Next Article in Journal
Enhancing Uranium Extraction Efficiency Using Protonated Amines and Quaternary Ammoniums-Based Ionic Liquids: Mechanistic Insights and Nonlinearities Analysis
Next Article in Special Issue
HPLC and SFC Enantioseparation of (±)-Trans-β-Lactam Ureas on Immobilized Polysaccharide-Based Chiral Stationary Phases—The Introduction of Dimethyl Carbonate as an Organic Modifier in SFC
Previous Article in Journal
Microalgae Biomass Harvesting Using Chitosan Flocculant: Optimization of Operating Parameters by Response Surface Methodology
Previous Article in Special Issue
Green Synthesis of Zinc Oxide Nanoparticles Using Aqueous Extracts of Hibiscus cannabinus L.: Wastewater Purification and Antibacterial Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Simultaneous Determination of Nine Furocoumarins in Angelica dahurica Using UPLC Combined with the QAMS Approach and Novel Health Risk Assessment Based on the Toxic Equivalency Factor

National Institutes for Food and Drug Control, No. 31 Huatuo Road, Daxing District, Beijing 102629, China
*
Authors to whom correspondence should be addressed.
Separations 2023, 10(9), 508; https://doi.org/10.3390/separations10090508
Submission received: 15 August 2023 / Revised: 4 September 2023 / Accepted: 5 September 2023 / Published: 15 September 2023
(This article belongs to the Special Issue Advances in Chromatographic Analysis of Bioactive Compounds)

Abstract

:
Objective: This study aimed to provide data for the type and content of linear furocoumarins (FCs) in Angelica dahurica (AD) in order to assess their cumulative risks and provide a scientific basis for the rational use and quality evaluation of the medicinal AD to improve public health. Methods: A UPLC method was developed for the simultaneous determination of nine FCs initially by using imperatorin (Im) as the internal standard substance, including Im, phellopterin (Ph), isoimperatorin (Is), oxypeucedanin hydrate (Oh), byakangelicin (Bn), xanthotoxin (8-MOP), bergapten (5-MOP), byakangelicol (Bl), and oxypeucedanin (Op) in two species of Angelica dahurica (AD). And, the risk assessment for the total FCs in AD was explored using the hazard index combined with the toxic equivalency factor (TEF-HI) strategy for the first time. Results: The established method revealed acceptable applicability, and there were no significant differences compared with the external standard method (ESM). The quantitative results demonstrated that the total content of FCs in Angelica dahurica (BZ) were higher than that in Angelica dahurica var. formosana (HBZ), and there was a great difference between the Bl and Op. Moreover, the risk assessment data revealed that the risk of total FCs in AD to human health was low. Conclusions: The established UPLC method that determined nine FCs in AD using a single marker could solve the problem of difficulty in obtaining a chemical reference substance with high purity and requiring a long determination time. And, the TEF-HI risk assessment approach associated with FCs in ADs could guide the rational utilization of toxic FCs in ADs in the progress of improving public health safety. In short, the whole systematic strategy provides a scientific basis for rational quality evaluation and the healthy use of related herbal medicines.

1. Introduction

Furocoumarins (FCs) are a class of natural chemical compounds that widely exists in many herbs, including some commonly used as food, cosmetics, flavorings, or herbal medicines by humans [1]. Just as its name implies, these compounds have a skeleton formed via a coumarin unit fused to a furan ring and can be divided into linear or angular isomers depending on the position of the furan ring [2]. Although FCs are widely used for medicine and food purposes, every coin has two sides, and it is reported that linear furanocoumarins, which are predominant in natural sources such as 5-methoxypsoralen (5-MOP) and 8-methoxypsoralen (8-MOP), are phototoxic and genotoxic [3,4,5,6]. And, furanocoumarin-induced hepatotoxicity has been reported to occasionally occur in humans [7]. It was once believed that FCs could interact with proteins, lipids in membranes, ribosomes, mitochondria, and, in particular, mono- and diadducts with DNA or RNA, which are the main contributors to toxicity in humans and animals [8,9,10]. In addition, certain FCs have been publicized to act as greatly potent inhibitors of a quantity of cytochromes P450 mono oxygenases [11], which can be interpreted as hepatotoxicity. These phenomena are defined as co-carcinogenic conditions [12]. Therefore, the U.S. Food and Drug Administration (FDA) had proscribed the use of FCs as food additives. As some research has reported, the family of FCs comprises more than 90 individual compounds, and the individual FCs shown differ in photomutagenic potency [13]. FCs are abundantly found in herbal products, especially in herbal medicine belonging to Angelica L., and these various FCs occur in one-plant ingredients, inevitably raising the problem of combination effects or interactions. Therefore, it is urgently required to establish feasible and reliable methods for the combined risk assessment of FCs’ components in herbal medicine.
Traditional Chinese medicine (TCM) Angelica dahurica (AD, called BaiZhi in Chinese) is the dried root of Angelica dahurica (Fisch. ex Hoffm.) Benth. et. Hook. f. or Angelica dahurica (Fisch. ex Hoffm.) Benth. et Hook. f. var. formosana (Boiss.) Shan et Yuan (Fam. Umbelliferae) [14]. It is a tall perennial herb mainly distributed in the provinces of Sichuan, Hebei, Henan, Zhejiang, Anhui, Shandong, Neimenggu, Fujian, and Taiwan of China [15]. As a typical medicinal and food plant, it has the effects of expelling wind, dehumidifying, relieving pain, and relieving itching [16]. The major chemical constituents of it comprise polysaccharide, coumarins, alkaloids, benzofurans, volatile oil, fatty acids, and amino acids, and the total content of coumarins in the active ingredient of AD is more than 60% [17,18,19]. Modern pharmacology has shown that these chemical constituents, especially FCs, have efficacy in treating tumors, antioxidants, etc. [20,21]. But, because of the photogenotoxicity and photocarcinogenicity of FCs, the application of AD for external use is limited in China [22]. And, in October 2007, the European Medicines Agency (EMA) suggested a risk-management strategy for herbal preparations, which is rich in FCs in the paper titled “Reflection Paper on the Risk Associated with Furocoumarins Contained in Preparations of Angelica archangelica L.” [12]. However, there are limited reports about the relevant toxicity risk assessment of AD for oral usage, and no official proposal on the potential consumption of FCs is available in China.
Nowadays, a lot of research on the identification and isolation of FCs from AD has been reported. Components in AD have been authenticated or quantified via HPLC, GC, LC-MS, GC-MS, NMR, and high-resolution mass spectrometry, such as ion trap MS and TOF-MS [23,24,25,26]. However, there has been a deficiency report about the simultaneous determination of nine FCs in AD via a single marker combined with UPLC. And, information regarding the detailed quantitative analysis of FCs in the two pieces of AD is limited. As for the risk assessments about FCs contained in plants, although EMEA has proposed some strategies, currently, the available assessment is primarily based on the data of 5-MOP and 8-MOP, owing to the fact that toxicological figures on other individual FCs in these plants are almost lacking. But, using 5-MOP and 8-MOP as a model for the risk assessment of plants which FCs are rich in might make the assessment inaccurate. Fortunately, recent researchers have proposed the concept of the toxic equivalency factor (TEF) based on photomutagenic, photocytotoxic, and photoclastogenic properties in a V79 cells experiment, which suggested that the photogenotoxic and phototoxic properties of individual furocoumarins may diverge widely but showed relatively strict additivity once occurring as multifarious mixtures [27,28]. Moreover, in 2008, the European Food Safety Authority (EFSA) conducted a series of research in experimental animals, declared that the carcinogenic influence was not triggered via a genotoxic mechanism, and then proposed the tolerable daily intake (TDI) values (0.1 mg coumarin/kg bw/day) based on the non-observed adverse effect level (NOAEL) for liver toxicity in a 2-year dog study [29]. Thus, it is significant to determine the content of various FCs in AD and to evaluate the related health risks when using medicine in a more scientific approach.
In this study, a practical QAMS method was adopted, wherein nine FCs could be determined simultaneously by using a single internal standard reference substance. And, a new risk assessment strategy for assessing the human health risk related to the consumption of AD via TEF-HI was provided to suggest the safety usage of FCs in TCM.

2. Materials and Methods

2.1. Chemicals and Reagents

Acetonitrile (MS-grade) was obtained from Merck (Overijse, Belgium). Ultrapure water was prepared by a Milli-Q system (Millipore, Billerica, MA, USA). The rest of the chemicals (analytical grade) were obtained from Sinopharm Chemical Reagent Beijing (Beijing, China). The reference standard of imperatorin (Im) and isoimperatorin (Is) were provided by NIFDC. Standard phellopterin (Ph), oxypeucedanin hydrate (Oh), byakangelicin (Bn), bergapten (5-MOP), xanthotoxin (8-MOP), byakangelicol (Bl), and oxypeucedanin (Op) were obtained from Chengdu DeSiTe Biological Technology Co., Ltd. (Chengdu, China), all with the purity above 98%.
In total, 20 batches of AD plants including 10 batches of Angelica dahurica (Hoffm.) Benth. & Hook. f. ex Franch. & Sav. (BZ, called BaiZhi in Chinese, Species No. 1) and 10 batches of Angelica dahurica var. formosana (Boissieu) Yen (HBZ, called HangBaiZhi in Chinese, Species No. 2) were collected from Bozhou Anhui, Anguo Hebei, Shehong Sichuan, Suining Sichuan of China, and Hong Kong, respectively, in August 2021. They were originally authenticated by Dr. Kang Shuai of the National Institutes for Food and Drug Control (NIFDC, Beijing, China). All specimens had been stored in the Chinese Herb medicine museum of NIFDC.

2.2. Instrumentation and Chromatographic Condition

An ultra-high-performance liquid chromatographic (UHPLC) system (Shimadzu LC_30AD, Shimadzu, Tokyo, Japan) equipped with a DAD detector was used. An ultrasonic bath (300 W) was also used in the process.
Chromatography was performed with a Waters ACQUITY UPLC® CSHTM C18 Column (2.1 mm × 100 mm, 1.7 μm) at 25 °C with a flow rate of 0.3 mL·min−1, and 254 nm was chosen as the detection wavelength. Mobile phases A and B consisted of water and acetonitrile. A gradient elution (0–10 min, 30–40% B; 10–17 min, 40–80% B; 17–20 min, 80% B) was adopted, and the injection volume was 2 μL.

2.3. Preparation of Standard Solutions

Nine standard FC substances were dissolved in methanol, respectively, and the stock solutions were made to be about 1 mg/mL. Then, the mixed reference stock solution was prepared comprising all standards including Oh, Bn, 8-MOP, 5-MOP, Bl, Op, Im, Ph, and Is at the concentration of 158.7, 49.6, 57.3, 53.6, 90.15, 258.75, 102.0, 60.1, and 102.5 mg/L, respectively. The working solutions were kept at −20 °C and were newly prepared each day via the appropriate dilution of the stock with methanol.

2.4. Preparation of BZ Samples

Before preparation, cut the 20 batches of BZ samples into small pieces and powder them through a No. 2 sieve, respectively. Then, accurately weigh 0.5 g of powdered sample into a 50 mL conical flask, add 20 mL of 70% methanol, and sonicate (300 W) the mixture for 20 min. Filter and transfer the filtrate to a 50 mL volumetric flask. Repeat the steps once. Then, wash the residue, filter paper, and the funnel with the appropriate volume of 70% methanol and combine the solutions into the same volumetric flask. Increase the volume of the volumetric flask up to the mark with 70% methanol. Filter the solution through a 0.22 µm nylon filter before analysis.

2.5. Method Validation

The established UPLC method was validated through complete methodology, including specificity, accuracy rate (using recovery rate), precision, reproducibility, linearity (or calibration curves), covers, limits of detection (LOD), limits of quantification (LOQ), stability, and durability.
Among these, Radix Ophiopogonis (one commonly used TCM) was chosen as a blank matrix to perform the specificity, LOD, and LOQ tests by adding an appropriate amount of the relevant reference. The LOD and LOQ values for the nine FCs were defined as signal-to-noise (S/N) ratios of 3 and 10, respectively. The mixed reference stock solution was diluted to six concentration levels, and calibration curves (Y = aX + b) were established where the X- and Y-axes represented the peak area and the concentration of every furocoumarin, respectively. The correlation coefficient (r) was calculated to inspect the linearity of every calibration curve. The precision was evaluated using six consecutive injections of the same mixed standard solutions. The repeatability was measured by analyzing six independently prepared sample solutions of the same batch. The stability was assessed by injecting the same prepared sample solution at different times (0, 2, 4, 8, 12, 24 h). The relative standard deviation (RSD %) was analyzed at the same time to estimate the results of precision, repeatability, and stability. Finally, the recovery test was performed to verify the accuracy of the method. The mixed reference stock solution of nine FCs at certain levels were precisely added to 0.25 g of the above sample power. Then, six parallel solutions were created according to the above sample preparation requirements, calculating the average recovery rate (n = 6) and analyzing the RSD of every component.

2.6. Theory of QAMS

The principle of the QAMS method is that the content of the component is proportional to the detector response under particular conditions, and different effective components of TCM exist on intrinsic functions and proportional relations, which make the calculation of relative correction factors (RCFs) feasible [14]. Based on the RCFs, the content of each analyst can be determined independently according to an internal standard substance. In this case, the calculation of RCFs is crucial in this study. First, Im was chosen as the internal standard substance, and then the RCFs for the rest of the FCs were calculated using the following multipoint method (Equation (1)). Thereafter, the content of the other FCs was determined according to Equation (2) [30].
The equation for RCFs was calculated as follows,
fs/i = fi/fs = Ai × Cs/(As × Ci)
The quantification of the other investigated FC components is calculated as follows,
Ci = (Ai × Cs)/(fs/i × As)
where As and Ai represent the peak areas of the internal standard (Im) and other FCs in standard solutions or in the AD samples, respectively. fs/i represents the average of the RCFs between Im and each of the other FC components. Cs and Ci represent the concentrations of Im and the other investigated FC components in standard solutions or in the AD samples.
Additionally, for the location of target chromatographic peaks, the relative retention time (RRT) was introduced to position the chromatographic peaks of the other investigated FC components according to Equation (3) [31],
ti/s = ti/ts
where ti/s is the RRT of the analysts and ti and ts are the retention times of the other investigated FC components and Im, respectively.

2.7. Quantification of AD Samples

Both the newly established QAMS and ESM methods were used to determine and analyze the samples. Each sample was determined 3 times, and then the mean was calculated. The accuracy of the two methods was assessed using standardized mean difference (SMD), which was calculated according to Equation (4) [32],
SMD = [(CESM − CQAMS)/CESM] × 100%
where CESM and CQAMS represent the mean content of the FCs determined by QAMS and ESM, respectively.

2.8. Healthy Risk Assessment

2.8.1. Assessment of Daily Intake of AD Based on the EQF

To examine the health risk of FCs in AD, a real-life exposure situation was considered by estimating the EDI (μg/kg bw/day) based on the EQF as in Equation (5) [33],
EDI = EF × Ed × IR × C × EQF/W × AT
where EDI is the estimated daily intake of FCs in AD; EF is the abbreviation of exposure frequency, which is 90 days/year [33]; and Ed represents exposure time, which according to our previous study is 20 years [33]; and IR is short for the daily intake rate of AD (g/day). Based on the guidelines of ChP [14], the mean IR is 6.5 g/day. C represents the content of the FCs in AD (mg/g) and EQF is the equivalency factor. It was suggested that the value for 5-MOP should be set at 1.00, then the value for Oh, Bn, 8-MOP, Bl, Op, Im, Ph, and Is would be 0.16, 0.01, 0.20, 0.01, 0.076, 0.15, 0.01, and 0.15, respectively. Then, W symbolizes the average of body weight, which is 70 kg based on the date given in adults by the EFSA and AT represents the mean duration of exposure to AD, which is usually 365 days/year × 70 years.

2.8.2. Risk Characterization Based on Health-Based Guidance Values

Risk characterization is commonly expressed by calculating risk metrics including hazard index (HI), Margins of Exposure (MoE), or Reference Point Index (RPI). For hazardous substances with health-based guideline values, an HI strategy is recommended. As the EFSA had allocated a TDI for coumarins via diet, the HI value can be calculated according to Equation (6) [33].
HI   = i = 1 n EDI i × 10 / TDI
In Equation (6), HI represents the hazard index. i = 1 n EDI i is the calculated daily intake of the nine FCs in AD and 10 is the safety factor, which indicates that the daily intake of FCs from TCM and its products is not greater than 10% of the daily exposure (including diet). TDI is the tolerable daily intake value (mg/kg bw/day). The total TDI of nine FCs in each sample was calculated in this study. According to the research of EFSA, the TDI of coumarins is 0.1 mg/kg bw/day in this study. Once the HI value is >1, the exposure risks of the analyst should not be ignored.

3. Results and Discussion

3.1. Extraction Procedures and UPLC Condition Optimization

To extract coumarins from AD more completely, the extraction solvents (100%, 70%, 50%, and 30% ethanol), extraction methods (ultrasound and reflux), extraction times (20, 30, and 40 min), and number of extractions were investigated. The extraction efficiency showed that adding 20 mL of 70% methanol, sonication (300 W) for 20 min, and repeating the extraction once could extract the coumarins exhaustively.
First, for chromatographic conditions, the chromatographic columns of ACQUITY UPLC® BEH Amide (2.1 mm × 100 mm, 1.7 µm), ACQUITY UPLC® CSHTM C18 (2.1 mm × 100 mm, 1.7 µm), CORTECS® UPLC® C18 (2.1 mm × 100 mm, 1.6 µm), ACQUITY UPLC® BEH SHIELD (2.1 × 50 mm, 1.7 µm), Thermo Syncronis aQ (2.1 mm × 100 mm, 1.7 µm), and Phenomenex OOD-4475-AN Kinetex C18 100A (2.1 mm × 100 mm, 1.7 µm) were investigated for studying the separation of complex components in AD. ACQUITY UPLC® CSHTM C18, CORTECS® UPLC® C18, and Thermo Syncronis aQ were selected for the establishment of the QAMS method. A CSH column had the best separation effect and was selected for its stability and excellent reproducibility for both high- and low-pH compounds. The methodology and durability of this experiment were studied with a Shimadzu UPLC instrument (LC-30AD), an ultrahigh performance liquid chromatograph.
Second, the DAD was used to scan the combined working solution at full wavelength to obtain better sensitivity, and the maximum absorption wavelength for each of the nine FCs was obtained. It is shown that, at 254 nm, the nine FC components exhibited relatively good absorbance.
Third, different mobile phases, for example, 0.1% formic acidacetonitrile, wateracetonitrile, and watermethanol with different proportional gradient elutions, were compared. The baseline condition showed that the mobile phase system of acetonitrile was better than that of methanol. The adoption of a 0.1% formic acid solution had no obvious effect on the CSH column; therefore, wateracetonitrile was used as the mobile phase. At the same time, the design of several chromatographic gradient methods, flow rates (0.2, 0.3, and 0.5 mL/min), and column temperatures (25 °C, 30 °C, and 35 °C) were optimized. Finally, we confirmed an acetonitrile water system for the mobile phase at 25 °C with a flow rate of 0.3 mL/min for analysis. Under the optimized conditions, the nine FCs were all separated from the baseline within 20 min. The UPLC chromatograms of the nine mixed standard solutions and the representative AD samples are shown in Figure 1.

3.2. Method Validation

The established UPLC protocol was evaluated for linearity, the limit of detection (LOD), the limit of quantification (LOQ), precision, stability, and accuracy. The correlation coefficients of the nine FCs were >0.9999 within the respective detection test ranges. The LOD and LOQ values of each of the FCs varied from 0.01–0.02 mg/mL to 0.04–0.07 mg/mL (Table 1). The RSD values of precision, repeatability, and stability for all nine FCs were <2.75% (Table 2). The overall recovery rate (n = 6) ranged from 90.07% to 107.94%, with RSD values of <3.00% (Table 2). These results indicated that the proposed method was precise, accurate, and efficient for simultaneously qualifying the nine FCs.

3.3. Quantitative Analyses of Multiple Components via Single Marker

The UPLC chromatograph detected nine FCs in the two species of AD. The chemical structures of the nine FCs are shown in Figure 2. For the QAMS method, selecting a proper internal standard substance for the calculation of RCFs and RRTs of the rest of the FCs in AD is vital. In this study, Im was selected as an internal standard substance owing to its high content, good stability, relatively moderate retention value, and easy availability in AD. Therefore, it met the requirements of QAMS. The calculated RCFs of eight FCs compared with Im are shown in Table 3. The results indicated that the RSDs of these FCs were <3%, calculated from the slope at various concentrations. Moreover, the mean RCFs were 0.891, 0.585, 1.136, 1.046, 0.587, 0.968, 1.051, and 1.015.
Further, the system suitability and durability of QAMS were evaluated. First, different instruments and diverse columns were investigated by calculating the RSD values of each RCF. Subsequently, the Shimadzu UPLC instrument with a CSH column was selected to assess the effects of changed flow rates (0.28, 0.30, and 0.32 mL/min) and column temperatures (23 °C, 25 °C, and 27 °C) on RCFs. The results show that these potential factors, including instruments, columns, flow rates, and column temperatures, displayed no substantial influence on RCFs based on the RSDs of < 3% (Table 4), which demonstrated that Im and the other eight FCs were robust under these conditions. Finally, three chromatographic columns with two different UPLC instruments assessed the RRTs. The results showed that all RSDs were <3%, implying that the RRT method could be used to locate the chromatographic peak component of nine FCs in AD (Table 5). The RRTs were 0.224, 0.237, 0.401, 0.518, 0.703, 0.726, 1.055, and 1.085.

3.4. Quantification and Method Assessment

BZ and HBZ are the two original plant sources of AD in China. Both the ESM and QAMS methods were performed simultaneously to determine the nine FCs in 20 AD samples. Subsequently, the SMD was calculated to assess the differences between the two methods. The results are shown in Supplementary Table S1. The absolute SMD values ranged from 0.02 % to 1.32 %, demonstrating that the accuracy of the content results obtained using QAMS could be accepted. Hence, it is feasible to determine the nine FCs in AD using QAMS simultaneously.
The results show that all samples of AD contain nine FCs, and the mean content of Oh, Bn, 8-MOP, 5-MOP, Bl, Op, Im, Ph, and Is are 0.47, 0.37, 0.08, 0.19, 0.37, 1.11, 1.95, 0.57, and 0.66 mg/g, respectively. As shown in Figure 3, the total content of the nine FCs varied from 4.38 to 9.37 mg/g, with those in BZ being relatively higher than in HBZ. Moreover, the most abundant ingredient of the nine quantified FCs in the two types of samples is Im. The content of Bl and Op were found to be substantially different in both samples.

3.5. Risk Assessment

In 1986, the IARC3 working group concluded that 8-MOP combined with ultraviolet radiation was carcinogenic to humans, based on sufficient evidence from both animal and human data [34]. The safety evaluation of furocoumarin via diet has been a concern for a long time. However, because of the toxicological data on all individual FCs being almost absent, this assessment mainly used the data on 5-MOP and 8-MOP, which are both used as the basis for risk assessment. To the best of our knowledge, furocoumarin is never found in plants as a single compound but always as complex mixtures of many individual furocoumarins. An overvalued scenario may emerge if a risk assessment is performed using only one type of furocoumarin, such as 8-MOP, as a basis for calculating the total toxic equivalent. However, diverse results are obtained when genuine mixtures of FCs in given herbal preparations are studied. Recently, the Raquet and Schrenk Group proposed an equivalency factor for phototoxicity and genotoxicity of furocoumarin mixtures. This provides the possibility for a toxicity estimation of different FCs and can be considered as the TEF. Herein, we calculated the EDI values based on the TEF. 5-MOP was selected as a basis or called a reference compound for the total risk assessment of FCs in AD owing to its strong photomutagenicity, as testified in previous studies and through its abundance in medicinal plants and food [12]. The TEF values of Oh, Bn, 8-MOP, Bl, Op, Im, Ph, and Is calculated based on research in the literature were 0.16, 0.01, 0.20, 0.01, 0.076, 0.15, 0.01, and 0.15, respectively [28]. The results showed that the EDI values of the total FCs in AD calculated without the TEF (0.029–0.061 mg/kg bw/day) were very close to the TDI values (0.1 mg/kg bw/day). However, after correction with the TEF, the EDI ranged from 0.004 to 0.008 mg/kg bw/day, revealing a reduced order of magnitude. According to the established HI-TEF strategy, the HI values of all the samples were <1, indicating that AD posed a low risk, which is consistent with the clinical and animal toxicity research [35,36]; the details are presented in Table 6.
The assessment in this study is quite conservative when compared with real clinical applications. AD as a part of TCM may rarely be used for a whole lifetime. In most cases, the duration of TCM usage will not exceed 2 years. In this exposure scenario, the EDI values of the total FCs in the 20 batches of AD samples are not more than 0.00023 mg/kg bw/day, which indicates that the risk of taking AD is extremely low. In addition, for the FCs with a TEF value of <0.01, we calculated under an assumption of a TEF equal to 0.01. Conversely, some uncertainties may cause underestimation in the risk calculation. For example, owing to the limited detection technology, some FCs of a low dose cannot yet be evaluated. In this study, we assumed that the toxicity of FCs in AD was the dose addition (DA) type. DA indicates that every single toxicant in the mixture contributes to the whole toxicity in proportion to its toxic unit, for example, its concentration or its potency. This implies that mixture toxicity will rise with the addition of individual agents. However, this is not the case. The joint toxicity of different FCs may be considerably larger or smaller than the toxicity (if any) associated with the NOAEL of each FC on its own [37,38]. Although there are numerous gaps and uncertainties in knowledge about the FCs from TCM, the established HI-TEF approach offers one possible strategy for the risk assessment of FCs originating from TCM.

4. Conclusions

In the present study, a fast, simple, credible, and sensitive evaluation method was developed for the accurate determination of nine FCs using single marks in 20 min based on UPLC. This method was successfully used for the content determination of AD samples in 20 batches. Thus, the problem of preparing a bulk of expense reference standards was effectively solved. Moreover, risk assessment methods for FCs from TCM in terms of toxicity were established for the first time, using AD as an example, based on the HI-TEF strategy. The cumulative risk assessment results revealed that the potential health risks of AD medicines were acceptable. Overall, the comprehensive application of the QAMS method alongside the HI-TEF risk assessment approach was meaningful, which could provide a significant reference for the rational utilization and quality control of FCs in TCM.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/separations10090508/s1, Table S1: Contents of the 9 FCs in AD determined by ESM and QAMS methods (mg/g, n = 3).

Author Contributions

Z.W.: conceptualization, formal analysis, data acquisition, examination, writing—original draft. K.Z.: methodology, validation. X.-W.H.: investigation. S.K.: collection and identification of samples. H.-L.L.: investigation. H.-Y.J.: investigation, data curation. T.-T.Z.: writing—review and editing. S.-C.M.: writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Traditional Chinese Medicine Modernization Project, under grant numbers 2022YFC3501505.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request. Others come from previously reported studies and datasets, which have been cited.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Melough, M.M.; Cho, E.; Chun, O.K. Furocoumarins: A review of biochemical activities, dietary sources and intake, and potential health risks. Food Chem. Toxicol. 2018, 113, 99–107. [Google Scholar] [CrossRef] [PubMed]
  2. Santana, L.; Uriarte, E.; Roleira, F.; Milhazes, N.; Borges, F. Furocoumarins in medicinal chemistry. Synthesis, natural occurrence and biological activity. Curr. Med. Chem. 2004, 11, 3239–3261. [Google Scholar] [CrossRef] [PubMed]
  3. Scott, B.R.; Pathak, M.A.; Mohn, G.R. Molecular and genetic basis of furocoumarin reactions. Mutat. Res. 1976, 39, 29–74. [Google Scholar] [CrossRef] [PubMed]
  4. Sayre, R.M.; Dowdy, J.C. The increase in melanoma: Are dietary furocoumarins responsible? Med. Hypotheses 2008, 70, 855–859. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, S.W.; Cho, E.; Feskanich, D.; Li, W.Q.; Sun, Q.; Han, J.L.; Qureshi, A.A. Citrus consumption and risk of basal cell carcinoma and squamous cell carcinoma of the skin. Carcinogenesis 2015, 36, 1162–1168. [Google Scholar] [CrossRef] [PubMed]
  6. Wu, S.W.; Han, J.L.; Feskanich, D.; Cho, E.; Stampfer, M.J.; Willett, W.C.; Qureshi, A.A. Citrus Consumption and Risk of Cutaneous Malignant Melanoma. J. Clin. Oncol. 2015, 33, 2500–2508. [Google Scholar] [CrossRef]
  7. Abraham, K.; Wöhrlin, F.; Lindtner, O.; Heinemeyer, G.; Lampen, A. Toxicology and risk assessment of coumarin: Focus on human data. Mol. Nutr. Food Res. 2010, 54, 228–239. [Google Scholar] [CrossRef]
  8. Müller, L.; Kasper, P.; Kersten, B.; Zhang, J. Photochemical genotoxicity and photochemical carcinogenesis—Two sides of a coin? Toxicol. Lett. 1998, 102–103, 383–387. [Google Scholar] [CrossRef]
  9. Kitamura, N.; Kohtai, S.; Nakagaki, R. Molecular aspects of furocoumarin reactions: Photophysics, photochemistry, photobiology, and structural analysis. J. Photochem. Photobiol. C Photochem. Rev. 2005, 6, 168–185. [Google Scholar] [CrossRef]
  10. Bordin, F. Photochemical and photobiological properties of furocoumarins and homologues drugs. Int. J. Photoenergy 1999, 1, 761394. [Google Scholar] [CrossRef]
  11. Guo, L.Q.; Yamazoe, Y. Inhibition of cytochrome P450 by furanocoumarins in grapefruit juice and herbal medicines. Acta Pharmacol. Sin. 2004, 25, 129–136. [Google Scholar] [PubMed]
  12. European Medicines Agency. Reflection Paper on the Risk Associated with Furocoumarins Contained in Preparations of Angelica archangelica L. EMEA/HMPC/317913/2006 [EB/OL]. 2007. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/final-reflection-paper-risks-associated-furocoumarins-contained-preparations-angelica-archangelica-l_en.pdf (accessed on 9 January 2021).
  13. Raquet, N.; Schrenk, D. Relative photomutagenicity of furocoumarins and limettin in the hypoxanthine phosphoribosyl transferase assay in V79 cells. Chem. Res. Toxicol. 2009, 22, 1639–1647. [Google Scholar] [CrossRef] [PubMed]
  14. Chinese Pharmacopeia Commission. Pharmacopoeia of People’s Republic of China; Chemical Medical Science Press: Beijing, China, 2020. [Google Scholar]
  15. Wang, Q.Q.; Li, Y.N.; Wang, S.G.; Xiang, Z.D.; Dong, W.C.; Li, X.Y.; Wei, Y.M.; Gao, P.; Dai, L. A review of the historical records, chemistry, pharmacology, pharmacokinetics and edibility of Angelica dahurica. Arab. J. Chem. 2023, 16, 1878–5352. [Google Scholar] [CrossRef]
  16. Zhu, C.; Wang, M.; Guo, J.; Su, S.L.; Yu, G.; Yang, Y.; Zhou, Y.; Tang, Z. Angelica dahurica Extracts Attenuate CFA Induced Inflammatory Pain via TRPV1 in Mice. Evid. Based Complement. Altern. Med. 2022, 2022, 4684830. [Google Scholar] [CrossRef] [PubMed]
  17. Xie, Y.; Zhao, W.; Zhou, T.; Fan, G.; Wu, Y. An efficient strategy based on MAE, HPLC-DAD-ESI-MS/MS and 2D-prep-HPLC-DAD for the rapid extraction, separation, identification and purification of five active coumarin components from Radix Angelicae Dahuricae. Phytochem. Anal. 2010, 21, 473–482. [Google Scholar] [CrossRef] [PubMed]
  18. Zhao, H.; Feng, Y.L.; Wang, M.; Wang, J.J.; Liu, T.; Yu, J. The Angelica dahurica: A Review of Traditional Uses, Phytochemistry and Pharmacology. Front. Pharmacol. 2022, 13, 896637. [Google Scholar] [CrossRef] [PubMed]
  19. Zhou, B.; Liu, P.; Chen, J.; Xu, Y.; Chen, D.W.; Duan, J.A. Analysis and evaluation of coumarins and polysaccharides in Angelica dahurica from different habitats. Acta. Univ. Med. 2015, 31, 68–73. [Google Scholar]
  20. Zhou, Y.; Na, L. Research progress on medicinal and food homology of Angelica dahurica. Asian-Pac. Tradit. Med. 2022, 18, 213–217. [Google Scholar]
  21. Pang, X.Y.; Jing, Y.S.; Zheng, Y.G.; Wu, L.F. Chemical constituents of Angelica dahurica and its therapeutic effect on nervous system diseases. Chin. J. Pharmacol. Toxicol. 2021, 35, 690–691. [Google Scholar]
  22. Liu, S.; Huang, X.; Lin, Y.; Lin, H.; Wang, Q.; Liu, Z. Skin phototoxicity study of radix Angelicae dahuricae water extract using rats by oral administration. J. Hunan Agric. Univ. Nat. Sci. 2018, 44, 424–429. [Google Scholar]
  23. Kang, J.; Zhou, L.; Sun, J.; Han, J.; Guo, D.A. Chromatographic fingerprint analysis and characterization of furocoumarins in the roots of Angelica dahurica by HPLC/DAD/ESI-MSn technique. J. Pharm. Biomed. Anal. 2008, 47, 778–785. [Google Scholar] [CrossRef]
  24. Zhang, H.; Gong, C.G.; Lv, L.; Xu, Y.J.; Zhao, L.; Zhu, Z.Y.; Chai YFZhang, G.Q. Rapid separation and identification of furocoumarins in Angelica dahurica by high-performance liquid chromatography with diode-array detection, time-of-flight mass spectrometry and quadrupole ion trap mass spectrometry. Rapid Commun. Mass Spectrom. 2009, 23, 2167–2175. [Google Scholar] [CrossRef] [PubMed]
  25. Yang, W.Q.; Zhu, Z.X.; Song, Y.L.; Qi, B.W.; Wang, J.; Su, C.; Tu, P.F.; Shi, S.P. Dimeric furanocoumarins from the roots of Angelica dahurica. Nat. Prod. Res. 2017, 31, 870–877. [Google Scholar] [CrossRef] [PubMed]
  26. Shu, P.; Li, J.P.; Fei, Y.Y.; Zhu, H.Q.; Yu, M.Z.; Liu, A.Q.; Niu, H.Y.; Zou, S.M.; Wei, X.L.; Ju, Z.Y.; et al. Isolation, structure elucidation, tyrosinase inhibitory, and antioxidant evaluation of the constituents from Angelica dahurica roots. J. Nat. Med. 2020, 74, 456–462. [Google Scholar] [CrossRef] [PubMed]
  27. Lohr, C.; Raquet, N.; Schrenk, D. Application of the concept of relative photomutagenic potencies to selected furocoumarins in V79 cells. Toxicol. Vitr. 2010, 24, 558–566. [Google Scholar] [CrossRef] [PubMed]
  28. Raquet, N.; Schrenk, D. Application of the equivalency factor concept to the phototoxicity and -genotoxicity of furocoumarin mixtures. Food Chem. Toxicol. 2014, 68, 257–266. [Google Scholar] [CrossRef] [PubMed]
  29. EFSA. Scientific opinion of the panel on food additives, flavourings, processing aids and materials in contact with food on a request from the European Commission on Coumarin in flavourings and other food ingredients with flavouring properties. EFSA J. 2008, 6, 793–808. [Google Scholar]
  30. Huang, J.; Yin, L.; Dong, L.; Quan, H.F.; Chen, R.; Hua, S.Y.; Ma, J.H.; Guo, D.Y.; Fu, X.Y. Quality evaluation for Radix Astragali based on fingerprint, indicative components selection and QAMS. Biomed. Chromatogr. 2018, 32, 4343. [Google Scholar] [CrossRef] [PubMed]
  31. Peng, Y.; Dong, M.; Zou, J.; Liu, Z. Analysis of the HPLC fingerprint and QAMS for Sanhuang gypsum soup. J. Anal. Methods Chem. 2018, 2018, 5890973. [Google Scholar] [CrossRef]
  32. Dong, Y.; Guo, Q.; Liu, J.; Ma, X. Simultaneous determination of seven phenylethanoid glycosides in Cistanches Herba by a single marker using a new calculation of relative correction factor. J. Sep. Sci. 2018, 41, 1913–1922. [Google Scholar] [CrossRef]
  33. Zuo, T.T.; Jin, H.Y.; Zhang, L.; Liu, Y.L.; Nie, J.; Chen, B.L.; Fang, C.F.; Xue, J.; Bi, X.Y.; Li, Z.; et al. Innovative health risk assessment of heavy metals in Chinese herbal medicines based on extensive data. Pharmacol. Res. 2020, 159, 104987–104999. [Google Scholar] [CrossRef]
  34. International Agency for Research on Cancer (IARC). 5-Methoxypsoralen [A]//IARC Monographs on the Evaluation of Carcinogenic Risks to Humans; World Health Organization: Lyon, France, 1986. [Google Scholar]
  35. Zhao, W.H.; Cao, Y.X.; Liu, J.; Gao, Y.W.; He, J.Y.; Cao, L.Q. Research on chronic toxicology of Compound Radix Angelicae Dahuricae capsule. J. Shanxi Med. Univ. 2006, 37, 160–163. [Google Scholar]
  36. Zheng, G.Y.; Ma, Y.Y.; Lu, X.L.; Hai, M.Z.; Guo, P.L. Effect of sulfuring on the acute toxicity of the angelica extract. Pharm. Clin. Chin. Mater. Medica 2012, 3, 43–50. [Google Scholar]
  37. Kortenkamp, A.; Evans, R.; Faust, M.; Kalberlah, F.; Scholze, M.; Wolz, S.U. Investigation of the state of the science on combined actions of chemicals in food through dissimilar modes of action and proposal for science-based approach for performing related cumulative risk assessment. EFSA Support. Publ. 2012, 9, 1–300. [Google Scholar] [CrossRef]
  38. European Food Safety Authority. Opinion of the Scientific Panel on food additives, flavourings, processing aids and materials in contact with food (AFC) related to Flavouring Group Evaluation 6 (FGE.06): Straight-and branched-chain aliphatic unsatured primary alcohols, aldehydes, carboxylic acids, and esters from chemical groups 1 and 4. EFSA J. 2004, 2, 108. [Google Scholar]
Figure 1. Representative UPLC chromatograms of the standards (A), the AD sample (BZ, Species No. 1) (B), and the AD sample (HBZ, Species No. 2) (C). Peak identification: oxypeucedanin hydrate (Oh, 1), byakangelicin (Bn, 2), xanthotoxin (8-methoxypsoralen, 8-MOP, 3), bergapten (5-methoxypsoralen, 5-MOP, 4), byakangelicol (Bl, 5), oxypeucedanin (Op, 6), imperatorin (Im, 7), phellopterin (Ph, 8), and isoimperatorin (Is, 9).
Figure 1. Representative UPLC chromatograms of the standards (A), the AD sample (BZ, Species No. 1) (B), and the AD sample (HBZ, Species No. 2) (C). Peak identification: oxypeucedanin hydrate (Oh, 1), byakangelicin (Bn, 2), xanthotoxin (8-methoxypsoralen, 8-MOP, 3), bergapten (5-methoxypsoralen, 5-MOP, 4), byakangelicol (Bl, 5), oxypeucedanin (Op, 6), imperatorin (Im, 7), phellopterin (Ph, 8), and isoimperatorin (Is, 9).
Separations 10 00508 g001
Figure 2. Chemical structures of FCs analyzed in AD: oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), imperatorin (7), phellopterin (8), and isoimperatorin (9).
Figure 2. Chemical structures of FCs analyzed in AD: oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), imperatorin (7), phellopterin (8), and isoimperatorin (9).
Separations 10 00508 g002
Figure 3. Comparison of FCs in 20 batches of samples. Ci represents the average content of oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), imperatorin (7), phellopterin (8), and isoimperatorin (9).
Figure 3. Comparison of FCs in 20 batches of samples. Ci represents the average content of oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), imperatorin (7), phellopterin (8), and isoimperatorin (9).
Separations 10 00508 g003
Table 1. Linearity (regression equation, linear range, and correlation coefficient) and sensitivity (LOD and LOQ) of the method determination of the eight FCs.
Table 1. Linearity (regression equation, linear range, and correlation coefficient) and sensitivity (LOD and LOQ) of the method determination of the eight FCs.
AnalytesCalibration Curvesr/μg·mL−1/μg·mL−1/μg·mL−1
1Y = 24489800X + 4561.670.99991.270–158.70.010.05
2Y = 16185800X + 850.890.99990.3970–49.600.010.04
3Y = 31411100X + 1986.140.99990.4580–57.280.010.04
4Y = 29319800X + 1010.050.99990.4290–53.650.010.04
5Y = 16335500X + 1443.080.99990.7210–90.150.020.07
6Y = 26801600X + 5930.630.99992.070–258.80.020.07
7Y = 27494800X + 3435.560.99990.8160–102.00.010.04
8Y = 28897035X + 1351.040.99990.8000–100.500.020.07
9Y = 28331800X + 597.950.99990.8200–102.500.010.04
‘1–9’ represents the analytes of oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), imperatorin (7), phellopterin (8), and isoimperatorin (9).
Table 2. Precision, stability, repeatability, and recovery results of nine FCs.
Table 2. Precision, stability, repeatability, and recovery results of nine FCs.
AnalytesPrecision (RSD/%, n = 6)Stability (RSD/%, n = 6)Repeatability (RSD/%, n = 6)Recoveries (n = 6)
Mean/%RSD/%
10.020.822.0197.572.98
20.090.761.89107.92.59
30.521.102.75107.92.98
40.191.142.07106.92.91
50.231.071.9390.072.98
60.020.931.9192.092.66
70.011.041.9793.763.00
80.210.892.10101.52.62
90.010.631.9891.972.16
‘1–9’ represents the analytes of oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), imperatorin (7), phellopterin (8), and isoimperatorin (9).
Table 3. Relative correction factor (RCF) values of the eight FCs of the AD calculated via slope and concentration method (n = 3).
Table 3. Relative correction factor (RCF) values of the eight FCs of the AD calculated via slope and concentration method (n = 3).
RCFCalculated by SlopeabcdefMeanRSD
(%)
f7/10.8910.8900.8920.8930.8920.8910.8890.8910.17
f7/20.5890.5740.5830.5850.5870.5880.5890.5850.91
f7/31.1421.1241.1311.1361.1371.1401.1421.1360.58
f7/41.0661.0161.0251.0411.0541.0601.0611.0461.85
f7/50.5940.5680.5840.5890.5900.5930.5930.5871.57
f7/60.9750.9610.9650.9670.9680.9700.9710.9680.46
f7/81. 0511.0551.0531.0521.0491.0481.0511. 0510.25
f7/91.0301.0081.0091.0111.0121.0141.0181.0150.76
fi’ represents the RCFs of oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), phellopterin (8), and isoimperatorin (9) compared with imperatorin (7). ‘a–f’ indicates six different concentration levels. Taking imperatorin (7) as an example, its concentration levels are 0.816 μg/mL, 2.04 μg/mL, 4.08 μg/mL, 10.2 μg/mL, 20.4 μg/mL, and 102 μg/mL, respectively.
Table 4. Effects of different instruments, columns, column temperatures, and flow rates on RCFs of nine FCs (n = 3).
Table 4. Effects of different instruments, columns, column temperatures, and flow rates on RCFs of nine FCs (n = 3).
ConditionsItemsf7/1f7/2f7/3f7/4f7/5f7/6f7/8f7/9
Waters ACQUITY UPLC H-ClassColumn 10.8980.5821.1101.0510.5990.9731.0551.018
Column 20.8900.5411.1251.0820.5470.9671.0491.042
Column 30.8810.5911.0411.0450.5970.9661.0621.021
Shimadzu UPLC Instrument (LC-30AD)Column 10.8910.5851.1361.0460.5870.9681.0511.015
Column 20.8780.5231.1211.1030.5710.9781.0481.038
Column 30.9020.5761.0641.0450.5900.9941.0491.016
 Mean0.8900.5661.1001.0620.5820.9741.0521.025
 RSD (%)1.054.883.472.323.391.100.511.16
Column temperature (°C)230.8900.6031.1411.0240.5770.9711.05181.018
250.8910.5851.1361.0460.5870.9681.0501.015
270.8910.5821.1221.0530.5940.9621.04751.014
Mean0.8910.5901.1331.0410.5860.9671.0501.016
RSD (%)0.071.920.871.461.460.470.210.21
Flow rates (mL/min)0.280.8720.5691.1141.0410.5790.9661.0491.006
0.300.8910.5851.1361.0460.5870.9681.0501.015
0.320.9250.6011.1511.0520.5950.9711.05241.034
Mean0.8960.5851.1341.0460.5870.9681.0511.018
RSD (%)3.002.741.640.531.360.260.171.41
Column 1—ACQUITY UPLC® CSHTM C18(2.1 mm × 100 mm, 1.7 µm). Column 2—CORTECS® UPLC® C18 Column (2.1 mm × 100 mm, 1.6 µm). Column 3—Thermo Syncronis aQ (2.1 mm × 100 mm, 1.7 µm).
Table 5. Relative retention values measured in different instruments and different columns (n = 3).
Table 5. Relative retention values measured in different instruments and different columns (n = 3).
InstrumentColumnt7/1t7/2t7/3t7/4t7/5t7/6t7/8t7/9
Waters ACQUITY UPLC H-ClassZORBAX Eclipse XDB-C180.2250.2390.4010.5090.7010.7301.0491.087
ZORBAX Eclipse plus C180.2210.2350.3970.5170.7060.7251.0511.084
ACQUITY UPLC HSS, T30.2230.2350.3990.5290.7030.7191.0641.081
Shimadzu UPLC Instrument (LC-30AD)ZORBAX Eclipse XDB-C180.2290.2410.4140.5110.7050.7261.0521.089
ZORBAX Eclipse plus C180.2220.2360.3980.5180.7060.7261.0521.084
ACQUITY UPLC HSS, T30.2230.2340.3980.5220.6980.7281.0631.083
mean 0.2240.2370.4010.5180.7030.7261.0551.085
RSD/% 1.291.141.611.410.460.510.620.27
‘ti’ represents the relative retention time (RRT) of oxypeucedanin hydrate (1), byakangelicin (2), xanthotoxin (3), bergapten (4), byakangelicol (5), oxypeucedanin (6), phellopterin (8), and isoimperatorin (9) compared with imperatorin (7).
Table 6. Estimated daily intake (EDI) and hazard index (HI) in 20 batches of AD samples.
Table 6. Estimated daily intake (EDI) and hazard index (HI) in 20 batches of AD samples.
Batch No.SourceSpeciesEDI1EDI2EDI3EDI4EDI5EDI6EDI7EDI8EDI9ΣEDI Based on TEFΣEDI without TEFHI Values Based on TEFHI Values without TEF
B1Bozhou, Anhui10.000840.000050.000290.001940.000050.000720.002980.000060.001090.00800.06130.806.13
B2Anguo, Hebei10.000450.000030.000050.001050.000030.001180.002200.000060.000750.00580.05060.585.06
B3Anguo, Hebei10.000280.000020.000060.001160.000040.000870.001790.000040.000690.00500.04110.504.11
B4Anguo, Hebei10.000530.000040.000110.001570.000060.001180.001680.000040.000770.00600.05070.605.07
B5Anguo, Hebei10.000300.000020.000050.001030.000050.000950.002030.000030.000590.00500.04360.504.36
B6Jiaozuo, Hebei10.000200.000010.000250.001850.000020.000560.001470.000030.000550.00490.03160.493.16
B7Hong Kong10.000210.000010.000270.001940.000020.000600.001530.000040.000590.00520.03330.523.33
B8Hong Kong10.000620.000020.000050.000850.000020.000490.001610.000030.000600.00430.03360.433.36
B9Hong Kong10.000580.000020.000060.000830.000010.000390.001600.000030.000590.00410.03080.413.08
B10Hong Kong10.000630.000020.000070.000860.000020.000410.001890.000030.000620.00460.03510.463.51
B11Shehong, Sichuan20.000410.000020.000100.000920.000010.000410.001670.000030.000500.00410.02930.412.93
B12Suining, Sichuan20.000630.000020.000180.001250.000010.000380.001470.000030.000530.00450.03060.453.06
B13Suining, Sichuan20.000820.000050.000190.001120.000010.000190.001570.000030.000480.00450.03250.453.25
B14Suining, Sichuan20.000480.000010.000070.000810.000010.000400.001660.000030.000480.00400.02860.402.86
B15Suining, Sichuan20.000700.000020.000040.000830.000010.000350.001640.000030.000590.00420.03090.423.09
B16Suining, Sichuan20.000410.000020.000100.001300.000030.000540.002320.000040.000670.00540.03950.543.95
B17Hong Kong20.000480.000020.000000.001080.000020.000310.002410.000040.000530.00490.03570.493.57
B18Hong Kong20.000460.000030.000000.001290.000020.000350.002500.000040.000790.00550.04020.554.02
B19Hong Kong20.000520.000030.000000.001300.000020.000390.001990.000040.000700.00500.03630.503.63
B20Hong Kong20.000340.000020.000070.001400.000030.000340.002190.000040.000730.00520.03610.523.61
‘B1–B20’ represents the batch numbers of AD samples. ‘1,2’ represents the species numbers of AD samples. 1—Angelica dahurica (Hoffm.) Benth. & Hook. f. ex Franch. & Sav. 2—Angelica dahurica var. formosana (Boissieu) Yen.
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

Wang, Z.; Zan, K.; Hu, X.-W.; Kang, S.; Li, H.-L.; Zuo, T.-T.; Jin, H.-Y.; Ma, S.-C. The Simultaneous Determination of Nine Furocoumarins in Angelica dahurica Using UPLC Combined with the QAMS Approach and Novel Health Risk Assessment Based on the Toxic Equivalency Factor. Separations 2023, 10, 508. https://doi.org/10.3390/separations10090508

AMA Style

Wang Z, Zan K, Hu X-W, Kang S, Li H-L, Zuo T-T, Jin H-Y, Ma S-C. The Simultaneous Determination of Nine Furocoumarins in Angelica dahurica Using UPLC Combined with the QAMS Approach and Novel Health Risk Assessment Based on the Toxic Equivalency Factor. Separations. 2023; 10(9):508. https://doi.org/10.3390/separations10090508

Chicago/Turabian Style

Wang, Zhao, Ke Zan, Xiao-Wen Hu, Shuai Kang, Hai-Liang Li, Tian-Tian Zuo, Hong-Yu Jin, and Shuang-Cheng Ma. 2023. "The Simultaneous Determination of Nine Furocoumarins in Angelica dahurica Using UPLC Combined with the QAMS Approach and Novel Health Risk Assessment Based on the Toxic Equivalency Factor" Separations 10, no. 9: 508. https://doi.org/10.3390/separations10090508

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