpenoids, particularly in the root extracts, highlights the potential for underexplored pharmacological effects unique to this species. These results emphasize the importance of expanding phytochemical research within the Achillea genus to include lesser-studied species like A. nobilis, which may serve as reservoirs of novel bioactive metabolites with therapeutic relevance [65,66,67].
The GC–MS analysis of the aerial ethanol extract of A. nobilis identified various pharmacologically active compounds that contribute to the plant’s traditional and modern medicinal relevance. Among them, 2,3-butanediol demonstrated a broad range of biological activities, including antitumor and immunomodulatory effects, cryoprotective properties, and utility as a drug carrier, alongside anti-inflammatory, neuroprotective, and antimicrobial potential [26,27]. 1,3-Propanediol was also identified, known for its application in pharmaceutical formulations as a solvent, humectant, and stabilizer [28]. Notably, butanoic acid, a histone deacetylase (HDAC) inhibitor, links to epigenetic modulation and potential anticancer effects [33]. Octadecanoic acid displayed a wide therapeutic range including antimicrobial, anti-inflammatory, antioxidant, anticancer, emollient, cholesterol-lowering, and immunomodulatory properties [34]. Other relevant compounds included dianhydromannitol, known for diuretic, osmoprotective, and excipient-related functions [40], γ-butyrolactone, which acts on the central nervous system as a sedative and anxiolytic but warrants caution due to its association with GHB-related dependence [43], and eugenol, a well-studied bioactive with antibacterial, antifungal, antiviral, anticancer, anti-inflammatory, and antioxidant activity [45].
In contrast, the aerial oil extract exhibited a distinct phytochemical profile, rich in lipophilic bioactives. For instance, α-tocopherol is a potent antioxidant with additional anti-inflammatory, cytoprotective, neuroprotective, and mitochondrial stabilizing properties; it also modulates signal transduction and lipid metabolism, and protects non-cancerous cells during chemotherapy [57,58,59,60]. Linoleic acid, a major polyunsaturated fatty acid, was detected, with confirmed anti-proliferative, pro-apoptotic, antioxidant, and anti-inflammatory functions, along with EMT and angiogenesis inhibition and immune modulation via mitochondrial biogenesis pathways (PGC-1α/NRF1/TFAM) [34,35]. Squalene, a triterpene, is known for its antioxidant, anti-inflammatory, anticancer, and antidiabetic effects [52]. Chondrillasterol demonstrated antimicrobial activity, adding to the therapeutic profile of the oil extract [56]. Another major compound, camphor, has been shown to possess antifungal activity against Rhizoctonia solani and other pathogens [53], antiviral action against orthopoxviruses [54], and cytotoxicity toward cancer cells through various mechanistic pathways [55].
The GC–MS examination of the root extract from A. nobilis identified various unique bioactive compounds with specific pharmacological significance. Among these, 2-furanmethanol was recognized, a substance known for its antimicrobial, antifungal, and anticancer properties, indicating its possible role in the plant’s conventional medicinal uses [31]. Furthermore, 4-cyclopentene-1,3-dione was exclusively detected in the root extract and is recognized for its significant antifungal characteristics, further reinforcing the antifungal potential of constituents derived from the root [42]. Another notable compound, 2,3-dimethylhydroquinone, showed antioxidant and antimicrobial properties, as well as cytotoxic and redox-modulating actions, suggesting a potential function in managing oxidative stress and pathways linked to cancer [49]. The identification of uric acid is especially significant, as it functions as a biomarker for cardiovascular and metabolic diseases, and contributes to inflammation in conditions like gout due to crystal-induced inflammation in hyperuricemic conditions [50]. Ultimately, uracil was identified and is linked to a wide variety of pharmacological effects, encompassing antiviral, anticancer, and antimicrobial actions, as well as its role in DNA and RNA synthesis, enzyme inhibition, radiosensitization, and immunomodulation [51]. Together, these compounds underscore the therapeutic potential of A. nobilis roots, which have been relatively overlooked, and stress the necessity for additional pharmacological and mechanistic studies regarding their effectiveness as treatments.
The GC–MS examination of A. nobilis identified a set of bioactive compounds typically found in both aerial and root extracts, all exhibiting a broad range of pharmacological effects. Glycerin acts as a powerful humectant and lubricant, commonly utilized for its ability to retain moisture in pharmaceutical and cosmetic uses [29]. In a similar vein, triethylene glycol demonstrates significant antimicrobial and antiviral properties, as well as disinfectant and air-purifying capabilities, establishing it as a versatile agent with pharmaceutical importance [30]. Propanoic acid offers antimicrobial and anti-inflammatory properties, and additionally influences lipid metabolism regulation, modulates gut microbiota, and inhibits HDAC, thus reinforcing its possible anticancer and immune-modulatory functions [32]. Additionally, 4-hydroxybutanoic acid is recognized for its sedative, muscle relaxant, and central nervous system depressant properties, along with its therapeutic application in narcolepsy and alcohol dependence [37]. Guaiacol is notable among phenolic compounds for its expectorant, analgesic, and antiseptic qualities, as well as its anti-inflammatory and antioxidant capabilities [44]. Succinimide, found in both sections, is known for its anticonvulsant, sedative, and enzyme-inhibiting properties, particularly in neurological conditions [41]. Phenol, a thoroughly researched antiseptic and disinfectant, offers extra antibacterial, antifungal, and pain-relieving effects [46]. Additionally, 2-methoxy-4-vinylphenol and hydroquinone exhibit potent antioxidant, anti-inflammatory, and anticancer properties, with hydroquinone also recognized for its ability to depigment skin and inhibit melanin production [47,48], further underscoring the therapeutic potential of these constituents in dermatological, oncological, and inflammatory conditions, and supporting the pharmacological relevance of A. nobilis as a source of multifunctional bioactive compounds.
Compared to earlier pharmacological research on A. nobilis and its subspecies, our results provide significant new information, especially about the antimicrobial effectiveness of ethanol extracts from both aerial and root sections. Our findings highlighted that the aerial extract displayed more potent inhibitory effects on all examined microorganisms, with the lowest MIC values recorded against MRSA (0.5 mg/mL), C. albicans (0.75 mg/mL), and C. neoformans (0.85 mg/mL), suggesting noteworthy antibacterial and antifungal activity. Additionally, the aerial oil extract showed moderate antimicrobial potency, with MICs ranging from 0.9 mg/mL against MRSA to 3.2 mg/mL against P. aeruginosa. The lowest MICs of the oil extract were also observed against MRSA (0.9 mg/mL), C. albicans (1.2 mg/mL), and C. neoformans (1.4 mg/mL), supporting its potential as a lipophilic antimicrobial agent. This enhanced activity is likely associated with the higher abundance of key bioactive compounds identified by GC–MS. In particular, 4-hydroxybutanoic acid (7.76% in aerial vs. 2.34% in root), guaiacol (2.40% vs. 0.72%), and hydroquinone (1.74% vs. 1.35%), all of which are well-supported in the literature for their antimicrobial mechanisms. 4-hydroxybutanoic acid not only exerts central nervous system depressant effects and is used clinically in narcolepsy and alcohol dependence, but also modulates innate immunity by upregulating cathelicidin LL-37, activating GPR109A, and inducing MAP kinase/NF-κB pathways, thereby increasing resistance to microbial infections [38,39]. Guaiacol is recognized for its antiseptic, expectorant, and anti-inflammatory effects, and acts as a local anesthetic and antioxidant that contributes to membrane disruption in pathogens [44]. Hydroquinone, known for its melanin-inhibiting and antioxidant properties, also exhibits broad-spectrum antibacterial and antifungal activities and anti-inflammatory effects [49,67]. While these three compounds were highlighted due to both their abundance and mechanistic relevance, it is also likely that other phytochemicals—such as phenol, isosorbide, and 2-methoxy-4-vinylphenol—act synergistically to enhance the antimicrobial efficacy of the aerial extract. These results are consistent with earlier studies on essential oils and extracts of A. nobilis subspecies, including sipylea and neilreichii, which have shown strong activity against C. albicans, E. faecalis, and P. vulgaris, likely due to the presence of 1,8-cineole, fragranol, linalool, α-bisabolol, and fragranyl acetate [9,15,20,67]. Moreover, antibacterial effects against E. coli, K. pneumoniae, P. aeruginosa, and S. aureus were linked to high levels of α-thujone, artemisia ketone, and camphor [15], while phenolic-rich Soxhlet extracts of A. nobilis subsp. neilreichii demonstrated low MIC values against S. aureus, further supporting the antimicrobial potential of the genus [20].
Compared to other Achillea species, Achillea nobilis demonstrates notable but moderately selective antimicrobial activity, which can be attributed to its major compounds such as α-thujone, fragranol, and fragranyl acetate. However, when contrasted with Achillea clavennae, which exhibited the strongest antimicrobial activity among four tested species (A. clavennae, A. holosericea, A. lingulata, and A. millefolium), the extract of A. clavennae was more potent and yielded structurally diverse compounds including guaiane sesquiterpenes (rupicolin A and B, and their peroxide derivatives) and flavonoids like apigenin and centaureidin—compounds with well-documented antimicrobial and anti-inflammatory properties [68]. Furthermore, A. atrata showed superior bioactivity against both Propionibacterium acnes and S. epidermidis, with significant anti-MRSA potential. Its chemical profile was rich in polar phenolics such as syringetin-3-O-glucoside, mearnsetin-hexoside, and nevadensin, which were absent in A. nobilis [69]. Similarly, A. wilhelmsii, collected from Iran, exhibited a unique chemotype rich in oxygenated monoterpenes, particularly fragranol (33.2%) and fragranyl acetate (16.2%), and showed a broad antimicrobial spectrum including C. albicans, S. aureus, Acinetobacter baumannii, and Shigella dysenteriae. Its inhibitory halos (~9–10 mm) were comparable to those of rifampin, emphasizing its pharmacological potential [70]. While A. nobilis shares some chemical similarities with A. wilhelmsii (e.g., fragranol and fragranyl acetate), its antimicrobial range appears more limited in scope and intensity. Therefore, although A. nobilis exhibits valuable antimicrobial properties, other species such as A. clavennae, A. atrata, and A. wilhelmsii may offer broader or stronger bioactivity due to the presence of distinct sesquiterpenes and phenolic derivatives.
Leveraging the encouraging phytochemical and antimicrobial discoveries related to A. nobilis, our research group plans to continue experimental investigations aimed at enhancing its therapeutic capabilities through various specific strategies. As part of our ongoing and future work, bioassay-guided fractionation of ethanol extracts will be performed to isolate and identify the particular bioactive compounds responsible for the potent antimicrobial effects, specifically against MRSA, C. albicans, and C. neoformans. In vitro cytotoxicity studies and selectivity index evaluations will support these efforts to guarantee safety and therapeutic significance. Moreover, combinatory tests will be conducted to investigate potential synergistic interactions with traditional antimicrobial drugs. Transcriptomic and metabolomic analyses will be utilized to enhance the comprehension of the plant’s bioactivity by exploring the biosynthetic pathways that contribute to the production of pharmacologically important secondary metabolites. Seasonal and environmental factors affecting phytochemical content will be evaluated to determine ideal collection conditions for optimal effectiveness. Sophisticated computational methods like molecular docking and in silico ADMET modeling will be utilized to forecast the drug-likeness and action mechanisms of important compounds. In the end, the development of formulations using standardized extracts or purified components will be investigated for topical and oral therapeutic uses. These studies are part of our ongoing research agenda, and we are committed to validating A. nobilis as a scientifically reliable source of new antimicrobial compounds and promoting its inclusion in contemporary phytopharmaceutical advancement by integrating preclinical efficacy data, pharmacokinetic profiling, and scalable extraction techniques to facilitate its transition from traditional use to evidence-based medical applications.

4. Materials and Methods

4.1. Plant Collection and Identification

On 21 July 2024, plant samples of A. nobilis were methodically gathered from its native environment in the Aktobe region, situated in the western Kazakhstan, a semi-arid zone noted for varied steppe flora. Samples comprised both above-ground structures and root systems, collected specifically at the peak flowering period to guarantee optimal phytochemical yield, since this developmental stage is recognized for having the greatest concentration of secondary metabolites. After collection, all plant materials were thoroughly washed with distilled water to eliminate soil and debris, and then air-dried in the shade at room temperature (22–25 °C) for 10–14 days to protect thermolabile compounds. The dried substance was then ground with a mechanical grinder to achieve a uniform fine powder, which was kept in sealed containers in dark, dry conditions until extraction. This preparatory process was performed following standard pharmacognostic protocols to preserve chemical integrity and guarantee the reproducibility of subsequent phytochemical analyses.

4.2. Chemicals and Reagents

All chemicals and reagents used in this research were of analytical quality to guarantee the consistency and repeatability of experimental findings. Ethanol (96%, v/v) served as the main solvent for extracting phytoconstituents, owing to its effectiveness in dissolving various polar and moderately non-polar substances. For qualitative phytochemical analysis, reagents were prepared following standard procedures: Dragendorff’s reagent (Sigma-Aldrich, St. Louis, MO, USA) was used to identify alkaloids, gelatin solution for tannins, ferric chloride (Sigma-Aldrich, St. Louis, MO, USA) for flavonoids, Liebermann–Burchard reagent (Sigma-Aldrich, St. Louis, MO, USA) for triterpenoids, and the Keller–Killiani test (using glacial acetic acid and ferric chloride from Merck, Darmstadt, Germany) for cardiac glycoside detection. Reagents were freshly made and kept in amber containers under regulated laboratory conditions to avoid photodegradation. For antimicrobial tests, culture media such as Mueller–Hinton Broth (MHB), Mueller–Hinton Agar (MHA), Sabouraud Dextrose Agar (SDA), and RPMI-1640 medium were obtained from HiMedia Laboratories (Mumbai, India), guaranteeing standardization in microbial susceptibility assessments. Dimethyl sulfoxide (DMSO, ≥99.9%, Sigma-Aldrich, St. Louis, MO, USA) was utilized as the solvent to create extract stock solutions for antimicrobial assessment, due to its capacity to dissolve a wide range of organic substances without affecting microbial growth. Helium gas of high purity (99.999%, Linde Gas, Munich, Germany) served as the carrier gas in the gas chromatography–mass spectrometry (GC–MS) analysis, guaranteeing superior resolution and sensitivity. The identification of compounds in GC–MS was aided by comparing spectra with the mass spectral libraries of the National Institute of Standards and Technology (NIST’02, Gaithersburg, MD, USA) and Wiley 7. All solutions and media were prepared with double-distilled water and stored under suitable laboratory conditions (4–8 °C for reagents and 20–25 °C for solvents and media) to maintain their chemical stability and analytical integrity.

4.3. Vortex-Assisted Extraction Method (VAM)

The VAM was utilized to obtain bioactive compounds from botanical sources. In this process, 70 g of finely ground raw materials was utilized, with the solvent comprising 340 mL of 96% ethyl alcohol. The extraction procedure was conducted at room temperature for a period of 110 min. Throughout this period, the vortex mixer enabled swift mixing of the solvent and plant material, aiding in efficient mass transfer and improving the solubility of the desired compounds. The rapid vortex motion guaranteed that the extraction solvent consistently engaged with the raw materials, enhancing the extraction efficiency. Following the extraction phase, the resulting solution underwent filtration with either standard filter paper or a vacuum filtration system to separate the intended extract. The filtrate was gathered, and the leftover solid material was thrown away. Following the filtration of the extract acquired via the VAM procedure, the filtrate underwent pre-concentration with a rotary evaporator at a temperature that did not surpass 40 °C under reduced pressure to eliminate most of the ethanol solvent. This temperature was carefully selected based on prior studies demonstrating that key phytochemical constituents, including flavonoids, triterpenoids, and glycosides, remain stable below 40°C during evaporation, minimizing degradation and ensuring the integrity of thermolabile compounds [71,72]. The concentrated extract was frozen at −80 °C for 12 h to guarantee full solidification. The frozen extract was subjected to freeze-drying (Labconco FreeZone 2.5, Kansas City, MO, USA), enabling solvent sublimation under a vacuum of around 0.05 Mbar at a chamber temperature of −50 °C. Remaining moisture was eliminated during the secondary drying stage at ~20 °C. The obtained powdered extract was kept in airtight containers and safeguarded from moisture for later phytochemical and bioactivity evaluations.

4.4. Oil-Based Maceration Extraction

To obtain the oil extract of the aerial parts of A. nobilis, a cold maceration technique was employed using sunflower oil as the solvent, in accordance with established phytochemical extraction protocols [73,74]. Briefly, 50 g of air-dried and finely powdered aerial plant material was immersed in 500 mL of refined sunflower oil in a sealed glass container. The mixture was kept at room temperature (25 ± 2 °C) for 14 days, protected from direct sunlight, and stirred gently once daily to facilitate diffusion of lipophilic phytochemicals into the solvent. Following the extraction period, the macerate was filtered through muslin cloth and subsequently through Whatman No. 1 filter paper to remove solid residues. The resulting oil extract was stored in amber glass bottles at 4 °C until further phytochemical analysis. This extraction method was specifically chosen to isolate non-polar, lipid-soluble bioactive constituents such as fatty acids, terpenes, sterols, and hydrocarbons, which are not efficiently recovered through ethanol-based extractions. In addition to its chemical relevance, the oil extract also aligns with traditional ethnopharmacological uses of A. nobilis for dermal applications, serving as a practical basis for topical phytotherapeutic formulations.

4.5. Preliminary Qualitative Analysis

The initial phytochemical analysis of A. nobilis aerial and root ethanol extracts was conducted utilizing standardized colorimetric and precipitation assays to qualitatively identify prominent secondary metabolites, as delineated by Barros et al. [75], with minor adaptations. The presence of alcohols was indicated by the emergence of a pronounced blue or green hue upon reaction with ferric chloride, while the presence of aldehydes resulted in a pink-to-magenta coloration upon treatment with Schiff’s reagent. The detection of amines was achieved through the use of ninhydrin reagent, which elicits a purple coloration in the presence of either primary or secondary amines. The confirmation of amides was accomplished by heating the extract with sodium hydroxide, with the liberation of gas or an ammonia-like odor serving as an indicator of a positive outcome. Flavonoids were evaluated utilizing ferric chloride, followed by the addition of hydrochloric acid, where the fleeting yellow coloration that subsequently dissipated validated their presence. The assessment of tannins was conducted using gelatin reagent, where the formation of dirty brownish-green precipitates was indicative of a positive test result. Alkaloids were identified through the development of a reddish-orange precipitate following the addition of Dragendorff’s reagent. The confirmation of triterpenoids was achieved via the Liebermann–Burchard reaction, characterized by the appearance of a brown ring, while glycosides were detected through the Keller–Killiani test, which resulted in a reddish-brown layer. All assays were executed in triplicate, and the results were interpreted visually based on distinct colorimetric changes or the formation of precipitates.

4.6. GC–MS Profiling

GC–MS analyses were performed separately for the ethanol extracts from the aerial and root parts, as well as for the essential oil extracted from the aerial part of A. nobilis. Each sample type was analyzed using optimized conditions tailored to its physicochemical properties to ensure accurate compound separation and identification.
The ethanol extracts of both aerial and root parts were analyzed using a Shimadzu GC–MS system (Shimadzu Corporation, Kyoto, Japan) equipped with a DB-35 MS ultra-inert capillary column (30 m × 0.25 mm, film thickness: 0.25 μm). The oven temperature program was set to begin at 40 °C (held for 3 min), followed by a linear increase to 280 °C at a rate of 5 °C/min, and then held isothermally for an additional 15 min. The injection port was maintained at 280 °C, with helium (99.999%) as the carrier gas at a constant flow rate of 1.4 mL/min. Extracts were diluted to 1% (v/v) and injected in split mode (15:1), with a sample volume of 1 μL. The mass spectrometer operated in electron impact ionization mode (70 eV), with the ion source and interface temperatures set at 220 °C and 280 °C, respectively. Full scan mode was used over the m/z range of 34–750. Compound identification was based on spectral comparison with reference databases (NIST’02 and Wiley 7) using Agilent MSD ChemStation (version 1701EA, Santa Clara, CA, USA).
The aerial oil extract was analyzed using a system configured with an Rtx-100DHA capillary column (30 m × 0.25 mm, film thickness: 0.5 μm, Restek, Bellefonte, PA, USA). The temperature program was initiated at 60 °C and increased to 300 °C at 8 °C/min. The injector temperature was set at 280 °C, while the ion source and quadrupole temperatures were maintained at 230 °C and 150 °C, respectively. Helium served as the carrier gas at a column pressure of 2 psi. A 0.2 μL sample was injected in split mode, and mass spectra were recorded in full scan mode. The total runtime for analysis was 32 min. Compound identification was performed using the NIST 08 spectral library, and data processing was carried out using GS-MSD Data Analysis software (version G1701EA E.02.02.1431, Agilent Technologies, Santa Clara, CA, USA). The relative abundance of compounds was estimated using a semi-quantitative approach based on peak area percentages.

4.7. Antimicrobial Activity

The antimicrobial properties of ethanol extracts derived from the aerial and root components of A. nobilis were assessed against a range of eight clinically significant microbial strains, including both bacterial and fungal pathogens. The organisms tested comprised methicillin-resistant S. aureus (MRSA, ATCC 43300), vancomycin-resistant E. faecalis (VRE, ATCC 51299), E. coli (ATCC 25922), P. aeruginosa (ATCC 27853), K. pneumoniae (ATCC 700603), C. albicans (ATCC 90028), A. fumigatus (ATCC 1022), and C. neoformans (ATCC 208821). The broth microdilution method was employed to ascertain the MICs of the extracts, following the Clinical and Laboratory Standards Institute (CLSI) guidelines released in 2018. Bacterial strains were preserved on MHA, whereas fungal isolates were cultivated on Sabouraud Dextrose Agar (SDA), with all cultures incubated at 37 °C for 24 to 48 h to promote optimal growth. Microbial inocula were adjusted to a turbidity matching 0.5 McFarland standard, which is roughly 1 × 108 colony-forming units per milliliter (CFU/mL), and then diluted to obtain a final working concentration of 1 × 105 CFU/mL in every well. Two-fold serial dilutions of the plant extracts, from 90 to 5000 µg/mL, were made in sterile 96-well U-bottom microplates using MHB for bacterial tests and RPMI-1640 medium for fungal tests [76,77]. Prior to MIC testing, the solubility of the extracts at the highest concentration (5 mg/mL) in the respective media was visually confirmed to ensure complete dissolution and avoid precipitation during the assay. The experimental design featured negative controls (extract in medium absent of microorganisms) and positive controls (medium containing microbial inoculum but lacking extract). After incubation at 35 °C for 24 to 48 h, MIC values were noted as the minimum extract concentrations that entirely prevented observable microbial growth under typical laboratory lighting conditions. This method facilitated a comparative evaluation of the inhibitory capacity of aerial and root-derived extracts against multidrug-resistant and opportunistic pathogens, while the use of RPMI-1640 for fungal MIC determination was based on CLSI M27-A3 [78] guidelines, which recommend this medium for standardizing antifungal susceptibility testing of yeasts and molds [77].

4.8. Statistical Analysis

All data are expressed as mean values accompanied by their corresponding standard deviations (mean ± SD). Statistical significance of the results was determined using Student’s t-test and one-way analysis of variance (ANOVA) to assess differences between experimental groups.

5. Conclusions

This research provides a comprehensive phytochemical and antimicrobial assessment of A. nobilis, incorporating for the first time a comparative analysis of ethanol extracts from both aerial and root parts alongside a macerated aerial oil extract. Through qualitative screening and GC–MS profiling, a chemically diverse array of bioactive secondary metabolites was identified, including phenolics, alcohols, organic acids, lactones, and lipophilic compounds such as sterols, terpenoids, and unsaturated fatty acids. Many of these constituents are known for their antimicrobial, antioxidant, anti-inflammatory, and neuroprotective effects.
Significantly, the aerial ethanol extract demonstrated the most potent antimicrobial activity, particularly against critical pathogens such as MRSA, C. albicans, and C. neoformans, supporting its promising therapeutic relevance. The root ethanol extract, while exhibiting comparatively lower potency, still revealed noteworthy antimicrobial effects, consistent with the traditional holistic use of the entire plant. Importantly, the inclusion of the aerial oil extract revealed a distinct and pharmacologically meaningful profile rich in lipid-soluble compounds, including α-tocopherol, linoleic acid, camphor, and squalene—substances known for their strong antioxidant, anti-inflammatory, and antimicrobial activities. While its antimicrobial activity was moderate compared to the ethanol extract, the oil extract’s chemical profile aligns with its traditional use in topical and dermal applications and suggests specific potential in the development of lipid-based formulations.
Taken together, this study highlights the complementary therapeutic value of both polar and non-polar extracts from A. nobilis and expands the understanding of its bioactive spectrum. The findings are consistent with prior pharmacological reports on A. nobilis and its subspecies, further supporting its ethnomedicinal importance and broad biological potential, including anticonvulsant, antioxidant, antispasmodic, and anti-inflammatory effects. These results not only validate the traditional uses of A. nobilis but also establish a strong foundation for future research focused on bioassay-guided fractionation, elucidation of molecular mechanisms, and the development of both systemic and topical phytotherapeutic products. In conclusion, A. nobilis remains an underexplored medicinal plant with considerable pharmacological promise, meriting continued investigation for its integration into evidence-based herbal medicine and its role in the discovery of novel natural antimicrobial agents.

Author Contributions

Conceptualization, A.B. (Aiman Berdgaleeva), Z.Z., A.S. (Akzharkyn Saginbazarova), and A.S. (Aigul Sartayeva); Methodology, G.T., D.Z., A.B. (Aliya Bazargaliyeva), and Z.K.; Software, Z.K., S.S., A.R., and A.B. (Akzhamal Bilkenova); Validation, Z.K., S.S., A.R., and A.B. (Aliya Bazargaliyeva); Formal analysis, G.T., A.R., and A.B. (Akzhamal Bilkenova); Investigation, A.B. (Aiman Berdgaleeva), Z.Z., A.S. (Akzharkyn Saginbazarova), G.T., and D.Z.; Resources, A.B. (Aliya Bazargaliyeva), Z.K., and S.S.; Data curation, D.Z., A.B. (Aliya Bazargaliyeva), and A.R.; Writing—original draft preparation, A.B. (Aiman Berdgaleeva), Z.Z., A.S. (Akzharkyn Saginbazarova), and G.T.; Writing—review and editing, A.S. (Aigul Sartayeva), S.S., and A.R.; Visualization, Z.K., A.R., and A.B. (Akzhamal Bilkenova); Supervision, A.S. (Aigul Sartayeva), S.S., and A.B. (Aliya Bazargaliyeva); Project administration, A.B. (Aiman Berdgaleeva), A.S. (Aigul Sartayeva), and Z.Z.; Funding acquisition, A.S. (Aigul Sartayeva). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

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. Shevchenko, A.; Akhelova, A.; Nokerbek, S.; Kaldybayeva, A.; Sagyndykova, L.; Raganina, K.; Dossymbekova, R.; Meldebekova, A.; Amirkhanova, A.; Ikhsanov, Y.; et al. Phytochemistry, Pharmacological Potential, and Ethnomedicinal Relevance of Achillea nobilis and Its Subspecies: A Comprehensive Review. Molecules 2025, 30, 2460. [Google Scholar] [CrossRef] [PubMed]
  2. Strzępek-Gomółka, M.; Gaweł-Bęben, K.; Kukula-Koch, W. Achillea species as sources of active phytochemicals for dermatological and cosmetic applications. Oxid. Med. Cell. Longev. 2021, 2021, 6643827. [Google Scholar] [CrossRef]
  3. USDA; NRCS. The PLANTS Database; National Plant Data Team: Greensboro, NC, USA, 2025. Available online: http://plants.usda.gov (accessed on 17 April 2025).
  4. Barda, C.; Grafakou, M.-E.; Tomou, E.-M.; Skaltsa, H. Phytochemistry and Evidence-Based Traditional Uses of the Genus Achillea L.: An Update (2011–2021). Sci. Pharm. 2021, 89, 50. [Google Scholar] [CrossRef]
  5. Solomko, O.V.; Dzhumyrko, S.F.; Kompantsev, V.A. Flavonoids of Achillea nobilis. Chem. Nat. Compd. 1978, 14, 224. [Google Scholar] [CrossRef]
  6. Adekenov, S.M.; Mukhametzhanov, M.N.; Kagarlitskii, A.D.; Turmukhambetov, A.Z. A chemical investigation of Achillea nobilis. Chem. Nat. Compd. 1984, 20, 568–571. [Google Scholar] [CrossRef]
  7. Soliman, G.A.; Abood, S.H.; Hassen, H.F.; El-Kased, R.F. The Potential Anticonvulsant Activity of the Ethanolic Extracts of Achillea nobilis and Momordica charantia in Rats. J. Pharm. Pharmacogn. Res. 2016, 4, 107–114. [Google Scholar] [CrossRef]
  8. Karabay-Yavaşoğlu, N.Ü.; Karamenderes, C.; Baykan, S.; Apaydın, S. Antinociceptive and Anti-Inflammatory Activities and Acute Toxicity of Achillea nobilis subsp. neilreichii Extract in Mice and Rats. Pharm. Biol. 2007, 45, 162–168. [Google Scholar] [CrossRef]
  9. Karamenderes, C.; Apaydın, S. Antispasmodic Effect of Achillea nobilis L. subsp. sipylea (O. Schwarz) Bässler on the Rat Isolated Duodenum. J. Ethnopharmacol. 2003, 84, 175–179. [Google Scholar] [CrossRef]
  10. Konyalıoğlu, S.; Karamenderes, C. The Protective Effects of Achillea L. Species Native in Turkey against H2O2-Induced Oxidative Damage in Human Erythrocytes and Leucocytes. J. Ethnopharmacol. 2005, 102, 221–227. [Google Scholar] [CrossRef]
  11. Trudybekov, K.M.; Turmukhambetov, A.Z.; Adekenov, S.M.; Struchkov, Y.T. Anolide—A new guaianolide from Achillea nobilis. Chem. Nat. Compd. 1994, 30, 460–463. [Google Scholar] [CrossRef]
  12. Kastner, U.; Breuer, J.; Glasl, S.; Baumann, A.; Robien, W.; Jurenitsch, J.; Kubelka, W. Guaianolide-Endoperoxide and Monoterpene-Hydroperoxides from Achillea nobilis. Planta Medica 1995, 61, 83–85. [Google Scholar] [CrossRef] [PubMed]
  13. Turmukhambetov, A.Z.; Buketova, G.K.; Gafurov, N.M.; Adekenov, S.M. Chrysartemin A and canin from Achillea nobilis. Chem. Nat. Compd. 1999, 35, 102. [Google Scholar] [CrossRef]
  14. Krenn, L.; Miron, A.; Pemp, E.; Petr, U.; Kopp, B. Flavonoids from Achillea nobilis L. Z. Naturforsch. C J. Biosci. 2003, 58, 11–16. [Google Scholar] [CrossRef] [PubMed]
  15. Palić, R.; Stojanović, G.; Nasković, T.; Ranelović, N. Composition and Antibacterial Activity of Achillea crithmifolia and Achillea nobilis Essential Oils. J. Essent. Oil Res. 2003, 15, 434–437. [Google Scholar] [CrossRef]
  16. Serkerov, S.V.; Mustafaeva, S.J. Detection of acetyleucanbin in Achillea nobilis. Chem. Nat. Compd. 2010, 46, 666. [Google Scholar] [CrossRef]
  17. Azizi, M.; Chizzola, R.; Ghani, A.; Oroojalian, F. Composition at different development stages of the essential oil of four Achillea species grown in Iran. Nat. Prod. Commun. 2010, 5, 283–290. [Google Scholar] [CrossRef]
  18. Rustaiyan, A.; Masoudi, S.; Ezatpour, L.; Danaii, E.; Taherkhani, M.; Aghajani, Z. Composition of the essential oils of Anthemis hyalina DC., Achillea nobilis L., and Cichorium intybus L.—Three Asteraceae herbs growing wild in Iran. J. Essent. Oil Bear. Plants 2011, 14, 472–480. [Google Scholar] [CrossRef]
  19. Karaalp, C.; Yurtman, A.N.; Karabay Yavasoglu, N.U. Evaluation of antimicrobial properties of Achillea L. flower head extracts. Pharm. Biol. 2009, 47, 86–91. [Google Scholar] [CrossRef]
  20. Taşkın, D.; Taşkın, T.; Rayaman, E. Phenolic composition and biological properties of Achillea nobilis L. subsp. neilreichii (Kerner) Formanek. Ind. Crops Prod. 2018, 111, 555–562. [Google Scholar] [CrossRef]
  21. Ozdemir, F.A. Potential effects of essential oil compositions on antibacterial activities of Achillea nobilis L. subsp. neilreichii. J. Essent. Oil Bear. Plants 2019, 22, 574–580. [Google Scholar] [CrossRef]
  22. Moradi, M.T.; Rafieian-Koupaei, M.; Imani-Rastabi, R.; Nasiri, J.; Shahrani, M.; Rabiei, Z.; Alibabaei, Z. Antispasmodic effects of yarrow (Achillea millefolium L.) extract in the isolated ileum of rat. Afr. J. Tradit. Complement. Altern. Med. 2013, 10, 499–503. [Google Scholar] [CrossRef] [PubMed]
  23. Lu, Y.; Luo, Q.; Cui, H.; Deng, H.; Kuang, P.; Liu, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; et al. Sodium fluoride causes oxidative stress and apoptosis in the mouse liver. Aging 2017, 9, 1623–1639. [Google Scholar] [CrossRef] [PubMed]
  24. Seca, A.M.L.; Pinto, D.C.G.A. Plant Secondary Metabolites as Anticancer Agents: Successes in Clinical Trials and Therapeutic Application. Int. J. Mol. Sci. 2018, 19, 263. [Google Scholar] [CrossRef] [PubMed]
  25. Song, C.W.; Park, J.M.; Chung, S.C.; Lee, S.Y.; Song, H. Microbial production of 2,3-butanediol for industrial applications. J. Ind. Microbiol. Biotechnol. 2019, 46, 1583–1601. [Google Scholar] [CrossRef]
  26. Yang, Z.; Zhang, Z. Recent Advances on Production of 2,3-Butanediol Using Engineered Microbes. Biotechnol. Adv. 2019, 37, 569–578. [Google Scholar] [CrossRef]
  27. Akdemir, H.; Liu, Y.; Zhuang, L.; Zhang, H.; Koffas, M.A. Utilization of Microbial Cocultures for Converting Mixed Substrates to Valuable Bioproducts. Curr. Opin. Microbiol. 2022, 68, 102157. [Google Scholar] [CrossRef]
  28. Becker, L.C.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G., Jr.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of Glycerin as Used in Cosmetics. Int. J. Toxicol. 2019, 38 (Suppl. S3), 6S–22S. [Google Scholar] [CrossRef]
  29. Goldman, E.; Choueiri, T.K.; Mainelis, G.; Ramachandran, G.; Schaffner, D.W. Triethylene Glycol Can Be Predeployed as a Safe Virus-Killing Indoor Air Treatment. J. Infect. Dis. 2022, 226, 2040–2041. [Google Scholar] [CrossRef]
  30. Lee, S.J.; Moon, T.W.; Lee, J. Increases of 2-Furanmethanol and Maltol in Korean Red Ginseng during Explosive Puffing Process. J. Food Sci. 2010, 75, C147–C151. [Google Scholar] [CrossRef]
  31. Dhall, H.; Sikka, P.; Kumar, A.; Mishra, A.K. Recent Advancements and Biological Activities of Aryl Propionic Acid Derivatives: (A Review). Orient. J. Chem. 2016, 32, 1831–1838. [Google Scholar] [CrossRef]
  32. Wang, X.; Fang, Y.; Liang, W.; Wong, C.C.; Qin, H.; Gao, Y.; Liang, M.; Song, L.; Zhang, Y.; Fan, M.; et al. Fusobacterium nucleatum Facilitates Anti-PD-1 Therapy in Microsatellite Stable Colorectal Cancer. Cancer Cell 2024, 42, 1729–1746.e8. [Google Scholar] [CrossRef] [PubMed]
  33. Zhen, Z.; Xi, T.F.; Zheng, Y.F. Surface Modification by Natural Biopolymer Coatings on Magnesium Alloys for Biomedical Applications. In Surface Modification of Magnesium and Its Alloys for Biomedical Applications; Woodhead Publishing: Cambridge, UK, 2015; pp. 301–333. [Google Scholar]
  34. Martiryan, A.I.; Galstyan, A.S.; Tadevosyan, L.G.; Petrosyan, I.A. Synthesis of γ-Hydroxy Acid Hydrazides of a New Structure and Study of Their Antioxidant Properties. Proc. YSU B Chem. Biol. Sci. 2020, 54, 188–195. [Google Scholar] [CrossRef]
  35. Liu, J.; Jiang, Y.; Zhang, Q.; Qin, Y.; Li, K.; Xie, Y.; Zhang, T.; Wang, X.; Yang, X.; Zhang, L.; et al. Linoleic Acid Promotes Mitochondrial Biogenesis and Alleviates Acute Lung Injury. Clin. Respir. J. 2024, 18, e70004. [Google Scholar] [CrossRef] [PubMed]
  36. Nava Lauson, C.B.; Tiberti, S.; Corsetto, P.A.; Conte, F.; Tyagi, P.; Machwirth, M.; Ebert, S.; Loffreda, A.; Scheller, L.; Sheta, D.; et al. Linoleic Acid Potentiates CD8+ T Cell Metabolic Fitness and Antitumor Immunity. Cell Metab. 2023, 35, 633–650.e9. [Google Scholar] [CrossRef]
  37. Okumu, A.; Lu, Y.; Dellos-Nolan, S.; Papa, J.L.; Koci, B.; Cockroft, N.T.; Gallucci, J.; Wozniak, D.J.; Yalowich, J.C.; Mitton-Fry, M.J. Novel Bacterial Topoisomerase Inhibitors Derived from Isomannide. Eur. J. Med. Chem. 2020, 199, 112324. [Google Scholar] [CrossRef]
  38. Pineda Molina, C.; Hussey, G.S.; Eriksson, J.; Shulock, M.A.; Cárdenas Bonilla, L.L.; Giglio, R.M.; Gandhi, R.M.; Sicari, B.M.; Wang, D.; Londono, R.; et al. 4-Hydroxybutyrate Promotes Endogenous Antimicrobial Peptide Expression in Macrophages. Tissue Eng. Part A 2019, 25, 693–706. [Google Scholar] [CrossRef]
  39. Molina, C.P.; Hussey, G.S.; Liu, A.; Eriksson, J.; D’Angelo, W.A.; Badylak, S.F. Role of 4-Hydroxybutyrate in Increased Resistance to Surgical Site Infections Associated with Surgical Meshes. Biomaterials 2021, 267, 120493. [Google Scholar] [CrossRef]
  40. Jarzyński, S.; Rapacz, A.; Dziubina, A.; Pękala, E.; Popiół, J.; Piska, K.; Wojtulewski, S.; Rudolf, B. Mechanochemical Synthesis and Anticonvulsant Activity of 3-Aminopyrrolidine-2,5-dione Derivatives. Biomed. Pharmacother. 2023, 168, 115749. [Google Scholar] [CrossRef]
  41. Mokhtari, M.; Jackson, M.D.; Brown, A.S.; Ackerley, D.F.; Ritson, N.J.; Keyzers, R.A.; Munkacsi, A.B. Bioactivity-Guided Metabolite Profiling of Feijoa (Acca sellowiana) Cultivars Identifies 4-Cyclopentene-1,3-dione as a Potent Antifungal Inhibitor of Chitin Synthesis. J. Agric. Food Chem. 2018, 66, 5531–5539. [Google Scholar] [CrossRef]
  42. Poma, P.; Rigogliuso, S.; Labbozzetta, M.; Carfì Pavia, F.; Carbone, C.; Ma, J.; Cusimano, A.; Notarbartolo, M. Antimigratory Effects of a New NF-κB Inhibitor, (S)-β-Salicyloylamino-α-exo-methylene-γ-butyrolactone, in 2D and 3D Breast Cancer Models. Biomed. Pharmacother. 2024, 180, 117552. [Google Scholar] [CrossRef]
  43. Premkumar, J.; Sampath, P.; Sanjay, R.; Chandrakala, A.; Rajagopal, D. Synthetic Guaiacol Derivatives as Promising Myeloperoxidase Inhibitors Targeting Atherosclerotic Cardiovascular Disease. ChemMedChem 2020, 15, 1187–1199. [Google Scholar] [CrossRef] [PubMed]
  44. Gao, T.; Zhang, Y.; Shi, J.; Mohamed, S.R.; Xu, J.; Liu, X. The Antioxidant Guaiacol Exerts Fungicidal Activity against Fungal Growth and Deoxynivalenol Production in Fusarium graminearum. Front. Microbiol. 2021, 12, 762844. [Google Scholar] [CrossRef] [PubMed]
  45. Azuma, Y.; Ozasa, N.; Ueda, Y.; Takagi, N. Pharmacological Studies on the Anti-Inflammatory Action of Phenolic Compounds. J. Dent. Res. 1986, 65, 53–56. [Google Scholar] [CrossRef] [PubMed]
  46. Lee, J.; Morshidi, N.A.A.B.; Lee, J.; Sim, W.; Kim, J.H. 2-Methoxy-4-vinylphenol Mitigates Malignancy of Cholangiocarcinoma Cells through the Blockade of Sonic Hedgehog Signalling. Biochem. Biophys. Res. Commun. 2025, 754, 151515. [Google Scholar] [CrossRef]
  47. Sathiyamoorthi, E.; Faleye, O.S.; Lee, J.H.; Lee, J. Hydroquinone Derivatives Attenuate Biofilm Formation and Virulence Factor Production in Vibrio spp. Int. J. Food Microbiol. 2023, 384, 109954. [Google Scholar] [CrossRef]
  48. Ma, C.; He, N.; Zhao, Y.; Xia, D.; Wei, J.; Kang, W. Antimicrobial Mechanism of Hydroquinone. Appl. Biochem. Biotechnol. 2019, 189, 1291–1303. [Google Scholar] [CrossRef]
  49. Pałasz, A.; Cież, D. In Search of Uracil Derivatives as Bioactive Agents. Uracils and Fused Uracils: Synthesis, Biological Activity and Applications. Eur. J. Med. Chem. 2015, 97, 582–611. [Google Scholar] [CrossRef]
  50. Benedek, B.; Kopp, B. Achillea millefolium L. s.l.–Is the Anti-inflammatory Activity Mediated by Protease Inhibition? J. Ethnopharmacol. 2007, 113, 312–317. [Google Scholar] [CrossRef]
  51. Gaweł-Bęben, K.; Strzępek-Gomółka, M.; Czop, M.; Sakipova, Z.; Głowniak, K.; Kukula-Koch, W. Achillea millefolium L. and Achillea biebersteinii Afan. Hydroglycolic Extracts–Bioactive Ingredients for Cosmetic Use. Molecules 2020, 25, 3368. [Google Scholar] [CrossRef]
  52. Widyawati, T.; Syahputra, R.A.; Syarifah, S.; Sumantri, I.B. Analysis of Antidiabetic Activity of Squalene via In Silico and In Vivo Assay. Molecules 2023, 28, 3783. [Google Scholar] [CrossRef]
  53. Zhang, L.; Huang, Y.; Duan, X.; Si, H.; Luo, H.; Chen, S.; Liu, L.; He, H.; Wang, Z.; Liao, S. Antifungal Activity and Mechanism of Camphor Derivatives against Rhizoctonia solani: A Promising Alternative Antifungal Agent for Rice Sheath Blight. J. Agric. Food Chem. 2024, 72, 11415–11428. [Google Scholar] [CrossRef] [PubMed]
  54. Sokolova, A.S.; Kovaleva, K.S.; Yarovaya, O.I.; Bormotov, N.I.; Shishkina, L.N.; Serova, O.A.; Sergeev, A.A.; Agafonov, A.P.; Maksuytov, R.A.; Salakhutdinov, N.F. (+)-Camphor and (−)-Borneol Derivatives as Potential Anti-Orthopoxvirus Agents. Arch. Pharm. 2021, 354, e2100038. [Google Scholar] [CrossRef] [PubMed]
  55. Singh, H.; Kumar, R.; Mazumder, A.; Salahuddin; Yadav, R.K.; Chauhan, B.; Abdulah, M.M. Camphor and Menthol as Anticancer Agents: Synthesis, Structure-Activity Relationship and Interaction with Cancer Cell Lines. Anticancer Agents Med. Chem. 2023, 23, 614–623. [Google Scholar] [CrossRef] [PubMed]
  56. Mozirandi, W.; Tagwireyi, D.; Mukanganyama, S. Evaluation of Antimicrobial Activity of Chondrillasterol Isolated from Vernonia adoensis (Asteraceae). BMC Complement. Altern. Med. 2019, 19, 249. [Google Scholar] [CrossRef]
  57. Chen, M.; Ghelfi, M.; Poon, J.F.; Jeon, N.; Boccalon, N.; Rubsamen, M.; Valentino, S.; Mehta, V.; Stamper, M.; Tariq, H.; et al. Antioxidant-Independent Activities of Alpha-Tocopherol. J. Biol. Chem. 2025, 301, 108327. [Google Scholar] [CrossRef]
  58. La Torre, M.E.; Cianciulli, A.; Monda, V.; Monda, M.; Filannino, F.M.; Antonucci, L.; Valenzano, A.; Cibelli, G.; Porro, C.; Messina, G.; et al. α-Tocopherol Protects Lipopolysaccharide-Activated BV2 Microglia. Molecules 2023, 28, 3340. [Google Scholar] [CrossRef]
  59. Uchihara, Y.; Ueda, F.; Tago, K.; Nakazawa, Y.; Ohe, T.; Mashino, T.; Yokota, S.; Kasahara, T.; Tamura, H.; Funakoshi-Tago, M. Alpha-Tocopherol Attenuates the Anti-Tumor Activity of Crizotinib against Cells Transformed by NPM-ALK. PLoS ONE 2017, 12, e0183003. [Google Scholar] [CrossRef]
  60. Kopańska, M.; Batoryna, M.; Banaś-Ząbczyk, A.; Błajda, J.; Lis, M.W. The Effect of α-Tocopherol on the Reduction of Inflammatory Processes and the Negative Effect of Acrylamide. Molecules 2022, 27, 965. [Google Scholar] [CrossRef]
  61. Frański, R.; Beszterda-Buszczak, M. Comment on Villalva et al. Antioxidant, Anti-Inflammatory, and Antibacterial Properties of an Achillea millefolium L. Extract and Its Fractions Obtained by Supercritical Anti-Solvent Fractionation against Helicobacter pylori. Antioxidants 2022, 11, 1849. Antioxidants 2023, 12, 1226. [Google Scholar] [CrossRef]
  62. El-Kalamouni, C.; Venskutonis, P.R.; Zebib, B.; Merah, O.; Raynaud, C.; Talou, T. Antioxidant and Antimicrobial Activities of the Essential Oil of Achillea millefolium L. Grown in France. Medicines 2017, 4, 30. [Google Scholar] [CrossRef]
  63. Yesil-Celiktas, O.; Sevimli, C.; Bedir, E.; Vardar-Sukan, F. Inhibitory Effects of Rosemary Extracts, Carnosic Acid and Rosmarinic Acid on the Growth of Various Human Cancer Cell Lines. Plant Foods Hum. Nutr. 2010, 65, 158–163. [Google Scholar] [CrossRef] [PubMed]
  64. Saeidnia, S.; Gohari, A.; Mokhber-Dezfuli, N.; Kiuchi, F. A review on phytochemistry and medicinal properties of the genus Achillea. Daru 2011, 19, 173–186. [Google Scholar] [PubMed] [PubMed Central]
  65. Sökmen, A.; Sökmen, M.; Daferera, D.; Polissiou, M.; Candan, F.; Unlü, M.; Akpulat, H.A. The in vitro antioxidant and antimicrobial activities of the essential oil and methanol extracts of Achillea biebersteinii Afan. (Asteraceae). Phytother. Res. 2004, 18, 451–456. [Google Scholar] [CrossRef]
  66. Plaskova, A.; Mlcek, J. New insights of the application of water or ethanol-water plant extract rich in active compounds in food. Front. Nutr. 2023, 10, 1118761. [Google Scholar] [CrossRef] [PubMed]
  67. Demirci, F.; Guven, K.; Demirci, B.; Dadalioglu, I.; Baser, K.H.C. Characterization and Biological Activity of Achillea teretifolia Willd. and A. nobilis L. subsp. neilreichii (Kerner) Formanek Essential Oils. Turk. J. Biol. 2009, 33, 129–136. [Google Scholar] [CrossRef]
  68. Stojanović, G.; Radulović, N.; Hashimoto, T.; Palić, R. In Vitro Antimicrobial Activity of Extracts of Four Achillea Species: The Composition of Achillea clavennae L. (Asteraceae) Extract. J. Ethnopharmacol. 2005, 101, 185–190. [Google Scholar] [CrossRef]
  69. Apel, L.; Lorenz, P.; Urban, S.; Sauer, S.; Spring, O.; Stintzing, F.C.; Kammerer, D.R. Phytochemical Characterization of Different Yarrow Species (Achillea sp.) and Investigations into Their Antimicrobial Activity. Z. Naturforsch. C J. Biosci. 2020, 76, 55–65. [Google Scholar] [CrossRef]
  70. Ghavam, M.; Castangia, I.; Manconi, M.; Bacchetta, G.; Manca, M.L. Chemical Composition and Antimicrobial Activity of a Newly Identified Chemotype of Achillea wilhelmsii K. Koch from Kashan, Iran. Sci. Rep. 2024, 14, 22655. [Google Scholar] [CrossRef]
  71. Abubakar, A.R.; Haque, M. Preparation of Medicinal Plants: Basic Extraction and Fractionation Procedures for Experimental Purposes. J. Pharm. Bioallied Sci. 2020, 12, 1–10. [Google Scholar] [CrossRef]
  72. Gazizova, A.; Datkhayev, U.; Amirkhanova, A.; Ustenova, G.; Kozhanova, K.; Ikhsanov, Y.; Kapsalyamova, E.; Kadyrbayeva, G.; Allambergenova, Z.; Kantureyeva, A.; et al. Phytochemical Profiling of Mentha asiatica Boriss. Leaf Extracts: Antioxidant and Antibacterial Activities. ES Food Agrofor. 2025, 19, 1355. [Google Scholar] [CrossRef]
  73. Bitwell, C.; Indra, S.S.; Luke, C.; Kakoma, M.K. A review of modern and conventional extraction techniques and their applications for extracting phytochemicals from plants. Sci. Afr. 2023, 19, e01585. [Google Scholar] [CrossRef]
  74. Azwanida, N.N. A Review on the Extraction Methods Used in Medicinal Plants, Principle, Strength, and Limitation. Med. Aromat. Plants 2015, 4, 196. [Google Scholar] [CrossRef]
  75. Barros, L.; Oliveira, S.; Carvalho, A.M.; Ferreira, I.C. In vitro antioxidant properties and characterisation in nutrients and phytochemicals of six medicinal plants from the Portuguese folk medicine. Ind. Crops Prod. 2010, 32, 572–579. [Google Scholar] [CrossRef]
  76. Wiegand, I.; Hilpert, K.; Hancock, R.E. Agar and Broth Dilution Methods to Determine the Minimal Inhibitory Concentration (MIC) of Antimicrobial Substances. Nat. Protoc. 2008, 3, 163–175. [Google Scholar] [CrossRef]
  77. Balouiri, M.; Sadiki, M.; Ibnsouda, S.K. Methods for In Vitro Evaluating Antimicrobial Activity: A Review. J. Pharm. Anal. 2016, 6, 71–79. [Google Scholar] [CrossRef]
  78. CLSI M27-A3; Reference Method for Broth Dilution Antifungal Susceptibility Testing of Yeasts. Approved Standard-Third Edition; CLSI: Wayne, PA, USA, 2008.
Figure 1. Ethnomedicinal applications and phytochemical discoveries of A. nobilis across cultures and time [1,5,6,11,12].
Figure 1. Ethnomedicinal applications and phytochemical discoveries of A. nobilis across cultures and time [1,5,6,11,12].
Molecules 30 02957 g001
Figure 2. GC–MS chromatogram of aerial ethanol extract constituents of A. nobilis.
Figure 2. GC–MS chromatogram of aerial ethanol extract constituents of A. nobilis.
Molecules 30 02957 g002
Figure 3. GC–MS chromatogram of the root ethanol extract of A. nobilis.
Figure 3. GC–MS chromatogram of the root ethanol extract of A. nobilis.
Molecules 30 02957 g003
Figure 4. GC–MS chromatogram of the aerial oil extract of A. nobilis.
Figure 4. GC–MS chromatogram of the aerial oil extract of A. nobilis.
Molecules 30 02957 g004
Figure 5. Chemical structures of common compounds identified in the aerial and root extracts of A. nobilis. Here, the numbering follows the one used in Table 6.
Figure 5. Chemical structures of common compounds identified in the aerial and root extracts of A. nobilis. Here, the numbering follows the one used in Table 6.
Molecules 30 02957 g005aMolecules 30 02957 g005bMolecules 30 02957 g005c
Table 1. Phytochemical screening of aerial and root extracts of A. nobilis.
Table 1. Phytochemical screening of aerial and root extracts of A. nobilis.
Phytochemical GroupsReagentObservationAerial Ethanol Extract Aerial Oil ExtractRoot Ethanol Extract
AlcoholsFerric chlorideIntense blue or green coloration+++
AldehydesSchiff’s reagentPink to magenta color formation+++
AminesNinhydrin reagentPurple coloration++
AmidesSodium hydroxide + heatAmmonia-like smell or evolution of gas++
FlavonoidsFerric chlorideYellowish appearance clears after acid (HCl) addition++
TanninsGelatinDirty (brownish) green precipitates+
AlkaloidsDragendorff’sReddish-orange precipitate++
TriterpenoidsLiebermann–BurchardBrown ring+++
GlycosidesKeller–KillianiReddish-brown layer++
“+” indicates the confirmed presence, while “–“ denotes the absence of the respective phytochemical group in the aerial and root extracts of A. nobilis based on preliminary phytochemical screening.
Table 2. Characterization of aerial ethanol extract constituents of A. nobilis through the GC–MS technique.
Table 2. Characterization of aerial ethanol extract constituents of A. nobilis through the GC–MS technique.
No.NameMolecular FormulaMolecular Mass, g/molRetention Indices (RI)Retention Time (min)PubChem
Compound CID
SimilaritiesArea, %
1Propanoic acidC3H6O274.0887810.581032930.92
22,3-ButanediolC4H10O290.12105511.40262966.08
3R-(–)-1,2-propanediolC3H8O276.0994611.62259994910.89
4Gamma-ButyrolactoneC4H6O286.0996012.327302960.95
5Butanoic acidC4H8O288.1195012.40264951.89
64-Hydroxybutanoic acidC4H8O3104.1102612.9910413967.76
7(L)-α-TerpineolC10H18O154.25119514.22443162921.53
8N-NitrosohexamethyleneimineC6H12N2O128.17112014.3513613900.19
91,2-CyclopentanedioneC5H6O298.198515.28566657890.54
10Methyl N-hydroxybenzenecarboximidateC8H9NO2151.16121015.589602988982.69
11GuaiacolC7H8O2124.14110517.01460852.40
12Ethanol, 2,2’-oxybis-C4H10O3106.12103519.14161927871.82
13PhenolC6H6O94.11108019.77996902.01
14Phenol, 4-ethyl-2-methoxy-C9H12O2152.19125520.3562465871.08
152-PyrrolidinoneC4H7NO85.1101020.6512025973.22
161,3-PropanediolC3H8O276.0992022.5710442952.20
17EugenolC10H12O2164.2135622.753314871.49
182-Methoxy-4-vinylphenolC9H10O2150.17132023.43332853.26
19DianhydromannitolC6H10O4146.14135023.6623619611951.07
20Phenol, 2,6-dimethoxy-C8H10O3154.16128024.667041852.85
21GlycerinC3H8O392.09115025.047538615.97
22Triethylene glycolC6H14O4150.17117525.338172912.80
231,4:3,6-Dianhydro- α-d-glucopyranoseC6H8O4144.13138526.3522213879871.76
24Benzofuran, 2,3-dihydro-C8H8O120.15115026.7510329880.60
255-tert-ButylpyrogallolC10H14O3182.22140027.08597592830.60
26SuccinimideC4H5NO299.0999027.6211439940.48
27IsosorbideC6H10O4146.14131029.5312597907.91
283’,5’-DimethoxyacetophenoneC10H12O3180.2145029.7395997862.54
29Tetraethylene glycolC8H18O5194.23122531.208200963.22
303-Methyl-4-phenyl-1H-pyrroleC11H11N157.21142031.9815164561850.61
31HydroquinoneC6H6O2110.11115535.97785891.74
323-Isobutylhexahydropyrrolo [1,2-a]pyrazine-1,4-dioneC11H18N2O2210.27132536.69102892871.27
33Hexaethylene glycolC12H26O7282.33127537.1817472883.56
34Octadecanoic acidC18H36O2284.48198037.755281931.42
35Pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)-C10H16N2O2196.25135038.6598951842.19
36β-D-Glucopyranose, 1,6-anhydro-C6H10O5162.14140039.6611947765831.39
37Pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-C7H10N2O2154.17129541.55193540841.07
384-MethyleneprolineC6H9NO2127.14123045.85558375865.63
Table 3. GC–MS identification of phytochemicals in the root ethanol extract of A. nobilis.
Table 3. GC–MS identification of phytochemicals in the root ethanol extract of A. nobilis.
No.NameMolecular FormulaMolecular Mass, g/molRetention Indices (RI)Retention Time (min)PubChem
Compound CID
SimilaritiesArea, %
1Propanoic acidC3H6O274.0888010.521032850.96
24-Cyclopentene-1,3-dioneC5H4O296.0890011.2770258800.81
34-Hydroxybutanoic acidC4H8O3104.1092012.2210413822.34
42-Propenoic acidC3H4O272.0694012.486581851.16
52-FuranmethanolC5H6O298.1096012.927361861.14
62(5H)-Furanone, 3-methyl-C5H6O298.1098014.1630945820.39
72,4-Dimethyl-2-oxazoline-4-methanolC6H11NO2129.16100014.2698073803.01
82(5H)-FuranoneC4H4O284.07102014.6910341820.97
91,2-CyclopentanedioneC5H6O298.1104015.16566657852.55
101,2-Cyclopentanedione, 3-methyl-C6H8O2112.13106016.5461209940.69
11GuaiacolC7H8O2124.14108016.98460880.72
12Benzaldehyde, 3-hydroxy-, oximeC7H7NO2137.14110017.499603073866.65
132-Cyclopenten-1-one, 3-ethyl-2-hydroxy-C7H10O2126.15112017.8462752820.54
14Ethanone, 1-(1H-pyrrol-2-yl)-C6H7NO109.13114019.1414079800.45
15PhenolC6H6O94.11116019.75996820.61
162-PyrrolidinoneC4H7NO85.10118020.6812025850.65
172(3H)-Furanone, 5-heptyldihydro-C11H18O2182.26120020.867714940.70
18Cyclopropyl carbinolC4H8O72.11122021.0475644881.06
191,3-Dioxol-2-one,4,5-dimethyl-C5H6O3114.10124021.79142210901.39
20α-Hydroxy-gamma-butyrolactoneC4H6O3102.09126022.5319444851.26
21Allyl acetateC5H8O2100.12128022.6111584800.85
222-Methoxy-4-vinylphenolC9H10O2150.17130023.38332821.10
232,3-DimethylhydroquinoneC8H10O2138.17132023.9569100850.41
244H-Pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6-methyl-C6H6O4142.11134024.12119838949.39
251,2,3-Propanetriol, 1-acetateC5H10O4134.13136024.5833510880.42
26Phenol, 2,6-dimethoxy-C8H10O3154.16138024.657041801.57
27GlycerinC3H8O392.09140025.217538226.73
28Triethylene glycolC6H14O4150.17142025.368172851.05
291,4:3,6-Dianhydro-α-d-glucopyranoseC6H8O4144.13144026.3539923607940.40
30Benzofuran, 2,3-dihydro-C8H8O120.15146026.7210329850.41
31AcetaminophenC8H9NO2151.16148026.911983880.09
32Benzoic acid, 3-pyridyl esterC12H9NO2199.20150027.10569697871.88
33SuccinimideC4H5NO299.09152027.5911439760.34
34(S)-(+)-2’,3’-DideoxyribonolactoneC4H6O3102.09154027.7632780890.96
352-Aminopyrimidine-1-oxideC4H5N3O111.10156028.04139694800.53
362,5-Dimethyl-4-phenylpyridineC13H13N183.25158028.64603086820.24
373-Pyridinol, 6-methyl-C6H7NO109.13160028.8114275850.32
38Butyl 9-decenoateC14H26O2226.36162029.1717825102941.58
39IsosorbideC6H10O4146.14164029.5412597880.31
403’,5’-DimethoxyacetophenoneC10H12O3180.20166029.6895997809.97
41DL-Proline, 5-oxo-, methyl esterC6H9NO3143.14168029.99500249820.12
42Tetraethylene glycolC8H18O5194.23170031.208200851.95
433-Methyl-4-phenyl-1H-pyrroleC11H11N157.21172031.9715164561860.48
443-(1H-Pyrrol-3-yl)propionic acid, methyl esterC8H11NO2153.18174032.09556813920.46
452,6-Dimethylphenyl isocyanateC9H9NO147.17176032.1998787860.46
46Ethyl N-(o-anisyl)formimidateC10H13NO2179.22178032.43601627900.38
472-NaphthalenamineC10H9N143.18180033.407057830.19
48Benzaldehyde, 4-hydroxy-3,5-dimethoxy-C9H10O4182.17182035.068655980.38
49HydroquinoneC6H6O2110.11184035.95785931.35
50Pentaethylene glycolC10H22O6238.28186037.1862551890.50
51Uric acidC5H4N4O3168.11188037.831175930.70
52Pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)-C11H18N2O2210.27190037.9998951861.55
534-((1E)-3-Hydroxy-1-propenyl)-2-methoxyphenolC10H12O3180.20192039.021549095880.59
54β-D-Glucopyranose, 1,6-anhydro-C6H10O5162.14194039.6611947765890.33
55Pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-C7H10N2O2154.17196041.53193540930.78
56Hexaethylene glycolC12H26O7282.33198042.4317472860.45
57UracilC4H4N2O2112.09200045.191174880.56
584-MethyleneprolineC6H9NO2127.14202045.88558375982.25
Table 4. Characterization of aerial oil extract constituents of A. nobilis through the GC–MS technique.
Table 4. Characterization of aerial oil extract constituents of A. nobilis through the GC–MS technique.
No.NameMolecular FormulaMolecular Mass, g/molRetention Indices (RI)Retention Time (min)PubChem
Compound CID
SimilaritiesArea, %
1CamphorC10H16O152.23117310.072537931.41
22,4-DecadienalC10H16O152.23128012.615283349960.46
32-Furanacetaldehyde, α-propyl-C10H12O2152.19107612.99557292910.69
43-(Hydroxymethylene)indolin-2-oneC9H7NO2161.16149016.21595118960.66
5HedycaryolC15H26O222.37153418.676432240950.70
66-Methyl-2,4(1H,3H)-pteridinedioneC7H6N4O2178.15159020.27601068960.57
7Tridecanoic acid, methyl esterC4H28O2228.37160822.0315608920.92
8Hexadecanoic acid, ethyl esterC18H36O2284.50199422.8712366861.15
911,14-Octadecadienoic acid, methyl esterC19H34O2294.50208924.03 5365677 900.68
104-Methyl-3-pentenalC6H10O98.1494224.13 21457 920.60
116-TetradecyneC14H26194.36139524.431380278714.99
12Linoleic acidC18H32O2280.40209524.52 5280450 858.08
13Linoleic acid ethyl esterC20H36O2308.50214524.80 5282184 931.75
14 Oleic acid ethyl ester C20H38O2310.50217524.89 5363269 882.28
152,2,2-Trifluoro-N-(hydroxymethyl)acetamideC3H4F3NO2143.06219725.66 3084931 953.35
16Palmitoyl chlorideC16H34ClO274.90225626.19 8206 912.50
17α -Tocopherol C29H50O2430.70310027.26 14985 868.36
18E,Z-1,3,12-NonadecatrieneC19H34262.50191627.44 5365680 8822.25
19Oleic acid, 3-hydroxypropyl esterC21H40O3340.50207627.51 5352775 906.92
202-DodecylcyclobutanoneC16H30O238.41160027.77 161875 901.24
21Isopropyl linoleateC21H38O2322.50215027.96 5352860 8114.64
22cis-13,16-Docasadienoic acidC22H40O2336.60256628.28 5312554 911.42
231-(Trimethylsilyl)-1-propyneC6H12Si112.240250930.71 80363 850.49
24ChondrillasterolC29H48O412.70338030.86 5283663 891.54
25SqualeneC30H50410.70281431.65 638072 902.35
Table 5. Identified phytochemicals in aerial and root extracts of A. nobilis.
Table 5. Identified phytochemicals in aerial and root extracts of A. nobilis.
No.Chemical ClassSubclassNameKnown Pharmacological Activities
1.AlcoholDiol2,3-Butanediol aAntitumor activity, immunomodulatory effects, cryoprotective agent, solvent and drug carrier, anti-inflammatory properties, neuroprotective effects, probiotic metabolite potential, inhibitory effect on certain pathogens [26,27]
2.DiolR-(–)-1,2-propanediol a
3.Diol1,3-Propanediol aSolvent and drug delivery agent, moisturizing and humectant properties, stabilizing agent [28]
4.Monoterpene alcohol(L)-α-Terpineol a
5.TriolGlycerin a,rHumectant and lubricant [29]
6.Polyether diolTriethylene glycol a,rAntimicrobial activity, antiviral activity, disinfectant properties, low toxicity profile, plasticizer and solvent in pharmaceuticals, air sanitizing agent, humectant in topical formulations [30]
7.Polyether diolTetraethylene glycol a,r
8.Polyether diolHexaethylene glycol a,r
9.Furan derivative2-Furanmethanol rAntimicrobial, antifungal, and anticancer [31]
10.Cyclopropyl alcoholCyclopropyl carbinol r
11.PolyetherPentaethylene glycol r
12.AldehydeOximeBenzaldehyde, 3-hydroxy-,
oxime r
13.α,β-Unsaturated aliphatic aldehyde2,4-Decadienal b
14.α,β-Unsaturated aldehyde4-Methyl-3-pentenal b
15.Phenolic aldehydeBenzaldehyde, 4-hydroxy-3,5-dimethoxy- r
16. Furan derivative2-Furanacetaldehyde, α-propyl- b
17.Aromatic compoundBenzofuran derivativeBenzofuran, 2,3-dihydro- a,r
18.Amino acidNon-proteinogenic amino acid4-Methyleneproline b
19.Amino acid derivativeLactam esterDL-Proline, 5-oxo-, methyl ester r
20.AmineAromatic amine2-Naphthalenamine r
21.AmideAniline derivativeAcetaminophen r
22.Trifluoroacetamide derivative2,2,2-Trifluoro-N-(hydroxymethyl)acetamide b
23.Carboxylic acidShort-chain fatty acidPropanoic acid a,rAntimicrobial activity, anti-inflammatory properties, anticancer potential, lipid metabolism regulation, gut microbiota modulation, histone deacetylase (HDAC) inhibition, immune response modulation [32]
24.Short-chain fatty acidButanoic acid aHDAC inhibition [33]
25.Alpha, beta-unsaturated acid2-Propenoic acid r
26.Saturated fatty acidOctadecanoic acid aAntimicrobial activity, anti-inflammatory properties, antioxidant activity, anticancer potential, emollient and skin-conditioning agent, cholesterol-lowering effects, immune-modulating activity [34]
27.Polyunsaturated fatty acidLinoleic acid bAnti-proliferative, anti-invasive, pro-apoptotic, cell cycle arrest (G1 phase), ROS-inducing, mitochondrial membrane potential disruption, anti-inflammatory, antioxidant, epithelial-mesenchymal transition (EMT) inhibition, angiogenesis inhibition, immune modulation, mitochondrial biogenesis stimulation, PGC-1α/NRF1/TFAM pathway activation [34,35]
28.Polyunsaturated fatty acidcis-13,16-Docasadienoic acid b
29.γ-hydroxy acid4-hydroxybutanoic acid a,rCNS depressant activity, sedative effects, anesthetic properties, muscle relaxant, euphoric effects [36], treatment of narcolepsy, treatment of alcohol dependence, potential neuroprotective effects [37], upregulation of expression of the Cramp gene (encoding cathelicidin LL-37) in murine bone marrow-derived macrophages [38], promotion of endogenous antimicrobial peptide expression in macrophages [39]
30.Carboxylic acid esterPyrrole derivative3-(1H-Pyrrol-3-yl)propionic acid, methyl ester r
31. Saturated methyl esterTridecanoic acid, methyl ester b
32. Saturated ethyl esterHexadecanoic acid, ethyl ester b
33. Polyunsaturated methyl ester11,14-Octadecadienoic acid, methyl ester b
34. Polyunsaturated ethyl esterLinoleic acid ethyl ester b
35. Monounsaturated ethyl esterOleic acid ethyl ester b
36. Polyunsaturated esterIsopropyl linoleate b
37. Monounsaturated esterOleic acid, 3-hydroxypropyl ester b
38.CarbohydrateDianhydrosugar alcoholDianhydromannitol aDiuretic activity, osmotic laxative effect, low toxicity, potential use as a pharmaceutical excipient, stabilizing agent, osmoprotective properties [40]
39.Sugar derivative1,4:3,6-Dianhydro-α-d-glucopyranose a,r
40.Sugar alcohol derivativeIsosorbide a,r
41.Monosaccharide derivativeβ-D-Glucopyranose, 1,6-anhydro- a,r
42.CarbonateDioxolone1,3-Dioxol-2-one,4,5-dimethyl- r
43.EtherDiol etherEthanol, 2,2’-oxybis- a
44.Allyl esterAllyl acetate r
45.Acylglycerol1,2,3-Propanetriol, 1-acetate r
46.Pyridine esterBenzoic acid, 3-pyridyl ester r
47.Fatty acid esterButyl 9-decenoate r
48.HeterocycleLactam2-Pyrrolidinone a,r
49.Pyrrole derivative3-Methyl-4-phenyl-1H-pyrrole a,r
50.Lactam3- Isobutylhexahydropyrrolo[1,2-a]pyrazine-1,4-dione a
51.LactamPyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)- a,r
52.LactamPyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro- a,r
53.Oxazoline derivative2,4-Dimethyl-2-oxazoline-4-methanol r
54.Pyrimidine derivative2-Aminopyrimidine-1-oxide r
55.Pyridine derivative2,5-Dimethyl-4-phenylpyridine r
56. Indole derivative3-(Hydroxymethylene)indolin-2-one b
57. Pteridine derivative6-Methyl-2,4(1H,3H)-pteridinedione b
58.ImidateAromatic imidateMethyl N-hydroxybenzenecarboximidate a
59.Aromatic imidateEthyl N-(o-anisyl)formimidate r
60.ImideCyclic imideSuccinimide a,rAnticonvulsant activity, antiepileptic effects, central nervous system depressant, muscle relaxant properties, sedative effects, enzyme inhibitor potential [41]
61.IsocyanateAromatic isocyanate2,6-Dimethylphenyl isocyanate r
62.KetoneDiketone1,2-Cyclopentanedione a
63.Acetophenone derivative3’,5’-Dimethoxyacetophenone a,r
64.Cyclic diketone4-Cyclopentene-1,3-dione rAntifungal [42]
65.Cyclic diketone1,2-Cyclopentanedione r
66.Cyclic diketone1,2-Cyclopentanedione, 3-methyl- r
67.Hydroxycyclopentenone2-Cyclopenten-1-one, 3-ethyl-2-hydroxy- r
68.Aryl ketoneEthanone, 1-(1H-pyrrol-2-yl)- r
69. Cyclobutanone derivative2-Dodecylcyclobutanone b
70.LactoneCyclic esterγ-Butyrolactone aCNS depressant activity, sedative and hypnotic effects, anxiolytic properties, anesthetic effects, muscle relaxant, prodrug of gamma-hydroxybutyric acid (GHB), treatment of narcolepsy (via GHB), potential abuse and dependence liability [43]
71.Furanone2(5H)-Furanone, 3-methyl- r
72.Furanone derivative2(3H)-Furanone, 5-heptyldihydro- r
73.Furanone2(5H)-Furanone r
74.Hydroxybutyrolactone-Hydroxy-γ-butyrolactone r
75.Sugar lactone(S)-(+)-2’,3’-Dideoxyribonolactone r
76.NitrosoamineCyclic nitrosamineN-Nitrosohexamethyleneimine a
77.PhenolMethoxyphenolGuaiacol a,rExpectorant activity, antiseptic, analgesic, anti-inflammatory, antioxidant, local anesthetic, antimicrobial [44]
78.Allyl-substituted methoxyphenolEugenol aAntibacterial, antiviral, antifungal, anticancer, anti-inflammatory and antioxidant [45]
79.MonohydroxybenzenePhenol a,rAntiseptic, anesthetic, antibacterial, antifungal, antiparasitic, disinfectant, cauterizing agent, local analgesic [46]
80.Alkylated methoxyphenolPhenol, 4-ethyl-2-methoxy- a
81.Vinyl-substituted methoxyphenol2-Methoxy-4-vinylphenol a,rAntioxidant, anti-inflammatory, antimicrobial, anticancer, antiplatelet, hepatoprotective, cytoprotective [47]
82.DimethoxyphenolPhenol, 2,6-dimethoxy- a,r
83.Trihydroxybenzene derivative5-tert-Butylpyrogallol a
84.DihydroxybenzeneHydroquinone a,rSkin depigmenting agent, antioxidant activity, anticancer potential, antibacterial activity, antifungal activity, melanin synthesis inhibition, anti-inflammatory [47,48]
85.Hydroquinone derivative2,3-Dimethylhydroquinone rAntioxidant activity, antimicrobial activity, cytotoxic effects, potential anticancer activity, redox-modulating [49]
86.Pyridinol3-Pyridinol, 6-methyl- r
87.Lignan derivative4-((1E)-3-Hydroxy-1-propenyl)-2-methoxyphenol r
88.PyranoneHydroxypyranone4H-Pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6-methyl- r
89.Purine derivativeHeterocyclic compoundUric acid rPotential biomarker for cardiovascular and metabolic disorders, pro-inflammatory effects in hyperuricemia, crystal-induced inflammation (e.g., gout) [50]
90.Pyrimidine baseNucleobaseUracil rAntiviral, anticancer, enzyme inhibition, involvement in DNA/RNA synthesis, radiosensitizing, antimicrobial, immunomodulatory [51]
91.HydrocarbonAlkyne6-Tetradecyne b
92. Polyunsaturated alkeneE,Z-1,3,12-Nonadecatriene b
93.TerpeneTriterpeneSqualene bAnticancer, antiinflammatory, antioxidant, and antidiabetic [52]
94. TerpenoidCamphor bAntifungal, antiviral, and anticancer pharmacological activities, including strong inhibitory effects against Rhizoctonia solani and other fungal pathogens [53], antiviral activity against orthopoxviruses [54], and cytotoxic effects on various cancer cell lines via modulation of cellular pathways and structure–activity relationships [55]
95. TerpenoidHedycaryol b
96.PhytosterolSterolChondrillasterol bAntibacterial activity against Staphylococcus aureus (25% inhibition), Klebsiella pneumoniae (38% inhibition), and Pseudomonas aeruginosa (65% inhibition); complete biofilm disruption of P. aeruginosa at 1.6 μg/mL; complete inhibition of biofilm formation at 100 μg/mL [56]
97.VitaminTocopherol antioxidantα-Tocopherol bAntioxidant activity, anti-inflammatory effects, gene-regulatory activity, neuroprotective activity, cytoprotective effects, mitochondrial protection, anti-apoptotic effects, modulation of signal transduction pathways, suppression of endoplasmic reticulum stress, selective protection of non-cancerous cells during chemotherapy, attenuation of drug-induced cytotoxicity, modulation of lipid metabolism, reduction of hepatic steatosis, prevention of nonalcoholic steatohepatitis progression, interference with anti-cancer drug efficacy [57,58,59,60]
98.OrganosiliconSilylated alkyne1-(Trimethylsilyl)-1-propyne b
99.Acyl chlorideFatty acid derivativePalmitoyl chloride b
Here, “a” indicates compounds identified from the aerial ethanol extract, “b” refers to compounds from the aerial oil extract, “r” represents compounds from the root ethanol extract, “a,r” represents compounds from both the aerial and root ethanol extracts, and “–“ denotes a lack of reported pharmacological activity.
Table 6. Common phytochemicals in ethanol extracts and major constituents of the aerial oil extract of A. nobilis.
Table 6. Common phytochemicals in ethanol extracts and major constituents of the aerial oil extract of A. nobilis.
Comp.NameAerial Ethanol ExtractRoot Ethanol ExtractComp.NameAerial Oil Extract
Area, %Area, %Area, %
1Propanoic acid0.920.9623Camphor1.41
24-hydroxybutanoic acid7.762.3424Linoleic acid8.08
3Guaiacol2.400.72252,2,2-Trifluoro-N-(hydroxymethyl)acetamide3.35
4Phenol2.010.6126Hexadecanoic acid, ethyl ester1.15
52-Pyrrolidinone3.220.65276-Tetradecyne14.99
62-Methoxy-4-vinylphenol3.261.1028Palmitoyl chloride2.50
7Phenol, 2,6-dimethoxy-2.851.5729Linoleic acid ethyl ester1.75
8Glycerin15.9726.7330 Oleic acid ethyl ester 2.28
9Triethylene glycol2.801.0531cis-13,16-Docasadienoic acid1.42
101,4:3,6-Dianhydro-α-d-glucopyranose1.760.4032α -Tocopherol 8.36
11Benzofuran, 2,3-dihydro-0.600.4133E,Z-1,3,12-Nonadecatriene22.25
12Succinimide0.480.3434Isopropyl linoleate14.64
13Isosorbide7.910.3135Oleic acid, 3-hydroxypropyl ester6.92
143’,5’-Dimethoxyacetophenone2.549.97362-Dodecylcyclobutanone1.24
15Tetraethylene glycol3.221.9537Chondrillasterol1.54
163-Methyl-4-phenyl-1H-pyrrole0.610.4838Squalene2.35
17Hydroquinone1.741.35
18Hexaethylene glycol3.560.45
19Pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)-2.191.55
20β-D-Glucopyranose, 1,6-anhydro-1.390.33
21Pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-1.070.78
224-Methyleneproline5.632.25
Table 7. MICs of extracts against various microbial strains.
Table 7. MICs of extracts against various microbial strains.
MicroorganismsAerial Ethanol Extract, mg/mLAerial Oil Extract, mg/mLRoot Ethanol Extract, mg/mL
C. albicans0.751.201.25
A. fumigatus1.502.502.00
C. neoformans0.851.401.50
MRSA0.500.901.00
E. coli1.001.501.80
P. aeruginosa2.003.203.00
K. pneumoniae1.202.102.00
VRE1.001.601.80
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

Berdgaleeva, A.; Zhalimova, Z.; Saginbazarova, A.; Tulegenova, G.; Zharylkassynova, D.; Bazargaliyeva, A.; Kuanbay, Z.; Sakhanova, S.; Ramazanova, A.; Bilkenova, A.; et al. Comparative Phytochemical Analysis and Antimicrobial Properties of Ethanol and Macerated Extracts from Aerial and Root Parts of Achillea nobilis. Molecules 2025, 30, 2957. https://doi.org/10.3390/molecules30142957

AMA Style

Berdgaleeva A, Zhalimova Z, Saginbazarova A, Tulegenova G, Zharylkassynova D, Bazargaliyeva A, Kuanbay Z, Sakhanova S, Ramazanova A, Bilkenova A, et al. Comparative Phytochemical Analysis and Antimicrobial Properties of Ethanol and Macerated Extracts from Aerial and Root Parts of Achillea nobilis. Molecules. 2025; 30(14):2957. https://doi.org/10.3390/molecules30142957

Chicago/Turabian Style

Berdgaleeva, Aiman, Zere Zhalimova, Akzharkyn Saginbazarova, Gulbanu Tulegenova, Dana Zharylkassynova, Aliya Bazargaliyeva, Zhaidargul Kuanbay, Svetlana Sakhanova, Akmaral Ramazanova, Akzhamal Bilkenova, and et al. 2025. "Comparative Phytochemical Analysis and Antimicrobial Properties of Ethanol and Macerated Extracts from Aerial and Root Parts of Achillea nobilis" Molecules 30, no. 14: 2957. https://doi.org/10.3390/molecules30142957

APA Style

Berdgaleeva, A., Zhalimova, Z., Saginbazarova, A., Tulegenova, G., Zharylkassynova, D., Bazargaliyeva, A., Kuanbay, Z., Sakhanova, S., Ramazanova, A., Bilkenova, A., & Sartayeva, A. (2025). Comparative Phytochemical Analysis and Antimicrobial Properties of Ethanol and Macerated Extracts from Aerial and Root Parts of Achillea nobilis. Molecules, 30(14), 2957. https://doi.org/10.3390/molecules30142957

Article Metrics

Back to TopTop