Next Article in Journal
Unveiling the Hidden Therapeutic Potential of Carnosine, a Molecule with a Multimodal Mechanism of Action: A Position Paper
Next Article in Special Issue
Croargoids A–G, Eudesmane Sesquiterpenes from the Bark of Croton argyratus
Previous Article in Journal
Polyphenols and Maillard Reaction Products in Dried Prunus spinosa Fruits: Quality Aspects and Contribution to Anti-Inflammatory and Antioxidant Activity in Human Immune Cells Ex Vivo
Previous Article in Special Issue
Systematic Screening of Chemical Constituents in the Traditional Chinese Medicine Arnebiae Radix by UHPLC-Q-Exactive Orbitrap Mass Spectrometry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Chemical Constituent Analysis of Ranunculus sceleratus L. Using Ultra-High-Performance Liquid Chromatography Coupled with Quadrupole-Orbitrap High-Resolution Mass Spectrometry

1
School of Pharmacy, Anhui Medical University, Hefei 230032, China
2
Anhui Institute for Food and Drug Control, Hefei 230051, China
3
The Center for Scientific Research of Anhui Medical University, Hefei 230032, China
*
Authors to whom correspondence should be addressed.
Submission received: 4 May 2022 / Revised: 17 May 2022 / Accepted: 19 May 2022 / Published: 20 May 2022

Abstract

:
Ranunculus sceleratus L.(RS) has shown various pharmacological effects in traditional Chinese medicine. In our previous study, the positive therapeutic effect on α-naphthylisothiocyanate induced intrahepatic cholestasis in rats was obtained using TianJiu treatment with fresh RS. However, the chemical profile of RS has not been clearly clarified, which impedes the research progress on the therapeutic effect of RS. Herein, an ultra-high performance liquid chromatography coupled with quadrupole Orbitrap high-resolution mass spectrometry (UHPLC-Q-Orbitrap HRMS) method was developed to rapidly separate and identify multiple constituents in the 80% methanol extract of RS. A total of sixty-nine compounds (19 flavonoids, 22 organic acids, 6 coumarins, 4 lignans, 14 nitrogenous compounds, and 4 anthraquinones) were successfully characterized. A total of 12 of these compounds were unambiguously identified by standard samples. Their mass spectrometric fragmentation pathways were investigated. It is worth noting that flavonoids and lignans were identified for the first time in RS. In this study, we successfully provide the first comprehensive report on identifying major chemical constituents in RS by UHPLC-Q-Orbitrap HRMS. The obtained results enrich the RS chemical profile, paving the way for further phytochemical study, quality control, and pharmacological investigation of RS.

Graphical Abstract

1. Introduction

Ranunculus sceleratus L. (RS), an annual herbaceous plant belonging to the Ranunculus L. family, has been listed among the top herbs in the Shennong Traditional Herbal Scriptures, written in the Western Han Dynasty. Compendium of Materia Medica notes that fresh RS can be pasted onto the acupuncture point of Cunkou overnight, (hyperemia and blistering in the skin), to cure jaundice induced by malaria. TianJiu treatment with fresh RS patches exhibited a positive therapeutic effect on α-naphthylisothiocyanate-intrahepatic cholestasis in rats by pasting on the acupuncture points of Dazhui, Ganshu (both sides), and Jizhong in our previous study [1]. The protoanemonin of fresh RS is commonly used as a robust blistering agent. It has been reported that protoanemonin is poisonous, but toxins will be destroyed when fresh RS is heated or dried. Moreover, the anti-inflammatory activity of the extract of RS administered orally at a dose of 100 mg·kg−1 was obtained in Wistar rats by inhibiting the induced hind paw edema [2]. A dosage between 3 and 9 g is recorded in Zhonghuabencao when taken orally as a decoct soup to clear heat, reduce swelling, disperse the knot, and relieve pain. However, little is known about the chemical basis of the therapeutic effect of RS. Over the past few years, several chemical constituents have been isolated from RS, such as emodin, scoparone, isoscopoletin, protocatechualdehyde, protocatechuic acid, hexadecanoic acid, β-sitosterol, stigmast-4-ene-3,6-dione, stigmasterol, 1-docosene, and stigmast-5-en-3-ol [3,4]. In our previous study, 31 volatile compounds were identified in fresh RS by GC-MS analysis. The main chemical constituents were lactones and phenolic acids, including protoanemonin, 2,5-furandione, 2-propanedioic acid, and phenacetic acid [1]. Considering the positive therapeutic application of RS, it is of great importance to establish a rapid and reliable method for chemical profiling RS.
Ultra-high performance liquid chromatography coupled with quadrupole Orbitrap high-resolution mass spectrometry (UHPLC-Q-Orbitrap HRMS), with mass accuracy and high sensitivity for precursor and product ions, exhibits high resolution. Conventional separation and identification processes are time and plant-material-consuming. In contrast, the UHPLC-Q-Orbitrap HRMS method has shown high efficiency in the separation, identification, and analysis of multiple chemical constituents of traditional Chinese medicines (TCMs) [5,6,7]. Therefore, this method can tentatively identify components without reference standards according to exact MS data. The UHPLC-Q-Orbitrap MS method combined the inclusion list, and data-dependent acquisition was established to achieve a comprehensive characterization of the TCM formula Kai-Xin-San, and 211 compounds were identified by comparison with reference standards, literature data, and databases [8]. A total of 18 phenolic acids were identified in Lycium ruthenicum Murray by comparing the retention time and the exact m/z in authentic standards using UPLC-Q-Orbitrap MS [9].
To the best of our knowledge, the comprehensive identification of multiple chemical constituents in RS has not been reported. A series of studies were carried out for the rapid characterization of the chemical profile in RS. First, we established a chemical database for the compounds of Ranunculus L. family plants and collected as many standards as possible. Second, samples were analyzed by UHPLC-Q-Orbitrap HRMS technology for full-spectrum scanning, and the spectrum information was obtained by Xcalibur 4.1 software. Finally, after characterizing the diagnostic ions and fragmentation rules for the standards, the fragment information in the mass spectrum was compared with the literature to confirm the chemical constituents of RS. In the present study, 19 flavonoids, 22 organic acids, 6 coumarins, 4 lignans, 14 nitrogenous compounds, and 4 anthraquinones from RS were identified by UHPLC-Q-Orbitrap HRMS, providing in-depth knowledge of its chemical constituents and offering valuable information regarding its quality control and further pharmacological study.

2. Results and Discussion

2.1. Optimization of UHPLC-Q-Orbitrap HRMS Conditions

The composition of the UHPLC mobile phase should be systemically optimized to achieve the best chromatographic and mass spectrometric properties for separation and analysis. To assess the resulting peak shape and signal strength, different mobile systems that consisted of methanol−0.1% formic acid aqueous solution or acetonitrile−0.1% formic acid aqueous solution were investigated. In the result, acetonitrile−0.1% formic acid aqueous solution was chosen as the UHPLC mobile phase for gradient elution. In terms of sample processing, 60%, 80%, and 100% methanol (v/v) were examined to obtain the optimal RS extract that contains the more chemical constituents of RS. It was found that 80% methanol showed the best extraction efficiency based on the number of peaks and was selected as the extraction solvent for RS. An 80% methanol extract of RS was then analyzed in the positive and negative ion modes. Most chemical constituents showed higher responses in negative ionization mode than in positive ionization mode. In addition, to improve the sensitivity and accuracy, the ion source voltage and capillary temperature were optimized. The optimal conditions are described in Section 3.3.

2.2. Identification of Chemical Constituents in RS

Under the optimal UHPLC-Q-Orbitrap HRMS conditions, the base peak chromatograms obtained in positive and negative ionization modes for the 80% methanol extract of RS are shown in Figure 1. As shown in Table 1, a total of 69 compounds (19 flavonoids, 22 organic acids, 6 coumarins, 4 lignans, 14 nitrogenous compounds, and 4 anthraquinones) in the 80% methanol extract of RS were either unambiguously identified (12 compounds) or tentatively characterized (57 compounds). Reference standards of isoscopoletin, scopoletin, and scoparone were detected in positive ionization mode (Figure S1), and reference standards of aesculetin, quercetin, protocatechuic acid, salicylic acid, ferulic acid, luteolin, caffeic acid, emodin, and oleanic acid were detected in negative ionization mode (Figure S2). Next, the names and chemical structures of the other 57 compounds were preliminarily inferred by comparing MS ion fragmentation information with relevant literature. Altogether, the chemical structures of identified 69 compounds are summarized in Figure S3.

2.2.1. Flavonoids

Flavonoids refer to a class of naturally occurring bioactive compounds in herbal medicine. The principal mass spectrometric fragmentation mechanisms for flavonoids are loss of neutral fragments, such as H2O, CH3, CO, CO2, and the cleavage of retro-Diels-Alder (RDA) at glycosyl bonds. The product ion spectrum of compound 39 shown in Figure 2A is representative. In ESI, compound 39 produced the deprotonated molecular ion at m/z 463.0882 [M − H], which could form the fragment ion m/z 301.0350 [M − H − C6H10O5] by losing one glucose sugar group (C6H10O5). After undertaking the RDA reaction, a series of product ions could be produced, such as m/z 151.0023 and 107.0123. By comparing their molecular formulas and fragmentation patterns with those reported in the literature [29], compound 39 was tentatively identified as hyperoside, reported here for the first time in RS. Similarly, the [M − H] ion of compound 47 was shown at m/z 445.0752, and was tentatively identified as apigenin-7-O-glucuronide. Characteristic fragment ions m/z 269.0452 were produced by successive loss of glucuronide [28]. Glucuronide residues at m/z 175.0233 and m/z 113.0229 could always be observed in glycosides. The mass spectra and proposed major fragmentations with structures are shown in Figure 2B. Except for luteolin (55) and quercetin (56), confirmed by reference standards, all other flavonoids were identified similarly. The other flavonoids identified according to their molecular mass, formulas, MS/MS fragments, and related literature studies, including kaempferol-3,7-di-O-glucoside (26), kaempferol-O-sophoroside (30), quercetin-O-(pentosyl) hexoside isomer (33), isoorientin (35), isorhamnetin (41), kaempferol (42), isorhamnetin-3-O-glucoside isomer (43), luteoloside (45), luteolin-7-O-glucuronide (48), apigenin (58), chrysoeriol (59), diosmetin (61), rhamnetin (62), pinocembrin (64), 7,3’-dihydroxy-8,4’-dimethoxyisoflavone/isomer (65). Flavonoids were identified for the first time in RS, and pharmacological studies have demonstrated that flavonoids have good efficacy in treating cholestatic liver disease [39].

2.2.2. Organic Acids

Organic acids widely occur in natural plants, especially in herbs. In negative ionization mode, organic acids exhibited deprotonated molecular peaks and easily lost CO2 and CO to generate corresponding fragment ion peaks. Moreover, the losses of small molecules (like H2O) or radicals (like CH3) sometimes occurred. For instance, protocatechuic acid (compound 21) showed a molecular formula of C7H6O4 and a deprotonated molecule [M − H] peak at m/z 153.0181. The deprotonated molecule lost a CO2 moiety to form a fragment ion [M − H − CO2] at m/z 109.0280. Then, it was dehydrated to form the [M − H − CO2 − H2O] fragment ion of m/z 91.0176. The fragmentation pathways of compound 21 are shown in Figure 3A. Compound 21 was unambiguously identified as protocatechuic acid by comparing its MS/MS fragmentation pattern and retention time of reference standard. Likewise, compounds 32, 46, 53, and 69 were identified as caffeic acid, ferulic acid, salicylic acid, and oleanic acid by reference standards. The deprotonated molecule [M − H] of compound 25 peak was at m/z 299.0777, and its chemical structure and fragmentation pathway are shown in Figure 3B. Compound 25 was identified as salicylic acid-O-glucopyranoside according to the literature [19]. The other compounds were identified according to their molecular mass, formulas, MS/MS fragments, and related literature studies, including gluconic acid (5), malic acid (8), aconitic acid (12), furoic acid (13), citric acid (14), succinic acid (16), protocatechuic acid-O-glucose (18), vanillic acid-O-glucopyranoside (19), hydroxytyrosol-1-glucopyranoside (20), caffeic acid-O-glucopyranoside (22), protocatechualdehyde (27), p-coumaric acid-O-glucopyranoside (29), suberic acid (36), coumaric acid (40), azelaic acid (50), and p-hydroxybenzene propanoic acid (51).

2.2.3. Coumarins and Lignans

Six coumarins were identified from the 80% methanol extract of RS, including esculin (24), scopolin (28), aesculetin (31), isoscopoletin (37), scopoletin (44), and scoparone (54). Their fragmentation patterns in mass spectrometry were investigated, and neutral losses of CO and CO2 could be commonly observed. A typical coumarin, scopoletin (44), was taken as an example to investigate the MS/MS fragmentation pattern of coumarin in RS. The protonated molecular ion of compound 44 was m/z 193.0493 [M + H]+ in positive ESI mode. Scopoletin produced a fragment ion [M + H − CH3]+ at m/z 178.0258 by demethylation, which further lost one CO moiety, and the fragment ions of [M + H − CH3 − CO]+ at m/z 150.0309 were generated. Scopoletin also directly lost one CH4 and CO2 neutral moiety to generate a product ion [M + H −CH4 − CO2]+ at m/z 133.0283, indicating the C-6 methoxy substituents and lactone structures. The possible fragmentation pathway for scopoletin is proposed in Figure 4A. The esculin (24) and scopolin (28) were identified according to related literature studies [20,23]. Compounds 31, 37, 44, and 54 were confirmed by comparison with the available reference standards.
Lignans are a class of natural compounds synthesized by polymerizing two phenylpropanoid derivatives (C6-C3 monomers). Compounds 38, 49, 52, and 57 were furofuran-type lignans. Taking Compound 38 as an example, the deprotonated molecular ion m/z 535.1772 was detected in the spectrum. Its MS/MS fragment ions at m/z 373.1284 [M − H − C6H10O5], m/z 355.1182 [M − H − C6H10O5 − H2O], and m/z 343.1182 [M − H − C6H10O5 − H2CO] were observed in negative ionization mode. Compound 38 was identified as 1-hydroxylpinoresinol 4′-O-glucopyranoside according to the literature [19]. The possible fragmentation mechanism of compound 38 is depicted in Figure 4B. Compounds 49, 52, and 57 were tentatively identified as matairesinoside, 1-hydroxypinoresinol, and matairesinol according to their MS/MS fragments and related literature studies [19].

2.2.4. Nitrogenous Compounds

Amino acids, nucleobases, and other nitrogenous compounds respond strongly in positive ionization mode, and most of the second mass spectra are broken in the center of N+. In the present study, 3 nucleobases, 7 amino acids, and other nitrogenous compounds in RS were characterized. The usual fragmentation pathways, including the losses of NH3, H2O, and HCOOH, were observed in these compounds. Compound 6 was identified as proline based on short retention time and specific fragments. It has a pseudomolecular ion of m/z 116.0704, indicative of the molecular formula C5H9NO2. The fragment ion at m/z 70.0656 for [M + H − HCOOH]+ agrees with the literature studies due to losing one carboxyl group [11]. The chemical structures and fragmentation pathways are shown in Figure 5. In this way, other amino acids can be successfully characterized according to related literature studies, including asparagine (1), glutamic acid (2), threonine (3), pyroglutamic acid (4), phenylalanine (17), and tryptophan (23). In addition, nucleobases (compounds 7, 9, and 15) and other nitrogenous compounds (compounds 10, 11, 34, and 67) were also detected and tentatively identified based on databases and the literature.

2.2.5. Anthraquinonoids

By comparing the retention time and MS spectrum with authentic standards, compound 68 was identified as emodin. The protonated molecular ion of emodin was m/z 269.0455 [M − H] in negative ionization modes. Its MS/MS fragment ions were at m/z 241.0504 [M − H − CO] and m/z 225.0552 [M − H − CO2]. The chemical structures and fragmentation pathways are shown in Figure 6. In addition, compounds 60, 63, and 66 were tentatively identified as 7-hydroxy-emodin, 1-O-methyl-emodin, and physcion according to their MS/MS fragments and related literature studies [34].

3. Materials and Methods

3.1. Chemicals and Reagents

High-performance liquid chromatography (HPLC)-grade acetonitrile and formic acid were purchased from Sigma (Sigma Aldrich, St. Louis, MO, USA). Analytical-grade methanol was purchased from Chinasun Specialty Products Co., Ltd. (Jiangsu, China). The aesculetin (Batch No. 5483, 99%), isoscopoletin (Batch No. 3620, 98%), scopoletin (Batch No. 5257, 98%), quercetin (Batch No. 1115, 98%), scoparone (Batch No. 1902, 98%), protocatechuic acid (Batch No. 5809, 99%), salicylic acid (Batch No. 5328, 99%), caffeic acid (Batch No. 2681, 98%), ferulic acid (Batch No. 8042, 99%), and emodin (Batch No. 8171, 98%) were purchased from Shanghai Standard Technology Co., Ltd. (Shanghai, China). Standards of luteolin (Batch No. C11352540, 98%) and oleanic acid (Batch No. M180850633, 98%) were obtained from Shanghai Macklin Biochemical Co., Ltd. (Shanghai, China). Ultrapure water (18.2 MΩ·cm−1) was purified by a Millipore system (Millipore Corp., Burlington, MA, USA).
The batch of Ranunculus sceleratus L. was collected in the Xinhe community of Feidong County (Hefei, China) and authenticated by Professor Huasheng Peng (School of Pharmacy, Anhui University of Chinese Medicine). Voucher specimens (Batch No. 20210401) were deposited in the herbarium of the School of Pharmacy, Anhui Medical University (Hefei, China).

3.2. Preparation of Sample and Standard Solutions

An aliquot of 0.1 g fine powder (<65 mesh) of RS samples was accurately weighed and placed in a 50 mL of a conical flask, and then 10 mL of 80% methanol−water (v/v) was added into the conical flask. After sonication for 30 min, the sample solution was cooled to room temperature. All the standards of aesculetin, isoscopoletin, scopoletin, quercetin, scoparone, protocatechuic acid, salicylic acid, ferulic acid, luteolin, caffeic acid, emodin and oleanic acid were dissolved in 80% methanol−water (v/v) at a concentration of 10 µg·mL−1 to prepare standard solutions. All the solutions were filtered through a 0.22 μm filter membrane (Bandao Corp., Shanghai, China) before analysis.

3.3. UHPLC-Q-Orbitrap HRMS System and Conditions

A UHPLC Dionex Ultimate 3000 (Thermo Scientific, San Jose, CA, USA) equipped with a cooling autosampler and column oven was utilized. The separation was performed on a Shim-pack GISS UHPLC C18 column (100 mm × 2.1 mm, 1.9 μm) (Shimadzu, Japan) with a column temperature maintained at 30 °C at a flow rate of 0.2 mL·min−1. Binary mobile solvents consisted of acetonitrile (A) and water containing 0.1% formic acid (B), and the following gradient elution program was used: 0−3 min, 2% A; 3–5 min, 2–30% A; 5–12 min, 30–70% A; 12–14 min, 70–95% A; 14–18 min, 95% A; 18–20 min, 95–2% A. The injection volume was set at 2 µL.
A Q-Exactive plus hybrid quadrupole-orbitrap mass spectrometer (Thermo Fisher Scientific, San Jose, CA, USA) with heat electrospray ionization (HESI) was employed. The mass conditions were set as follows: capillary temperature, 320 °C; auxiliary gas heater temperature, 200 °C; spray voltage, 4 kV/3.5 kV (positive/negative); Apex trigger, 2–6 s; Loop count, 5; S-lens RF level, 50 V. Full MS/dd-MS2 scan mode conditions were set as follows: Scan range, 75–1125 m/z; Full MS resolution, 70,000; dd-MS2 resolution, 17,500; Maximum injection time (IT), 50 ms; Isolation window, 1.0 m/z; Normalized collision energy (NCE), 20/40/60 eV; Automatic gain control (AGC) target, 1.0 × 105; Dynamic exclusion, 10 s. Nitrogen was used for spray stabilization, for collision-induced dissociation experiments in the HCD cell, and as the damping gas in the C-trap.

3.4. Data Processing and Analysis

Tune 2.9 (Thermo Fisher Scientific, San Jose, CA, USA) was used to control the mass spectrometer, and Xcalibur 4.1 software (Thermo Fisher Scientific, San Jose, CA, USA) was used to control the instrument for data acquisition and analysis. The mass tolerance of MS and MS2 was within 5 ppm. The chemical formulas for all parent and fragment ions were calculated according to the exact mass, and the parameters are set as follows: C (0–60), H (0–120), O (0-60), and N (0–10).

4. Conclusions

The inherent variety of natural products in TCM has presented a big challenge in separation and detection techniques for the rapid characterization of its chemical profiling. In the present study, the chemical constituents of RS extract were determined by UHPLC-Q-Orbitrap HRMS. A total of 69 compounds, including 19 flavonoids, 22 organic acids, 6 coumarins, 4 lignans, 14 nitrogenous compounds, and 4 anthraquinones, were identified based on the comparison of their accurate masses, fragment ions, literature studies, and standard samples. Isoscopoletin, scopoletin, scoparone, aesculetin, quercetin, protocatechuic, salicylic acid, ferulic acid, luteolin, caffeic acid, emodin and oleanic acid were identified by standard samples. It is worth noting that flavonoids and lignans were identified for the first time in RS. This work can provide an essential chemical basis for quality control and further studies on the pharmacological and clinical application of RS.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/molecules27103299/s1. Figure S1: Product ion spectra of standard samples of isoscopoletin (A), scopoletin (B), and scoparone (C) in positive ionization mode. Figure S2: Product ion spectra of standard samples of protocatechuic acid (A), aesculetin (B), caffeic acid (C), ferulic acid (D), salicylic acid (E), luteolin (F), quercetin (G), emodin (H) and oleanic acid (I) in negative ionization mode. Figure S3: Chemical structures of 69 compounds identified in the 80% methanol extract of RS.

Author Contributions

Writing—original draft and formal analysis, S.C.; data curation, M.H.; formal analysis, L.Y. and F.C.; resources, M.L.; methodology, Z.S.; writing—review & editing, W.C.; supervision, Y.Z.; software, S.W.; project administration and writing—review & editing, Q.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors would like to thank the Center for Scientific Research of Anhui Medical University for valuable help in our experiments.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Zhang, Z.Y.; Miao, Y.R.; Xu, M.; Cheng, W.M.; Yang, C.Y.; She, X.J.; Geng, Q.Q.; Zhang, Q.L. Tianjiu therapy for alpha-naphthyl isothiocyanate-induced intrahepatic cholestasis in rats treated with fresh Ranunculus sceleratus L. J. Ethnopharmacol. 2020, 248, 112310. [Google Scholar] [CrossRef] [PubMed]
  2. Prieto, J.M.; Recio, M.C.; Giner, R.M.; Rios, J.L. Pharmacological approach to the pro- and anti-inflammatory effects of Ranunculus sceleratus L. J. Ethnopharmacol. 2003, 89, 131–137. [Google Scholar] [CrossRef]
  3. Zhou, C.X.; Zhang, S.L.; Yao, W.; Zhao, Y. Studies on the chemical constituents in herb of Ranunculus sceleratus L. China J. Chin. Mater. Med. 2005, 2, 45–47. [Google Scholar]
  4. Peng, T.; Xing, Y.J.; Zhang, Q.J.; Kang, W.Y. Chemical constituents in Ranunculus sceleratus L. Chin. J. Exp. Tradit. Med. Formulae 2011, 17, 66–67. [Google Scholar]
  5. Zuniga-Lopez, M.C.; Maturana, G.; Campmajo, G.; Saurina, J.; Nunez, O. Determination of bioactive compounds in sequential extracts of chia leaf (Salvia hispanica L.) using UHPLC-HRMS (Q-Orbitrap) and a global evaluation of antioxidant in vitro capacity. Antioxidants 2021, 10, 1151. [Google Scholar] [CrossRef]
  6. Ma, Q.; Cai, S.B.; Liu, X.J.; Shi, J.Y.; Yi, J.J. Characterization of phytochemical components and identification of main antioxidants in Crateva unilocalaris Buch. shoots by UHPLC-Q-Orbitrap-MS2 analysis. Food Res. Int. 2021, 143, 110264. [Google Scholar] [CrossRef]
  7. Chen, M.; Wang, P.L.; Li, T.; Li, L.S.; Li, J.F.; Bai, H.; Lei, H.M.; Ma, Q. Comprehensive analysis of Huanglian Jiedu decoction: Revealing the presence of a self-assembled phytochemical complex in its naturally-occurring precipitate. J. Pharm. Biomed. Anal. 2021, 195, 113820. [Google Scholar] [CrossRef]
  8. Lin, R.M.; Yin, J.X.; Wu, M.F.; Ding, H.; Han, L.F.; Yang, W.Z.; Song, X.B.; Bei, S.T.; Wang, C.H.; He, Y.Z.; et al. Global identification and determination of the major constituents in Kai-Xin-San by ultra-performance liquid chromatography-quadrupole-orbitrap mass spectrometry and gas chromatography-mass spectrometry. J. Pharm. Biomed. Anal. 2021, 206, 114385. [Google Scholar] [CrossRef]
  9. Zhang, G.; Chen, S.S.; Zhou, W.; Meng, J.; Deng, K.; Zhou, H.N.; Hu, N.; Sou, Y.R. Rapid qualitative and quantitative analyses of eighteen phenolic compounds from Lycium ruthenicum Murray by UPLC-Q-Orbitrap MS and their antioxidant activity. Food Chem. 2018, 269, 150–156. [Google Scholar] [CrossRef]
  10. Jeong, H.C.; Kim, T.; Yang, D.H.; Shin, K.H. Development of a UPLC-MS/MS method for the therapeutic monitoring of L-asparaginase. Transl. Clin. Pharmacol. 2018, 26, 134–140. [Google Scholar] [CrossRef]
  11. Kang, H.J.; Yang, H.J.; Kim, M.J.; Han, E.S.; Kim, H.J.; Kwon, D.Y. Metabolomic analysis of meju during fermentation by ultraperformance liquid chromatography-quadrupole-time of flight mass spectrometry (UPLC-Q-TOF MS). Food Chem. 2011, 127, 1056–1064. [Google Scholar] [CrossRef] [PubMed]
  12. Quirantes-Pine, R.; Lozano-Sanchez, J.; Herrero, M.; Ibanez, E.; Segura-Carretero, A.; Fernandez-Gutierrez, A. HPLC-ESI-QTOF-MS as a powerful analytical tool for characterising phenolic compounds in olive-leaf extracts. Phytochem. Anal. 2013, 24, 213–223. [Google Scholar] [CrossRef] [PubMed]
  13. Ruf, A.; Lange, J.; Eddhif, B.; Geffroy, C.; d’Hendecourt, L.L.; Poinot, P.; Danger, G. The challenging detection of nucleobases from pre-accretional astrophysical ice analogs. Astrophys. J. Lett. 2019, 887, L31. [Google Scholar] [CrossRef]
  14. Smith, K.M.; Wilson, I.D.; Rainville, P.D. Sensitive and reproducible mass spectrometry-compatible RP-UHPLC analysis of tricarboxylic acid cycle and related metabolites in biological fluids: Application to human urine. Anal. Chem. 2021, 93, 1009–1015. [Google Scholar] [CrossRef]
  15. Kong, X.B.; Liu, C.X.; Liu, P.; Guo, Y.Z.; Zhao, C.C.; Yang, Y.Y.; Bo, Z.C.; Wang, F.Y.; Peng, Y.Y.; Meng, J.Y. Combination of UPLC-Q-TOF/MS and network pharmacology to reveal the mechanism of qizhen decoction in the treatment of colon cancer. ACS Omega 2021, 6, 14341–14360. [Google Scholar] [CrossRef]
  16. Carpi, F.M.; Cortese, M.; Orsomando, G.; Polzonetti, V.; Vincenzetti, S.; Moreschini, B.; Coleman, M.; Magni, G.; Pucciarelli, S. Simultaneous quantification of nicotinamide mononucleotide and related pyridine compounds in mouse tissues by UHPLC–MS/MS. Sep. Sci. Plus 2018, 1, 22–30. [Google Scholar] [CrossRef]
  17. Wang, Y.F.; Li, H.Y.; Zhen, Y.; Zhang, W.J.; Hua, J. Simultaneous determination of furfural and its degradation products, furoic acid and maleic acid, in transformer oil by the reversed-phase vortex-assisted liquid-liquid microextraction followed by high-performance liquid chromatography. J. Sep. Sci. 2017, 40, 4805–4812. [Google Scholar] [CrossRef]
  18. Chen, L.; Yao, C.L.; Li, J.Y.; Wang, J.; Yao, S.; Shen, S.J.; Yang, L.; Zhang, J.; Wei, W.L.; Bi, Q.R.; et al. Systematic characterization of chemical constituents in Mahuang decoction by UHPLC tandem linear ion trap-Orbitrap mass spectrometry coupled with feature-based molecular networking. J. Sep. Sci. 2021, 44, 2717–2727. [Google Scholar] [CrossRef]
  19. Sun, H.Y.; Liu, M.X.; Lin, Z.T.; Jiang, H.; Niu, Y.Y.; Wang, H.; Chen, S.Z. Comprehensive identification of 125 multifarious constituents in Shuang-huang-lian powder injection by HPLC-DAD-ESI-IT-TOF-MS. J. Pharm. Biomed. Anal. 2015, 115, 86–106. [Google Scholar] [CrossRef]
  20. Chen, Y.Y.; Cai, X.J.; Li, G.W.; He, X.Y.; Yu, X.X.; Yu, X.W.; Xiao, Q.M.; Xiang, Z.; Wang, C.J. Chemical constituents of radix Actinidia chinensis planch by UPLC-QTOF-MS. Biomed. Chromatogr. 2021, 35, e5103. [Google Scholar] [CrossRef]
  21. Nie, R.; Zhang, Y.R.; Jin, Q.Z.; Zhang, S.; Wu, G.C.; Chen, L.; Zhang, H.; Wang, X.G. Identification and characterisation of bioactive compounds from the seed kernels and hulls of Paeonia lactiflora Pall by UPLC-QTOF-MS. Food Res. Int. 2021, 139, 109916. [Google Scholar] [CrossRef] [PubMed]
  22. Liu, R.X.; Zhao, Z.Y.; Dai, S.J.; Che, X.; Liu, W.H. Identification and quantification of bioactive compounds in Diaphragma juglandis fructus by UHPLC-Q-Orbitrap HRMS and UHPLC-MS/MS. J. Agric. Food Chem. 2019, 67, 3811–3825. [Google Scholar] [CrossRef] [PubMed]
  23. Yao, T.; Zhu, L.; Tang, Y.N.; Zhang, J.Y.; Liang, Z.T.; Xu, J.; Zhao, Z.Z.; Yu, Z.L.; Bian, Z.X.; Yang, Z.J.; et al. An integrated strategy based on UPLC-DAD-QTOF-MS for metabolism and pharmacokinetic studies of herbal medicines: Tibetan “Snow Lotus” herb (Saussurea laniceps), a case study. J. Ethnopharmacol. 2014, 153, 701–713. [Google Scholar]
  24. Hegazi, M.N.; Khattab, A.R.; Frolov, A.; Wessjohann, L.A.; Farag, M.A. Authentication of saffron spice accessions from its common substitutes via a multiplex approach of UV/VIS fingerprints and UPLC/MS using molecular networking and chemometrics. Food Chem. 2022, 367, 130739. [Google Scholar] [CrossRef]
  25. Zengin, G.; Mahomoodally, M.F.; Paksoy, M.Y.; Picot-Allain, C.; Glamocilja, J.; Sokovic, M.; Diuzheva, A.; Jeko, J.; Cziaky, Z.; Rodrigues, M.J.; et al. Phytochemical characterization and bioactivities of five apiaceae species: Natural sources for novel ingredients. Ind. Crops Prod. 2019, 135, 107–121. [Google Scholar] [CrossRef]
  26. El-Newary, S.A.; Afifi, S.M.; Aly, M.S.; Ahmed, R.F.; El Gendy, A.E.-N.G.; Abd-ElGawad, A.M.; Farag, M.A.; Elgamal, A.M.; Elshamy, A.I. Chemical profile of Launaea nudicaulis ethanolic extract and its antidiabetic effect in streptozotocin-induced rats. Molecules 2021, 26, 1000. [Google Scholar] [CrossRef]
  27. Mathias, M.D.; de Oliveira, R.R. Differentiation of the phenolic chemical profiles of Cecropia pachystachya and Cecropia hololeuca. Phytochem. Anal. 2019, 30, 73–82. [Google Scholar] [CrossRef] [Green Version]
  28. Uysal, A.; Zengin, G.; Mahomoodally, M.F.; Picot-Allain, C.; Jeko, J.; Cziaky, Z.; Rodrigues, M.J.; Ak, G.; Polat, R.; Urusan, Z.; et al. A comparative study on biological properties and chemical profiles of different solvent extracts from Centaurea bingoelensis, an endemic plant of Turkey. Process. Biochem. 2021, 102, 315–324. [Google Scholar] [CrossRef]
  29. Li, W.J.; Zhang, Y.; Shi, S.J.; Yang, G.; Liu, Z.H.; Wang, J.M.; Kang, W.Y. Spectrum-effect relationship of antioxidant and tyrosinase activity with Malus pumila flowers by UPLC-MS/MS and component knock-out method. Food Chem. Toxicol. 2019, 133, 110754. [Google Scholar] [CrossRef]
  30. Wang, X.; Zhong, X.J.; Zhou, N.; Cai, N.; Xu, J.H.; Wang, Q.B.; Li, J.J.; Liu, Q.; Lin, P.C.; Shang, X.Y. Rapid characterizaiton of chemical constituents of the tubers of Gymnadenia conopsea by UPLC-Orbitrap-MS/MS analysis. Molecules 2020, 25, 898. [Google Scholar] [CrossRef] [Green Version]
  31. Tian, D.; Yang, Y.; Yu, M.; Han, Z.Z.; Wei, M.; Zhang, H.W.; Jia, H.M.; Zou, Z.M. Anti-inflammatory chemical constituents of Flos chrysanthemi indici determined by UPLC-MS/MS integrated with network pharmacology. Food Funct. 2020, 11, 6340–6351. [Google Scholar] [CrossRef] [PubMed]
  32. Fu, S.; Cheng, R.R.; Deng, Z.X.; Liu, T.G. Qualitative analysis of chemical components in Lianhua Qingwen capsule by HPLC-Q Exactive-Orbitrap-MS coupled with GC-MS. J. Pharm. Anal. 2021, 11, 709–716. [Google Scholar] [CrossRef] [PubMed]
  33. Yuan, J.Q.; Wang, Y.T.; Mi, S.Q.; Zhang, J.Y.; Sun, Y.X. Rapid screening and characterization of caffeic acid metabolites in rats by UHPLC-Q-TOF mass spectrometry. Trop. J. Pharm. Res. 2022, 20, 389–401. [Google Scholar] [CrossRef]
  34. Huang, Z.H.; Xu, Y.; Wang, Q.; Gao, X.Y. Metabolism and mutual biotransformations of anthraquinones and anthrones in rhubarb by human intestinal flora using UPLC-Q-TOF/MS. J. Chromatogr. B 2019, 1104, 59–66. [Google Scholar] [CrossRef] [PubMed]
  35. Areche, C.; Hernandez, M.; Cano, T.; Ticona, J.; Cortes, C.; Simirgiotis, M.; Caceres, F.; Borquez, J.; Echeverria, J.; Sepulveda, B. Corryocactus brevistylus (K. Schum. ex Vaupel) Britton & Rose (Cactaceae): Antioxidant, gastroprotective effects, and metabolomic profiling by ultrahigh-pressure liquid chromatography and electrospray high resolution orbitrap tandem mass spectrometry. Front. Pharmacol. 2020, 11, 417. [Google Scholar] [PubMed]
  36. Fu, X.Z.; Belwal, T.; He, Y.H.; Xu, Y.Q.; Li, L.; Lou, Z.S. UPLC-Triple-TOF/MS characterization of phenolic constituents and the influence of natural deep eutectic solvents on extraction of Carya cathayensis Sarg. peels: Composition, extraction mechanism and in vitro biological activities. Food Chem. 2022, 370, 131042. [Google Scholar] [CrossRef]
  37. Mi, N.; Cheng, T.F.; Li, H.L.; Yang, P.M.; Mu, X.M.; Wang, X.Y.; Zu, X.P.; Qi, X.P.; Guo, X.; Ye, J.; et al. Metabolite profiling of traditional Chinese medicine formula DanZhi Tablet: An integrated strategy based on UPLC-QTOF/MS combined with multivariate statistical analysis. J. Pharm. Biomed. Anal. 2019, 164, 70–85. [Google Scholar] [CrossRef]
  38. Qin, Y.; Zhou, R.R.; Jin, J.; Xie, J.; Liu, H.; Cai, P.; Shu, J.; Zhao, Y.H.; Huang, L.Q.; Zhang, S.H. UPLC-ESI-Q-TOF-MS/MS analysis of anticancer fractions from Ophiocordyceps xuefengensis and Ophiocordyceps sinensis. Biomed. Chromatogr. 2020, 34, e4841. [Google Scholar] [CrossRef]
  39. Sanchez-Salgado, J.C.; Estrada-Soto, S.; Garcia-Jimenez, S.; Montes, S.; Gomez-Zamudio, J.; Villalobos-Molina, R. Analysis of flavonoids bioactivity for cholestatic liver disease: Systematic literature search and experimental approaches. Biomolecules 2019, 9, 102. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Base peak chromatograms of RS in positive ionization mode (A) and negative ionization mode (B).
Figure 1. Base peak chromatograms of RS in positive ionization mode (A) and negative ionization mode (B).
Molecules 27 03299 g001
Figure 2. Characteristic of MS/MS spectra and possible fragmentation pathways of hyperoside (A) and apigenin-7-O-glucuronide (B).
Figure 2. Characteristic of MS/MS spectra and possible fragmentation pathways of hyperoside (A) and apigenin-7-O-glucuronide (B).
Molecules 27 03299 g002
Figure 3. Characteristic of MS/MS spectra and possible fragmentation pathways of protocatechuic acid (A) and salicylic acid-O-glucopyranoside (B).
Figure 3. Characteristic of MS/MS spectra and possible fragmentation pathways of protocatechuic acid (A) and salicylic acid-O-glucopyranoside (B).
Molecules 27 03299 g003
Figure 4. Characteristic of MS/MS spectra and possible fragmentation pathways of scopoletin (A) and hydroxylpinoresinol 4′-O-glucopyranoside (B).
Figure 4. Characteristic of MS/MS spectra and possible fragmentation pathways of scopoletin (A) and hydroxylpinoresinol 4′-O-glucopyranoside (B).
Molecules 27 03299 g004
Figure 5. Characteristic of MS/MS spectra and possible fragmentation pathways of proline.
Figure 5. Characteristic of MS/MS spectra and possible fragmentation pathways of proline.
Molecules 27 03299 g005
Figure 6. Characteristic of MS/MS spectra and possible fragmentation pathways of emodin.
Figure 6. Characteristic of MS/MS spectra and possible fragmentation pathways of emodin.
Molecules 27 03299 g006
Table 1. 69 compounds identified in the 80% methanol extract of RS and their UHPLC-Q-Orbitrap HRMS data.
Table 1. 69 compounds identified in the 80% methanol extract of RS and their UHPLC-Q-Orbitrap HRMS data.
NO.Retention Time (min)Ionization ModeMolecular
Formula
Theoretical (m/z)Measured (m/z)Error (ppm)Identified CompoundsMS/MS Fragments (m/z)Ref.
11.37[M + H]+C4H8N2O3133.0607133.0604−2.25Asparagine133.0604, 115.0391, 97.0286, 87.0558[10]
21.40[M + H]+C5H9NO4148.0604148.0598−4.05Glutamic acid148.0598, 102.0550, 84.0447[11]
31.40[M + H]+C4H9NO3120.0655120.06550.00Threonine120.0655, 102.0553, 74.0607, 56.0503[11]
41.40[M + H]+C5H7NO3130.0498130.04990.77Pyroglutamic acid130.0499, 102.0551, 84.0449[11]
1.40[M − H]C5H7NO3128.0342128.0340−1.56Pyroglutamic acid128.0339, 85.0279, 68.1163
51.42[M − H]C6H12O7195.0499195.05011.03Gluconic acid195.0501, 129.018, 75.0072[12]
61.47[M + H]+C5H9NO2116.0706116.0704−1.72Proline116.0704, 70.0656[11]
71.51[M + H]+C4H5N3O112.0505112.05071.78Cytosine112.0507, 95.0244, 69.0455[13]
81.57[M − H]C4H6O5133.0131133.0129−1.50Malic acid133.0129, 115.0023, 71.0124[14]
91.91[M + H]+C5H5N5O152.0567152.0565−1.32Guanine152.0565, 135.0300, 110.0351[13]
101.91[M + H]+C6H5NO2124.0393124.03940.81Nicotinic acid124.0394, 106.0291, 96.0448, 80.0501[15]
111.99[M + H]+C6H6N2O123.0553123.05540.81Nicotinamide123.0554, 80.0501[16]
122.13[M − H]C6H6O6173.0080173.00821.16Aconitic acid173.0082, 129.0179, 111.0072, 85.0279[14]
132.17[M − H]C5H4O3111.0076111.0073−2.70Furoic acid111.0073, 83.0122, 67.0174[17]
142.24[M − H]C6H8O7191.0186191.01870.52Citric acid191.0187, 111.0073, 87.0072[14]
152.62[M + H]+C4H4N2O2113.0345113.03471.77Uracil113.0347, 96.0084, 70.0294[11]
162.64[M − H]C4H6O4117.0182117.0179−2.56Succinic acid117.0179, 99.0073, 73.0280[15]
175.05[M + H]+C9H11NO2166.0862166.0861−0.60Phenylalanine166.0861, 120.0809, 103.0545[11]
187.09[M − H]C13H16O9315.0711315.07223.49Protocatechuic acid-O-glucose315.0722, 153.0545, 109.0280[18]
197.28[M − H]C14H18O9329.0867329.0858−2.73Vanillic acid-O-glucopyranoside329.0858, 167.033, 123.0437[19]
207.28[M − H]C14H20O8315.1074315.10925.71Hydroxytyrosol-1-glucopyranoside315.1092, 153.0545[19]
217.37[M−H]C7H6O4153.0182153.0181−0.65Protocatechuic acid153.0181, 123.0437, 109.0280, 91.0176Standard
227.61[M−H]C15H18O9341.0867341.08731.76Caffeic acid-O-glucopyranoside341.0873, 179.0339, 135.0439[19]
237.61[M + H]+C11H12N2O2205.0972205.0970−0.98Tryptophan205.0970, 188.0703, 159.0914, 146.0598[11]
7.61[M − H]C11H12N2O2203.0815203.08181.48Tryptophan203.0818, 186.0551, 159.091, 142.0649
247.65[M − H]C15H16O9339.0711339.07182.06Esculin339.0718,177.0183[20]
257.67[M − H]C13H16O8299.0761299.07775.35Salicylic acid-O-glucopyranoside299.0777, 137.0231, 93.0331[19]
267.78[M − H]C27H30O16609.1450609.14642.30Kaempferol-3,7-di-O-glucoside609.1464, 447.0930, 285.0402, 255.0295, 151.0023[21]
277.82[M − H]C7H6O3137.0233137.0231−1.46Protocatechualdehyde137.0231, 109.0279, 108.0201, 93.0330[22]
287.89[M − H]C16H18O9353.0867353.0865−0.57Scopolin353.0865, 191.0553, 179.0341, 173.0450, 135.0439[23]
297.91[M − H]C15H18O8325.0918325.09283.08p-Coumaric acid-
O-glucopyranoside
325.0928, 163.0388, 119.0487,[19]
308.12[M − H]C27H30O16609.1450609.14530.49Kaempferol-O-sophoroside609.1453, 429.0818, 284.0323, 255.0295, 227.0343[24]
318.16[M − H]C9H6O4177.0182177.01820.00Aesculetin177.0182, 149.0231, 133.0280, 121.0280Standard
328.21[M − H]C9H8O4179.0339179.03421.68Caffeic acid179.0342, 135.0437, 90.9966Standard
338.25[M − H]C26H28O16595.1294595.13011.18Quercetin-O-(pentosyl)hexoside isomer595.1301, 300.0274, 271.0248, 255.0294, 151.0022, 135.0433[25]
348.51[M − H]C11H13NO3206.0811206.08130.97n-Acetyl-L-phenylalanine206.0813, 164.0705, 147.0438, 118.9914, 91.0536[26]
358.53[M − H]C21H20O11447.0922447.09291.57Isoorientin447.0929, 429.0821, 357.0613, 327.0508, 285.0420, 133.0281[27]
368.55[M − H]C8H14O4173.0808173.08080.00Suberic acid173.0808, 129.0906, 111.0800[28]
378.59[M + H]+C10H8O4193.0495193.0490−2.95Isoscopoletin193.0490, 178.0255, 133.0280Standard
388.60[M − H]C26H32O12535.1810535.18181.491-Hydroxylpinoresinol 4’-O-glucopyranoside535.1818, 373.1284, 343.1182[19]
398.62[M + H]+C21H20O12465.1027465.1024−0.65Hyperoside465.1024, 303.0494, 257.0439, 229.0493, 153.0181[29]
8.62[M − H]C21H20O12463.0871463.08822.38Hyperoside463.0882, 301.0350, 271.0244, 178.9974, 151.0023[29]
408.77[M − H]C9H8O3163.0389163.03900.61Coumaric acid163.0390, 119.0488[19]
418.88[M + H]+C16H12O7317.0656317.0649−2.21Isorhamnetin317.0649, 302.0416, 285.0388, 274.0467, 153.0180[30]
428.91[M + H]+C15H10O6287.0550287.0545−1.74Kaempferol287.0545, 269.0437, 231.0651, 213.0541, 153.0180, 121.0285[29]
438.92[M − H]C22H22O12477.1028477.10341.26Isorhamnetin-3-O-glucoside isomer477.1034, 314.0430, 285.0402, 271.0247, 243.0295[22]
448.94[M + H]+C10H8O4193.0495193.0493−1.04Scopoletin193.0493, 178.0258, 175.1479, 150.0309, 133.0283, 109.0857Standard
8.94[M − H]C10H8O4191.0339191.03400.52Scopoletin191.0340, 176.0105, 146.973,111.0074, 102.9474
458.96[M − H]C21H20O11447.0933447.0930−0.67Luteoloside447.0930, 285.0420, 255.0296, 241.0500, 217.05.01, 227.0343, 199.0395, 151.0023[19]
468.98[M − H]C10H10O4193.0495193.04950.00Ferulic acid193.0495, 178.0261, 149.0595,134.0360, 121.0282Standard
479.03[M + H]+C21H18O11447.0922447.09240.45Apigenin-7-O-glucuronide447.0924, 271.0595, 231.1145, 199.2475, 153.0180, 119.0490[28]
9.03[M − H]C21H18O11445.0765445.0752−2.92Apigenin-7-O-glucuronide445.0752, 269.0452, 175.0233, 113.0229
489.06[M − H]C21H18O12461.0714461.07272.82Luteolin-7-O-glucuronide461.0727, 285.0402, 151.0022, 133.0279[31]
499.10[M − H]C26H32O11519.1861519.18732.31Matairesinoside519.1972, 357.1340, 342.1098, 313.1464, 221.0804, 161.0595[19]
509.22[M − H]C9H16O4187.0964187.09661.07Azelaic acid187.0966, 125.0958, 97.0644[32]
519.31[M − H]C9H10O3165.0546165.0545−0.61p-Hydroxybenzene
propanoic acid
165.0545, 147.0439, 136.9310, 119.0488, 72.9916[33]
529.33[M − H]C20H22O7373.1282373.12891.881-Hydroxylpinoresinol373.1289, 343.1176, 313.1081, 298.0844, 147.0439, 123.0074, 109.0277[19]
539.68[M − H]C7H6O3137.0233137.0232−0.73Salicylic acid137.0232, 93.0331Standard
549.82[M + H]+C11H10O4207.0652207.0649−1.45Scoparone207.0649, 191.0337, 163.0388, 151.0752, 121.0648, 105.0703Standard
5510.28[M + H]+C15H10O6287.0550287.0545−1.74Luteolin287.0545, 153.0180, 131.0439Standard
10.28[M − H]C15H10O6285.0393285.04033.51Luteolin285.0403, 175.0387, 133.0281,121.0279, 107.0125, 83.0124
5610.32[M − H]C15H10O7301.0343301.03553.99Quercetin301.0355, 229.0504, 201.0565, 178.9975, 151.0024, 121.0281Standard
5710.83[M − H]C20H22O6357.1332357.13412.52Matairesinol357.1341, 342.1097, 313.0370, 283.0078,[19]
5811.06[M + H]+C15H10O5271.0601271.0595−2.21Apigenin271.0595, 153.0181, 119.0492[26]
11.06[M − H]C15H10O5269.0444269.04533.35Apigenin269.0453, 251.0590, 227.0341, 181.0644, 151.0025, 117.0332
5911.16[M − H]C16H12O6299.0550299.05603.34Chrysoeriol299.0560, 284.0326, 256.0372, 255.0293,
227.0345
[31]
6011.19[M − H]C15H10O6285.0394285.04033.167-Hydroxy-emodin285.0403, 257.0449, 211.0380[34]
6111.41[M − H]C16H12O6299.0550299.05613.68Diosmetin299.0561, 284.0325, 256.0378, 255.0296, 227.0344[31]
6211.51[M − H]C16H12O7315.0499315.05082.86Rhamnetin315.0508, 300.0271, 151.0024,107.0123[35]
6311.67[M − H]C16H12O5283.0601283.06113.531-O-Methyl-emodin283.0611, 268.0376, 239.0346, 211.0395[34]
6412.75[M − H]C15H12O4255.0651255.06593.14Pinocembrin255.0659, 213.0549, 171.0441, 151.0024[36]
6512.95[M + H]+C17H14O6315.0863315.0857−1.907,3′-Dihydroxy-8,4′-dimethoxyisoflavone/isomer315.0857, 300.0623[37]
6613.21[M − H]C16H12O5283.0601283.06103.18Physcion283.0610, 268.0377, 239.0346, 211.0393[34]
6713.77[M + H]+C18H39NO3318.3002318.2996−1.892-Amino-1,3,4-octadecanetriol318.2996, 300.2891, 282.2787, 264.2681, 60.0452[38]
6814.02[M − H]C15H10O5269.0444269.04554.09Emodin269.0455, 241.0504, 225.0552, 210.0320Standard
6917.25[M − H]C30H48O3455.3531455.3528−0.66Oleanic acid455.3528, 240.9500, 206.1664, 82.4031Standard
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cao, S.; Hu, M.; Yang, L.; Li, M.; Shi, Z.; Cheng, W.; Zhang, Y.; Chen, F.; Wang, S.; Zhang, Q. Chemical Constituent Analysis of Ranunculus sceleratus L. Using Ultra-High-Performance Liquid Chromatography Coupled with Quadrupole-Orbitrap High-Resolution Mass Spectrometry. Molecules 2022, 27, 3299. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27103299

AMA Style

Cao S, Hu M, Yang L, Li M, Shi Z, Cheng W, Zhang Y, Chen F, Wang S, Zhang Q. Chemical Constituent Analysis of Ranunculus sceleratus L. Using Ultra-High-Performance Liquid Chromatography Coupled with Quadrupole-Orbitrap High-Resolution Mass Spectrometry. Molecules. 2022; 27(10):3299. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27103299

Chicago/Turabian Style

Cao, Shanshan, Min Hu, Lingli Yang, Meiqin Li, Zhen Shi, Wenming Cheng, Yazhong Zhang, Fei Chen, Sheng Wang, and Qunlin Zhang. 2022. "Chemical Constituent Analysis of Ranunculus sceleratus L. Using Ultra-High-Performance Liquid Chromatography Coupled with Quadrupole-Orbitrap High-Resolution Mass Spectrometry" Molecules 27, no. 10: 3299. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27103299

Article Metrics

Back to TopTop