Metabolite Profile of Salidroside in Rats by ... - ACS Publications

Oct 2, 2015 - We are grateful to Ting Liu and Zhimin Long from SCIEX Shanghai Application Support Center for performing the Q-trap MS analysis...
0 downloads 0 Views 2MB Size
Article pubs.acs.org/JAFC

Metabolite Profile of Salidroside in Rats by Ultraperformance Liquid Chromatography Coupled with Quadrupole Time-of-Flight Mass Spectrometry and High-Performance Liquid Chromatography Coupled with Quadrupole-Linear Ion Trap Mass Spectrometry Zhiwei Hu,†,‡ Ziming Wang,†,‡ Yong Liu,†,‡,§ Yan Wu,§ Xuejiao Han,†,‡ Jian Zheng,†,‡ Xiufeng Yan,†,‡ and Yang Wang*,†,‡ †

Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin 150040, China Key Laboratory of Saline-Alkali Vegetation Ecology Restoration in Oil Field, Ministry of Education, Harbin 150040, China § Heilongjiang Entry-Exit Inspection and Quarantine Bureau, Harbin 150001, China ‡

ABSTRACT: In the present work, the salidroside metabolite profile in rat urine was investigated, and subsequently the metabolic pathways of salidroside were proposed. After administrations of salidroside at an oral dose of 100 or 500 mg/kg, rat urine samples were collected and pretreated with methanol to precipitate the proteins. The pretreated samples were analyzed by an Acquity ultraperformance liquid chromatography (UPLC) coupled with an HSS T3 column and detected by quadrupole timeof-flight mass spectrometry (Q-TOF-MS) or high-performance liquid chromatography coupled with hybrid triple-quadrupole linear ion trap mass spectrometry (HPLC/Q-trap-MS). A total of eight metabolites were detected and identified on the basis of the characteristics of their protonated ions in the urine samples. The results elucidated that salidroside was metabolized via glucuronidation, sulfation, deglycosylation, hydroxylation, methylation, and dehydroxylation pathways in vivo. KEYWORDS: salidroside, UPLC/Q-TOF-MS, HPLC/Q-trap-MS, metabolites, metabolic pathways



INTRODUCTION Rhodiola rosea is a plant distinguished by its propensity to growing in colder climates, such as the Rocky Mountains, and certain arctic regions. R. rosea is an adaptogen; it allows the mind to relax even in the face of immense mental stress and other conditions. As a supplement, Rhodiola extracts are used worldwide.1 Due to the recorded pharmacological effects and its safety in use, commercial interest in roseroot-based products has rapidly increased. Cultivation experiments have been performed in Russia, as well as in Sweden and Poland.2,3 Salidroside (structure shown in Figure 1), p-tyrosol glycoside, is a major phenolic glycoside in Rhodiola, and its content is

of products that are valued as improving physical and mental stamina and are selling on major Web sites (amazon.com, buy.com.) and in drug stores (CVS and GNC) all around the world. Studies on pharmacokinetics of salidroside have been carried out in beagle dogs and rats.15−19 p-Tyrosol was identified as the aglycone metabolite of salidroside in rat plasma after intravenous administration.18 p-Tyrosol is the main antioxidant phenolic constituent in olive oil.20 The transformation of ptyrosol was investigated in vitro, and a glucuronide of tyrosol has been identified.21,22 However, further understanding of the metabolites and metabolic patterns of salidroside in vivo are still lacking. Time-of-flight (TOF) analysis with higher mass resolution, sensitivity, and accuracy can provide accurate masses of ions and molecular formulas compared with other MS detectors.23,24 Ion trap mass spectrometry can get more fragmental information for better identifying the exact structure of unknown metabolites.25 The combination of quadrupole time-of-flight mass spectrometry (Q-TOF-MS) and quadrupole linear ion trap mass spectrometry (Q-trap-MS) provided more possibility for the in vivo qualitative analyses of phenyl flavonoids. Urine is recognized as an ideal biological sample to investigate in vivo metabolic patterns. In this study, eight metabolites of salidroside were detected and identified in rat

Figure 1. Chemical structure of salidroside.

often used as the evaluating criterion for Rhodiola. It has been shown to possess various activities including the ability to provide protective effects on anoxia/reoxygenation,4 anticancer,5−7 anti-inflammation,8 and antioxidation9,10 in vitro as well as hepatoprotective, antiaging, antiviral, and sedative hypnotic effects in animal models; the effective doses were 20−100 mg/kg.11−14 Salidroside is also an important ingredient © 2015 American Chemical Society

Received: Revised: Accepted: Published: 8999

June 4, 2015 October 1, 2015 October 2, 2015 October 2, 2015 DOI: 10.1021/acs.jafc.5b04510 J. Agric. Food Chem. 2015, 63, 8999−9005

Article

Journal of Agricultural and Food Chemistry

(Foster City, CA, USA). The Analyst software package (version 1.6) was used for system control, data acquisition, and processing. The separation was performed on an Aglient Proshell 120 column (2.7 μm, 100 mm × 2.1 mm; Palo Alto, CA, USA), with mobile phases of water (A) and acetonitrile (B) eluting in the following program: 0− 2 min, 2% B; 2−7 min, 5% B; 7−12 min, 5−10% B; 12−13 min, 10− 80% B; 13.0−13.1 min, 80−2% B; 13.1−16 min, 2% B. The flow rate was 0.3 mL/min, and the injection volume was 2 μL. The detection was performed under negative mode with a Turbo VTM Ion Source. Polypropylene glycol dilution solvent was used to turn the ion optics. The optimized MS conditions were as follows: nitrogen as the nebulizer, heater, curtain, and collision gas with nebulizer (GS1), heater (GS2), and curtain gas flow rates of 45, 45, and 25 psi, respectively; ion-spray needle voltage, −4500 V; and heater gas temperature, 550 °C. Multiple ion monitoring (MIM) mode was utilized to analyze the biological samples. A set of MIM ion transitions of the metabolites detected in Q-TOF-MS were introduced for the detection of potential metabolites of salidroside. The parameters were used as follows: The dwell time, declustering potential, and collision energy of each MIM ion transition were fixed at 30 ms, −40 V, and −10 eV, respectively; CE and collision energy spread (CES) of EPI were fixed at −40 and ±20 eV.

urine. The chemical structures were characterized on the basis of their MS and MS2 data. The principal metabolic pathways of salidroside were proposed as well.



MATERIALS AND METHODS

Chemicals. Salidroside was purchased from the National Institutes for Food and Drug Control (Beijing, China). p-Tyrosol was purchased from Sigma (St. Louis, MO, USA). HPLC grade acetonitrile and methanol were obtained from Fisher Scientific (Pittsburgh, PA, USA). HPLC grade water was purified by a Millipore Milli-Q purification system via deionized water (Billerica, MA, USA). All other reagents were of reagent grade. Animals and Drug Administration. Male Wistar rats (200 ± 20 g of body weight) were purchased from the Laboratory Animal Center of Jilin University (Changchun, Jilin Province, China). Animals were handled according to the standard procedures approved by the institutional animal care and use committee. All rats experienced an overnight fasting with free access to water before oral administration. Rats were acclimatized for 1 week before experiments. The rats were divided into three groups: salidroside administration groups (low and high doses) and control group; each group contained three rats. Salidroside dissolved in saline was orally administered to the rats at a single dose of 100 mg/kg of body weight for the Q-TOF-MS detection or at a single dose of 500 mg/kg of body weight for the Q-trap-MS detection. The rats in the control group were administered the solvent saline orally at the same volume as the treatment group. Sample Collection. Rats were housed in metabolic cages for 3 days. Urine samples were collected at intervals of 0−12, 12−24, 24− 48, and 48−72 h after salidroside administration. All samples were stored at −40 °C until further use. Sample Preparation. Briefly, 600 μL of methanol was added to 200 μL of urine followed by vortexing for 5 min and centrifugation at 15000g for 10 min. The supernatant of the sample was transferred to a clean test tube and evaporated to dryness under nitrogen. The residue was redissolved in 200 μL of water, and the supernatant was ready for the injection after centrifugation at 15000g for 10 min. UPLC/Q-TOF-MS Analysis. The analysis was performed on a Waters ACQUITY ultraperformance liquid chromatographic system (Waters Corp., Milford, MA, USA) coupled with a Waters ACQUITY Synapt G2 quadrupole time-of-flight (Q-TOF) tandem mass spectrometer (Waters Corp., Manchester, UK). System control and data acquisition were performed using MassLynx V4.1 software. Chromatographic separation was achieved on an ACQUITY UPLC HSS T3 column (1.7 μm, 50 mm × 2.1 mm; Waters Corp., Milford, MA, USA). The mobile phase system consisted of acetonitrile (A) and water (B) at a flow rate of 0.25 mL/min. The time program of the gradient was 0−2 min, 0% A; 2−5 min, 0−5% A; 5−10 min, 5−10% A; 10−15 min, 10−80% A; 15−18 min, 80−0% A; 18−20 min, 0% A for equilibration of the column. The column and autosampler temperatures were maintained at 40 and 4 °C, respectively. Metabolites were detected in negative ionization mode. The optimized MS conditions were as follows: capillary voltage, 2.5 kV; sample cone voltage, 40 V; extraction cone voltage, 4.0 V; source temperature, 120 °C; desolvation temperature, 400 °C; desolvation gas, 800 L/h nitrogen; and cone gas, 50 L/h nitrogen. Argon was used as collision gas. For accurate mass measurement, data were collected in centroid mode, and an external reference (LockSpray) at a concentration of 200 pg/mL in 50% acetonitrile−water solution (including 0.1% formic acid) was employed to generate [M − H]− ion in negative mode at m/z 554.2615 via a lock spray interface at a flow rate of 5 μL/min to acquire accurate mass during the analysis. MSE analysis was performed in two scan functions: 5 eV for the low collision energy scan and a collision energy ramp of 10−30 eV for the high-collision energy scan. To get more clean and accurate fragments for each metabolite, MS/MS analysis was performed with a collision energy ramp of 10−25 eV. HPLC/Q-Trap-MS Analysis. The analysis was performed on a Shimadzu LC 20AD system (Shimadzu, Kyoto, Japan) coupled with an Applied Biosystem Sciex API 4000 Q-trap mass spectrometer



RESULTS AND DISCUSSION Fragmentation of Salidroside Standard. Because metabolites can retain the base substructure of the parent compound, the first important step in the analysis of metabolites of salidroside in rat urine is to obtain its chromatogram and mass spectrum characterization. All of these data were the substructural “template” for interpreting the structures of the metabolites. The chromatographic and mass spectrometric conditions were optimized using salidroside. Under negative electrospray ionization mode, the salidroside molecule (C14H20O7, MW = 300.12 Da) could easily form a molecular ion of m/z 299.1127 ([M − H]−). The product ion spectrum of the molecular ion of salidroside and its predominant fragmentation patterns are illustrated in Figure 2. Fragmentation of the molecular ion of salidroside led to four major product ions at m/z 179.0562, 137.0599, 119.0496, and 113.0230. The product ions at m/z 179.0562 (C6H11O6) and m/z 137.0599 (C8H9O2) were ions of glucose and the aglycone p-tyrosol produced by the salidroside molecular ion. The product ion at m/z 119.0496 (C8H7O) was formed by the loss of a molecule of hydrone from the ion at m/z 137.0599. The product ion at m/z 113.0230 (C5H5O3) was inferred to be produced by the loss of one molecule of formaldehyde and two molecules of hydrone from the ion at m/z 179.05629, so it can be concluded that the ions at m/z 179.0562, 137.0599, 119.0496, and 113.0230 were the characteristic product ions of salidroside. In Vivo Metabolites of Salidroside. Metabolites of salidroside in rat urine were achieved by comparing the data of blank and salidroside-dosed samples. Unique precursor ions found in salidroside-dosed samples were considered as potential metabolites for MS2 analysis. Eight metabolites were thus identified (M1−M8). The retention times, measured and calculated masses, accurate masses, mass errors, and MS/MS product ions of salidroside (M0) and its speculated metabolites (M1−M8) are summarized in Table 1. The mass errors between the measured and the calculated values were < 6 ppm, which displays a high level of confidence in the compositions of the speculated metabolites. The chemical structures were proposed using MS and MS2 data. 9000

DOI: 10.1021/acs.jafc.5b04510 J. Agric. Food Chem. 2015, 63, 8999−9005

Article

Journal of Agricultural and Food Chemistry

as glucuronic acid molecular ion, displaying a strengthened abundance. The product ion at m/z 85.0297 was the loss of 18 Da (hydrone group) from the ion at m/z 113.0231. Furthermore, another diagnostic fragment ion at m/z 137.0607 was the aglycone p-tyrosol, the same as the product ion of parent drug M0. M1 was assessed as the glucuronidated product of M0. The MS2 fragmentation (Figure 4A) of M1 showed product ions at m/z 313.3 and 299.4 by Q-trap-MS, demonstrating the glucuronidation occurred at the hydroxyl group on the phenyl ring. Metabolite M2. M2 was detected by Q-TOF-MS with an [M − H]− ion at m/z 379.0710 (2.9 ppm, C14H19O10S), which was 80 Da higher than that of M0, implying sulfation of M0. QTOF MS2 fragmentation (Figure 3B) showed fragment ions at m/z 137.0583, 119.0498, 113.0235, and 79.9576. The diagnostic ion at m/z 79.9576 was the sulfuric acid group; the other product ions were identical with the fragment ions of M0. Accordingly, M2 was structurally identified as the sulfated conjugate of M0. The product ion at m/z 217.0 (Figure 4B) found by Q-trap-MS detection indicated the sulfation at the hydroxyl group on the phenyl ring. Metabolite M3. Metabolite M3 was detected with an accurate [M − H]− ion at m/z 137.0607 (2.9 ppm, elemental composition of C8H9O2). It was similar to the product ion at m/z 137.0599 of parent drug M0. The product ion at m/z 137.0599 was assigned as the aglycone, according to the structure of salidroside. The Q-TOF MS2 fragmentations of M3, shown in Figure 3C, were observed at m/z 119.0476, 106.0411, and 93.0329. These two characteristic fragment ions at m/z 119.0476 and 93.0329 were formed by the loss of 18 Da (hydrone group) and 44 Da (vinyl alcohol group) from the [M − H]− ion at m/z 137.0599, respectively. The MS2 product ion at m/z 106.0411 was formed by the loss of 13 Da (methenyl group) from the fragment ion at m/z 119.0476. According to our previous study, we also found the product ion at m/z 106.0411 from the aglycone standard.18 Hence, M3 was identified as p-tyrosol, the aglycone of M0. Metabolite M4. The Q-TOF MS spectrum of M4 (Figure 3D) provided an accurate [M − H]− ion at m/z 313.0927 (1.3 ppm, C14H17O8), which was 176 Da (glucuronic acid group) higher than that of M3. Three major fragment ions were found at m/z 175.0241 (the [M − H]− ion of glucuronic acid), 137.0607 ([M − H]− ion of the aglycone p-tyrosol), and 113.0231 (C5H5O3). According to the Q-trap MS2 spectrum of M4 (Figure 4C), the product ion at m/z 295.1 indicated the glucuronidation was at the hydroxyl group on the phenyl ring.

Figure 2. (A) MS2 spectrum of salidroside; (B) its proposed fragmentation pathways in negative ion mode with m/z 299.11 as the precursor ion.

Parent Drug M0. M0 was identified with an accurate [M − H]− ion of m/z 299.1127 (−1.3 ppm, C14H19O7). The productions of M0 were identical to those of salidroside standard. Therefore, M0 was designated salidroside. Metabolite M1. The Q-TOF MS spectrum of M1 provided an accurate [M − H]− ion at m/z 475.1456 (0.8 ppm, C20H27O13), which was 176 Da higher than that of M0. Further Q-TOF MS2 fragmentation (Figure 3A) showed fragment ions at m/z 175.0741, 137.0607, 113.0231, and 85.0297. The diagnostic ion at m/z 175.0741 was the [M − H]− ion of glucuronic acid. The product ion at m/z 113.0231 (C5H5O3) could be fragmentized from the salidroside fragment ion as well

Table 1. Retention Times, Measured and Calculated Masses, Accurate Masses, and MS/MS Product Ions of Salidroside (M0) and Its Metabolites (M1−M8) metabolite

retention time (min)

M0

7.23

M1 M2 M3 M4 M5 M6 M7 M8

elemental composition

measured mass [M − H]− (m/z)

predicted mass [M − H]− (m/z)

error (ppm)

C14H20O7

299.1127

299.1131

−1.3

1.90 3.18

C20H28O13 C14H20O10S

475.1456 379.0710

475.1452 379.0699

+0.8 +2.9

6.87 0.97 2.17 4.37 4.21 4.54

C8H10O2 C14H18O8 C8H10O5S C8H10O6S C9H12O5S C14H1807

137.0607 313.0927 217.0161 233.0107 231.0336 297.0985

137.0603 313.0923 217.0171 233.0120 231.0327 297.0974

+2.9 +1.3 −4.6 −5.6 +3.9 +3.7

9001

fragment ions (m/z) 179.0562, 137.0599, 119.0496, 113.0230 175.0741, 137.0607, 113.0231 137.0583, 119.0498, 113.0235, 101.0231, 79.9576 119.0476, 106.0411 175.0241, 137.0607, 113.0231 137.0607, 79.9576 153.0551, 123.0433, 79.9558 151.0757, 79.9576 175.0241, 121.0642, 113.0231

metabolite description null + GlcA + SO3 − − − − − −

Glc Glc Glc Glc Glc Glc

+ + + + +

GlcA SO3 SO3 + OH SO3 + CH3 GlcA − OH

DOI: 10.1021/acs.jafc.5b04510 J. Agric. Food Chem. 2015, 63, 8999−9005

Article

Journal of Agricultural and Food Chemistry

Figure 3. MS2 spectra of metabolites M1 (A), M2 (B), M3 (C), M4 (D), M5 (E), M6 (F), M7 (G), and M8 (H) by UPLC/Q-TOF-MS. The MS2 data were obtained with [M − H]− as the precursor ion.

z 153.0551 was also 16 Da more than the product ion at m/z 137.0583, and the ion at m/z 123.0433 was a further loss of hydroxymethyl (OCH2) from the side chain of the ion at m/z 153.0551. The existence of 123.0433 proved that the hydroxylation occurred on the phenyl ring. The product ion at m/z 189.1 (Q-trap MS2 spectrum, Figure 4E) of M6 further confirmed the sulfation position, the same as M5, was at the hydroxyl group on the phenyl ring. Hence, M6 was characterized as the hydroxylation product of M5. Metabolite M7. The Q-TOF MS spectrum of M7 provided an accurate [M − H]− ion at m/z 231.0336 (3.9 ppm, C9H11O5S). MS2 fragmentation (Figure 3G) showed fragment ions at m/z 151.0757 and 79.9576. The molecular ion at m/z 231.0336 and its MS2 fragment ion at m/z 151.0757 were 14 Da more than m/z 217.0161 (M5) and its product ion at m/z 137.0583. According to product ions at m/z 187.0 and 213.2 of M7 (Q-trap MS2 spectrum Figure 4F), the position of

M4 was identified as the glucuronidated product of M3 or the deglycosylation product of M1. Metabolite M5. Metabolite M5 (Figure 3E) was detected with an accurate [M − H]− ion at m/z 217.0161 (−4.6 ppm, C8H9O5S), which was 80 Da higher than that of M3, suggesting a sulfuric acid group was conjugated to M3. Fragment ions at m/z 137.0583 and 79.9576 of the Q-TOF MS2 fragmentation also proved the existence of the aglycone and sulfate. The product ion at m/z 173.1 (Q-trap MS2 spectrum, Figure 4D) of M5 indicated the sulfation was at the hydroxyl group on the phenyl ring. Therefore, M5 was the product from the sulfation of M3 or the deglycosylation of M2. Metabolite M6. M6 (Figure 3F) was detected with an accurate [M − H]− ion at m/z 233.0107 (−5.6 ppm, C8H9O6S). The molecular ion at m/z 233.0107 was 16 Da more than m/z 217.0161(M5), which suggested that M6 was a hydroxylation metabolite of M5. The fragment ion of M6 at m/ 9002

DOI: 10.1021/acs.jafc.5b04510 J. Agric. Food Chem. 2015, 63, 8999−9005

Article

Journal of Agricultural and Food Chemistry

Figure 4. MS2 spectra of metabolites M1 (A), M2 (B), M4 (C), M5 (D), M6 (E), and M7 (F) by HPLC/Q-trap-MS.

Figure 5. Proposed metabolite structures and predicted salidroside metabolic pathway: (a) glucuronidation; (b) sulfation; (c) deglycosylation; (d) hydroxylation; (e) methylation; (f) dehydroxylation.

9003

DOI: 10.1021/acs.jafc.5b04510 J. Agric. Food Chem. 2015, 63, 8999−9005

Journal of Agricultural and Food Chemistry methylation can only be asserted on the phenyl ring, and the sulfation occurred at the hydroxyl group on the phenyl ring. Hence, M7 was characterized as the methylation product of M5. Metabolite M8. M8 (Figure 3H) was detected at 4.54 min and with an accurate [M − H]− ion at m/z 297.0985 (3.7 ppm, C14H17O7), Further MS2 fragmentation (Figure 3H) showed fragment ions at m/z 175.0241, 121.0642, and 113.0231. The molecular ion at m/z 297.0985 and its MS2 fragment ion at m/z 121.0642 were 16 Da less than m/z 313.0923 (M4) and its product ion at m/z 137.0583, suggesting M8 was the dehydroxylation product of metabolite M4. The diagnostic ions of glucuronic acid at m/z 175.0741 and 113.0231 were detected, further proving the glucuronidation. We failed to get the signals of M8 by Q-trap MS detection. We could not identify the exact position of dehydroxylation. Metabolic Pathways of Salidroside in Rats. In the present study, metabolites in rat urine samples after oral administration of salidroside were detected and identified through UPLC/Q-TOF-MS and HPLC/Q-trap-MS. Eight metabolites have been characterized in the urine samples within 12 h after oral administration; most of these metabolites could not be detected after 12 h. p-Tyrosol (M3) was the only phase I metabolite detected, formed by deglycosylation of salidroside. Two phase II metabolites of salidroside were derived from glucuronidation (M1) and sulfation (M2). The other metabolites were phase II metabolites of p-tyrosol, including the direct glucuronidation (M4) and sulfation (M5) products. Puzzlingly, the sulfation of hydroxylated (M6) and methylated (M7) p-tyrosol and the glucuronidation of dehydroxylated p-tyrosol (M8) were identified, whereas the corresponding phase I metabolites were not detected. A possible explanation is that salidroside metabolizes to p-tyrosol, which probably experienced hydroxylation, methylation, and dehydroxylation in vivo and the phase I products readily converted to phase II metabolites M6, M7, and M8. We also tested the feces samples, but no metabolite was found. In our previous study, p-tyrosol was found in urine samples but not in plasma and feces samples after oral administration, which is consistent with our present results.18,26 The possible structures of metabolites and predicted metabolic pathway are speculated in Figure 5. In summary, salidroside and its deglycosylation phase I metabolite p-tyrosol were further metabolized to glucuronidation and sulfation products and mainly excreted through the urine excretion pathway.



Article



ACKNOWLEDGMENTS



REFERENCES

We are grateful to Ting Liu and Zhimin Long from SCIEX Shanghai Application Support Center for performing the Qtrap MS analysis.

(1) Khanum, F.; Bawa, A. S.; Singh, B. Rhodiola rosea: a versatile adaptogen. Compr. Rev. Food Sci. Food Saf. 2005, 4, 55−62. (2) Platikanov, S.; Evstatieva, L. Introduction of wild golden root (Rhodiola rosea L.) as a potential economic crop in Bulgaria. Econ Bot. 2008, 62, 621−627. (3) Furmanowa, M.; Kedzia, B.; Hartwich, M.; Kozlowski, J.; Krajewska-Patan, A.; Mscisz, A.; Jankowiak, J. Phytochemical and pharmacological properties of Rhodiola rosea L. Herba Pol. 1999, 45, 108−113. (4) Ye, Y. C.; Chen, Q. M.; Jin, K. P.; Zhou, S. X.; Chai, F. L.; Hai, P. Effect of salidroside on cultured myocardial cells anoxia/reoxygenation injuries. Acta Pharmacol. Sin. 1993, 14, 424−426. (5) Hu, X. L.; Lin, S. X.; Yu, D. H.; Qiu, S. F.; Zhang, X. Q.; Mei, R. H. A preliminary study: the anti-proliferation effect of salidroside on different human cancer cell lines. Cell Biol. Toxicol. 2010, 26, 499−507. (6) Fang, D. L.; Chen, Y.; Xu, B.; Ren, K.; He, Z. Y.; He, L. L.; Lei, Y.; Fan, C. M.; Song, X. R. Development of lipid-shell and polymer core nanoparticles with water-soluble salidroside for anti-cancer. Int. J. Mol. Sci. 2014, 15, 3373−3388. (7) Wang, J.; Li, J. Z.; Lu, A. X.; Zhang, K. F.; Li, B. J. Anticancer effect of salidroside on A549 lung cancer cells through inhibition of oxidative stress and phospho-p38 expression. Oncol. Lett. 2014, 7, 1159−1164. (8) Lanza, A. M. D.; Martinez, M. J. A.; Matellano, L. F.; Carretero, C. R.; Castillo, L. V.; Sen, A. M. S.; Benito, P. B. Lignan and phenylpropanoid glycosides from Phillyrea latifolia and their in vitro anti-inflammatory activity. Planta Med. 2001, 67, 219−223. (9) Qian, E. W.; Ge, D. T.; Kong, S. K. Salidroside protects human erythrocytes against hydrogen peroxide-induced apoptosis. J. Ethnopharmacol. 2011, 133, 308−314. (10) Zhang, L.; Yu, H. X.; Sun, Y.; Lin, X. F.; Chen, B.; Tan, C.; Cao, G. X.; Wang, Z. W. Protective effects of salidroside on hydrogen peroxide-induced apoptosis in SH-SY5Y human neuroblastoma cells. Eur. J. Pharmacol. 2007, 564, 18−25. (11) Wu, Y. L.; Piao, D. M.; Han, X. H.; Nan, J. X. Protective effects of salidroside against acetaminophen-induced toxicity in mice. Biol. Pharm. Bull. 2008, 31, 1523−1529. (12) Mao, G. X.; Deng, H. B.; Yuan, L. G.; Li, D. D.; Yvonne Li, Y. Y.; Wang, Z. Protective role of salidroside against aging in a mouse model induced by D-galactose. Biomed. Environ. Sci. 2010, 23, 161− 166. (13) Wang, H. B.; Ding, Y. Y.; Zhou, J.; Sun, X. L.; Wang, S. W. The in vitro and in vivo antiviral effects of salidroside from Rhodiola rosea L. against coxsackievirus B3. Phytomedicine 2009, 16, 146−155. (14) Li, T. L.; Xu, G. H.; Wu, L. L.; Sun, C. Y. Pharmacological studies on the sedative and hypnotic effect of salidroside from the Chinese medicinal plant Rhodiola sachalinensis. Phytomedicine 2007, 14, 601−604. (15) Mao, Y.; Zhang, X. R.; Zhang, X. D.; Lu, G. C. Development of an HPLC method for the determination of salidroside in beagle dog plasma after administration of salidroside injection: application to a pharmacokinetics study. J. Sep. Sci. 2007, 30, 3218−3222. (16) Chang, Y. W.; Yao, H. T.; Hsieh, S. H.; Lu, T. J.; Ye, T. K. Quantitative determination of salidroside in rat plasma by on-line solid-phase extraction integrated with high-performance liquid chromatography/electrospray ionization tandem mass spectrometry. J. Chromatogr. B: Anal. Technol. Biomed. Life Sci. 2007, 857, 164−169. (17) Yu, S.; Liu, L.; Wen, T.; Liu, Y. C.; Wang, D. L.; He, Y. X.; Liang, Y.; Liu, X. D.; Xie, L.; Wang, G. J.; Wei, W. Z. Development and validation of a liquid chromatographic/electrospray ionization mass spectrometric method for the determination of salidroside in rat

AUTHOR INFORMATION

Corresponding Author

*(Y.W.) Phone/fax: +86-451-82190052. E-mail: ywang1971@ hotmail.com. Funding

This research was financially supported by the Fundamental Research Funds for the Central Universities (DL13EA02) and Heilongjiang Provincial Natural Science Foundation of China (C201139). Notes

The authors declare no competing financial interest. 9004

DOI: 10.1021/acs.jafc.5b04510 J. Agric. Food Chem. 2015, 63, 8999−9005

Article

Journal of Agricultural and Food Chemistry plasma: application to the pharmacokinetics study. J. Chromatogr. B: Anal. Technol. Biomed. Life Sci. 2008, 861, 10−15. (18) Guo, N.; Hu, Z. W.; Fan, X. X.; Zheng, J.; Zhang, D. H.; Xu, T.; Yu, T.; Wang, Y.; Li, H. Y. Simultaneous determination of salidroside and its aglycone metabolite p-tyrosol in rat plasma by liquid chromatography-tandem mass spectrometry. Molecules 2012, 17, 4733−4754. (19) Zhang, Y.; Li, L. Q.; Lin, L.; Liu, J. X.; Zhang, Z. H.; Xu, D. J.; Xiang, F. J. Pharmacokinetics, tissue distribution, and excretion of salidroside in rats. Planta Med. 2013, 79, 1429−1433. (20) Vissers, M. N.; Zock, P. L.; Roodenburg, A. J.; Leenen, R.; Katan, M. B. Olive oil phenols are absorbed in humans. J. Nutr. 2002, 132, 409−417. (21) Corona, G.; Tzounis, X.; Assunta, D. M.; Deiana, M.; Debnam, E. S.; Visioli, F.; Spencer, J. P. The fate of olive oil polyphenols in the gastrointestinal tract: implications of gastric and colonic microfloradependent biotransformation. Free Radical Res. 2006, 40, 647−658. (22) Mateos, R.; Goya, L.; Bravo, L. Metabolism of the olive oil phenols hydroxytyrosol, tyrosol, and hydroxytyrosyl acetate by human hepatoma HepG2 cells. J. Agric. Food Chem. 2005, 53, 9897−9905. (23) Guo, J. M.; Xue, C. F.; Shang, E. X.; Duan, J. A.; Tang, Y. P.; Qian, D. W. Identification of hyperoside metabolites in rat using ultra performance liquid chromatography/quadrupole-time-of-flight mass spectrometry. J. Chromatogr. B: Anal. Technol. Biomed. Life Sci. 2011, 879, 1987−1992. (24) Cui, L.; Sun, E.; Zhang, Z. H.; Qian, Q.; Tan, X. B.; Xu, F. J.; Jia, X. B. Metabolite profiles of epimedin B in rats by ultra performance liquid chromatography/ quadrupole-time-of-flight mass spectrometry. J. Agric. Food Chem. 2013, 61, 3589−3599. (25) Xia, Y. Q.; Miller, J. D.; Bakhtiar, R.; Franklin, R. B.; Liu, D. Q. Use of a quadrupole linear ion trap mass spectrometer in metabolite identification and bioanalysis. Rapid Commun. Mass Spectrom. 2003, 17, 1137−1145. (26) Guo, N.; Zhu, M. X.; Han, X. J.; Sui, D.; Wang, Y.; Yang, Q. The metabolism of salidroside to its aglycone p-tyrosol in rats following the administration of salidroside. PLoS One 2014, 9, e103648.

9005

DOI: 10.1021/acs.jafc.5b04510 J. Agric. Food Chem. 2015, 63, 8999−9005