Next Article in Journal
Assessing the Benefits and Costs of the Hydrogen Cyanide Antiherbivore Defense in Trifolium repens
Next Article in Special Issue
Formulation and Biopharmaceutical Evaluation of Capsules Containing Freeze-Dried Cranberry Fruit Powder
Previous Article in Journal
Changes in Metal-Chelating Metabolites Induced by Drought and a Root Microbiome in Wheat
Previous Article in Special Issue
Antibacterial, Antioxidant Activities, GC-Mass Characterization, and Cyto/Genotoxicity Effect of Green Synthesis of Silver Nanoparticles Using Latex of Cynanchum acutum L
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Biological and Chemical Insights into Optimization of Chamomile Extracts by Using Artificial Neural Network (ANN) Model

1
Faculty of Technology, University of Novi Sad, Bulevar Cara Lazara 1, 21000 Novi Sad, Serbia
2
Department of Biology, Science Faculty, Selcuk University, Konya 42130, Turkey
3
Institute of Chemical Industry of Forestry Products, Chinese Academy of Forestry, Nanjing 210042, China
4
Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
5
Institute of Food Technology, University of Novi Sad, Bulevar Cara Lazara 1, 21000 Novi Sad, Serbia
*
Author to whom correspondence should be addressed.
Plants 2023, 12(6), 1211; https://doi.org/10.3390/plants12061211
Submission received: 24 November 2022 / Revised: 9 February 2023 / Accepted: 20 February 2023 / Published: 7 March 2023

Abstract

:
Chamomile is one of the most consumed medicinal plants worldwide. Various chamomile preparations are widely used in various branches of both traditional and modern pharmacy. However, in order to obtain an extract with a high content of the desired components, it is necessary to optimize key extraction parameters. In the present study, optimization of process parameters was performed using the artificial neural networks (ANN) model using a solid-to-solvent ratio, microwave power and time as inputs, while the outputs were the yield of the total phenolic compounds (TPC). Optimized extraction conditions were as follows: a solid-to-solvent ratio of 1:80, microwave power of 400 W, extraction time of 30 min. ANN predicted the content of the total phenolic compounds, which was later experimentally confirmed. The extract obtained under optimal conditions was characterized by rich composition and high biological activity. Additionally, chamomile extract showed promising properties as growth media for probiotics. The study could make a valuable scientific contribution to the application of modern statistical designs and modelling to improve extraction techniques.

1. Introduction

Chamomile (Chamomilla recutita L.) is an annual plant whose medicinal properties have been known to almost all civilizations of the world for centuries, and some ancient civilizations considered it as a sacred plant. Besides its healing properties, its mild effect on the body without toxicity is of paramount importance when it comes to using it as a tea and/or as a medicine for babies and adults [1]. Among all its uses, its use in the fight against gastrointestinal diseases is particularly important, and there is almost no such type of disease for which chamomile cannot be used as medicine or prevention. In addition, its role as an antidepressant, its effect against insomnia, its use for rinsing wounds, cuts, and rashes, have been noted [2]. Precisely because of the beneficial effect on the skin, there are numerous skin care preparations based on chamomile. Modern science has established its anti-inflammatory, antiphlogistic [3], anti-allergic [4], antibacterial [5,6,7], antispasmodic, antiseptic [8], as well as antioxidant activity through numerous in vitro and in vivo tests [9]. More than 100 different compounds make up its composition and contribute to its activity and wide use. Studies show that the correct choice of extraction technique, solvents, and extraction conditions can favor the content of individual components in the final extract, and that to some extent the content of certain bioactive components can be favored, which can increase the bioactivity of the final product [10].
Among modern extraction techniques, microwave-assisted extraction (MAE) plays a particularly important role in the laboratory and increasingly in industry. MAE is a modern technique of extraction that combines traditional (solid–liquid extraction) with microwave heating to achieve a higher process effect [1]. In addition, the extraction process can be enhanced by enzymes. In general, enzymes play a key role in extraction as biocatalysts of highly specific reactions under mild conditions that drive the reaction in the desired direction, in an environmentally friendly environment (water), which characterizes the technique as green. Enzymes can degrade or hydrolyze cell wall elements and membranes, allowing for the better release and more efficient extraction of bioactive molecules. Enzymes such as beta-glucosidase can hydrolyze the bound form of molecules (such as flavonoid glucosides) and further increase the levels of the free form of these molecules (aglycones). In this way, the amount of aglycones (which often can be more bioactive) is higher, thereby increasing the bioactivity of the final extract [9]. However, in order to lead the processes in the desired direction and to be economically viable, it is necessary to know exactly the connection and impact of all key extraction parameters and target compounds.
In MAE, it is very hard to find a theoretical model that explains the connection between input and outputs variables. Nowadays, with the evolution of hardware, software consequently becomes more powerful and data driven modelling such as artificial neural networks (ANNs) find an everyday use in science. ANN, as a direct and fast analysis of experimental data, can serve as an adequate alternative to models based on phenomenological hypotheses. It is worth nothing that the ANN models have already been applied in the extraction processes [11,12,13]. New and powerful optimization techniques have been developed parallel with data-driven modelling. Therefore, in order to overcome the possible local/minimal/global problems, optimization was settled. The two most used solvers in global optimization are the particle swarm and genetic algorithm. Each solver has its own characteristics that lead to different solutions. In order to improve solver effectiveness, many parameters need to be adjusted and different search functions have to be tested, so there is still place for progress in this scientific field. Particle swarm is a population-based algorithm. It is very similar to the genetic algorithm. Kennedy and Eberhart [14] introduced the particle swarm algorithm. Later, Mezura-Montes and Coello [15] improved the algorithm. It started by creating initial particles and assigning them the initial velocity values. A group of the particles move in steps through an area. The algorithm at each step determines the objective function of each particle. It determines the lowest function value and the best particle location. When the particles move, the algorithm reevaluates. It chooses new velocities based on the current velocity and the best location of the particles. It then iteratively updates the velocities and locations. This proceeds until the algorithm reaches a stopping criterion and finds the global optima.
One of the goals of this study was to optimize the microwave-assisted extraction process combined with enzymes in order to obtain the highest yield of phenols. The greater performance of prediction models is still the first-line goal in academia and the industrial community, so the ANN was used to algorithmically map input and outputs variables. The ANN is a black box model, and as the fitting function for optimization problems, it can lead to a solution that represents the local minimums. To overcome this, in the last phase we used a global optimization to find the best operating parameters for the microwave extraction. In terms of increasing the added value of the extracts, the growth of potential probiotic bacteria on the obtained extract was also evaluated.

2. Results and Discussion

2.1. Extraction and Optimization of the Extraction via ANN

One of the crucial moments in the process of obtaining natural products with added value is the process of extraction. The choice of extraction technique as well as the choice of solvent can significantly influence the presence and concentration of bioactive substances in the final extract. In this work, chamomile extraction was performed via MAE with ethanol. Ethanol is suitable for extracting plant matter because it is labeled as GRAS and can easily penetrate the pores of the plant matrix. MAE combined with ethanol is considered a green technique that requires less extraction time compared to traditional techniques [10]. Microwave heating also increases the temperature of the mixture, resulting in a better extraction performance. However, when extracting thermolabile compounds, high temperatures can lead to the decomposition of the components. The microwave power in the microwave extraction must be chosen sufficiently to avoid too-high temperatures, which leads to the decomposition of heat-sensitive substances [16,17,18]. Within this work, the ANN analysis was applied with the aim of determining the optimal extraction conditions and to gain an insight into the detailed influence of the extraction parameters, as well as to define the optimal extraction conditions for obtaining polyphenol-rich extracts.
Weights and bias initial values could significantly influence the result of the ANN. Also, the number of hidden neurons influence the predictability of the ANN. Hence, a different ANN architecture was tried, and the best ANN showed a good predictability performance. The best fitting ANN topology consisted of three layers with neurons: the input layer had three neurons, the hidden layer had eight neurons, and the output layer had one neuron [19]. The quality of the fit with respect to the coefficient of determination (R2) and mean absolute error (MAE) is presented in Figure 1, showing the parity plot of the experimental and predicted TPC yield. The parity plot demonstrated the generalization ability of the developed ANN. The ANNs were trained on the 70% randomly sampled points and were tested on the 30% remaining points.
In the next step, a developed ANN was incorporated into the Yoon’s model in order to determine the relative influence of the process parameters. The obtained relative importance values and the standard deviations are presented in Figure 2. A low variability in RI %, about 5%, makes the explanation of the input influence acceptable. Figure 2 shows that solid-to-solvent is the most influential parameter. The TPC yield decreased when decreasing the solid-to-solvent ratio from 1:80 to 1:40. From our experiments, it is important to note that the solid-to-solvent ratio of 1:40 produced the smallest TPC yield up to 46 mg/g. Hence, this leads to the conclusion that the solid-to-solvent ratio should not be smaller than 1:60. The maximal value for a TPC yield, of 59.56 mg/g, was obtained when the time of extraction was the longest, at 40 min, and the maximal microwave power was 800 W. Additionally, a similar TPC yield of 57.32 mg/g was obtained at 800 W and 20 min. So, this brings the conclusion that there is no need for a high microwave power to perform a long extraction exposure, because that increases the energy consumption. Moreover, from Figure 2 it can be seen that the microwave power has the smallest influence on the TPC yield. This is experimentally confirmed; the second highest value for TPC yield was 58.435 mg/g, regardless of the minimal microwave power of 400 W.
The ANN used in this study represents the black box model, so standard optimization methods such as response surface methodology can be trapped into the local minimum. This was reason to use global search solvers such as the particle swarm algorithm. Therefore, in the final stage, the particle swarm optimization determined the following operating parameters: 400 W, 30 min, and a 1:80 solid-to-solvent ratio. The predicted TPC yield obtained with the above-mentioned conditions was 59.56 mg/g. These results have been experimentally verified.

2.2. Chemical Profiling

Chamomile extract obtained under previously defined optimal conditions was exposed to the chemical profiling analysis. The first spectrophotometric assays were applied to determine the total phenol and flavonoid content, and the obtained results are presented in Figure 3. As can be seen, the total phenol content was 55.21 mg GAE/g, which was very close to the predicted value, confirming the ANN model. Analysis of the total amount of flavonoids showed the content of this bioactive compounds to be 44.98 mg RE/g, which represents more than 80% of the total polyphenolic compounds in the extracts, showing that flavonoids are the dominant group of polyphenols presented in chamomile.
Apart from the spectrophotometric analysis, an analysis of the polyphenolic profile of the extract obtained under the optimal extraction conditions was performed as well. For this purpose, the UHPLC-LTQ OrbiTrap MS technique was used and a total of 67 compounds were identified (Table 1).
Fifteen compounds were identified via a comparison with the available analytical standards, while the others were tentatively identified by the search for [M–H]− deprotonated molecules in combination with MS4 fragmentations. To facilitate the understanding of the obtained results, the identified compounds are divided into two groups: (1) phenolic acids and their derivatives (32 compounds) and (2) flavonoid glycosides and aglycones (35 compounds) (Table 1).
The most represented compounds from the group of phenolic acids and their derivatives were hexosides and quinic acid esters of hydroxycinnamic acids (p-coumaric, caffeic, and ferulic acids). A total of 22 hydroxycinnamates were identified, which is in line with previously published results [20]. The other 10 compounds were hydroxybenzoic acid derivatives. Among all compounds from this subgroup, it is interesting to single out compound 21 from Table 1. In the first stage of fragmentation (MS2 base peak) it gives 413 m/z, which can occur with a loss of 138 Da (mass of hydroxybenzoic acid). Further fragmentation (MS3 base peak) produces an ion at 137 m/z, which also corresponds to the mass of deprotonated hydroxybenzoic acid. The difference between 413 and 137 m/z gives a neutral loss of 276 m/z, which may correspond to deprotonated vicianose (arabinopyranosyl-glucopyranose) or sambubiose (xylosyl-glucopyranose). All these facts show that this compound probably contains two molecules of hydroxybenzoic acid: one pentose and one hexose unit.
Considering the identified flavonoid glycosides and aglycones, the most abundant were apigenin derivatives, as was expected [21]. Nine compounds were confirmed via a comparison with the standards (37, 42–44, 57, 59, 63, 65, and 66) and the others were tentatively identified via examination of the MS spectra. Some flavonoids specific for chamomile, such as 6-methoxyapigenin or hispidulin, 6-methoxyluteolin or nepetin [22], 6-hydroxyquercetin or quercetagetin [23], and 6-methoxyquercetin or patuletin [24], were also found in this study. Compound 50 was marked as an apigenin derivative, because its exact structure could not be suggested. Based on the isotopic composition and exact mass (569.12927 m/z), the chemical formula of this compound (C28H26O13) can be calculated. In the first fragmentation step, this compound loses the 300 Da and thus the MS2 base peak at 269 m/z was formed. Further MS3 and MS4 fragmentation confirms the presence of apigenin as one part of this compound, because it is in accordance with the fragmentation of compound 41 (apigenin 7-O-glucoside).
By using standards, quantification was performed for the 15 compounds, and the obtained results are presented in Table 2. Among the phenolic acids, p-hydroxybenzoic was presented in the highest concentration (1.619 mg/L), which was in accordance with the literature data, and which suggests that it is the most abundant compared to other phenolic acids present in chamomile. Regarding the flavonoids, as was expected, apigenin and apigenin-7-O-glucoside were the dominant compounds. Generally, apigenin is considered as one of the leading constituents of chamomile and responsible for many of its biological activities [9]. In addition to apigenin, the concentration of quercetin, which is considered an extremely potent biological molecule, was also very high (0.861 mg/L), and is of notable importance for the overall activity of the extracts. Namely, this compound is known as one of the most potent ingredients of many plants and possesses antioxidant, antimicrobial, antitumor, anti-inflammatory, and immunosuppressive effects, among many others [25]. Kaempferol was present in the extract with a concentration of 0.358 mg/L. The presence of kaempferol can have a significant role in the biological activity of chamomile extracts due to the wide range of its biological activities. This compound is used for reducing the risk of chronic diseases, such as cancer, liver injury, obesity, and diabetes [26,27]. Because of its anti-inflammatory potential, kaempferol is used for different inflammation-induced diseases, such as intervertebral disc degeneration and colitis, post-menopausal bone loss, and acute lung injury [28].

2.3. Biological Activity

2.3.1. Antioxidant Activity

When it comes to the biological activity of natural products, one of the fundamental activities is antioxidant activity. The very abundant presence of this activity is the basis for other biological activities such as antimicrobial and cardioprotective activities, etc. There are generally two ways in which the precursor compounds exert their antioxidant activity. Namely, they can prevent the oxidation process or neutralize free radicals. For this very reason, when determining the antioxidant capacity of natural products, it is desirable to use several different tests based on different mechanisms of action. In this study, the antioxidant capacity of the chamomile extract was tested using six different tests, which gives a more realistic insight into the potential of the obtained extract to be used as a source of antioxidants (Table 3). Two applied antiradical assays confirmed that the chamomile extract possessed a high potential to neutralize DPPH (60.24 mg TE/g) and ABTS (126.92 mg TE/g) free radicals. By applying two reduction assays, the high reduction ability of the extract was confirmed. Namely, by using CUPRAC (123.12 mgTE/g) and FRAP (95.37 mg TE/g) tests, the chamomile-optimized extract showed its high reduction capability. Results obtained for the chelating metal test (21.34 mg EDTAE/g) express higher activity than the activity of other plant species obtained via the same extraction technique. Namely, in our previous published data, Origanum vulgare (6.49 mg EDTAE/g) [29] and Tanacetum parthenium (10.83 mg EDTAE/g) [30] extracts obtained via the MAE extraction technique express lower antioxidant potential in comparison with MAE chamomile extract obtained in this study.

2.3.2. Enzyme-Inhibitory Activity

Enzymes are important therapeutic targets for the treatment of global health issues such as Alzheimer’s disease, diabetes, and obesity. Some enzymes can be targeted for this purpose, and their inhibition can reduce disease symptoms. For instance, amylase and glucosidase are known as anti-diabetic enzymes, and their inhibition can help diabetic patients control their blood glucose levels [31]. Several compounds have been synthesized in the pharmaceutical industry as enzyme inhibitors, but the majority of them have undesirable side effects. Thus, there is a great deal of interest in replacing natural enzyme inhibitors with synthetic ones. The enzyme inhibitory properties of the optimized chamomile extract against acetylcholinesterase (AChE), amylase, and glucosidase were investigated. The results are tabulated in Table 3. The optimized extracts exhibited inhibitory properties on the tested enzymes (AChE: 0.85 mg of GALAE/g; amylase: 0.18 mmol of ACAE/g; and glucosidase: 13.11 mmol of ACAE/g). The observed enzyme inhibitory abilities can be explained by the presence of some compounds in the optimized extract. For instance, several studies have been reported that apigenin has great potential as one of natural inhibitors against AChE [32], amylase, and glucosidase [33]. In addition to apigenin, the presence of caffeic and chlorogenic acids can be attributed to observed abilities [34,35]. Our results are comparable to the reported values in the literature. For example, in our previous study [36], the chamomile extracts from subcritical water extracts under different pressures exhibited inhibitory effects on amylase (0.44–0.46 mmol ACAE/g) and glucosidase (2.42–3.60 mmol ACAE/g). In another study [37], the amylase- and glucosidase-inhibitory effects of the optimized extract for apigenin isolation were 0.45 mmol of ACAE/g and 2.54 mmol of ACAE/g, respectively. The values were also reported as 0.94 mmol of ACAE/g and 3.24 mmol of ACAE/g for autofermented chamomile ligulate flowers [38]. Taken together, chamomile extracts have significant enzyme inhibitory properties on anti-diabetic enzymes. The results presented could contribute significantly to the design of natural enzyme inhibitors for pharmaceutical applications.

2.4. Evaluation of Potential of MAE for the Growth of La. rhamnosus ATCC 7469

Research shows that plant extracts that are rich in biologically active compounds in combination with probiotics show a significant effect against inflammatory diseases and disorders. Namely, the synergistic action of bioactives and probiotics express a promising potential in the amendment of inflammatory diseases and disorders [39,40]. From those reasons, the influence of chamomile extract on the growth of La. rhamnosus was determined.
As previously stated, since the MAE did not exhibit a significant antimicrobial effect, especially on bacteria (La. rhamnosus ATCC 7469 was also included in microwell dilution method), the potential of MAE and MAEC for growth of La. rhamnosus was investigated. The reducing sugar utilization after 24 h for MAE and MAEC was 68.4 and 82.4%, respectively, and at this point the increase in viability stopped. The initial viability (at 0 h) in all experiments was approximately 8 log CFU/mL, whereas after 24 h the significant increase in viability was observed. The final viability for MAE and MAEC was 9.2 log CFU/mL and 9.7 log CFU/mL, respectively, resulting in significant increases of 15 and 20%. The findings suggest that La. rhamnosus could be successfully grown on this type of chamomile extract. However, the production of lactic acid was not significant (data not shown), suggesting that MAE and MAEC are not complete mediums for lactic acid fermentation, since the reducing sugars concentration is relatively low for efficient lactic acid fermentation. Nevertheless, the MAE and MAEC could be used for enhancement (stimulation) of lactic acid bacteria growth and/or formulation of synergetic bioactive compound-rich probiotic preparations. Marhamatizadeh et al. [41] successfully used Chamomile Essence for enhancement of Bifidobacterium and Lactobacillus strains in milk and yoghurt. Echium amoenum extract was also successfully used for enhancement of the growth of Bifidobacterium and Lactobacillus strains in kefir [42]. Similar results were observed by Dimofte et al. [43] when fermentation media for Weissella confuse PP29 was enriched with deferment concentrations of anthocyanins from Hibiscus sabdariffa L. The fermentation media rich in anthocyanins (high concentrations) stimulated the growth of Weissella confuse and biosynthesis of exopolysaccharide, thus leading to augmented probiotic and prebiotic properties. Martinelli et al. [44] determined (during a clinical trial) that administration of M. chamomilla L., M. officinalis L., tyndallized L. acidophilus (HA122), and L. reuteri DSM 17938 was more effective than simethicone on an infant colic. Synergetic action of probiotic bacteria with herbal extracts (Chamomile (Matricaria chamomilla), fennel (Foeniculum vulgare), peppermint (Mentha piperita), and thyme (Thymus vulgaris) were proven as efficient antibacterial agents against Escherichia coli, Salmonella typhimurium, and Salmonella [45].
The connection of medical plants and probiotics is well known in the traditional medicine of many countries [46]. The current interest, with constantly increasing relevance in relation to healthy supplements and diets, indicates the potential of novel delivery systems that combine probiotics with plant extracts rich in bioactive compounds. This simultaneous and synergistic delivery system could be significantly beneficial for health. In this sense, research on the complementary benefits of herbal extracts and probiotics, as well research on the potential enrichment of food via the incorporation of these bioactive compounds and improvements in probiotic survivability, is highly valuable [47].

3. Material and Methods

3.1. Chemicals

Acetonitrile and formic acid (both of them LC/MS grade) were purchased from Merck (Darmstadt, Germany). Standards of phenolic compounds (gallic acid, protocatechuic acid, p-hydroxybenzoic acid, gentisic acid, caffeic acid, p-coumaric acid, apigenin 7-O-glucoside, luteolin, apigenin, chrysoeriol, quercetin 3-O-galactoside, isorhamnetin 3-O-glucoside, quercetin, and kaempferol) were supplied by Sigma Aldrich (Steinheim, Germany). The 3.5-dinitrosalicylic acid was also supplied by Sigma Aldrich (Steinheim, Germany). Syringe filters (25 mm, nylon membrane, 0.45 µm) were purchased from PSI Lab (Belgrade, Serbia). MRS broth and agar were supplied by HiMedia (Mumbai, India).

3.2. Plant Material

For the purpose of this study, ligulate flowers of chamomile (CLF) were used. Plant material was obtained from the Institute of Field and Vegetable Crops, Novi Sad, Serbia. The collected material was firstly dried (40 °C), and then CLF were separated from the tubular parts of the flower by sieving. Prior to extraction, Dried CLF were treated using a sodium–acetate buffer under the conditions which were optimal for the activity of β-glucosidase [9]. The conditions were kept constant for 72 h and afterwards dried at room temperature for five days. Such plant material was further extracted via ethanol by applying the MAE extraction technique.

3.3. Extraction, Experimental Design, and ANN Optimization

Microwave-assisted extraction (MAE) was performed in a modified domestic microwave oven (Figure 4). In order to define the best conditions for obtaining extract with the maximum phenol content, a 33 full factorial design was chosen. Plant material was mixed with different proportions of ethanol (1:40, 1:60, 1:80) and exposed to microwave irradiation of different powers (400, 600, 800 W) for different periods of time (20, 30, 40 min). These three parameters (solid-to-solvent ratio, microwave power, and time) represented the inputs and were used to feed the artificial neural network. The experimental design with 27 runs is shown in the Table 4. The feed-forward back propagation ANN was used to predict the experimental values for phenol yield outputs.
The transfer function was the linear (purelin) at the output layer and the tangent sigmoid function (tansig) at the hidden layer. The best predictive ability was achieved via the Levenberg–Marquad solver. Successful creation of the ANNs and obtained weight matrices provide the determination of the relative importance (RI) of the input values and its effect on the phenols yield by using a partitioning methodology. In this study, the following Yoon’s equation was used [48]:
R I i j   % = k = 0 n w i k   w k j i = 0 m a b s   k = 0 n w i k   w k j   100 %  
where RIij is the relative importance of the ith input variable on the jth output, wik is the weight between the ith input and the kth hidden neuron, and wkj is the weight between the kth hidden neuron and the jth output. This analysis proved that the decision variables have a conflicting influence on the performance parameters. Thus, to achieve the maximal microwave extraction yield, a global optimization with the particle swarm solver was used. All data analysis was performed using MATLAB 2016b.

3.4. Chemical Analysis of Extracts

3.4.1. Assays for Total Phenolic and Flavonoid Content

The content of two major groups of bioactive compounds (phenols–TPC and flavonoids–TFC) in obtained extracts was determined spectrophotometrically using appropriate Folin–Ciocalteu and aluminium chloride methods [49]. Expression of obtained results was performed using equivalent of standards–gallic acid (in thw case of TPC) and rutin (in the case of TFC).

3.4.2. UHPLC-LTQ OrbiTrap MS Analysis of Polyphenolic Compounds

Separation and tentative identification of bioactive compounds in tested chamomile extract were performed by using an ultra-high-pressure liquid chromatography (UHPLC) system (Accela 600, ThermoFisher Scientific, Bremen, Germany) coupled to linear ion trap/orbitrap mass spectrometer (LTQ OrbiTrap MS) from ThermoFisher Scientific, Bremen, Germany). All the details about chromatographic separation settings [50], electrospray ionization (ESI) source, and MS detector parameters [51] were previously described.
Identification of some compounds was achieved by comparison with the available standard, while other compounds were tentatively identified via high resolution mass spectrometry (HRMS) in combination with multistage mass spectrometry (MSn). The available literature about analyses of bioactive compounds in various chamomile species was used to confirm the identified compounds [20,21,52,53,54,55].

3.5. Determination of Biological Activity of Extracts

To provide comprehensive insights into the biopotential of the extracts, the antioxidant, anti-α-amylase, anti-α-glucosidase, and anti-cholinesterase activities were determined. Estimation of the anti-enzymatic activity of the extracts was conducted via in vitro assays previously described by Uysal et al. [56]. The data obtained via these assays were given as reference inhibitors equivalents: galantamine (GALAE) for acetylcholinesterase (AChE), acarbose (ACAE) for α-amylase and α-glucosidase. Measurements of the antioxidant and free radical-scavenging properties of the extracts, ferric reducing antioxidant power (FRAP), 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid) (ABTS), cupric reducing antioxidant capacity (CUPRAC), 2,2-diphenyl-1-picrylhydrazyl (DPPH), metal chelating, and phosphomolybdenum were performed. The data were given as reference compounds (Trolox (TE) and ethylenediaminetetraacetic (EDTAE) equivalents). Detailed description of applied assays was given previously [57,58,59].

3.6. Evaluation of Chamomile Potential for the Growth of La. rhamnosus ATCC 7469

3.6.1. Preparation of Growth Media and Inoculum, and Bacteria Growth Conditions

Since the MAE did not exert a significant antimicrobial effect, especially towards bacteria (preliminary research), the MAE was used as potential growth medium for potential prebiotic Lacticaseibacillus rhamnosus [60] (previously known as Lactobacillus rhamnosus) [61]. Sterilized MAE without and with prior concentration (MAEC, up to 50% of the original volume) were used as potential growth media of lactic acid bacteria. MAE and MAEC contained 0.7% and 1.3% of reducing sugars, respectively.
The stock culture of the homofermentative lactic acid (LA) strain La. rhamnosus (ATCC, Rockville, MD, USA) was activated as previously described [62]. The inoculums for free cell fermentations were prepared by transferring 3 mL of activated culture into 60 mL of MRS (de Man, Rogosa and Sharpe) broth. Inoculums for immobilization experiments were prepared by adding 7 mL of activated culture into 120 mL of MRS broth. In order to achieve high LAB viability (9 log CFU/mL), all inoculums were incubated at 37 °C for 24 h.
All LA fermentations were performed with shaking (150 rpm, Biosan shaking bath model ES-20, Biosan Ltd., Riga, Latvia). The experiments were performed in triplicates. The fermentations were performed in 300 mL Erlenmayer flasks with 200 mL of MAE or MAEC media. The fermentation was initiated by the addition of inoculum (5% v/v) and conducted at 37 °C. In order to neutralize the produced LA, sterile calcium carbonate (CaCO3, 0.5% (w/v)) was added to all fermentation media prior to inoculation.

3.6.2. Determination of Reducing Sugars Concentration and Viability of La. rhamnosus Cells

Reducing sugar concentration, calculated as glucose, was determined via the 3.5-dinitrosalicylic acid method [63] using UV/VIS Spectrometer (UV-1800, Shimadzu, Kyoto, Japan). A calibration curve was set at 570 nm using standard glucose solutions. A number of viable La. rhamnosus cells were determined using a pour plating method. Microaerophilic conditions were maintained during incubation in Petri plates using a double MRS agar medium layer. Samples were incubated for 48 h at 37 °C. The total viable cell number was expressed as log CFU/mL. All chemicals used in experiments were of analytical and microbiological grade.

4. Conclusions

The aim of this study was to use an environmentally friendly and sustainable extraction technique to isolate bioactive compounds from Matricaria chamomile. With this in mind, we selected ethanol and microwave extraction and optimized the procedure by applying the ANN methodology. To provide sufficient input experimental data to train the ANN, a 33 full factorial design for 3 factors was chosen. An experimental design with 27 runs combining extraction time, microwave irradiation power as well as solid-to-solvent ratio was therefore conducted. These data were used to feed the artificial neural network that will predict the desired outputs (total phenol content). The optimized conditions were 400 W, 30 min, and 1:80. The predicted TPC yield obtained with such conditions was 59.56 mg/g, and these results have been verified experimentally. The optimized chamomile extracts were rich in total phenols (55.21 mg CAE/g) and total flavonoids (44.98 mg RE/g). LC-MS analysis confirmed the presence of 67 polyphenolic compounds among which apigenin and apigenin-7-O-glucoside were the dominant. In addition, the bioactivity of the optimized extract was evaluated, showing a promising ability to inhibit antioxidants and glucosidase. The results could be valuable for further industrial applications where chamomile is used as a source of natural active ingredients such as antioxidants, antimicrobials, or enzyme inhibitors.

Author Contributions

Conceptualization, A.C.K., M.R. and P.K.; methodology, A.C.K., M.R. and P.K.; validation, A.C.K. and M.R.; formal analysis, A.C.K., M.R., G.Z., U.G. and P.K.; investigation, A.C.K., M.R., G.Z., L.Y., U.G. and P.K.; resources, A.C.K., L.Y. and Z.Z.; writing—original draft, A.C.K., M.R., G.Z., U.G. and P.K.; writing—review and editing, A.C.K., L.Y., A.T., M.B. and Z.Z.; supervision, A.C.K.; project administration, A.C.K., L.Y. and Z.Z.; funding acquisition, A.C.K., L.Y. and Z.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Fundamental Research Funds for the Central Non-profit Research Institution of CAF through the project “green extraction technology for functional active ingredients isolation from agricultural and forestry wastes” (Project No. CAFYBB2018GB001). The authors are also grateful to the Ministry of Education, Science and Technological Development of Republic of Serbia (Project No. -451-03-47/2023-01/200134, University of Novi Sad, Faculty of Technology; Project No. 451-03-68/2022-14/200007, University of Belgrade, Institute for Biological Research “Siniša Stanković”).

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful to Aleksandar Gajić for technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cvetanović, A. Optimization of Novel Extraction Techniques for Apigenin Isolation from Chamomile Flowers (Chamomilla recutita L.) and Characterization of Biological Activity of Obtained Extracts. Ph.D. Thesis, University of Novi Sad, Faculty of Technology Novi Sad, Novi Sad, Serbia, 2016. [Google Scholar]
  2. Newall, C.A.; Anderson, L.A.; Phillipson, J.D. Herbal Medicines: A Guide for Health-Care Professionals; Pharmaceutical Press: London, UK, 1996. [Google Scholar]
  3. Srivastava, J.K.; Shankar, E.; Gupta, S. Chamomile: A herbal medicine of the past with a bright future. Mol. Med. Rep. 2010, 3, 895–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Chandrashekhar, V.M.; Halagali, K.S.; Nidavani, R.B.; Shalavadi, M.H.; Biradar, B.S.; Biswas, D.; Muchchandi, I.S. Anti-allergic activity of German chamomile (Matricaria recutita L.) in mast cell mediated allergy model. J. Ethnopharmacol. 2011, 137, 336–340. [Google Scholar] [CrossRef] [PubMed]
  5. Lis-Balchin, M.; Deans, S.G.; Eaglesham, E. Relationship between bioactivity and chemical composition of commercial essential oils. Flavour Fragr. J. 1998, 13, 98–104. [Google Scholar] [CrossRef]
  6. Franke, R.; Schilcher, H. Chamomile: Industrial Profiles, Series: Medicinal and Aromatic Plants: Industrial Profiles; CRC Press/Taylor & Francis Group: New York, NY, USA, 2005. [Google Scholar]
  7. McKay, D.L.; Blumberg, J.B. A Review of the bioactivity and potential health benefits of chamomile tea (Matricaria recutita L.). Phyther. Res. 2006, 20, 519–530. [Google Scholar] [CrossRef]
  8. Sahebkar, A.; Emami, S.A. Medicinal plants for the treatment of uterus inflammation: Implications from iranian folk medicine. J. Acupunct. Meridian Stud. 2013, 6, 1. [Google Scholar] [CrossRef] [Green Version]
  9. Cvetanović, A.; Švarc-Gajić, J.; Zeković, Z.; Savić, S.; Vulić, J.; Mašković, P.; Ćetković, G. Comparative analysis of antioxidant, antimicrobiological and cytotoxic activities of native and fermented chamomile ligulate flower extracts. Planta 2015, 242, 721–732. [Google Scholar] [CrossRef]
  10. Cvetanović, A.; Švarc-Gajić, J.; Mašković, P.; Savić, S.; Nikolić, L. Antioxidant and biological activity of chamomile extracts obtained by different techniques: Perspective of using superheated water for isolation of biologically active compounds. Ind. Crops Prod. 2015, 65, 582–591. [Google Scholar] [CrossRef]
  11. Ameer, K.; Chun, B.-S.; Kwon, J.-H. Optimization of Supercritical Fluid Extraction of Steviol Glycosides and Total Phenolic Content from Stevia Rebaudiana (Bertoni) Leaves Using Response Surface Methodology and Artificial Neural Network Modeling. Ind. Crops Prod. 2017, 109, 672–685. [Google Scholar] [CrossRef]
  12. Sinha, K.; Chowdhury, S.; Saha, P.D.; Datta, S. Modeling of Microwave-Assisted Extraction of Natural Dye from Seeds of Bixa Orellana (Annatto) Using Response Surface Methodology (RSM) and Artificial Neural Network (ANN). Ind. Crops Prod. 2013, 41, 165–171. [Google Scholar] [CrossRef]
  13. Thakker, M.R.; Parikh, J.K.; Desai, M.A. Microwave Assisted Extraction of Essential Oil from the Leaves of Palmarosa: Multi-Response Optimization and Predictive Modelling. Ind. Crops Prod. 2016, 86, 311–319. [Google Scholar] [CrossRef]
  14. Kennedy, J.; Eberhart, R. Particle Swarm Optimization. In Proceedings of the ICNN’95—International Conference on Neural Networks, Perth, WA, Australia, 27 November—1 December 1995; Volume 4, pp. 1942–1948. [Google Scholar] [CrossRef]
  15. Mezura-Montes, E.; Coello, C.A.C. Constraint-Handling in Nature-Inspired Numerical Optimization: Past, Present and Future. Swarm Evol. Comput. 2011, 1, 173–194. [Google Scholar] [CrossRef]
  16. Font, N.; Hernández, F.; Hogendoorn, E.; Baumann, R.; van Zoonen, P. Microwave-assisted solvent extraction and reversed-phase liquid chromatography—UV detection for screening soils for sulfonylurea herbicides. J. Chromatogr. A. 1998, 798, 179–186. [Google Scholar] [CrossRef]
  17. Brachet, A.; Christen, P.; Veuthey, J.L. Focused microwave-assisted extraction of cocaine and benzoylecgonine from coca leaves. Phytochem. Anal. 2002, 13, 162–169. [Google Scholar] [CrossRef]
  18. Blekić, M.; Jambrak, A.R.; Chemat, F. Mikrovalna ekstrakcija bioaktivnih spojeva. Croat. J. Food Sci. Technol. 2011, 3, 32–47. [Google Scholar]
  19. Pavlić, A.; Kaplan, M.; Bera, O.; Olgun, E.E.; Canli, O.; Milosavljević, N.; Antić, B.; Zeković, Z. Microwave-assisted extraction of peppermint polyphenols—Artificial neural networks approach. Food Bioprod. Process. 2019, 118, 258–269. [Google Scholar] [CrossRef]
  20. Lin, L.Z.; Harnly, J.M. LC-PDA-ESI/MS identification of the phenolic components of three compositae spices: Chamomile, tarragon, and Mexican arnica. Nat. Prod. Commun. 2012, 7, 749–752. [Google Scholar] [CrossRef] [Green Version]
  21. Švehlíková, V.; Bennett, R.N.; Mellon, F.A.; Needs, P.W.; Piacente, S.; Kroon, P.A.; Bao, Y. Isolation, identification and stability of acylated derivatives of apigenin 7-O-glucoside from chamomile (Chamomilla recutita [L.] Rauschert). Phytochemistry 2004, 65, 2323–2332. [Google Scholar] [CrossRef]
  22. Franco, E.P.D.; Contesini, F.J.; Lima da Silva, B.; Alves de Piloto Fernandes, A.M.; Wielewski Leme, C.; Gonçalves Cirino, J.P.; Bueno Campos, P.R.; de Oliveira Carvalho, P. Enzyme-assisted modification of flavonoids from Matricaria chamomilla: Antioxidant activity and inhibitory effect on digestive enzymes. J. Enzym. Inhib. Med. Ch. 2020, 35, 42–49. [Google Scholar] [CrossRef] [Green Version]
  23. Dondi, F.; Kahie, Y.D.; Lodi, G.; Reschiglian, P.; Pietrogrande, C.; Bighi, C.; Cartoni, G.P. Comparison of the Sequential Simplex Method and Linear Solvent Strength Theory in HPLC Gradient Elution Optimization of Multicomponent Flavonoid Mixtures. Chromatographia 1987, 23, 844–849. [Google Scholar] [CrossRef]
  24. Petroianu, G.; Szoke, E.; Kalász, H.; Szegi, P.; Laufer, R.; Benko, B.; Darvas, F.; Tekes, K. Monitoring by HPLC of Chamomile Flavonoids Exposed to Rat Liver Microsomal Metabolism. Open J. Med. Chem. 2009, 3, 1–7. [Google Scholar] [CrossRef] [Green Version]
  25. Yang, D.; Wang, T.; Long, M.; Li, P. Quercetin: Its Main Pharmacological Activity and Potential Application in Clinical Medicine. Oxid. Med. Cell. Longev. 2020, 2020, 8825387. [Google Scholar] [CrossRef] [PubMed]
  26. Imran, M.; Rauf, A.; Shah, Z.A.; Saeed, F.; Imran, A.; Arshad, M.U.; Ahmad, B.; Bawazeer, S.; Peters, D.; Mubarak, M.S. Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res. 2019, 33, 263–275. [Google Scholar] [CrossRef] [PubMed]
  27. Wong, S.K.; Chin, K.Y.; Ima-Nirwana, S. The osteoprotective effects of kaempferol: The evidence from in vivo and in vitro studies. Drug Des. Devel. Ther. 2019, 13, 3497–3514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [Green Version]
  29. Zengin, G.; Cvetanović, A.; Gašić, U.; Dragičević, D.; Stupar, A.; Uysal, A.; Senkardes, I.; Ibrahime Sinan, K.; Picot-Allain, M.C.N.; Ak, G.; et al. UHPLC-LTQ OrbiTrap MS analysis and biological properties of Origanum vulgare subsp. viridulum obtained by different extraction methods. Ind. Crops Prod. 2020, 146, 112747. [Google Scholar] [CrossRef]
  30. Zengin, G.; Cvetanović, A.; Gašić, U.; Stupar, A.; Bulut, G.; Şenkardes, I.; Dogan, A.; Ibrahime Sinan, K.; Uysal, S.; Aumeeruddy-Elalfi, Z.; et al. Modern and traditional extraction techniques affect chemical composition and bioactivity of Tanacetum parthenium (L.) Sch.Bip. Ind. Crops Prod. 2020, 146, 112202. [Google Scholar] [CrossRef]
  31. Papoutsis, K.; Zhang, J.; Bowyer, M.C.; Brunton, N.; Gibney, E.R.; Lyng, J. Fruit, vegetables, and mushrooms for the preparation of extracts with α-amylase and α-glucosidase inhibition properties: A review. Food Chem. 2021, 338, 128119. [Google Scholar] [CrossRef]
  32. Karaoglan, E.S.; Koca, M. Tyrosinase and cholinesterase inhibitory activities and molecular docking studies on apigenin and vitexin. J. Pharm. Istanbul Univ. 2020, 50, 268–272. [Google Scholar] [CrossRef]
  33. Proença, C.; Freitas, M.; Ribeiro, D.; Tomé, S.M.; Oliveira, E.F.; Viegas, M.F.; Araújo, A.N.; Silva, A.M.S.; Fernandes, P.A.; Fernandes, E. Evaluation of a flavonoids library for inhibition of pancreatic α-amylase towards a structure–activity relationship. J. Enzym. Inhib. Med. Ch. 2019, 34, 577–588. [Google Scholar] [CrossRef] [Green Version]
  34. Agunloye, O.M.; Oboh, G.; Ademiluyi, A.O.; Ademosun, A.O.; Akindahunsi, A.A.; Oyagbemi, A.A.; Omobowale, T.O.; Ajibade, T.O.; Adedapo, A.A. Cardio-protective and antioxidant properties of caffeic acid and chlorogenic acid: Mechanistic role of angiotensin converting enzyme, cholinesterase and arginase activities in cyclosporine induced hypertensive rats. Biomed. Pharmacother. 2019, 109, 450–458. [Google Scholar] [CrossRef]
  35. Oboh, G.; Agunloye, O.M.; Adefegha, S.A.; Akinyemi, A.J.; Ademiluyi, A.O. Caffeic and chlorogenic acids inhibit key enzymes linked to type 2 diabetes (in vitro): A comparative study. J. Basic Clin. Physiol. Pharmacol. 2015, 26, 165–170. [Google Scholar] [CrossRef]
  36. Cvetanović, A.; Švarc-Gajić, J.; Zeković, Z.; Gašić, U.; Tešić, Ž.; Zengin, G.; Mašković, P.; Mahomoodally, M.F.; Đurović, S. Subcritical water extraction as a cutting edge technology for the extraction of bioactive compounds from chamomile: Influence of pressure on chemical composition and bioactivity of extracts. Food Chem. 2018, 266, 389–396. [Google Scholar] [CrossRef]
  37. Cvetanović, A.; Švarc-Gajić, J.; Gašić, U.; Tešić, Ž.; Zengin, G.; Zeković, Z.; Đurović, S. Isolation of apigenin from subcritical water extracts: Optimization of the process. J. Supercrit. Fluids 2017, 120, 32–42. [Google Scholar] [CrossRef] [Green Version]
  38. Cvetanović, A.; Zeković, Z.; Zengin, G.; Mašković, P.; Petronijević, M.; Radojković, M. Multidirectional approaches on autofermented chamomile ligulate flowers: Antioxidant, antimicrobial, cytotoxic and enzyme inhibitory effects. S. Afr. J. Bot. 2019, 120, 112–118. [Google Scholar] [CrossRef]
  39. Currò, D.; Ianiro, G.; Pecere, S.; Bibbò, S.; Cammarota, G. Probiotics, fibre and herbal medicinal products for functional and inflammatory bowel disorders. Br. J. Pharmacol. 2017, 174, 1426–1449. [Google Scholar] [CrossRef] [Green Version]
  40. Cocetta, V.; Catanzaro, D.; Borgonetti, V.; Ragazzi, E.; Giron, M.C.; Governa, P.; Carnevali, I.; Biagi, M.; Montopoli, M. A Fixed Combination of Probiotics and Herbal Extracts Attenuates Intestinal Barrier Dysfunction from Inflammatory Stress. Preprints 2017, 2017, 120199. [Google Scholar] [CrossRef]
  41. Marhamatizadeh, M.H.; Shahriarpoor, M.S.; Rezazadeh, S. Effects of Chamomile Essence on the Growth of Probiotic Bacteria, Bifidobacterium bifidum and Lactobacillus acidophilus in Milk and Yoghurt. Glob. Vet. 2012, 8, 605–611. [Google Scholar]
  42. Zarei, K.; Marhamatizadeh, M.H.; Javamardi, F. The Effect of Echium amoenum Hydro-alcoholic Extract on the Growth of Probiotic Bacteria Lactobacillus acidophilus and Bifidobacterium bifidum in Kefir. Alborz Univ. Med. Sci. J. 2020, 9, 219–230. [Google Scholar] [CrossRef]
  43. Dimofte, A.; Simionescu, N.; Petrovici, A.R.; Spiridon, I. Probiotic Properties of Weissella confusa PP29 on Hibiscus sabdariffa L. Media. Fermentation 2022, 8, 553. [Google Scholar] [CrossRef]
  44. Martinelli, M.; Ummarino, D.; Giugliano, F.P.; Sciorio, E.; Tortora, C.; Bruzzese, D.; De Giovanni, D.; Rutigliano, I.; Valenti, S.; Romano, C.; et al. Efficacy of a standardized extract of Matricariae chamomilla L., Melissa officinalis L. and tyndallized Lactobacillus acidophilus (HA 122) in infantile colic: An open randomized controlled trial. Neurogastroenterol. Motil. 2017, 29, e13145. [Google Scholar] [CrossRef]
  45. Hajqasemi, M.; Vaseji, N.; Iranmanesh, M.; Torshizi, K.; Mojgani, N. Synergistic Effects and Viability of Indigenous Probiotic Bacteria in the Presence of Herbal Extracts and Evaluations of their Minimum Inhibitory Concentrations in invitro Conditions. Anim. Sci. J. 2021, 34, 49–64. [Google Scholar] [CrossRef]
  46. Köberl, M.; Erschen, S.; Etemadi, M.; White, R.A.; El-Arabi, T.F.; Berg, G. Deciphering the microbiome shift during fermentation of medicinal plants. Sci. Rep. 2019, 9, 13461. [Google Scholar] [CrossRef] [PubMed]
  47. Holkem, A.T.; Silva, M.P.D.; Favaro-Trindade, C.S. Probiotics and plant extracts: A promising synergy and delivery systems. Crit. Rev. Food Sci. Nutr. 2022, 1–19. [Google Scholar] [CrossRef] [PubMed]
  48. Yoon, Y.; Swales, G., Jr.; Margavio, T.M. A comparison of discriminant analysis versus artificial neural networks. J. Oper. Res. Soc. 1993, 44, 51–60. [Google Scholar] [CrossRef]
  49. Cvetanović, A.; Zengin, G.; Zeković, Z.; Švarc-Gajić, J.; Ražić, S.; Damjanović, A.; Mašković, P.; Mitić, M. Comparative in vitro studies of the biological potential and chemical composition of stems, leaves and berries Aronia melanocarpa’s extracts obtained by subcritical water extraction. Food Chem. Toxicol. 2018, 121, 458–466. [Google Scholar] [CrossRef]
  50. Jurič, A.; Gašić, U.; Brčić Karačonji, I.; Jurica, K.; Milojković-Opsenica, D. The phenolic profile of strawberry tree (Arbutus unedo L.) honey. J. Serb. Chem. Soc. 2020, 85, 1011–1019. [Google Scholar] [CrossRef] [Green Version]
  51. Banjanac, T.; Dragićević, M.; Šiler, B.; Gašić, U.; Bohanec, B.; Nestorović Živković, J.; Trifunović, S.; Mišić, D. Chemodiversity of two closely related tetraploid Centaurium species and their hexaploid hybrid: Metabolomic search for high-resolution taxonomic classifiers. Phytochemistry 2017, 140, 27–44. [Google Scholar] [CrossRef]
  52. Mulinacci, N.; Romani, A.; Pinelli, P.; Vincieri, F.F.; Prucher, D. Characterization of Matricaria recutita L. Flower extracts by HPLC-MS and HPLC-DAD analysis. Chromatographia 2000, 51, 301–307. [Google Scholar] [CrossRef]
  53. Viapiana, A.; Struck-Lewicka, W.; Konieczynski, P.; Wesolowski, M.; Kaliszan, R. An Approach Based on HPLC-Fingerprint and Chemometrics to Quality Consistency Evaluation of Matricaria chamomilla L. Commercial Samples. Front. Plant Sci. 2016, 7, 1561. [Google Scholar] [CrossRef] [Green Version]
  54. Kováčik, J.; Klejdus, B. Dynamics of phenolic acids and lignin accumulation in metal-treated Matricaria chamomilla roots. Plant Cell Rep. 2008, 27, 605–615. [Google Scholar] [CrossRef]
  55. Nováková, L.; Vildová, A.; Mateus, J.P.; Gonçalves, T.; Solich, P. Development and application of UHPLC–MS/MS method for the determination of phenolic compounds in Chamomile flowers and Chamomile tea extracts. Talanta 2010, 82, 1271–1280. [Google Scholar] [CrossRef]
  56. Uysal, S.; Zengin, G.; Locatelli, M.; Bahadori, M.B.; Mocan, A.; Bellagamba, G.; De Luca, E.; Mollica, A.; Aktumsek, A. Cytotoxic and enzyme inhibitory potential of two Potentilla species (P. speciosa L. and P. reptans Willd.) and their chemical composition. Front. Pharmacol. 2017, 8, 290. [Google Scholar] [CrossRef]
  57. Grochowski, D.M.; Uysal, S.; Aktumsek, A.; Granica, S.; Zengin, G.; Ceylan, R.; Locatelli, M.; Tomczyk, M. In vitro enzyme inhibitory properties, antioxidant activities, and phytochemical profile of Potentilla thuringiaca. Phytochem. Lett. 2017, 20, 365–372. [Google Scholar] [CrossRef]
  58. Secci, D.; Carradori, S.; Bizzarri, B.; Chimenti, P.; De Monte, C.; Mollica, A.; Rivanera, D.; Zicari, A.; Mari, E.; Zengin, G.; et al. Novel 1,3-thiazolidin-4-one derivatives as promising anti-Candida agents endowed with anti-oxidant and chelating properties. Eur. J. Med. Chem. 2016, 117, 144–156. [Google Scholar] [CrossRef] [Green Version]
  59. Zengin, G.; Nithiyanantham, S.; Locatelli, M.; Ceylan, R.; Uysal, S.; Aktumsek, A.; Selvi, P.K.; Maskovic, P. Screening of in vitro antioxidant and enzyme inhibitory activities of different extracts from two uninvestigated wild plants: Centranthus longiflorus subsp. longiflorus and Cerinthe minor subsp. auriculata. Eur. J. Integr. Med. 2016, 8, 286–292. [Google Scholar] [CrossRef]
  60. Radosavljević, M.; Pejin, J.; Pribić, M.; Kocić-Tanackov, S.; Romanić, R.; Mladenović, D.; Djukić-Vuković, A.; Mojović, L. Utilization of brewing and malting by-products as carrier and raw materials in l-(+)-lactic acid production and feed application. Appl. Microbiol. Biotechnol. 2019, 103, 3001–3013. [Google Scholar] [CrossRef]
  61. Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; et al. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef]
  62. Pejin, J.; Radosavljević, M.; Mojović, L.; Kocić-Tanackov, S.; Djukić-Vuković, A. The influence of calcium-carbonate and yeast extract addition on lactic acid fermentation of brewer’s spent grain hydrolysate. Food Res. Int. 2015, 73, 31–37. [Google Scholar] [CrossRef]
  63. Miller, G. Use of dinitrosalicylic acid for determining reducing sugars. Anal. Chem. 1959, 31, 426–428. [Google Scholar] [CrossRef]
Figure 1. Parity plot of the experimental and predicted TPC yield.
Figure 1. Parity plot of the experimental and predicted TPC yield.
Plants 12 01211 g001
Figure 2. Relative importance of independent variables on the TPC.
Figure 2. Relative importance of independent variables on the TPC.
Plants 12 01211 g002
Figure 3. Total phenolics and flavonoids content in optimized chamomile extract.
Figure 3. Total phenolics and flavonoids content in optimized chamomile extract.
Plants 12 01211 g003
Figure 4. Schematic diagram of the microwave-assisted distillation apparatus 1. microwave oven, 2. glass flask, 3. irradiation power controller, 4. time controller, 5. display, 6. condenser, 7. inlet water, 8. outlet water.
Figure 4. Schematic diagram of the microwave-assisted distillation apparatus 1. microwave oven, 2. glass flask, 3. irradiation power controller, 4. time controller, 5. display, 6. condenser, 7. inlet water, 8. outlet water.
Plants 12 01211 g004
Table 1. Compounds identified in chamomile extract.
Table 1. Compounds identified in chamomile extract.
NotR, MinCompound NameMolecular Formula,
[M − H]
Calculated Mass,
[M − H]
Exact Mass,
[M − H]
Δ ppmMS2 Fragments, (% Base Peak)MS3 Fragments, (% Base Peak)MS4 Fragments, (% Base Peak)
Phenolic acids and their derivatives
11.95Gallic acid hexoside isomer 1C13H15O10331.06707331.066800.82211(15), 193(80), 175(30), 169(100), 151(50)151(100)110(10), 97(30), 81(100), 53(30)
22.39Gallic acid aC7H5O5169.01425169.013852.37125(100)107(100)
32.69Dihydroxybenzoic acid hexoside isomer 1C13H15O9315.07216315.072060.32153(100), 152(50), 109(15), 108(10)109(100)123(25), 109(10), 85(10), 81(100)
43.13Gallic acid hexoside isomer 2C13H15O10331.06707331.067020.15313(100), 169(25), 168(90), 151(10), 125(25)193(50), 151(100), 125(80)123(100), 107(90), 95(65)
53.82Dihydroxybenzoic acid hexoside isomer 2C13H15O9315.07216315.071213.02153(100), 135(10), 109(10)135(100), 109(50)91(100)
64.32Dihydroxybenzoic acid hexoside isomer 3C13H15O9315.07216315.071691.49153(100), 109(10)135(100), 109(50)91(100)
74.51Protocatechuic acid aC7H5O4153.01933153.018723.99109(100)81(60), 80(50), 67(30), 65(100)
84.38Caffeoylquinic acid hexoside isomer 1C22H27O14515.14008515.139281.55353(80), 341(5), 323(10), 191(100), 179(5)173(65), 127(90), 111(50), 93(55), 85(100)
94.613-O-Caffeoylquinic acidC16H17O9353.08781353.086762.97191(100), 179(35), 135(10)173(75), 127(100), 111(40), 93(60), 85(90)109(30), 99(40), 85(100)
104.68Caffeic acid hexoside isomer 1C15H17O9341.08781341.087161.91191(10), 179(100), 135(10)135(100)135(100), 107(50)
114.88Caffeoylquinic acid hexoside isomer 2C22H27O14515.14008515.139281.55353(15), 341(15), 323(100), 191(25), 179(5)161(100), 133(5)133(100), 117(20)
125.17Caffeic acid hexoside isomer 2C15H17O9341.08781341.087311.47179(100), 135(10)135(100)107(100), 79(20)
135.22Ferulic acid hexosylhexosideC22H29O14517.15628517.154663.13221(25), 193(100), 179(25), 161(10), 149(20)149(100)134(100)
145.304-O-Caffeoylquinic acidC16H17O9353.08781353.087490.91223(20), 191(50), 179(60), 173(100), 135(10)115(20), 111(50), 93(100), 71(20)
155.32Coumaric acid hexosideC15H17O8325.09289325.092830.18163(100), 119(10)119(100)
165.42p-Hydroxybenzoic acid aC7H5O3137.02442137.024201.61109(10), 93(100)66(100)
175.46Gentisic acid aC7H5O4153.01933153.018952.48109(100)95(10), 81(100), 65(35)
185.73Ferulic acid hexoside isomer 1C16H19O9355.10346355.101874.48193(100), 149(25)149(100)133(100)
195.783-O-Feruoylquinic acidC17H19O9367.10346367.102632.26193(100), 178(5), 173(5), 134(10)178(90), 149(40), 134(100)106(100)
205.82Caffeic acid aC9H7O4179.03498179.034592.18135(100)135(60), 117(15), 107(100), 91(55), 79(15)
215.86Hydroxybenzoic acid derivativeC25H27O14551.14063551.139771.56431(10), 413(100)281(5), 179(5), 137(100)93(100)
225.945-O-p-Coumaroylquinic acidC16H17O8337.09289337.092680.62191(100), 179(5), 163(10)173(75), 127(100), 111(40), 93(60), 85(90)109(30), 99(40), 85(100)
236.394-O-Feruoylquinic acid isomer 1C17H19O9367.10346367.102751.93193(5), 173(100), 155(5), 111(5)155(15), 111(40), 93(100), 71(10)
246.46Ferulic acid acetylhexoside isomer 1C18H21O10397.11402397.113461.41193(100), 149(30), 134(10)149(100)134(100)
256.51Ferulic acid hexoside isomer 2C16H19O9355.10346355.103061.13193(100), 149(10)149(100)133(100)
266.644-O-Feruoylquinic acid isomer 2C17H19O9367.10346367.102941.42193(5), 179(10), 173(100), 155(5), 111(10)155(15), 111(40), 93(100), 71(10)
276.72p-Coumaric acid aC9H7O3163.04007163.040060.06119(100)119(60), 101(20), 93(25), 91(100), 72(10)
286.94Dicaffeoylquinic acid isomer 1C25H23O12515.11950515.117493.90353(100), 355(20), 299(10), 191(10), 179(15)191(40), 179(70), 173(100), 135(10)155(10), 111(50), 93(100), 71(10)
297.09Ferulic acid acetylhexoside isomer 2C18H21O10397.11402397.114000.05193(100), 149(30), 134(10)149(100)134(100)
307.16Dicaffeoylquinic acid isomer 2C25H23O12515.11950515.117803.30353(100)191(100), 179(30), 173(5), 135(10)173(65), 127(90), 111(50), 93(55), 85(100)
317.33Dicaffeoylquinic acid isomer 3C25H23O12515.11950515.118741.48353(100), 355(5), 317(5), 299(10), 203(5)191(30), 179(60), 173(100), 135(10)155(20), 111(60), 93(100), 71(20)
328.05Ferulic acidC10H9O4193.05063193.049854.04178(70), 149(100), 134(40)134(100)
Flavonoid aglycones and glycosides
335.62Apigenin 6,8-di-C-hexosideC27H29O15593.15119593.150021.97533(10), 503(30), 473(100), 383(20), 353(35)383(20), 353(100)275(20), 265(60), 249(100), 221(35), 173(80)
346.186-Hydroxyquercetin 7-O-hexosideC21H19O13479.08311479.081762.82318(10), 317(100)299(40), 271(100), 167(75), 139(45)243(100), 227(30), 215(20), 199(50)
356.65Quercetin 3-O-galactoside aC21H19O12463.08820463.087531.45301(100), 300(30)273(25), 257(20), 179(100), 151(75)151(100)
366.74Luteolin 7-O-hexosideC21H19O11447.09329447.092621.50286(10), 285(100)257(30), 241(100), 217(75), 199(85), 175(95)241(5), 226(15), 213(30), 197(100)
376.796-Methoxyquercetin 7-O-hexosideC22H21O13493.09876493.097722.11477(20), 373(10), 331(100), 323(30), 316(5)316(100), 209(5), 181(5), 166(5)287(100), 271(60), 194(40), 166(70), 151(5)
386.92Apigenin 7-O-(6”-rhamnosyl)hexosideC27H29O14577.15628577.155760.90270(10), 269(100)225(100), 201(20), 197(30), 183(30), 149(25)225(5), 210(10), 197(100), 181(50), 169(40)
397.076-Methoxyapigenin 7-O-(6”-rhamnosyl)hexosideC28H31O15607.16684607.166560.46300(15), 299(100), 284(5)284(100)284(30), 256(100), 239(5), 227(15), 211(10)
407.22Isorhamnetin 3-O-glucoside aC22H21O12477.10385477.102782.24357(20), 315(50), 314(100), 300(5), 299(5)300(30), 285(100), 271(75), 257(10), 243(25)270(100)
417.27Apigenin 7-O-glucoside aC21H19O10431.09837431.098110.60270(10), 269(100)225(100), 201(15), 197(20), 183(30), 149(30)210(15), 197(80), 183(100), 181(30), 169(30)
427.30Naringenin aC15H11O5271.06120271.061130.26177(10), 151(100)107(100)65(100)
437.38Isorhamnetin 7-O-hexosideC22H21O12477.10385477.103850.00462(5), 357(10), 316(10), 315(100), 300(5)300(100), 285(5), 151(10)283(20), 272(65), 271(70), 227(30), 151(100)
447.436-Methoxyapigenin 7-O-hexosideC22H21O11461.10894461.108341.30446(80), 341(10), 299(100), 284(20)284(100)284(30), 256(100), 239(5), 227(15), 211(10)
457.516-Methoxyquercetin 7-O-(6”-caffeoyl)hexosideC31H27O16655.13046655.130430.05533(15), 505(10), 331(100), 323(20), 316(30)316(100), 209(5), 181(5), 166(5)287(100), 271(60), 194(40), 166(70), 151(5)
467.54Isorhamnetin 7-O-(6”-acetyl)hexosideC24H23O13519.11441519.113831.12357(5), 316(10), 315(100), 300(5), 285(5)300(100), 287(5), 272(10)272(30), 271(100), 255(50)
477.81Apigenin 7-O-acetylhexoside isomer 1C23H21O11473.10894473.107822.37413(15), 311(10), 270(15), 269(100), 268(60)225(100), 201(25), 197(35), 183(30), 149(40)210(20), 197(100), 183(30), 181(60), 169(30)
488.15Apigenin 7-O-acetylhexoside isomer 2C23H21O11473.10894473.106994.12413(30), 311(10), 270(10), 269(100), 268(40)225(100), 201(30), 197(30), 183(30), 149(35)207(10), 197(100), 183(30), 181(70), 169(40)
498.20Apigenin 7-O-(6”-caffeoyl)hexosideC27H29O15593.13006593.128482.66323(95), 269(100), 221(15), 179(10)225(100), 201(30), 183(10), 151(20), 149(35)225(10), 208(10), 197(40), 181(100), 169(15)
508.38Apigenin derivativeC28H25O13569.13006569.129271.39270(10), 269(100)225(100), 201(20), 197(30), 183(30), 149(25)225(5), 210(10), 197(100), 181(50), 169(40)
518.50Apigenin 7-O-acetylhexoside isomer 3C23H21O11473.10894473.107263.55413(5), 311(10), 270(15), 269(100), 268(50)225(100), 201(30), 197(25), 183(25), 149(30)197(70), 183(50), 181(100), 169(30)
528.55Apigenin 7-O-diacetylhexoside isomer 1C25H23O12515.11950515.117134.60455(30), 431(5), 413(10), 311(15), 269(100)225(100), 201(25), 197(25), 183(25), 149(30)210(10), 197(100), 183(30), 181(50), 169(40)
538.636-Methoxyapigenin 7-O-(6”acetyl)hexosideC24H23O12503.11950503.118162.66488(100), 299(20), 284(10)429(10), 327(10), 313(40), 283(100), 255(30)255(100)
548.686-MethoxyapigeninC16H11O6299.05611299.055691.40285(15), 284(100)256(100)238(30), 228(70), 211(60), 188(100)
558.67Luteolin aC15H9O6285.04046285.039921.89257(40), 241(100), 217(50), 199(70), 175(70)255(50), 227(100), 211(75), 197(35), 183(85)
568.74Apigenin 7-O-diacetylhexoside isomer 2C25H23O12515.11950515.118042.83455(20), 270(10), 269(100), 268(40)225(100), 201(30), 197(20), 183(25), 149(40)197(100), 183(50), 181(80), 169(30)
578.80Quercetin aC15H9O7301.03538301.034831.83271(50), 255(20), 179(100), 151(80), 107(5)151(100)107(100), 83(10)
588.846-MethoxyluteolinC16H11O7315.05103315.050631.27301(20), 300(100), 166(5)283(40), 272(70), 255(50), 243(40), 216(100)201(25), 188(100), 173(20)
599.05Apigenin 7-O-diacetylhexoside isomer 3C25H23O12515.11950515.118232.47455(20), 293(10), 270(10), 269(100), 268(20)225(100), 201(25), 197(35), 183(30), 149(40)210(20), 197(100), 183(30), 181(60), 169(30)
609.53Apigenin 7-O-diacetylhexoside isomer 4C25H23O12515.11950515.117613.67455(30), 413(10), 311(15), 270(15), 269(100)225(100), 201(30), 197(30), 183(30), 149(35)207(10), 197(100), 183(30), 181(70), 169(40)
619.56Apigenin aC15H9O5269.04554269.044493.90225(5), 177(15), 151(100)65(100)
629.68Apigenin 7-O-diacetylhexoside isomer 5C25H23O12515.11950515.118352.23455(10), 270(10), 269(100), 268(30)225(100), 201(30), 197(25), 183(25), 149(30)197(70), 183(50), 181(100), 169(30)
639.73Kaempferol aC15H9O6285.04046285.039692.70255(100), 227(10)211(100), 195(5), 167(15)211(40), 137(100)
649.81Chrysoeriol aC16H11O6299.05611299.055651.54285(10), 284(100)256(100)239(10), 227(100), 212(20), 200(15)
659.95IsorhamnetinC16H11O7315.05103315.050172.73301(20), 300(100)283(30), 271(100), 255(40), 227(50), 151(90)243(100), 227(50), 215(10), 199(20)
6610.32ChrysosplenolC18H15O8359.07724359.077030.58345(10), 344(100), 287(10), 240(5)329(100), 301(5)314(100), 301(20), 286(30), 270(5), 175(5)
6711.74ChrysosplenetinC19H17O8373.09289373.092770.32359(10), 358(100)343(100)328(100), 315(15), 300(30), 284(10), 272(10)
a Confirmed using available standards; all the other compounds were identified based on HRMS data. Bold numbers are peaks which were further fragmented in the MS3 and MS4 experiments.
Table 2. Concentration (mg/L) of individual phenolics found in optimal chamomile extracts.
Table 2. Concentration (mg/L) of individual phenolics found in optimal chamomile extracts.
Compound Namemg/L
Phenolic acids and their derivatives
Gallic acid0.072
Protocatechuic acid0.649
p-Hydroxybenzoic acid1.619
Gentisic acid0.510
Caffeic acid0.494
p-Coumaric acid0.762
Flavonoid aglycones and glycosides
Quercetin 3-O-galactoside0.283
Isorhamnetin 3-O-glucoside0.569
Apigenin 7-O-glucoside2.408
Naringenin0.142
Luteolin0.191
Quercetin0.861
Apigenin1.542
Kaempferol0.358
Chrysoeriol0.286
Table 3. Antioxidant potential of optimized MAE chamomile extract.
Table 3. Antioxidant potential of optimized MAE chamomile extract.
ParametersResults
Antioxidant assays
DPPH (mg TE/g)60.24 ± 2.79
ABTS (mg TE/g)126.92 ± 9.50
CUPRAC (mg TE/g)123.12 ± 0.93
FRAP (mg TE/g)95.37 ± 1.98
MCA (mg EDTAE/g)21.34 ± 0.06
PBD (mmol TE/g)1.42 ± 0.06
Enzyme inhibitory assays
AChE inhibition (mg GALAE/g)0.85 ± 0.11
Amylase inhibition (mmol ACAE/g)0.18 ± 0.01
Glucosidase inhibition (mmol ACAE/g)13.11 ± 0.72
Values are reported as mean ± SD of three parallel measurements. TE: Trolox equivalent; EDTAE: EDTA equivalent. GALAE: Galantamine equivalent; ACAE: Acarbose equivalent.
Table 4. Full factorial design 33 and experimentally observed response.
Table 4. Full factorial design 33 and experimentally observed response.
InputsOutput
Microwave Power (kW)Extraction Time (min)Solid-to-Solvent RatioTPC
400401:6046.04
400301:8046.16
400201:6042.85
400401:4044.18
400301:4037.99
400201:4035.54
400301:6058.435
400201:8056.75
400401:8054.14
600201:8050.68
600201:4035.95
600201:6040.65
600401:4038.47
600401:6048.5
600301:4038.07
600401:8047.46
600301:6054.51
600301:8051.86
800301:6046.9
800301:8041.6
800401:4046.29
800201:8057.32
800301:4044.34
800401:8059.56
800201:4037.17
800201:6047.88
800401:6049.36
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

Cvetanović Kljakić, A.; Radosavljević, M.; Zengin, G.; Yan, L.; Gašić, U.; Kojić, P.; Torbica, A.; Belović, M.; Zeković, Z. New Biological and Chemical Insights into Optimization of Chamomile Extracts by Using Artificial Neural Network (ANN) Model. Plants 2023, 12, 1211. https://doi.org/10.3390/plants12061211

AMA Style

Cvetanović Kljakić A, Radosavljević M, Zengin G, Yan L, Gašić U, Kojić P, Torbica A, Belović M, Zeković Z. New Biological and Chemical Insights into Optimization of Chamomile Extracts by Using Artificial Neural Network (ANN) Model. Plants. 2023; 12(6):1211. https://doi.org/10.3390/plants12061211

Chicago/Turabian Style

Cvetanović Kljakić, Aleksandra, Miloš Radosavljević, Gokhan Zengin, Linlin Yan, Uroš Gašić, Predrag Kojić, Aleksandra Torbica, Miona Belović, and Zoran Zeković. 2023. "New Biological and Chemical Insights into Optimization of Chamomile Extracts by Using Artificial Neural Network (ANN) Model" Plants 12, no. 6: 1211. https://doi.org/10.3390/plants12061211

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