Next Article in Journal
Metabarcoding Analysis Reveals Microbial Diversity and Potential Soilborne Pathogens Associated with Almond Dieback and Decline
Previous Article in Journal
Change in C, N, and P Characteristics of Hypericum kouytchense Organs in Response to Altitude Gradients in Karst Regions of SW China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Profiling Hydrophilic Cucurbita pepo Seed Extracts: A Study of European Cultivar Variability

1
Department of Pharmacognosy-Phytotherapy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Str. 16, 700115 Iasi, Romania
2
Max Zeller & Söhne AG, Seeblickstrasse 4, 8590 Romanshorn, Switzerland
*
Authors to whom correspondence should be addressed.
Plants 2025, 14(15), 2308; https://doi.org/10.3390/plants14152308
Submission received: 2 June 2025 / Revised: 20 July 2025 / Accepted: 21 July 2025 / Published: 26 July 2025
(This article belongs to the Section Phytochemistry)

Abstract

Cucurbita pepo (CP) seeds are traditionally used to alleviate lower urinary tract symptoms associated with benign prostatic hyperplasia and overactive bladder. While these effects are often attributed to lipophilic constituents, recent studies have highlighted the therapeutic potential of oil-free hydroethanolic extracts. However, their composition remains insufficiently characterized, considering the species’ significant phenotypic and phytochemical variability. This study aimed to characterize the phytochemical profile of hydrophilic hydroethanolic seed extracts from ten CP cultivars originating from different European regions, with a focus on compositional variability. The elemental composition, along with primary and secondary metabolites, was analyzed using established spectroscopic and chromatographic methods. The extracts showed considerable variation in protein (45.39 to 114.58 mg/g dw) and free amino acid content (46.51 to 111.10 mg/g dw), as well as differences in elemental composition. Principal component analysis revealed distinct clustering patterns, with several samples displaying metabolite profiles comparable to the Cucurbita pepo var. styriaca variety currently recommended by the European Pharmacopoeia (Ph. Eur.) and the Committee on Herbal Medicinal Products (HMPC). These findings open the possibility of using other CP varieties as alternative sources for extract preparation and offer novel insights into the composition of less explored hydrophilic extracts derived from CP seeds.

1. Introduction

Cucurbita pepo L. (CP), a versatile member of the Cucurbitaceae family, originating from Mexico [1], is known for its wide range of morphological varieties including pumpkins, squashes, and zucchini [2]. The plant is cultivated globally for culinary applications of both fruits and seeds [3], but it has gathered significant interest over the years, particularly for its therapeutic potential [4]. Pumpkin flesh and seed extracts have been investigated for their antidiabetic properties both in vitro and in vivo, demonstrating alpha-amylase and alpha-glucosidase inhibitory activities [5] and showing dose-dependent hypoglycemic effects when administered orally to diabetic rats [6]. Furthermore, pumpkin seeds and their extracts, whether whole extracts obtained from seeds [7], or cold pressed oil [8,9], particularly alleviated overactive bladder (OAB) [10] and benign prostatic hyperplasia (BPH) symptoms [9].
In recent years, aqueous extracts from CP seeds have gathered attention for their bioactivity, particularly in alleviating symptoms of OAB [11,12], a condition that affects both men and women globally. OAB is characterized by sudden, uncontrollable urges to urinate, often accompanied by increased frequency and nocturia, significantly impairing quality of life [13]. A randomized, double-blind, placebo-controlled trial demonstrated that a formulation combining oil-free pumpkin seed extract with soy germ extract significantly reduced urinary frequency, urgency, and incontinence in women with OAB over 12 weeks [14]. Another study showed that a similar product effectively improved symptoms of stress urinary incontinence in women over a six-week period [15]. Additionally, a three-month pilot study found that oil-free pumpkin seed extract, used as a monotherapy, improved lower urinary tract symptoms (LUTSs) in men with BPH without notable side effects [16]. These clinical findings suggest that constituents beyond phytosterols [17] and fatty acids [18] may contribute to the therapeutic effects observed in LUTSs.
In 2013, the Committee on Herbal Medicinal Products (HMPC) published a monograph recognizing the traditional use of CP seeds for the relief of LUTSs associated with BPH or OAB, based on long-standing medicinal use [19]. This monograph refers to the herbal substance as “whole, ripe, and dried seeds”, without specifying a particular variety. However, the accompanying assessment report indicates that CP convar. citrullina var. styriaca is typically used for pharmaceutical purposes [20]. Commonly known as the “Styrian oil pumpkin”, this variety arose in the 19th century from a natural mutation of a single recessive gene [21], resulting in seeds with a very thin outer layer that facilitated cold-pressed oil production [22]. Although a thin membrane remains, these seeds are generally referred to as “hull-less” or “naked” due to the absence of a lignified hull [23]. As of July 2023, the European Pharmacopoeia (Ph. Eur.) introduced a monograph on pumpkin seeds [24], defining the plant material as “whole, dried, ripe seeds of hull-less seeded varieties of Cucurbita pepo L.”, echoing the HMPC guidance but without restricting it to a specific cultivar [20].
CP exhibits substantial variability across its cultivars [25,26,27], both in morphological traits and chemical composition [28]. Numerous studies have reported differences in fatty acid profiles, phytosterol content, and other bioactive compounds, largely influenced by genetic diversity and environmental conditions [29,30]. This inherent variability poses a challenge to the reproducibility of hydrophilic extract compositions and the consistency of their associated biological activities across different CP varieties.
Although oil-free pumpkin seed extracts exhibited clinical benefits in managing LUTSs, pharmacopeial standards continue to focus primarily on lipophilic constituents such as fatty acids and phytosterols. In contrast, the hydrophilic fraction remains poorly characterized and is largely overlooked in standardization frameworks. This study aimed to comprehensively investigate the chemical composition of hydrophilic extracts from CP seeds, comparing two hull-less varieties with eight hulled ones. The objectives were to identify key components that could serve as chemical markers for standardizing hydrophilic extracts intended for medicinal use, to highlight major compositional differences between varieties and their implications for quality control, and to assess whether the styriaca variety possesses a distinct profile that justifies its preferential inclusion in official monographs. To accomplish our goal, the extracts were characterized starting with elemental analysis to understand the sample matrix. Broad profiling of hydrophilic constituents was achieved using high performance thin layer chromatography (HPTLC) and spectrophotometric methods. Subsequently, targeted analyses were conducted using ultra performance liquid chromatography (UPLC) coupled with a photodiode array detector (PDA), single quadrupole mass detector (QDA), high resolution mass spectrometry (HRMS), or evaporative light scattering detector (ELSD) to identify and quantify primary and secondary metabolites.

2. Results

2.1. Water Content and Elemental Analysis

2.1.1. Water Content

The water content of the extracts was determined prior to this study using Karl Fischer titration method and incorporated into data normalization. The results are presented for each sample in Table S1 (Supplementary Materials). Accordingly, all further results were expressed as mg/g of extract dry weight (dw), enabling direct comparison across samples, with the CP convar. citrullina var. styriaca (SK) extract used as a reference. The remaining nine extracts were coded as follows: SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), and HV (CP—Hungarian Cultivar).

2.1.2. Metal Analysis

Quantitative flame atomic absorption spectroscopy (FAAS) analysis revealed variations in elemental composition among hydrophilic extracts, as depicted in Figure 1A,B. Regarding the macroelements, relative to the SK sample (1962.92 µg/g dw), Na concentration was elevated in samples SS (2538.71 ± 22.85 µg/g dw) and HV (2419.78 ± 4.84 µg/g dw), in contrast with the lowest level in SW (464.76 ± 6.51 µg/g dw). K was present overall in high concentrations, with the SK sample (2041.56 ± 8.17 µg/g dw) being the most abundant, while the SW sample (1468.59 ± 10.28 µg/g dw) presented the lowest concentration. Mg levels also showed variation across samples, peaking in the HV sample (1988.81 ± 41.76 µg/g dw), as opposed to the RZ sample (1092.67 ± 48.08 µg/g dw).
The GA sample, compared to the SK control sample, had the highest level of Cu (1525.40 ± 6.10 vs. 692.50 ± 10.45 µg/g dw). Zn was quantified only in trace amounts in the SK sample (0.9 ± 0.00 µg/g dw), while markedly higher levels were observed in all the other samples, varying between SW and SS (117.31 ± 3.75 and 587.60 ± 4.08 µg/g dw). Cr displayed consistent distributions across varieties with the SK sample containing 96.94 ± 1.36 µg/g dw. Ni displayed variable accumulation in six (SK, LN, RZ, SS, GV, HV) of the ten tested extracts. Fe was sporadically detected in the SS (3.180 ± 0.38 µg/g dw), RZ (0.820 ± 0.08 µg/g dw) and LN samples (32.00 ± 0.38 µg/g dw), while Co and Mn levels were below the limit of quantification (LOQ) in all samples.

2.1.3. Total Carbon and Total Nitrogen Content

Analysis of the carbon and nitrogen contents of the extracts revealed variations among samples, as shown in Table 1. Notably, the GV sample exhibited higher levels of total carbon (TC) compared with the SK control sample (531.27 ± 0.74 vs. 452.32 ± 1.04 mg/g dw), while TC contents in GA and BBZ samples were lower (308.74 ± 0.40 and 304.38 ± 1.95 mg/g dw, respectively). Inorganic carbon (IC) level in the SK sample differed from that in the BBZ sample, which had the highest level (2.95 ± 0.17 vs. 7.05 ± 0.13 mg/g dw) among the tested extracts. Total nitrogen content (TN) in the SK sample was lower than those in SW and HV samples (14.32 ± 0.02 vs. 26.57 ± 0.03 and 27.09 ± 0.24 mg/g dw, respectively), while the differences between the control sample and the remaining samples were less pronounced.

2.2. LC-PDA-HRMS Analysis

Phytochemical screening of CP hydrophilic extracts began with HPTLC, revealing the presence of mono and oligosaccharides as well as amino acids, while phenolic acids and flavonoids were not detected (Figures S1–S3). UPLC-PDA-HRMS analysis confirmed a diverse phytochemical profile (Table A1). Cucurbitin, trigonelline, and nicotinic acid were consistently identified across all extracts, along with nucleosides such as adenosine and guanosine; inosine was absent. Phenolic glycosides (cucurbitosides A-M, except G) and gibberellins (A1, A8, A39) were uniformly present. Most flavonoids including epigallocatechin, catechin, quercetin derivatives, and kaempferol were not detected, while some samples only showed trace amounts of rutin (SK, RZ, SS, and GA), orientin, and homoorientin (SW, RZ, SS, GA, and BBZ). Chlorogenic acid was variably present in SK, LN, RZ, GA, and BBZ, while gallic and protocatechuic acids were undetected. Lignans such as secoisolariciresinol (Seco), lariciresinol (Lar), and pinoresinol (Pino) were found in several samples (LN, RZ, SS, GA, GV, BBZ, HV) but were absent in SK and SKR, suggesting seed coat localization. Cucurbitacins (A, D, E, I) were not detected, with only trace cucurbitacin B observed in LN.

2.3. Analysis of Carbohydrates

Carbohydrate composition was assessed via the phenol–sulfuric acid method and UPLC-ELSD, as shown in Figure 2A,C. Total carbohydrate content was highest in the SK sample (463.62 ± 8.18 mg/g dw) and lowest in the SW sample (352.41 ± 4.85 mg/g dw); SKR, RZ, and GV showed similar values to the SK sample. Sucrose, (Figure 2C) dominated across all samples, with the highest level in SK (268.63 ± 4.87 mg/g dw) and the lowest in SW (182.74 ± 3.73 mg/g dw). Stachyose varied widely, from 5.48 ± 0.82 mg/g dw (SW) to 117.00 ± 0.94 mg/g dw (SKR), while raffinose was notably high in GA and BBZ (72.10 ± 0.65 and 68.76 ± 1.56 mg/g dw).
Verbascose was present in minor but consistent amounts across all samples (1.32–3.93 mg/g dw). Glucose and fructose were mainly found in RZ, HV, and GV samples, but were absent or present in trace amounts in SK and SKR. A moderate positive correlation (Spearman ρ = 0.6606, p < 0.0438) was observed between total carbohydrate content and the sum of quantified saccharides (Figure 2B), supporting the consistency and representativeness of the analytical methods used.

2.4. Analysis of Proteins and Free Amino Acids

Total protein content, as determined by spectrophotometric analysis, is presented in Figure 3. The highest levels were observed in HV and RZ samples (111.83 ± 1.80 and 105.54 ± 2.16 mg/g dw, respectively), while SKR, GV, and SW showed the lowest contents (45.39–66.19 mg/g dw).
To support qualitative assessment, a rapid UPLC-UV method was developed for the simultaneous identification and quantification of twenty-three aminic compounds, including twenty proteinogenic amino acids, one non-proteinogenic amino acid (cucurbitin), and two amines (ethanolamine and GABA). Their distribution is presented as a heat map in Figure 4.
Glu, Arg, and Ala were the most abundant proteinogenic amino acids, while Met, Gly, and Cys (undetectable) were consistently low across all extracts. SKR showed a higher total aminic content than SK (119.01 ± 0.40 vs. 61.07 ± 0.28 mg/g dw). SW had notably high Gln and GABA levels (20.68 ± 1.88 and 15.38 ± 0.38 mg/g dw). Cuc was most concentrated in BBZ and RZ (18.07 ± 0.35 and 15.33 ± 1.44 mg/g dw), followed by SKR, GA, GV, and HV. Gly, Met, and His remained low across all samples. Tyr peaked in SKR (5.41 ± 0.54 mg/g dw), and Trp peaked in HV (6.02 ± 0.23 mg/g dw), exceeding levels in the SK control. These results highlight notable variation in nitrogen-containing compound profiles among the extracts.

2.5. Analysis of Lignans

Analysis of lignans revealed pronounced differences between the extracts, particularly in Lar levels, which peaked in RZ (506.27 ± 21.11 µg/g dw), followed by SS (374.95 ± 2.79 µg/g dw) and GA (368.38 ± 15.42 µg/g dw). Seco showed a similar distribution pattern, while Pino was more evenly distributed across samples, but much less concentrated. Lignans were absent in the extracts derived from hull-less varieties SK and SKR (Figure 5), while the SW sample showed only trace amounts below the LOQ.

2.6. Analysis of Trigonelline

The trigonelline content also varied considerably among the tested extracts (Table 2), ranging from 0.41 ± 0.01 mg/g dw in GV and BBZ samples to 3.45 ± 0.03 mg/g dw in SW. The highest concentration was determined in the SW extract, followed by the SS, LN, and SK samples (1.97 ± 0.00, 1.76 ± 0.02, and 1.76 ± 0.00 mg/g dw). Samples extracted from zucchini varieties (BBZ and RZ) exhibited a low trigonelline content, below 0.50 mg/g dw (0.41 ± 0.01 and 0.46 ± 0.01 mg/g dw, respectively).

2.7. Principal Component Analysis (PCA)

Prior to multivariate analysis, the contribution of quantified compounds to the total dry weight of CP seed hydrophilic extracts was assessed. As shown in Figure 6, carbohydrates consistently represented the major fraction across all samples, whereas differences in protein and free amino acids levels highlighted compositional variability among cultivars. Trigonelline and lignans, although present at relatively low levels, contributed to the further chemical differentiations. This heterogeneity in phytochemical composition supported the use of multivariate analysis in identifying compositional trends among the extracts.
As a methodological consideration, PCA was performed exclusively on quantitatively determined compounds, and therefore does not capture the potential contribution of trace-level constituents identified through LC-PDA-HRMS analysis. The first two principal components (PCs) accounted for 70.62% of the total variance (PC1: 46.68%, PC2: 23.94%; Figure 7A). The scree plot and parallel analysis (Figure 7B) confirmed that only PC1 and PC2 captured meaningful variation, as subsequent components showed eigenvalues below the significance threshold. Loading plots (Figure 7C) revealed that PC1 was positively associated with free amino acids (e.g., Val, Thr, Lys, Arg; loadings: 0.871 to 0.972) and negatively with lignans (Seco, Lar, Pino; loadings: −0.711 to −0.750). PC2 showed positive correlations with rhamnose, fructose, and glutamine (loadings: 0.701 to 0.792), and negative correlations with oligosaccharides (loadings: −0.841 to −0.875).
The PC1 vs. PC2 biplot (Figure 7D) showed a clear separation among samples, reflecting compositional differences among the extracts. The SW and SKR samples, positioned on the right side, had distinct profiles, with the SW sample (upper right quadrant) due to high amino acids and monosaccharides, and the SKR sample (lower right quadrant) due to elevated amino acids and oligosaccharides. The SK, LN, and BBZ samples clustered near the origin, indicating similar compositions. Hierarchical cluster analysis using Ward’s linkage method based on Euclidean distance (Figure 8) supported the PCA results, revealing two main clusters. Cluster I included most samples, with the following subgroups: GA, SS, and RZ (closely related); LN and HV (moderately similar); and SK, GV, and BBZ (maintaining internal similarity). Cluster II grouped the SKR and SW samples, highlighting their distinct metabolic profiles.

3. Discussion

CP, a widely cultivated [28] and polymorphic species [25,26] of the Cucurbitaceae family, is traditionally used for managing LUTSs associated with BPH and OAB [19,20]. While its medicinal effects are largely attributed to lipophilic compounds like phytosterols, fatty acids, and tocopherols [8], emerging evidence suggests hydrophilic constituents such as sugars, amino acids, and polar secondary metabolites may also contribute to its pharmacological activity [14,16]. However, the hydrophilic fraction remains poorly characterized, and differences among CP varieties are understudied. As seed composition is crucial for quality control and standardization, this study provides a comprehensive comparison of hydrophilic extracts from ten CP varieties, highlighting their chemical diversity and identifying potential alternatives to CP convar. citrullina var. styriaca.
The hydrophilic extracts showed a varied mineral profile with notable differences in both macro and microelements. Due to low extraction yields (1.67–2.5%) and selective solubility, mineral contents cannot be directly extrapolated to whole seeds. Na levels ranged from 464.76 to 2538.71 µg/g dw, while K and Mg levels (1468.59–2041.56 µg/g and 1092.67–1988.81 µg/g dw, respectively) were 3–8 times lower than in seeds or oil cakes [31], aligning with their aqueous extractability. Notably, Cr (82.24–100.55 µg/g dw) and Cu (376.77–1525.40 µg/g dw) levels were higher than the literature values [30,31,32]. However, at typical intake levels of 350–875 mg/day dry extract [11,12,16], the estimated amounts of both elements remain below the permitted daily exposure limits established by the International Council for Harmonisation (ICH) guidelines, specifically, 10,700 µg/day for Cr and 3,400 µg/day for Cu [33]. Therefore, despite elevated concentrations relative to the literature data, the estimated intakes are well within internationally accepted safety thresholds and do not pose a toxicological concern according to ICH guideline and Codex nutrient reference values [33,34].
Zn levels in the extracts ranged from 0.90 ± 0.00 to 587.60 ± 4.08 µg/g dw, suggesting wide variability that may reflect differences in extraction efficiency and cultivar genetics. Despite some higher values, all Zn concentrations remained below the adult recommended daily intake (8–11 mg/day) [34], posing no risk of excessive intake. Fe (0.020–31.90 µg/g dw) and Ni (3.64–32.39 µg/g dw) also showed noticeable variation across samples. The low Fe levels may reflect poor aqueous solubility or phytate binding [35], while Ni levels aligned with pumpkin seed oil values [36]. Hull-less varieties SK and SKR had the lowest Ni levels, and estimated intakes remained well below the permitted daily intake [33]. In contrast to earlier findings [35], Mn was undetectable, while Co was consistently absent, in line with previous reports [30]. Phosphorus was below LOQ in all samples, despite its known abundance in pumpkin seeds [30], likely due to insoluble forms such as phytates. These findings underscore the importance of systematic mineral profiling for quality control, especially in therapeutic applications.
Metabolite profiling revealed a diverse phytochemical composition across samples, with consistent detection of Cuc and nucleosides, aligning with the previous literature [37,38]. Chlorogenic and caffeic acids were selectively present, while gallic, protocatechuic acids, and flavonoids such as quercetin, catechin, epicatechin, and myricetin were absent. These results are in contrast with those of Macedo et al., who obtained higher yields from ultrasound (UAE) and microwave-assisted (MAE) methods [39], highlighting the impact of extraction technique on phenolic recovery. However, they also noted the absence of apigenin, kaempferol, quercetin, and quercitrin, while only trace amounts of rutin, orientin, and homoorientin were observed in the current study. p-Coumaric acid was present in all samples except GA, supporting findings that many phenolics exist in bound forms requiring alkaline hydrolysis for release [40]. Phenolic glycosides (cucurbitosides A-M, except G) were identified in all extracts, consistent with their proposed antioxidant roles [41,42]. Gibberellins were also consistently detected, indicating a conserved biosynthetic pathway in CP [43]. Cucurbitacins (A, D, E, I) were absent, confirming their localization in non-seed tissues [44], though trace cucurbitacin B was found in the LN sample.
Quantitative analysis of carbohydrates, proteins, and free amino acids revealed distinct patterns across the tested cultivars. Sucrose and stachyose emerged as the dominant carbohydrates, reaching maximum concentrations of 268.63 ± 4.87 mg/g dw and 117.00 ± 0.94 mg/g dw, respectively, in the SKR sample. Verbascose and raffinose were also consistently detected across all samples in lower concentrations, indicating a saccharide profile enriched in raffinose family oligosaccharides (RFOs), typically associated with seed storage [45]. While direct comparisons to whole seed composition are limited by methodological differences, the literature provides context: Mansour et al. reported high levels of sucrose (17.9 mg/g dw), stachyose (8.1 mg/g dw), and raffinose (4.1 mg/g dw) in defatted CP seed meal [46]. Similarly, Polyzos et al. found sucrose as the predominant carbohydrate in C. maxima seeds and seed cake (1.97–2.90 g/100 g dw), with much lower levels of trehalose, glucose, and fructose, and undetectable levels of raffinose and stachyose [47]. Glu and Arg were the most abundant amino acids, consistent with findings by Horak et al., who also reported high levels of Arg, Glu, and Ala in CP var. styriaca pressed cake [48]. The available literature links both Arg and Glu to cardiovascular modulation, while Cuc has been associated with antiparasitic activity [49]. Arg further contributes to lipid-lowering and vascular effects, particularly when delivered in the natural seed matrix [50]. Glu and Ala, notably high in SKR, support neurotransmission and nitrogen metabolism [32]. The distinct clustering of SK and SKR in PCA and hierarchical analysis reflects major differences in free amino acid and total protein content. Despite SKR having 47% less protein than SK, it showed markedly higher levels of nearly all amino acids, especially Glu, Arg, Ala, and Cuc, and a higher total nitrogen content (34.48%), indicating a shift toward free amino acids. As the quantification of free amino acids used 0.1 N HCl with limited impact on protein hydrolysis [51], this shift likely results from endogenous protein degradation or inherent varietal traits. While storage-induced proteolysis is a possible factor [52], similar saccharide profiles and unchanged total carbohydrate content suggest that pre-harvest conditions or genetic variation are more likely responsible for the differences.
Trigonelline, a hydrophilic pyridine alkaloid found in fenugreek and green coffee beans, is known for its antimicrobial, anticancer, and antihyperglycemic properties, and showed a strong safety profile in preclinical studies [53]. Li et al. reported trigonelline levels of 12.8–23.0 mg/100 g in CP seeds and pulp [54]. In our extracts, trigonelline ranged from 0.41 ± 0.01 to 3.45 ± 0.03 mg/g dw, equating to a safe daily intake of 0.14–3.02 mg based on proposed extract doses (350–875 mg). Based on the similarity in molecular formula of both trigonelline and p-aminobenzoic acid (PABA), we further investigated if PABA is present in our extracts. The absence of a second peak at m/z 138.055 confirmed that, between the two compounds, only trigonelline is present in the extracts. However, Appendino et al. have identified PABA as part of triterpenoid esters such as zucchini factor A and related multiflorane derivatives in the seeds of Cucurbita species [55,56]. These compounds include PABA covalently bound to complex triterpenoid scaffolds that are unlikely to be extracted or detected under our experimental conditions which were optimized for polar metabolites.
Lignan levels varied slightly, with higher concentrations in RZ, SS, and GA. Their absence in hull-less varieties (SK, SKR) may reflect limitations in extraction conditions, such as polarity or lack of hydrolysis, as Sicilia et al. detected Seco and Lar in CP var. styriaca using ethanolic extraction followed by hydrolysis [57]. Similarly, Milder et al. reported the presence of Lar (64 µg/100 g), Pino (37 µg/100 g), and Seco (18 µg/100 g) in CP. var melopepo seeds, following a hydro-methanolic extraction under mild alkaline conditions [58].
Overall, the comparative analysis revealed clear compositional differences across extract samples. Carbohydrates were the dominant constituents followed by proteins and free amino acids, while trigonelline and lignans were consistently low. Free amino acid levels varied considerably, with SKR and SW showing the highest content, and GA, GV, and SS showing the lowest content.
These findings underscore the necessity for a broader quality control framework that integrates both lipophilic and hydrophilic constituents to ensure a comprehensive assessment of extract composition and consistency across CP cultivars. Our results reveal substantial compositional variability of the extracts influenced by seed morphology and varietal differences, particularly in the content of amino acids, carbohydrates, trigonelline, and lignans. Based on our analyses, trigonelline and lignans may serve as useful markers for distinguishing between cultivars. The extract variability highlights the importance of detailed profiling, especially of lignans and other secondary metabolites, in future studies to better understand their contribution to extract quality and pharmacological potential.
While these findings provide important insights, several methodological limitations should be considered when interpreting the results. First, analyses were conducted on technical rather than biological replicates, which may limit the broader applicability of the findings across different cultivation conditions. Second, complete taxonomic identification of all CP varieties was not possible due to inconsistent labeling provided by seed suppliers, potentially affecting the reproducibility of cultivar specific comparisons. Third, the PCA analysis included only quantitatively determined compounds, excluding metabolites detected at trace levels, which may contribute to chemical variability and possibly to biological activity. Finally, as this study did not aim to identify pharmacologically active constituents, no functional assays were performed, warranting future studies integrating bioactivity-guided fractionation or receptor-based assays to assess the therapeutic relevance of specific hydrophilic components.
Importantly, our findings challenge the notion of treating CP as a uniform botanical entity. The substantial phytochemical diversity observed across varieties indicates that CP is not necessarily CP in a chemical or therapeutic sense. Therefore, monographs and regulatory texts should be revised or interpreted with caution, clearly specifying the CP variety used to avoid ambiguity in research, clinical application, and quality assurance.

4. Materials and Methods

4.1. Chemicals and Reagents

Common solvents, and general-purpose reagents used throughout the study, are listed in Appendix B. The section below details the reference substances and standards employed for compound identification and quantification. (–)-Secoisolariciresinol (cat. no. 80532), secoisolariciresinol diglucoside (cat. no. 80533), (+)-pinoresinol (cat. no. 89525), pinoresinol-4-O-glucoside (cat. no. 84251), pinoresinol diglucoside (cat. no. 89850), (−)-matairesinol (cat. no. 80497), olivil (cat. no. 83883), cucurbitacin A (cat. no. 84046), cucurbitacin B (cat. no. 82226), cucurbitacin D (cat. no. 80254), cucurbitacin E (cat. no. 80013), cucurbitacin E-2-O-glucoside (cat. no. 80613), cucurbitacin I (cat. no. 89464), cucurbitin chloride 58% (cat. no. 80034), vitexin-2-glucoside (cat. no. 84193), orientin (cat. no. 83529), vitexin-2-rhamnoside (cat. no. 89291), protocatechuic acid (cat. no. 89766), and epicatechin (cat. no. 89192) were obtained from Phytolab GmbH & Ko KG (Vestenbergsgreuth, Germany). (+)-Isolariciresinol (cat. no. CFN98915) was from ChemFaces (Wuhan, China), while (+)-lariciresinol (cat. no. 06892), (−) olivil 4″-O-glucoside (cat. no. SMB0026), naringin (cat. no. 71162), p-coumaric acid (cat. no. C9008), glucose (cat. no. 49139), fructose (cat. no. F0127), sucrose (cat. no. S9378), raffinose (cat. no. R0250), stachyose (cat. no. S4001), verbascose (cat. no. 56217), alanine (cat. no. A7627), tryptophan (cat. no. T0254), asparagine (cat. no. A0884), glutamine (cat. no. 49419), aspartic acid (cat. no. A8949), glutamic acid (cat. no. G1251), arginine (cat. no. A5131), ethanolamine (cat. no. E9508), gamma-aminobutyric acid (cat. no. A2129), nicotinic acid (cat. no. 72309), guanosine (cat. no. G6752), and inosine (cat. no. I4125) were obtained from Sigma-Aldrich (St. Louis, MO, USA). Vitexin (cat. no. 1235S), epicatechin gallate (cat. no. 0978S), homoorientin (cat. no. 1055S), epigallocatechin (cat. no. 0979S), quercitrin (cat. no. 1236S), isoquercitrin (cat. no. 1327S), myricetin (cat. no. 1127S), kaempferol (cat. no. 1124S), apigenin (cat. no. 1102S), myrcitrin (cat. no. 1029S), and 8-hydroxypinoresinol O-beta D-glucopyranoside (cat. no. SMB00145) were from Extrasynthese (Genay, France). Amino Acid Standard H (a mixture of 18 amino acids, cat. no. 20088) and Pierce™ BCA Protein Assay Kit (cat. no. A55865 containing BCA Reagent A, 500 mL; BCA Reagent B 25 mL, and prediluted Albumin Standard) were from Thermo Fisher Scientific (Waltham, MA, USA), while AccQ-Tag Ultra Derivatization Kit (cat. no. 186003836, containing AccQ-Tag Ultra Borate Buffer, AccQ-Tag Ultra Reagent Powder in Vial 1 A, and AccQ-Tag Ultra Reagent Diluent in Vial 2B) was from Waters (Milford, MA, USA).

4.2. Plant Material

The suppliers and characteristics of the dried seeds from ten cultivars of C. pepo are detailed in Table 3 and Figure 9. Voucher specimens were deposited in the Department of Pharmacognosy-Phytotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania.

4.3. Extract Preparation

Hydroethanolic extracts were prepared from each batch of seeds by mixing 3 L of 60% (v/v) ethanol with 600 g of plant material ground to a particle size of approximately 0.5 mm. The extraction was performed in a 5 L round-bottomed flask placed in a heating mantle (Biobase, Meihua Group Co., Ltd., Jinan, China) equipped with an overhead stirrer (250 rpm) and maintained at 50 °C for 3 h. Following extraction, the solution was filtered, and ethanol was removed under reduced pressure using a rotary evaporator (Büchi R300 Rotavapor, Büchi Labortechnik AG, Flawil, Switzerland). The oil phase obtained after ethanol evaporation was removed by liquid–liquid partitioning in a separatory funnel. The resulting extract was subsequently diluted to 3 L to achieve a final ethanol concentration of 8% (v/v) and filtered through a 1 μm pore membrane to eliminate residual particulates. The filtrate was further concentrated and freeze-dried (Alpha 1–2 LD plus freeze dryer, Martin Christ Gefriertrocknungsanlagen GmbH, Osterode am Harz, Germany), yielding dry extracts. The freeze-dried extracts were sealed in glass vials, coded according to Table 3, and stored at −20 °C until further use.

4.4. Water Content and Elemental Analysis

4.4.1. Water Content

The water content of the samples was determined on a 874 Metrohm oven sample processor (Metrohm, Herisau, Switzerland) coupled to a 917 Coulometer (Metrohm, Herisau, Switzerland), according to the method described by Faria-e-Silva et al. [59]. Results were expressed as wt%.

4.4.2. Metal Elements

Quantitative elemental analysis was performed using FAAS with a contrAA 300 spectrometer (Analytik Jena, Jena, Germany) equipped with a high-resolution Echelle double monochromator and a UV-sensitive charge-coupled device (CCD) detector. A pneumatic concentric jet nebulizer generated the aerosol. Each metal was measured at its optimal analytical wavelength, selected for sensitivity and minimal interference. Calibration curves (0.1–100 mg/L) were constructed from elemental standards. Sample solutions (5 mg/mL in 8% ethanol) were aspirated, aerosolized, and mixed with acetylene/air or acetylene/nitrous oxide, depending on the element.

4.4.3. TC and TN Content

Total organic carbon (TOC), IC, TC, and TN were quantified using a Multi N/C 3100 TOC Analyzer (Analytik Jena, Jena, Germany) with a chemiluminescence detector (CLD) for TN analysis. Samples (25 µg/mL in ultrapure water) were burned in an oxygen rich atmosphere at 800 °C in the presence of a catalyst, converting carbon to CO2 and nitrogen containing compounds to NO. Resulting gasses were detected and measured quantitatively. Results were expressed in mg/g of dry extract.

4.5. HPTLC Analysis

The analysis of primary and secondary metabolites in the studied samples was initially performed using several HPTLC methods (See Supplementary Materials).

4.6. LC-PDA-HRMS Analysis

Phytochemical screening was conducted using LC-PDA-HRMS on a Waters Acquity H-Class UPLC coupled to a SYNAPT G2-S Q-TOF mass spectrometer (Milford, MA, USA) with an ESI source. Analyses were performed in both positive and negative ion modes (100–900 Da), using the following conditions: capillary voltage of 2.5 V, cone voltage of 20 V, source temperature 150 °C, and desolvation temperature 350 °C. Extracts (20 mg/mL in 30% ethanol) were injected (10 µL) and separated on a Waters XSelect HSS T3 column (250 × 4.6 mm, 5 µm) using the following gradient: 0.1% formic acid in water (A), 0.1% formic acid in acetonitrile (B): 0–5 min, 0% B; 5–10 min, 0–5% B; 10–25 min, 5–10% B, 25–50 min, 10–20% B, 50–60 min, 20–40% B, 60–80 min, 40–98% B. The presence of lignans, flavonoids, phenolic acids, nucleosides, epigallocatechin, orientin, vitexin derivatives, quercetin and its glycosides, isolariciresinol, matairesinol, Pino, Lar, Seco, and salicylic acid was evaluated using reference standards (0.1 mg/mL in 70% methanol). Identification was based on retention time, UV spectra, accurate mass, fragmentation, and comparison with standards and the literature using MassLynx 4.1.

4.7. Total Carbohydrate Content

The total carbohydrate content was measured using the phenol–sulfuric acid method [60] with a few modifications. Briefly, dry extracts were dissolved in water (1 mg/mL) and a 0.05 mL aliquot was mixed with 0.55 mL water and 1.8 mL sulfuric acid in a heat-resistant tube, followed by the immediate addition of 0.36 mL of 5% (m/v) phenol solution. After vortexing (5 s), the mixture was incubated at 90 °C for 5 min and cooled. The absorbance was measured at 590 nm using a Specord 210 plus UV-Vis spectrophotometer equipped with ASpect UV 2.0 software (Analytik Jena, Jena, Germany). A calibration curve prepared under the same conditions with D-glucose as standard (0.1–1 mg/mL) was used for quantification.

4.8. UPLC Analysis of Saccharides

Monosaccharides and oligosaccharides were analyzed using a Waters Acquity H-Class UPLC system coupled with a 1260 Infinity II ELSD (Agilent, Santa Clara, CA, USA). Separation was performed on a Phenomenex Luna Omega Sugar column (150 × 3.6 mm, 3 µm) following the manufacturer’s guidelines with minor modifications [61]. The mobile phases used were water (A) and acetonitrile–isopropanol–water, 90:5:5, v/v/v (B). Two separate isocratic methods were used, each operating at a flow rate of 1 mL/min. Monosaccharides were separated using an A–B ratio of 8:92 for 18 min, while oligosaccharides were analyzed using an A–B ratio of 29:71 for 17 min. Samples (5 and 10 mg/mL in 50% acetonitrile, respectively) were injected (10 µL) and the column temperature was maintained at 30 °C. ELSD settings included an evaporator temperature of 90 °C, nebulizer temperature of 40 °C, and gas flow rate of 1.5 L/min. Compound identification was achieved by comparing retention times with those of reference standards using Empower 3 software (Waters, Milford, MA, USA). Calibration curves were prepared using standard solutions (0.01–2.00 mg/mL) of rhamnose, glucose, fructose, sucrose, raffinose, stachyose, and verbascose, all dissolved in 50% acetonitrile.

4.9. Total Protein Content

A Spectro Star Nano microplate reader equipped with reader control software (BMG LABTECH, Ortenberg, Germany) was employed for the total protein quantification using the BCA assay kit, according to the manufacturer’s protocol [62]. Briefly, 25 µL of extract (2.5 mg/mL in PBS) was mixed with 200 µL of BCA working reagent and incubated at 37 °C for 30 min. After cooling for 5 min at room temperature, the absorbance was measured at 562 nm. Total protein content was expressed as mg bovine serum albumin (BSA) equivalents per gram of extract (mg BSA/g extract).

4.10. HPLC Analysis of Free Amino Acids

Free amino acids in CP seed extracts were analyzed using an Acquity H-Class UPLC system with PDA and QDA detectors following derivatization with the ACCQ-Tag Ultra kit (Waters, Milford, MA, USA), per the manufacturer’s protocol [63]. Extracts (10 mg/mL in 0.1 N HCl) were derivatized with 70 µL borate buffer and 20 µL reagent, vortexed, incubated at room temperature, then heated at 55 °C for 10 min. After centrifugation (5000 rpm, 10 min), 2 µL of the supernatant was injected. The derivatizing agent, 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate (ACQ), binds to primary and secondary amines to form stable carbamate derivatives with peak absorbance at 248 nm. Excess reagent reacts with water to form 6-aminoquinoline (AMQ), which can form bis-derivatives.
Analytes were identified by their retention times and corresponding m/z values using reference standards. All compounds produced single peaks, except Cuc, which formed both single- and double-derivatized products due to its two reactive amino groups. Separation was performed on a Waters BEH C18 column (150 × 2.1 mm, 1.7 µm) at 45 °C, with a flow rate of 0.4 mL/min. A gradient elution was applied using mobile phase A (ammonium acetate, acetonitrile, acetic acid, water; pH 4.3) and B (acetonitrile) following the program 0–1.35 min, 0.5–0.5% B; 1.35–18 min, 0.5–8% B; 18–21 min, 8–19.5% B; 21–24 min, 19.5% B, 24–24.5 min, 19.5–59% B; 24.5–26 min, 59% B, 26–26.5 min, 59–0.5% B, 26.5–33 min, 0.5% B. Calibration curves were built using a standard mix of 18 amino acids (2.5 µmol/mL), and for additional compounds (Asp, Glu, Trp, Cuc, GABA, Eta), individual standards were prepared (0.6–0.005 mg/mL in 0.1 N HCl).

4.11. UPLC Analysis of Lignans

Lignans Seco, Lar, Pino were identified and quantified using the same Waters Acquity H-Class UPLC system coupled with PDA and QDA detectors and equipped with a Waters Cortecs C18 column (150 × 3.0 mm, 1.6 µm). Identification was confirmed by comparing the retention time, UV (280 nm) and MS spectra with reference substances, while quantification relied on six-point calibration curves (1–500 µg/mL). The mobile phase consisted of 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B): 0–17.9 min, 15–45% B; 17.9–19.1 min, 45–99% B; 19.1–19.4 min, 99–15% B; 19.4–24 min, 15% B. The gradient ran at a flow rate of 0.4 mL/min, with the column temperature set at 35 °C. Dry extracts (20 mg/mL in 80% methanol) were ultrasonicated for 15 min, and centrifuged at 10,000 rpm for 10 min. Finally, a volume of 2 µL of supernatant was injected.

4.12. UPLC Analysis of Trigonelline

Trigonelline identification and quantification was conducted using the same system and detectors as mentioned in Section 4.10. Chromatographic separation was performed on a Waters X Select HSS T3 column (250 × 4.6 mm, 5 µm) maintained at room temperature (24.5 °C). The mobile phase consisted of water with 0.1% formic acid (A) and acetonitrile with 0.1% formic acid (B). The gradient program started with an isocratic step for 7 min at 0% B; 7–10 min, 0–98% B; 10–20 min, 98% B; 20–22 min, 98–0% B; 22–32 min, 0% B. Samples (25 mg/mL in 8% ethanol) were injected (5 µL) and separated at a flow rate of 0.8 mL/min. Quantification was carried out using a six-point calibration curve (1–500 µg/mL). The UV detection was set at 263 nm, whereas a single ion recording (SIR) channel was set at m/z 138. A manual splitter directed 25% of the flow to the QDa detector.

4.13. Multivariate Analysis

Quantitative phytochemical analyses were performed for each sample in triplicate and results were reported as mean ± standard deviation (SD). A multivariate analysis was used to investigate the quantitative composition of the hydrophilic extracts of CP seeds. PCA was performed to reduce dimensionality and identify patterns in the chemical composition of the tested extracts. The PCA considered 35 quantified variables, including total protein, individual sugars, free amino acids, and secondary metabolites. The dataset was standardized using a Z-score standardization, and the principal components were computed, together with their eigenvalues, proportion of variance, and cumulative variance, to determine the optimal number of components for an accurate interpretation. This analysis was carried out using GraphPad Prism software version 10.04 (GraphPad Software, Boston, MA, USA) and SPSS Statistics version 26.0 (SPSS Inc., Chicago, IL, USA).

5. Conclusions

In conclusion, our findings reveal notable variation in the chemical composition of hydroethanolic seed extracts across various CP cultivars, underscoring the need for a comprehensive standardization strategy. While CP convar. citrullina var. styriaca is currently recommended by the HMPC for use in herbal medicinal products, the Ph. Eur. refers only to hull-less seeded varieties without specifying a cultivar. Our data show that CP var. styriaca, cultivar ‘Gleisdorfer Rustikal’ (SKR), exhibits a hydrophilic metabolite profile comparable to that of the reference convar. citrullina var. styriaca (SK), with even higher levels of certain amino acids and oligosaccharides.
These results highlight the influence of genetic background on extract composition and emphasize the urgent need for regulatory alignment between HMPC and Ph. Eur., including the establishment of a clear and cultivar-specific framework for the standardization of CP seed extracts.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/plants14152308/s1, Figure S1: HPTLC chromatogram of free amino acids; Figure S2: HPTLC chromatogram of saccharides; Figure S3: HPTLC chromatogram of flavonoids and phenolic acids, Table S1: Water content of hydrophilic Cucurbita pepo seed extracts determined by Karl Fischer titration (wt%).

Author Contributions

Conceptualization, V.B., C.H., A.S. and A.M.; investigation, A.-E.G.; visualization, A.-E.G. and R.S.; writing—original draft, A.-E.G.; writing—review and editing: V.B., A.S. and C.H.; supervision, V.B., A.M. and A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

Max Zeller Söhne AG is gratefully acknowledged for supporting this work. We also thank Amin Chaanin from Vitaplant AG for his help in procuring the seeds, Maria Ignat and Florica Doroftei from Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania, for their valuable assistance with the elemental analysis, and Carmen Pintilescu from Alexandru Ioan Cuza University of Iasi, Romania, for her kind support with the Principal Component Analysis.

Conflicts of Interest

V.B., C.H., R.S., and A.S. are employed at Max Zeller Söhne AG, CH-8590 Romanshorn, Switzerland. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Appendix A

Table A1. Qualitative composition of Cucurbita pepo hydrophilic extracts using UHPLC-PDA-HRMS.
Table A1. Qualitative composition of Cucurbita pepo hydrophilic extracts using UHPLC-PDA-HRMS.
No. RT
(min)
Compound NameChemical
Formula
Chemical ClassCalculated Mass Main
Fragment
Ion TypeOther
Fragments
Identified Not IdentifiedReference
12.47Cucurbitin C5H10N2O2amino acid130.0742131.0819[M+H]+85.07661–10 [37]
24.17TrigonellineC7H7NO2alkaloid137.0477138.0557[M+H]+94.06011–10 [54]
38.594Nicotinic acidC6H5NO2pyridine-3-carboxylic acid123.0320124.0399[M+H]+106.02711–10 [54]
414.93Adenosine C10H13N5O4nucleoside267.0968268.1045[M+H]+136.06241–10 [38]
515.37Guanosine C10H13N5O5nucleoside283.0917282.0841[M-H]152.0531
135.0305
1–10 [38]
616.32Gallic acid C7H6O5phenolic acids 170.0200 169.0133[M-H]125.0236 1–10[39,64]
724.39Protocatechuic acid C7H6O4phenolic acids 154.0266 153.0188[M-H]109.0292 1–10[39,40]
825.16Cucurbitoside LC24H29NO12phenolic glycoside523.1689522.1614[M-H]457.1924
411.1865
122.0247
1–10 [42]
934.07Epigallocatechin C15H14O7flavonoid306.0739307.0818[M+H]+289.0715
165.0218
1–10
1035.61Chlorogenic acidC16H18O9phenolic acid 354.0951 353.0811[M-H]191.0560, 375.0696, 443.0560, 511.0439,1–3, 5–7, 94, 8, 10[39]
1135.87Catechin C15H14O6flavonoid 290.0790289.0715[M-H]357.0585,
245.0805
1–10[39,64]
1239.41Cucurbitoside HC25H31NO12phenolic glycoside537.1846536.1776[M-H]383.2079
122.0247
1–10 [41]
1339.42Caffeic acid C9H8O4phenolic acids 180.0423 179.0349[M-H]153.0190, 135.0448, 109.02902, 3, 5, 6, 101, 2, 4, 7–9[39,40]
1440.69Cucurbitoside MC25H31NO12phenolic glycoside537.1846536.1776[M-H]399.1300
136.0402
1–10 [42]
1540.98Gibberelin A 39C20H26O8terpenoid394.1627393.1555[M-H]319.1184,
290.1255,
231.1384
1–10 [43]
1642.81Epicatechin C15H14O6flavonoid 290.0790289.0715[M-H]245.0808 1–10[39]
1744.37Gibberelin A 8 C19H24O7terpenoid 364.152 363.1449[M-H]275.1650,
257.1543
1–10 [43]
1844.98Cucurbitoside DC25H30O13phenolic glycoside538.1686537.1608[M-H]186.1130
137.0236
1–10 [41]
1946.21Cucurbitoside IC26H33NO12phenolic glycoside551.2003550.1920[M-H]413.14531–10 [42]
2046.92Gibberelin A 1C19H24O6terpenoid348.1572347.1491[M-H]241.01261–10 [43]
2148.97Homoorientin C21H20O11flavonoid 448.1006 449.1081[M+H]+471.0893; 431.0974; 329.06574, 5, 7, 91–3, 6, 8, 10
2249.38p-coumaric acid C9H8O3phenolic acid 164.0473 163.0395[M-H]119.04991–6, 8–107[39]
2349.77Secoisolariciresinol-diglucoside C20H22O6lignan 686.2786 327.159[M+H]+704.3115
295.1328
163.0755
137.0594
3–7, 9, 101, 2, 8
2450.42OrientinC21H20O11flavonoid 448.1006 449.1075[M+H]+431.0966 329.0656 4, 5, 7, 91–3, 6, 8, 10
2548.70Hydroxypinoresinol glucopyranoside C26H32O12lignan536.1894559.1791 (Na adduct)[M+H]+163.07543, 101, 2, 4–9
2651.09Cucurbitoside BC26H32O13phenolic glycoside552.1843551.1759[M-H]137.02341–10 [41]
2751.76Vitexin-2-glucoside C27H30O15flavonoid glycoside 594.1585 595.1657[M+H]+617.1475, 475.3251 453.3431 1–10
2853.84Rutin C27H30O16flavonoid 610.1534 609.1453[M-H]677.13351, 5–82–4, 9, 10[39,64]
2954.71Olivil C20H24O7lignan 376.1522 399.1412 (Na adduct)[M+H]+341.1387
323.1279
187.0756
3–5, 7–91, 2, 6, 10
3055.33Vitexin-2-rhamnoside C27H30O14flavonoid glycoside 578.1636 579.171[M+H]+601.1523 1–10
3155.36VitexinC21H20O10flavonoid 432.1056 433.1129[M+H]+415.1014 1–10[39]
3254.41Myrcitrin C21H20O12flavonoid 464.0955463.087[M-H]509.09381, 62–5, 7–10
3354.84Cucurbitoside KC23H34O12phenolic glycoside502.205501.1969[M-H]399.1287
123.0447
1–10 [42]
3454.91Hyperoside C21H20O12flavonoid 464.0955463.0876[M-H]301.0354, 151.0014 1–10
3555.12Epicatechin gallateC22H18O10flavonoid 442.09441.0819[M-H]473.0710 1–10
3655.67Isoquercitrin C21H20O12flavonoid 464.0955463.0871[M-H]301.0324, 151.0018 1–10
3756.38Cucurbitoside CC25H30O12phenolic glycoside522.1737521.1652[M-H]121.02911–10 [41]
3854.47Cucurbitoside EC24H28O12phenolic glycoside508.1581507.1497[M-H]121.02911–10 [41]
3957.50Naringin C27H32O14flavanone 7-O glycoside 580.1792 579.1414[M-H]647.1518 1–10[39,64]
4057.58Cucurbitoside JC25H38O13phenolic glycoside546.2312545.2236[M-H]353.16921–10 [42]
4157.70Cucurbitoside AC26H32O12phenolic glycoside536.1894535.1805[M-H]413.1449
347.1494
121.0295
1–10 [41]
4257.72Quercitrin C21H20O11flavonoid 448.1006447.0929[M-H]303.0469 1–10
4358.18Cucurbitoside FC23H36O12phenolic glycoside516.2207515.2123[M-H]413.1451
137.0606
1–10 [42]
4459.78Myricetin C15H10O8flavonoid 318.0297 317.0298[M-H]287.0165 1–10[64]
4559.87Secoisolariciresinol C20H26O6lignan 362.1729 327.1593[M+H]+385.1620 295.1328 163.0758 137.05983–101, 2[65]
4660.15Salicylic acid C7H6O3carboxylic acid 138.0317 137.0239[M-H]297.03721–10 [66]
4760.80Lariciresinol C20H24O6lignan360.1573219.1019[M+H]+189.0912
721.1034
3–101,2[57]
4863.37Daidzein C15H10O4isoflavone 254.0579253.0504[M-H]223.0413 1–10[65]
4959.29Quercetin C15H10O7flavonoid 302.0427 301.0349[M-H]179.0108
125.0114
1–10[64]
5063.94IsolariciresinolC32H46O16lignan360.1573361.1646[M+H]+209.0814 1–10
5164.96Pinoresinol C20H22O6lignan358.1416341.1385[M+H]+323.1273 175,07563–101, 2[67]
5265.16Cucurbitacin E-2-O glucoside C38H54O13cucurbitacin type triterpene718.3564741.3465 [M+H]+479.2796 461.268931, 2, 4–10[67]
5365.86Cucurbitacin AC32H46O9cucurbitacin type triterpene574.3142597.3035 [M+H]+515.3007
467.2791
1–10[67]
5465.60Apigenin C15H10O5flavonoid270.0528271.0609[M+H]+158.0123 1–10[39]
5566.49Cucurbitacin D C30H44O7cucurbitacin type triterpene516.3087539.2978 [M+H]+481.2951
499.3055
1–10[44]
5666.50Kaempferol C15H10O6flavonoid 286.0477285.0402[M-H]164.9982
119.0323
1–10[64]
5768.14Cucurbitacin I C30H42O7cucurbitacin type triterpene514.2931537.2821 [M+H]+97.2896
479.2794
461.2686
1–10[44]
5871.11Cucurbitacin B C32H46O8cucurbitacin type triterpene558.3193581.3089 [M+H]+481.295
463.2846
31, 2, 4–10[44]
5975.51Matairesinol C20H22O6lignan358.1416359.2412[M+H]+375.2146
175.1334
103.0756
1–10

Appendix B. List of Routine Solvents and General Laboratory Reagents

Acetonitrile (LC-MS grade cat. no. AE70.2) was provided by Roth (Karlsruhe, Germany); acetonitrile HPLC grade (cat. no. 83639.329), ethanol absolute (cat. no.20821.330), methanol (cat. no. 20864.420), LC-MS grade formic acid (cat. no. 84865.260), ethyl acetate (cat. no. 83621.320), and isopropanol (cat. no. 20842.330) were purchased from VWR (Radnor, PA, USA). Toluene (cat. no. 1.08325.2500), 1-butanol (cat. no. 1.01990.2500), boric acid (cat. no. 1.00165.0100), ninhydrin (cat. no. 1.06762.0010), disodium hydrogen phosphate hydrate (cat. no. 1.06580.0500), phenol (cat. no. 16016), and sulfuric acid (cat. no. 1007131) were from Merck (Darmstadt, Germany). Ultrapure water was obtained from ELGA Ultrapure water system PURELAB® 7120 (ELGA LabWater, High Wycombe, UK).

References

  1. Castellanos-Morales, G.; Ruiz-Mondragon, K.Y.; Hernandez-Rosales, H.S.; Sanchez-de la Vega, G.; Gamez, N.; Aguirre-Planter, E.; Montes-Hernandez, S.; Lira-Saade, R.; Eguiarte, L.E. Tracing back the origin of pumpkins (Cucurbita pepo ssp. pepo L.) in Mexico. Proc. Biol. Sci. 2019, 286, 20191440. [Google Scholar] [CrossRef]
  2. Robinson, R.; Munger, H.; Whitaker, T.; Bohn, G. Genes of the Curcurbitaceae. Hort. Sci. 1976, 11, 554–568. [Google Scholar]
  3. Batool, M.; Ranjha, M.; Roobab, U.; Manzoor, M.F.; Farooq, U.; Nadeem, H.R.; Nadeem, M.; Kanwal, R.; AbdElgawad, H.; Al Jaouni, S.K.; et al. Nutritional Value, Phytochemical Potential, and Therapeutic Benefits of Pumpkin (Cucurbita sp.). Plants 2022, 11, 1394. [Google Scholar] [CrossRef]
  4. Ratnam, N.; Naijibullah, M.; Ibrahim, M. A review on Cucurbita pepo. Int. J. Pharm. Phytochem. Res. 2017, 9, 1190–1194. [Google Scholar] [CrossRef]
  5. Thanh, T.T.T.; Quach, T.T.M.; Yuguchi, Y.; Nguyen, N.T.; Van Ngo, Q.; Van Bui, N.; Kawashima, S.; Ho, C.D. Molecular structure and anti-diabetic activity of a polysaccharide extracted from pumpkin Cucurbita pepo. J. Mol. Struct. 2021, 1239, 130507. [Google Scholar] [CrossRef]
  6. Sedigheh, A.; Jamal, M.S.; Mahbubeh, S.; Somayeh, K.; Mahmoud, R.; Azadeh, A.; Fatemeh, S. Hypoglycaemic and hypolipidemic effects of pumpkin (Cucurbita pepo L.) on alloxan-induced diabetic rats. Afr. J. Pharm. Pharmacol. 2011, 5, 2620–2626. [Google Scholar] [CrossRef]
  7. Vahlensieck, W.; Theurer, C.; Pfitzer, E.; Patz, B.; Banik, N.; Engelmann, U. Effects of pumpkin seed in men with lower urinary tract symptoms due to benign prostatic hyperplasia in the one-year, randomized, placebo-controlled GRANU study. Urol. Int. 2015, 94, 286–295. [Google Scholar] [CrossRef]
  8. Gossell-Williams, M.; Davis, A.; O’Connor, N. Inhibition of testosterone-induced hyperplasia of the prostate of sprague-dawley rats by pumpkin seed oil. J. Med. Food 2006, 9, 284–286. [Google Scholar] [CrossRef]
  9. Zerafatjou, N.; Amirzargar, M.; Biglarkhani, M.; Shobeirian, F.; Zoghi, G. Pumpkin seed oil (Cucurbita pepo) versus tamsulosin for benign prostatic hyperplasia symptom relief: A single-blind randomized clinical trial. BMC Urol. 2021, 21, 147. [Google Scholar] [CrossRef]
  10. Nishimura, M.; Ohkawara, T.; Sato, H.; Takeda, H.; Nishihira, J. Pumpkin Seed Oil Extracted From Cucurbita maxima Improves Urinary Disorder in Human Overactive Bladder. J. Tradit. Complement. Med. 2014, 4, 72–74. [Google Scholar] [CrossRef]
  11. Sogabe, H.; Terado, T. Open clinical study of effects of pumpkin seed extract/soybean germ extract vixture-containing processed food on nocturia. Jpn. J. Med. Pharm. Sci. 2001, 46, 727–737. [Google Scholar]
  12. Terado, T.; Sogabe, H.; Saito, K. Clinical Study of mixed processed foods containing of pumpkin seed extract and soybean germ extract on pollakiuria in night infielder men. Jpn. J. Med. Pharm. Sci. 2004, 52, 551–561. [Google Scholar]
  13. Abrams, P.; Kelleher, C.; Kerr, L.A.; Rogers, R.G. Overactive bladder significantly affects quality of life. Am. J. Manag. Care. 2000, 6, S580–S590. [Google Scholar]
  14. Shim, B.; Jeong, H.; Lee, S.; Hwang, S.; Moon, B.; Storni, C. A randomized double-blind placebo-controlled clinical trial of a product containing pumpkin seed extract and soy germ extract to improve overactive bladder-related voiding dysfunction and quality of life. J. Funct. Foods 2014, 8, 111–117. [Google Scholar] [CrossRef]
  15. Yanagisawa, E.; Satoh, I. Clinical study of mixed processed foods containing of pumpkin seed extract and soybean germ extract on stress urinary incontinence (SUI) in women. Jpn. J. Med. Pharm. Sci. 2003, 50, 313–322. [Google Scholar]
  16. Leibbrand, M.; Siefer, S.; Schon, C.; Perrinjaquet-Moccetti, T.; Kompek, A.; Csernich, A.; Bucar, F.; Kreuter, M.H. Effects of an oil-free hydroethanolic pumpkin seed extract on symptom frequency and severity in men with benign prostatic hyperplasia: A pilot study in humans. J. Med. Food 2019, 22, 551–559. [Google Scholar] [CrossRef]
  17. Heim, S.; Seibt, S.; Stier, H.; Moré, M.I. Uromedic® pumpkin seed derived Δ7-sterols, extract and oil inhibit 5α-reductases and bind to androgen receptor in vitro. Pharmacol. Pharm. 2018, 9, 193. [Google Scholar] [CrossRef]
  18. Ramak, P.; Mahboubi, M. The beneficial effects of pumpkin (Cucurbita pepo L.) seed oil for health condition of men. Food Rev. Int. 2019, 35, 166–176. [Google Scholar] [CrossRef]
  19. EMA; HMPC (Committee on Herbal Medicinal Products). Final Community Herbal Monograph on Cucurbita pepo L., semen; European Medicines Agency: Amsterdam, The Netherlands, 2013. [Google Scholar]
  20. Widy-Tyszkiewicz, E.; Matlawska, I.; Bylka, W. Assessment Report on Cucurbita pepo L., Semen; EMA/HMPC/136022/2010; Committee on Herbal Medicinal Products (HMPC): Amsterdam, The Netherlands, 2012. [Google Scholar]
  21. Chahal, G.K.; Kaur, A.; Dhatt, A.S. A single-gene mutation changed the architecture of pumpkin seed: A review. J. Plant Growth Regul. 2022, 41, 113–118. [Google Scholar] [CrossRef]
  22. Sedláčková, V.H.; Avagyan, A. Morphological characteristics of selected fruit parts and naked seeds of Cucurbita pepo var. styriaca. Agrobiodivers. Improv. Nutr. Health Life Qual. 2022, 6, 251–261. [Google Scholar] [CrossRef]
  23. Lelley, T.; Loy, B.; Murkovic, M. Hull-Less Oil Seed Pumpkin. In Oil Crops; Vollmann, J., Rajcan, I., Eds.; Springer: New York, NY, USA, 2010; pp. 469–492. [Google Scholar]
  24. EDQM. Pumpkin seed (2941). In European Pharmacopoeia (Ph. Eur.), 11th ed.; Council of Europe, Ed.; Council of Europe: Strasbourg, France, 2023; pp. 4858–4859. [Google Scholar]
  25. Paris, H.S.; Burger, Y.; Schaffer, A.A. Genetic variability and introgression of horticulturally valuable traits in squash and pumpkins of Cucurbita pepo. Isr. J. Plant Sci. 2006, 54, 223–231. [Google Scholar] [CrossRef]
  26. Lebeda, A.; Křístková, E.; Paris, H. Variation for morphological traits within and among Cucurbita pepo genotypes. Acta. Hortic. 2010, 871, 211–217. [Google Scholar] [CrossRef]
  27. Tomić, D.; Radovanović, M.; Marjanović, M.; Đurović, V.; Stevović, V.; Pavlović, N.; Lazarević, Đ.; Zornić, V.; Madić, M. Divergence analysis of Cucurbita pepo L. population for seed oil production. Not. Bot. Horti. Agrobot. Cluj-Napoca 2024, 52, 13907. [Google Scholar] [CrossRef]
  28. Meru, G.; Fu, Y.; Leyva, D.; Sarnoski, P.; Yagiz, Y. Phenotypic relationships among oil, protein, fatty acid composition and seed size traits in Cucurbita pepo. Sci. Hortic. 2018, 233, 47–53. [Google Scholar] [CrossRef]
  29. Younis, Y.M.; Ghirmay, S.; al-Shihry, S.S. African Cucurbita pepo L.: Properties of seed and variability in fatty acid composition of seed oil. Phytochem. Anal. 2000, 54, 71–75. [Google Scholar] [CrossRef]
  30. Seymen, M.; Uslu, N.; Türkmen, Ö.; Al Juhaimi, F.; Özcan, M.M. Chemical compositions and mineral contents of some hull-less pumpkin seed and oils. J. Am. Oil. Chem. Soc. 2016, 93, 1095–1099. [Google Scholar] [CrossRef]
  31. Sinkovic, L.; Kolmanic, A. Elemental composition and nutritional characteristics of Cucurbita pepo subsp. pepo seeds, oil cake and pumpkin oil. J. Elem. 2021, 26, 97–107. [Google Scholar] [CrossRef]
  32. Glew, R.H.; Glew, R.S.; Chuang, L.T.; Huang, Y.S.; Millson, M.; Constans, D.; Vanderjagt, D.J. Amino acid, mineral and fatty acid content of pumpkin seeds (Cucurbita spp) and Cyperus esculentus nuts in the Republic of Niger. Plant Foods Hum. Nutr. 2006, 61, 51–56. [Google Scholar] [CrossRef]
  33. Intrenational Council for Harmonisation (ICH). Guideline for Elemental Impurities Q3D (R2); European Medicines Agency: Amsterdam, The Netherlands, 2022. [Google Scholar]
  34. Lewis, J. Codex Nutrient Reference Values; FAO-WHO: Rome, Italy, 2019. [Google Scholar]
  35. Elinge, C.; Muhammad, A.; Atiku, F.; Itodo, A.; Peni, I.; Sanni, O.; Mbongo, A. Proximate, mineral and anti-nutrient composition of pumpkin (Cucurbita pepo L) seeds extract. Int. J. Plant. Res. 2012, 2, 146–150. [Google Scholar] [CrossRef]
  36. Cindric, I.J.; Zeiner, M.; Steffan, I. Trace elemental characterization of edible oils by ICP–AES and GFAAS. Microchem. J. 2007, 85, 136–139. [Google Scholar] [CrossRef]
  37. Grzybek, M.; Kukula-Koch, W.; Strachecka, A.; Jaworska, A.; Phiri, A.M.; Paleolog, J.; Tomczuk, K. Evaluation of Anthelmintic Activity and Composition of Pumpkin (Cucurbita pepo L.) Seed Extracts-In Vitro and in Vivo Studies. Int. J. Mol. Sci. 2016, 17, 1456. [Google Scholar] [CrossRef]
  38. Wang, D.C.; Xiang, H.; Li, D.; Gao, H.Y.; Cai, H.; Wu, L.J.; Deng, X.M. Purine-containing cucurbitane triterpenoids from Cucurbita pepo cv dayangua. Phytochemistry 2008, 69, 1434–1438. [Google Scholar] [CrossRef]
  39. Macedo, C.; Silva, A.M.; Ferreira, A.S.; Moreira, M.M.; Delerue-Matos, C.; Rodrigues, F. Microwave- and ultrasound-assisted extraction of Cucurbita pepo seeds: A Comparison study of antioxidant activity, phenolic profile, and in vitro cells effects. Appl. Sci. 2022, 12, 1763. [Google Scholar] [CrossRef]
  40. Peričin, D.; Krimer, V.; Trivić, S.; Radulović, L. The distribution of phenolic acids in pumpkin’s hull-less seed, skin, oil cake meal, dehulled kernel and hull. Food Chem. 2009, 113, 450–456. [Google Scholar] [CrossRef]
  41. Koike, K.; Li, W.; Liu, L.; Hata, E.; Nikaido, T. New phenolic glycosides from the seeds of Cucurbita moschata. Chem. Pharm. Bull. 2005, 53, 225–228. [Google Scholar] [CrossRef]
  42. Li, W.; Koike, K.; Tatsuzaki, M.; Koide, A.; Nikaido, T. Cucurbitosides F−M, Acylated Phenolic Glycosides from the Seeds of Cucurbita pepo. J. Nat. Prod. 2005, 68, 1754–1757. [Google Scholar] [CrossRef]
  43. Lange, T.; Kappler, J.; Fischer, A.; Frisse, A.; Padeffke, T.; Schmidtke, S.; Lange, M.J. Gibberellin biosynthesis in developing pumpkin seedlings. Plant Physiol. 2005, 139, 213–223. [Google Scholar] [CrossRef]
  44. Mejía-Morales, C.; Rodríguez-Macías, R.; Salcedo-Pérez, E.; Zamora-Natera, J.; Rodríguez-Zaragoza, F.; Molina-Torres, J.; Délano-Frier, J.; Zañudo-Hernández, J. Contrasting metabolic fingerprints and seed protein profiles of Cucurbita foetidissima and C. radicans fruits from feral plants sampled in central Mexico. Plants 2021, 10, 2451. [Google Scholar] [CrossRef]
  45. Salvi, P.; Varshney, V.; Majee, M. Raffinose family oligosaccharides (RFOs): Role in seed vigor and longevity. Biosci. Rep. 2022, 42, BSR20212896. [Google Scholar] [CrossRef]
  46. Mansour, E.H.; Dworschák, E.; Lugasi, A.; Barna, É.; Gergely, A. Nutritive value of pumpkin (Cucurbita pepo Kakai 35) seed products. J. Sci. Food. Agric. 1993, 61, 73–78. [Google Scholar] [CrossRef]
  47. Polyzos, N.; Fernandes, Â.; Calhelha, R.C.; Petrović, J.; Soković, M.; Ferreira, I.C.; Barros, L.; Petropoulos, S.A. Biochemical composition of pumpkin seeds and seed by-products. Plants 2024, 13, 2395. [Google Scholar] [CrossRef]
  48. Horak, J.; Gerhardt, H.; Theiner, J.; Lindner, W. Correlation between amino acid racemization and processing conditions for various wheat products, oil seed press cakes and lignin samples. Food Bioprod. Process. 2014, 92, 355–368. [Google Scholar] [CrossRef]
  49. Krimer-Malešević, V. Pumpkin seeds: Phenolic acids in pumpkin seed (Cucurbita pepo L.). In Nuts and Seeds in Health and Disease Prevention; Preedy, V.R., Watson, R.R., Eds.; Academic Press: London, UK, 2020; pp. 533–542. [Google Scholar]
  50. Abuelgassim, A.O.; Al-Showayman, S.I. The effect of pumpkin (Cucurbita pepo L.) seeds and L-arginine supplementation on serum lipid concentrations in atherogenic rats. Afr. J. Tradit. Complement. Altern. Med. 2012, 9, 131–137. [Google Scholar] [CrossRef]
  51. Mustățea, G.; Ungureanu, E.L.; Iorga, E. Protein acidic hydrolysis for amino acids analysis in food-progress over time: A short review. J. Hyg. Eng. Des. 2019, 26, 81–87. [Google Scholar]
  52. Müller, A.; Nunes, M.T.; Maldaner, V.; Coradi, P.C.; de Moraes, R.S.; Martens, S.; Leal, A.F.; Pereira, V.F.; Marin, C.K. Rice drying, storage and processing: Effects of post-harvest operations on grain quality. Rice Sci. 2022, 29, 16–30. [Google Scholar] [CrossRef]
  53. Nguyen, V.; Taine, E.G.; Meng, D.; Cui, T.; Tan, W. Pharmacological activities, therapeutic effects, and mechanistic actions of trigonelline. Int. J. Mol. Sci. 2024, 25, 3385. [Google Scholar] [CrossRef]
  54. Li, Y.; Ramaswamy, S.H.; Li, J.; Gao, Y.; Yang, C.; Zhang, X.; Irshad, A.; Ren, Y. Nutrient evaluation of the seed, pulp, flesh, and peel of spaghetti squash. Food Sci. Technol. 2021, 42, 1–9. [Google Scholar] [CrossRef]
  55. Appendino, G.; Jakupovic, J.; Belloro, E.; Marchesini, A. Triterpenoid p-aminobenzoates from the seeds of zucchini. Fitoterapia 2000, 71, 258–263. [Google Scholar] [CrossRef]
  56. Appendino, G.; Jakupovic, J.; Belloro, E.; Marchesini, A. Multiflorane triterpenoid esters from pumpkin. An unexpected extrafolic source of PABA. Phytochemistry 1999, 51, 1021–1026. [Google Scholar] [CrossRef]
  57. Sicilia, T.; Niemeyer, H.B.; Honig, D.M.; Metzler, M. Identification and stereochemical characterization of lignans in flaxseed and pumpkin seeds. J. Agric. Food Chem. 2003, 51, 1181–1188. [Google Scholar] [CrossRef]
  58. Milder, I.E.; Arts, I.C.; van de Putte, B.; Venema, D.P.; Hollman, P.C. Lignan contents of Dutch plant foods: A database including lariciresinol, pinoresinol, secoisolariciresinol and matairesinol. Br. J. Nutr. 2005, 93, 393–402. [Google Scholar] [CrossRef]
  59. Faria-e-Silva, A.; Heckel, L.; Belli, R.; Lohbauer, U. Determination of water content in direct resin composites using coulometric Karl Fischer Titration. Materials 2022, 15, 8524. [Google Scholar] [CrossRef]
  60. Masuko, T.; Minami, A.; Iwasaki, N.; Majima, T.; Nishimura, S.; Lee, Y.C. Carbohydrate analysis by a phenol-sulfuric acid method in microplate format. Anal. Biochem. 2005, 339, 69–72. [Google Scholar] [CrossRef]
  61. Phenomenex Inc. Luna Omega Sugar Application Guide; Phenomenex Inc.: Torrance, CA, USA, 2018; p. 23. [Google Scholar]
  62. Smith, P.K.; Krohn, R.I.; Hermanson, G.; Mallia, A.; Gartner, F.; Provenzano, M.; Fujimoto, E.; Goeke, N.; Olson, B.; Klenk, D. Measurement of protein using bicinchoninic acid. Anal. Biochem. 1985, 150, 76–85. [Google Scholar] [CrossRef]
  63. Guba, A.; Bába, O.; Tőzsér, J.; Csősz, É.; Kalló, G. Fast and sensitive quantification of AccQ-Tag derivatized amino acids and biogenic amines by UHPLC-UV analysis from complex biological samples. Metabolites 2022, 12, 272. [Google Scholar] [CrossRef]
  64. Švarc-Gajić, J.; Rodrigues, F.; Moreira, M.M.; Delerue-Matos, C.; Morais, S.; Dorosh, O.; Silva, A.M.; Bassani, A.; Dzedik, V.; Spigno, G. Chemical composition and bioactivity of oilseed cake extracts obtained by subcritical and modified subcritical water. Bioresour. Bioprocess. 2022, 9, 114. [Google Scholar] [CrossRef]
  65. Murkovic, M.; Piironen, V.; Lampi, A.M.; Kraushofer, T.; Sontag, G. Changes in chemical composition of pumpkin seeds during the roasting process for production of pumpkin seed oil (Part 1: Non-volatile compounds). Food Chem. 2004, 84, 359–365. [Google Scholar] [CrossRef]
  66. Abdelmonsef, M.; Shawky, E.; Ghareeb, D.A.; El Naggar, E.M.B.; El Newehy, N.M. Comprehensive metabolomics and chemometrics unravel potential anti-diabetic metabolites of pumpkin (Cucurbita pepo L.) fruits through UPLC-QqQ-MS and GC–MS analyses. Food Res. Int. 2024, 192, 114771. [Google Scholar] [CrossRef]
  67. Ghallab, D.S.; Ghareeb, D.A. UPLC-MS/MS-based metabolomics and chemometrics unveil metabolic patterns and their regulatory pathways over the course of pumpkin seeds (Cucurbita maxima) germination. Sci. Hortic. 2024, 337, 113563. [Google Scholar] [CrossRef]
Figure 1. Bar chart of mean elemental concentrations (µg/g dw) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates). (A) Higher-abundance elements (Na, K, Mg, Cu > 500 µg/g dw in at least one extract). (B) Lower-abundance elements (Zn, Cr, Ni, Fe, Co, Mn). Grouping was based on concentration range allowing appropriate scaling and clearer visualization of differences across samples. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 1. Bar chart of mean elemental concentrations (µg/g dw) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates). (A) Higher-abundance elements (Na, K, Mg, Cu > 500 µg/g dw in at least one extract). (B) Lower-abundance elements (Zn, Cr, Ni, Fe, Co, Mn). Grouping was based on concentration range allowing appropriate scaling and clearer visualization of differences across samples. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g001
Figure 2. (A) Total carbohydrate content in CP seed hydrophilic extracts expressed as mean ± SD (n = 3 technical replicates). (B) Dot plot revealing the Spearman correlation between the sum of quantified saccharides and the total carbohydrate content. (C) Bar chart showing the content of individually quantified saccharides in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 2. (A) Total carbohydrate content in CP seed hydrophilic extracts expressed as mean ± SD (n = 3 technical replicates). (B) Dot plot revealing the Spearman correlation between the sum of quantified saccharides and the total carbohydrate content. (C) Bar chart showing the content of individually quantified saccharides in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g002
Figure 3. Bar chart of mean protein contents (mg/g dw) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 3. Bar chart of mean protein contents (mg/g dw) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g003
Figure 4. Heat map illustrating the concentrations of free amino acids (mg/g dw) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean (n = 3 technical replicates). The color gradient reflects the variation in concentration across the samples, with red indicating higher concentrations and blue indicating lower concentrations. Analyzed amino acids include the following: asparagine (Asn), serine (Ser), aspartic acid (Asp), glutamine (Gln), glycine (Gly), histidine (His), glutamic acid (Glu), arginine (Arg), threonine (Thr), alanine (Ala), proline (Pro), tyrosine (Tyr), methionine (Met), valine (Val), cucurbitin (Cuc), lysine (Lys), isoleucine (Ile), phenylalanine (Phe), leucine (Leu), tryptophan (Trp), ethanolamine (Eta), and gamma-aminobutyric acid (GABA). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 4. Heat map illustrating the concentrations of free amino acids (mg/g dw) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean (n = 3 technical replicates). The color gradient reflects the variation in concentration across the samples, with red indicating higher concentrations and blue indicating lower concentrations. Analyzed amino acids include the following: asparagine (Asn), serine (Ser), aspartic acid (Asp), glutamine (Gln), glycine (Gly), histidine (His), glutamic acid (Glu), arginine (Arg), threonine (Thr), alanine (Ala), proline (Pro), tyrosine (Tyr), methionine (Met), valine (Val), cucurbitin (Cuc), lysine (Lys), isoleucine (Ile), phenylalanine (Phe), leucine (Leu), tryptophan (Trp), ethanolamine (Eta), and gamma-aminobutyric acid (GABA). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g004
Figure 5. Lignans quantification in hydrophilic extracts of Cucurbita pepo (CP) seeds, expressed as mean ± SD (n = 3 technical replicates). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 5. Lignans quantification in hydrophilic extracts of Cucurbita pepo (CP) seeds, expressed as mean ± SD (n = 3 technical replicates). Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g005
Figure 6. Contribution of various phytochemicals to the total mass of Cucurbita pepo (CP) seed hydrophilic extracts. Minor compounds below 0.5% at the bar-top region are shown as an inset. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 6. Contribution of various phytochemicals to the total mass of Cucurbita pepo (CP) seed hydrophilic extracts. Minor compounds below 0.5% at the bar-top region are shown as an inset. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g006
Figure 7. Principal component analysis (PCA) showing the variability of the phytochemical composition of Cucurbita pepo (CP) seed hydrophilic extracts. (A) Bar chart showing the percentage of explained variance for each principal component. (B) Eigenvalues of each PC. (C) Loading plot displaying the relationship between phytochemicals and the two significant PCs. (D) Score plot showing the samples distribution in the two-dimensional plans obtained from two significant PCs. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 7. Principal component analysis (PCA) showing the variability of the phytochemical composition of Cucurbita pepo (CP) seed hydrophilic extracts. (A) Bar chart showing the percentage of explained variance for each principal component. (B) Eigenvalues of each PC. (C) Loading plot displaying the relationship between phytochemicals and the two significant PCs. (D) Score plot showing the samples distribution in the two-dimensional plans obtained from two significant PCs. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g007
Figure 8. Hierarchical clustering analysis of Cucurbita pepo (CP) seed hydrophilic extracts based on 35 phytochemical variables. Clustering was performed using Ward’s linkage method and Euclidean distance. Two main clusters were identified, with cluster I further divided into three sub-clusters, reflecting varying degrees of metabolic similarity among the samples. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 8. Hierarchical clustering analysis of Cucurbita pepo (CP) seed hydrophilic extracts based on 35 phytochemical variables. Clustering was performed using Ward’s linkage method and Euclidean distance. Two main clusters were identified, with cluster I further divided into three sub-clusters, reflecting varying degrees of metabolic similarity among the samples. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g008
Figure 9. Cucurbita pepo (CP) seeds used in the study. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Figure 9. Cucurbita pepo (CP) seeds used in the study. Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Plants 14 02308 g009
Table 1. Levels (mg/g dw) of total organic carbon (TOC), inorganic carbon (IC), total carbon (TC), and total nitrogen (TN) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates).
Table 1. Levels (mg/g dw) of total organic carbon (TOC), inorganic carbon (IC), total carbon (TC), and total nitrogen (TN) in Cucurbita pepo (CP) seed hydrophilic extracts, expressed as mean ± SD (n = 3 technical replicates).
Nr.SampleTOC [mg/g]IC [mg/g]TC [mg/g]TN [mg/g]
1SK449.37 ± 1.052.95 ± 0.17452.32 ± 1.0414.32 ± 0.02
2SKR340.61 ± 3.343.68 ± 0.07344.29 ± 3.3419.25 ± 0.16
3LN394.42 ± 0.682.35 ± 0.03396.77 ± 0.6716.22 ± 0.07
4SW337.48 ± 1.682.46 ± 0.01339.94 ± 1.6826.57 ± 0.03
5RZ397.61 ± 3.082.08 ± 0.01399.69 ± 3.0814.39 ± 0.18
6SS341.83 ± 1.914.55 ± 0.03346.37 ± 1.9113.32 ± 0.07
7GA306.43 ± 0.432.31 ± 0.15308.74 ± 0.4012.59 ± 0.09
8GV528.16 ± 0.753.11 ± 0.13531.27 ± 0.7413.59 ± 0.02
9BBZ297.33 ± 1.957.05 ± 0.13304.38 ± 1.9514.81 ± 0.14
10HV444.47 ± 1.103.29 ± 0.24447.76 ± 1.0727.09 ± 0.24
Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Table 2. Trigonelline contents expressed as mean (mg/g dw) ± SD (n = 3 technical replicates) in the studied hydrophilic extracts of Cucurbita pepo (CP) seeds.
Table 2. Trigonelline contents expressed as mean (mg/g dw) ± SD (n = 3 technical replicates) in the studied hydrophilic extracts of Cucurbita pepo (CP) seeds.
NumberExtractTrigonelline Content
[mg/g]
±SD
1SK1.7640.004
2SKR1.6850.002
3LN1.7650.023
4SW3.4470.027
5RZ0.4590.008
6SS1.9750.001
7GA0.9850.002
8GV0.4130.016
9BBZ0.4070.006
10HV1.3330.002
Sample codifications are as follows: SK (CP convar. citrullina var. styriaca), SKR (CP var. styriaca cultivar Gleisdorfer Rustikal), LN (CP—Lady Nail), SW (CP—Snow White), RZ (CP var. giromontia—Radu), SS (CP—Shine Skin), GA (CP—Greek Cultivar), GV (CP—Grey Volga), BBZ (CP var. cylindrica—Black Beauty), HV (CP—Hungarian Cultivar).
Table 3. Description of Cucurbita pepo seeds used in the study.
Table 3. Description of Cucurbita pepo seeds used in the study.
Nr.NameCodeExtraction Yield [g]Country of OriginDistributorBatchMacroscopy
1C. pepo convar. citrullina, var styriacaSK10.25AustriaEstyria Naturprodukte GmbH21-039676 0001Medium, dark green seeds, with a very thin shell and a pointed tip. Length 1.3–1.7 cm, width 0.4–0.6 cm.
2C. pepo var.
styriaca cultivar Gleisdorfer Rustikal
SKR10.01SwitzerlandVitaplant AGCUP 22_2202Medium, dark green seeds, with a very thin shell and a pointed tip. Length 1.2–2.1 cm, width 0.5–0.7 cm.
3C. pepo—Lady NailLN11.21SwitzerlandVitaplant AGCUP 22_2021Medium, sharp light-yellow seeds. Length 1.5–2.0 cm, width 0.4–0.5 cm.
4C. pepo—Snow WhiteSW15.63SwitzerlandVitaplant AGCUP 22_2103Large, with a thick shell, and light-yellow seeds. Length 2.0–2.5 cm, width 1.0–1.4 cm.
5C. pepo var. giromontia -RaduRZ10.35RomaniaAgrosem Impex2MS1196Small, round, light yellow seeds.
Length 0.8–1.5 cm, width 0.5–0.6 cm.
6C. pepo—Shine SkinSS13.37UkraineM.A.R. Pumpkin SeedsCUS18006/20Medium, with a thick shell, pointed tip, and light-yellow seeds. Length 1.5–2.5 cm, width 0.8–1.0 cm.
7C. pepo -Greek CultivarGA10.21GreeceAntonio FoodsCUS19001(S/K/H)Medium, narrow and sharp, light-yellow seeds. Length 1.4–2.1 cm, width 0.4–0.5 cm.
8C. pepo—Grey VolgaGV12.44UkraineM.A.R. Pumpkin SeedsCUS18005/20Medium, with a soft smooth shell, and light-yellow seeds.
Length 1.5–2.2 cm, width 0.9–1.0 cm.
9C. pepo var. cylindrica—Black BeautyBBZ10.33RomaniaAgrosem Impex2MS1197Small, round, light yellow seeds. Length 0.9–1.2 cm, width 0.5–0.6 cm.
10C. pepo—Hungarian CultivarHV14.67HungaryH.F.I. Hungarian Food IngredientsCUS18008/20Medium, round, with a thick orange shell. Length 1.5–2.2 cm, width 0.7–1.0 cm.
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

Grasu, A.-E.; Senn, R.; Halbsguth, C.; Schenk, A.; Butterweck, V.; Miron, A. Profiling Hydrophilic Cucurbita pepo Seed Extracts: A Study of European Cultivar Variability. Plants 2025, 14, 2308. https://doi.org/10.3390/plants14152308

AMA Style

Grasu A-E, Senn R, Halbsguth C, Schenk A, Butterweck V, Miron A. Profiling Hydrophilic Cucurbita pepo Seed Extracts: A Study of European Cultivar Variability. Plants. 2025; 14(15):2308. https://doi.org/10.3390/plants14152308

Chicago/Turabian Style

Grasu, Adina-Elena, Roman Senn, Christiane Halbsguth, Alexander Schenk, Veronika Butterweck, and Anca Miron. 2025. "Profiling Hydrophilic Cucurbita pepo Seed Extracts: A Study of European Cultivar Variability" Plants 14, no. 15: 2308. https://doi.org/10.3390/plants14152308

APA Style

Grasu, A.-E., Senn, R., Halbsguth, C., Schenk, A., Butterweck, V., & Miron, A. (2025). Profiling Hydrophilic Cucurbita pepo Seed Extracts: A Study of European Cultivar Variability. Plants, 14(15), 2308. https://doi.org/10.3390/plants14152308

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

Article Metrics

Back to TopTop