Jaboticabin and Related Polyphenols from ... - ACS Publications

Jan 9, 2019 - ... Center, The City University of New York, 365 Fifth Avenue, New York City, New York ... Department of Medicine, College of Physicians...
1 downloads 0 Views 1005KB Size
Article Cite This: J. Agric. Food Chem. XXXX, XXX, XXX−XXX

pubs.acs.org/JAFC

Jaboticabin and Related Polyphenols from Jaboticaba (Myrciaria cauliflora) with Anti-inflammatory Activity for Chronic Obstructive Pulmonary Disease Da-Ke Zhao,†,‡,§,∥ Ya-Na Shi,†,⊥ Vanya Petrova,#,∇ Grace G. L. Yue,○ Adam Negrin,#,∇ Shi-Biao Wu,∇ Jeanine M. D’Armiento,◆ Clara B. S. Lau,○ and Edward J. Kennelly*,#,∇

J. Agric. Food Chem. Downloaded from pubs.acs.org by UNIV OF WINNIPEG on 01/24/19. For personal use only.



Biocontrol Engineering Research Center of Plant Disease & Pest, §Biocontrol Engineering Research Center of Crop Disease & Pest, and ∥School of Life Science, Yunnan University, Kunming, Yunnan 650504, People’s Republic of China ⊥ Institute of Medicinal Plants, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan 650200, People’s Republic of China # Ph.D. Program in Biology, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York City, New York 10016, United States ∇ Department of Biological Sciences, Lehman College, The City University of New York, 250 Bedford Park Boulevard West, Bronx, New York 10468, United States ○ Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong ◆ Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, P&S 9-449, New York City, New York 10032, United States S Supporting Information *

ABSTRACT: Myrciaria cauliflora (jaboticaba) is an edible fruit common in Brazil that has been used for treating respiratory diseases, including chronic tonsillitis and asthma. This study explores the distribution of an anti-inflammatory depside, jaboticabin, in different parts of the jaboticaba plant as well as major polyphenols from the wood of jaboticaba, some with biological activity similar to jaboticabin. The peel of the fruit was found to be the major source of jaboticabin. This is the first phytochemical study of the wood of M. cauliflora. The antioxidant-activity-guided fractionation strategy successfully identified 3,3′-dimethylellagic acid-4-O-sulfate from jaboticaba wood. This ellagic acid derivative, in a manner similar to jaboticabin, showed antiradical activity and inhibited the production of the chemokine interleukin-8 after treating the human small airway epithelial cells with cigarette smoke extract. The human intestinal Caco-2 cell studies demonstrated the jaboticabin transport in vitro. The polyphenols, jaboticabin and 3,3′-dimethyellagic acid-4-O-sulfate, from jaboticaba were both found to exhibit anti-inflammatory activities, thus suggesting the potential use of these compounds or even the fruits themselves for chronic obstructive pulmonary disease. KEYWORDS: jaboticaba, Myrciaria caulif lora, Myrtaceae, jaboticabin, depside, COPD



INTRODUCTION

interleukin 8 (IL-8) by 81.3% in untreated small airway epithelial (SAE) cells and also lowered the production by 47.3% in SAE cells treated by 5% cigarette smoke extract (CSE).10−12 M. cauliflora extract was reported to improve diabetic nephropathy by suppressing oxidative stresses and inflammation of the tested mice13 and function as a vasorelaxant and hypotension factor.14 The phenolic secondary metabolites should, in part, confer to the protective effects.15 The main anthocyanins in jaboticaba peels include delphinidin-3-Oglucoside and cyanidin-3-O-glucoside.16 Other related polyphenols in jaboticaba are isoquercitrin, gallic acid, quercetin, myricitrin, ellagic acid, quercitrin, and quercimeritrin.10,16 Some of these compounds have been explored as potential treatments for COPD.10,17 Among the identified compounds from

Chronic obstructive pulmonary disease (COPD) is a multidimensional lung disease characterized by unalterable airflow obstruction caused by inflammation. The disease is one of the leading causes of death throughout the world1,2 and frequently occurs with comorbidities.3 COPD has recently been recognized as central to the progression of other related chronic diseases, as seen in a longitudinal exploration of the Danish population.4,5 The proteolysis and inflammation characteristics in COPD patients are secondary to the ordinary inflammatory reaction with cigarette smoke that is the major etiological agent related to the disease.6,7 Jaboticaba (Myrciaria cauliflora), native to tropical Brazil, is a tree with dark-colored and pleasant-tasting fruits, and the plant belongs to the Myrtaceae family.8,9 The fruits of jaboticaba are reported to have powerful anti-inflammatory and antioxidant activity. The 1,1-diphenyl-2-picrylhydrazyl (DPPH) assay value of the fresh fruit extract was 0.282 mg/mL (IC50), and one of the major constituents, jaboticabin, reduced the production of © XXXX American Chemical Society

Received: October 24, 2018 Revised: January 9, 2019 Accepted: January 9, 2019

A

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry jaboticaba fruits, jaboticabin, a depside polyphenol first reported in jaboticaba fruits by our group, showed promise in in vitro results for the treatment of COPD,11,18 indicating that jaboticaba-related polyphenols could be useful for treating COPD lung health. This minor bioactive depside was successfully synthesized in our laboratory for future in vivo and clinical studies.19 To localize the production of the anti-inflammatory depside jaboticabin in M. cauliflora plants, the leaves, wood, bark, and fruit components (peel, pulp, and seed) were compared individually. Previous investigations focused mainly on the whole fruits, the peel, seeds,20 non-fermented and fermented jaboticaba fruits,21 or commercial products from the fruits.10 Until now, no phytochemical investigation is available on the other organs, except the fruits, which could potentially be exploited in the future.10 To identify the candidate compounds with biological activity similar to jaboticabin, the wood of this plant has now been studied chemically for the first time, combined with the antioxidant-activity-guided strategy. In the present study, ultraperformance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry (UPLC−ESI−QTOF−MS), together with nuclear magnetic resonance (NMR), was used to identify the constituents of jaboticaba wood. One of the major constituents, 3,3′dimethyellagic acid-4-O-sulfate (9), was evaluated for its cytokine inhibition in smoke-exposed SAE cells for the first time. While some of the compounds from the wood displayed anti-inflammatory activity, none was as active as the previously identified jaboticabin. While jaboticabin has been studied in vitro, there have been no studies in vivo to examine its bioavailability. Thus, the Caco-2 transport investigations for jaboticabin were employed to predict its absorption in vivo.



LC−TQD−MS analysis was carried out on the Waters Acquity UPLC triple quadrupole mass spectrometer. A Phenomenex Kinetex C18 UPLC column (1.7 μm particle size, 50 × 2.1 mm inner diameter, 100 Å) was employed coupled to a Phenomenex UPLC C18 SecurityGuard ULTRA cartridge (2.1 mm inner diameter). The mobile phase employed used LC−MS-grade aqueous formic acid at 0.1% as solvent A and LC−MS-grade MeCN as solvent B. Its flow was run at 0.5 mL/min, and the C-18 column of LC−MS was kept at 40 °C for analysis. MS parameters for LC−TQD−MS analysis employed during the analysis was shown as follows: 3.0 kV capillary voltage, 150 °C source temperature, 450 °C desolvation temperature, nitrogen selected as the desolvation gas with a flow of 800 L/h, cone gas flow at 50 L/h, and collision gas using argon (flow rate at 0.15 mL/min). Quantification of jaboticabin was achieved using optimized multiple reaction monitoring (MRM) parameters of parent to daughter ion transitions detected. Suitable MRM methods were developed in positive and negative modes, but positive was chosen as a result of higher response during ionization. Three MRM transitions were optimized, and two were used for the analysis: 335.1 > 137.1 (quantitation MRM) and 335.1 > 109.0 (confirmation MRM). A calibration curve was prepared using jaboticabin standard to quantify jaboticabin in extracted tissues at concentrations from 13.2 to 105.6 ng/mL. The lower detection limit for jaboticabin was established to be below 7.5 ng/mL with a signal-to-noise (S/N) ratio of 15:1 using a peak-topeak integration method. Analyses of calibration curve standards and jaboticaba tissue extracts were performed in triplicate. Isolating 3,3′-Dimethyellagic Acid-4-O-sulfate. Dried jaboticaba wood (200 g) was extracted 2 times with 80% methanol (2 × 1 L, total amount of 2 L) at 25 °C. The solvent was evaporated under reduced pressure, and 15 g of a brown residue remained. The entire crude extracts were loaded to vacuum liquid chromatography (VLC, 40 × 8 cm column) over silica gel (600 g) with a gradient solvent from hexane (300 mL), ethyl acetate (800 mL), acetone (400 mL), and methanol (300 mL) to yield nine fractions (1−9, each 200 mL solvent). The nine fractions were measured for the antioxidant roles by the DPPH assay, and fractions 7−9 displayed better activities (Figure S1 of the Supporting Information). The following study further indicated that the nonpolar last fraction 9 had the strongest activity (unpublished data). Therefore, its major compound (compound 9) was isolated by semi-preparative Waters HPLC with a Alliance 2695 system, 2695 separation system, 2996 photodiode array (PDA) detector, and 10 × 250 mm, 4 μm Phenomenex HPLC column from fraction 9 (380 mg). The mobile phase consisted of 1% formic acid (aqueous) solution as solvent A and acetonitrile as solvent B. The gradient conditions of HPLC were used as 21% B maintaining for 5 min, then from 21 to 25% B until 42 min, then increased B up to 95% in 3 min, and maintained at 95% B until 50 min. The flow rate was 3 mL/min, and the injection volume was 100 μL. Compound 9 (2.2 mg) was eluted in 40 min. Sample and column temperatures were both at 25 °C. Compound 9 was identified on the basis of exact mass molecular weight, MS fragmentations, and analysis of 1H, 13C, and two-dimensional (2D) NMR spectra. UPLC Quadrapole Time of Flight Mass Analysis. LC−MS analysis with high resolution was conducted on a Waters Acquity UPLC Xevo G2 quadrapole time-of-flight (QTOF) mass spectrometer with Masslynx version 4.1 software. A Phenomenex Kinetex C18 UPLC column (50 × 2.1 mm inner diameter, 1.7 μm, 100 Å) was employed coupled to a Phenomenex UPLC C18 SecurityGuard ULTRA cartridge (2.1 mm inner diameter). The mobile phase employed used LC−MS-grade 0.1% aqueous formic acid as solvent A and LC−MS-grade MeCN with formic acid (0.1%) as solvent B. The HPLC flow rate was performed at 0.5 mL/min, and the column temperature was set at 40 °C during the whole analysis. MS parameters for UPLC−QTOF−MS analysis employed during the analysis were as follows: capillary voltage kept at 3.0 kV, source temperature using 110 °C, desolvation temperature of 300 °C, desolvation gas of N2 with a flow rate of 600 L/h, cone gas flow at 50 L/h, and collision gas of argon, with a flow rate of 0.15 mL/min. IL-8 Immunoassay. On the basis of the instructions of the supplier (Lonza, Walkersville, MD, U.S.A.), SAE cells were cultured and

MATERIALS AND METHODS

Reagents. 1,1-Diphenyl-2-picrylhydrazyl (DPPH) was ordered from MilliporeSigma. High-performance liquid chromatography (HPLC)-grade solvents, such as acetonitrile and formic acid, were ordered from J.T. Baker Chemicals, and methanol [guaranteed reagent (GR) grade] was from VWR International, Inc. Ultrapure water (UPW) was generated from one Millipore system that is MilliRO 12 Plus, MilliporeSigma. Plant Materials. The wood, leaves, and fresh fruits of M. cauliflora were sourced from the Fruit and Spice Park (Homestead, FL, U.S.A.) in 2011 and 2013. Voucher specimens of these plant parts were deposited at the herbarium of the New York Botanical Garden (NYBG, NY, U.S.A.). The plant materials were identified by Chris Rollins and Eric McDonough from the Fruit and Spice Park in Homestead, FL, U.S.A. Wood was crushed to powder and stored at room temperature before extraction. Ripe fruits were sampled and then kept frozen at −20 °C prior to extraction. Quantification of Jaboticabin. Quantification of jaboticabin in M. cauliflora samples was achieved using liquid chromatography tandem quadrupole detector mass spectrometry (LC−TQD−MS). Samples of jaboticaba fruits, leaves, wood, and bark (n = 3) were collected, stored at −80 °C, and freeze-dried prior to processing. Jaboticaba fruits were combined from each respective collection and dissected to separate peel, pulp, and seeds for extraction. All samples were then pulverized using a commercial tissue grinder to produce a fine homogeneous powder. Each sample (ca. 100 mg) was weighed and extracted with 10 mL of LC−MS-grade 70% MeOH and 30% water for 15 min at 20 °C using an ultrasonicator bath. Then, the extracts were subjected to centrifugation at 2800 rpm for 4 min, and the supernatant was decanted into 20 mL borosilicate glass scintillation vials. For the extraction procedure, it was conducted repeatedly, and the resulting extracts were combined. Samples were evaporated to dryness under N2 gas. All samples were kept at −20 °C before analysis. B

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry maintained under controlled air with 5% CO2 at 37 °C. A total of 80% of confluent SAE cells at passages 2−5 were selected in this study. CSE was prepared as follows: the Barnet vacuum pump that was operating with constant flow was adopted to draw smoke from a 3R4F research-grade cigarette (University of Kentucky, Lexington, KY, U.S.A.) via phosphate-buffered saline of Dulbecco (25 mL). The solution with 100% CSE was regulated to pH 7.4, filtered, diluted to a final concentration (5%) by small airway growth medium, and then immediately added to cells. The cells were then treated with 5% CSE and test chemicals (50 μM). On the basis of the specifications of the manufacturer (Invitrogen, Carlsbad, CA, U.S.A.), the viability of cells was estimated following the CSE exposure with alamarBlue kit. IL-8 in the supernatants was measured after 24 h by enzyme-linked immunosorbent assay (ELISA, BioLegend, San Diego, CA, U.S.A.) after normalization to the protein content by a bicinchoninic acid (BCA) assay (Pierce, Rockford, IL, U.S.A.). Student’s t test in a twosided way was selected for statistical analyses and defined at the 5% level. The purities (%) of test compounds ranged from 97 to 99% as determined by analytical HPLC and LC−MS. Caco-2 Transport Studies. For the cultures of Caco-2 human colonic cells, refer to our previous study.22 In brief, the target Caco-2 cells seeded on six-well plates with Transwell inserts (Corning, MA, U.S.A.) were cultured for 21 days to form the monolayers. With the epithelial voltohmmeter (World Precision Instruments, Sarasota, FL, U.S.A.), the integrity for the newly formed monolayer was monitored via surveying the transepithelial electrical resistance (TEER). Transport tests on monolayers were performed with the TEER over 600 Ω cm2. The TEER of the monolayer was surveyed before and after tests on transport. To conduct this transport test, Transwell inserts were washed twice and then equilibrated with 37 °C Hank’s balanced salt solution (HBSS) transport buffer at pH 6.8 for 15 min ahead of this transport experiment. After that, jaboticabin (100 mg/mL) was supplemented to the apical side of the monolayer and then incubated at 37 °C. Aliquots of 500 μL were withdrawn from the basolateral side of the monolayer after 30, 60, 90, and 120 min. The withdrawn samples were replaced by the prewarmed HBSS transport buffer in equal volumes and then frozen for LC−MS analysis at −20 °C. The purity of jaboticabin was 99% as determined by LC−QTOF−MS and NMR. Two reference compounds, atenolol (purity of 98%) and propranolol (purity of 98%), were used as positive controls.21 The apparent permeability coefficient (Papp) was measured as the protocol described by Artursson and Karlsson23

Papp =

Figure 1. LC−TQD−MS chromatograms of jaboticabin reference standard and different jaboticaba plant parts [A, extracted ion chromatogram (EIC) of jaboticabin based on daughter ion 137.1; B, EIC of jaboticabin based on daughter ion 109.0; C, EIC of jaboticabin from peel extract; D, EIC of jaboticabin from pulp extract; and E, EIC of jaboticabin from seed extract].

dC /dtV AC

where “dC/dt” indicates the alteration for drug density over time in the receiver chambers, “V” displays the solution volume in the receiver chambers in cm3, “A” is the surface area of the membrane in cm2, and “C” represents the original density in donor chambers. The Papp value of the low-permeability compound atenolol (positive control) was 0.79 ± 0.24 × 10−6, while that of the high-permeability compound propranolol (another positive control) was 32.5 ± 2.53 × 10−6.

Figure 2. Quantification of jaboticabin in different jaboticaba plant parts.



A powder of jaboticaba peel has been shown to have strong antioxidant activity in vitro as well as in vivo,9,26 and its intake attenuates oxidative stress resulting from obesity.12 A clinical study found that the consumption of jaboticaba peel powder strengthened the serum antioxidant capacity toward peroxyl radicals in healthy volunteers.27 These reports indicated that, in addition to anthocyanins, e.g., compound 13 and cyanidin3-O-glucoside (12),11 depside jaboticabin (11) may contribute to the excellent antioxidant activity of the jaboticaba peel. Leaf, wood, and bark extracts that did not yield a detectable signal by LC−TQD−MS for jaboticabin upon initial analysis were further processed using solid-phase extraction to yield concentrated sample extracts for additional analysis. In our study, we failed to detect jaboticabin in extracts of the leaves, wood, and bark even after repeated analysis of concentrated SPE extracts. Thus, fruits are the best natural source of jaboticabin as well as

RESULTS AND DISCUSSION To localize the production of the anti-inflammatory depside jaboticabin in M. cauliflora plant compounds, the leaves, wood, bark, and fruit components (peel, pulp, and seed) were compared separately. The results of LC−TQD−MS revealed that jaboticabin was found in all parts of the fruit (Figures 1 and 2) but in the highest level in the peels. Peels of fruits are frequently known to be rich sources of bioactive compounds, such as the well-studied antioxidant resveratrol found in the highest concentrations in grape skins.24,25 The analysis showed that jaboticabin was present in the highest concentrations in fruit peels (3.04−4.35 μg of jaboticabin/g of dry weight), followed by fruit pulp (0.89−3.00 μg/g) and seed (0.34− 1.11 μg/g). C

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Figure 3. Structures of the compounds identified from jaboticaba wood and fruits.

(7 and 8) in Myciaria species, and the first report of compounds 9 and 10 in M. cauliflora. These ellagic acid derivatives or sulfates are only detected in the jaboticaba wood and bark but not found in the fruits. Furthermore, the major compounds found in the fruits, such as anthocyanins and flavonoids, were not found in its wood or bark.10,11 Jaboticabin has been studied for its therapeutic potential for COPD, which is comprised of two conditions: emphysema and chronic bronchitis. COPD is usually steroid-resistant; furthermore, anti-inflammatories that target chemokine pathways are considered as the new potential therapies for this disease. IL-8 is one cytokine implicated in various chronic inflammatory diseases. Moreover, jaboticaba fruit polyphenol compounds, such as the depside jaboticabin (11) and compounds 12 and 13, were reported to inhibit the CSE-induced IL-8 in SAE cells by our group previously.11 Ellagic acid (6) was also shown to inhibit matrix metalloproteinase 1 (MMP-1) and IL-8 expression in vitro.17 In the current study, jaboticaba wood was extracted, fractionated, and tested for anti-inflammatory activity, and subfractions 7−9 were found to inhibit the IL-8 expression in cigarettesmoke-exposed SAE cells to the greatest extent. Fraction 9 showed the strongest antioxidant potential in the DPPH assay (unpublished data). One of its major compounds, compound 9, was isolated and tested for its anti-inflammatory activity. Jaboticabin (11), previously shown to decrease CSE-induced IL-8 levels, was included in the study as a positive control.11 Similar to jaboticabin (523.28 pg/mL when CSE-induced and 190.50 pg/mL when non-induced), compound 9 significantly decreased IL-8 levels in CSE-exposed SAE cells (337.73 pg/mL when CSE-induced and 180.63 pg/mL when non-induced) (Figure 5). Compound 9 did not show any significant cytotoxic

a renewable resource to obtain this bioactive depside from these small trees. The genus of Myciaria contains approximately 99 known species,28 including species allied to jaboticaba, such as Myciaria vexator and Myciaria dubia (camu camu). Jaboticabin was detected in the fruits of M. vexator9,17 but not in any other related species. Further investigations of the fruits of Myciaria species may find other sources for jaboticabin and other bioactive constituents. Only a handful of reports on jaboticaba chemical constituents are available.10,11 This study reports the chemical constituents of jaboticaba wood for the first time. Five gallotannins (compounds 1−5) were tentatively identified from the wood and bark. Among them, castalagin (1) was reported in camu camu fruit (M. dubia) before,29 and casuarinin (4) and casuarictin (5) have also been reported in jaboticaba fruits.6 Ellagic acid A (6), delphinidin-3-O-glucoside (13), and compound 12 have been isolated and reported from this species by our group as well.11 A rare ellagic acid derivative, 3,3′dimethyellagic acid-4-O-sulfate (compound 9; Figure 3), was isolated and identified for the first time in this species. Its structure was determined by 1H and 13C NMR spectra. This compound has been previously reported once before from the leaves of Euphorbia soongarica.30 Besides compound 9, other ellagic acid derivatives or sulfates, such as 3-O-methylellagic acid (7), 3-methyellagic acid-4-O-sulfate (8), and 3,3′-dimethyellagic acid (10), were also detected from the MeOH extract of jaboticaba wood, and their UPLC−QTOF−MS data, such as retention time, exact mass, and adduct and fragmental ion exact masses in positive mode were used for tentative identifications (Figure 4 and Table 1). Sulfuric acid losses from compounds 7 and 8 were both detected under negative and positive modes. This is the first report of these two sulfates D

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Figure 4. LC−TOF−MS analysis of MeOH extracts of jaboticaba wood, bark, and leaves in negative mode [A, total ion chromatogram (TIC) of wood extract; B, TIC of bark extract; and C, TIC of leaves extract].

effect on SAE cells. These results showed that compound 9 was able to decrease inflammation secondary to the exposure of smoke and may confer a therapeutic effect with regard to COPD. While jaboticabin has been studied in vitro, there have been no studies in vivo to examine its bioavailability. Therefore, the transport of jaboticabin was examined by the Caco-2 cell monolayer test. Dimethyl sulfoxide (DMSO) and jaboticabin

at concentrations in the assay were not observed to change the values of TEER (unpublished data) after incubation for 120 min, indicating the intact integrity for these Caco-2 cell monolayers. After the absorptive transport from the apical side to basolateral side of the cell monolayer, jaboticabin was observed in the sample collected from the basolateral side, suggesting that jaboticabin could pass across the Caco-2 cell E

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry Table 1. Identification of Compounds from Jaboticaba Wood by UPLC−QTOF−MS compound number

RT (min)

[M]+, [M + H]+, or [M − H]− (MF, ppm)

adduct and fragmental ion exact masses [M − X]+ or [M − X]− (MF, ppm)

note or reference

castalagin

Fracassetti et al.29

eugenigrandin A

Koenig et al.31

1

0.98

935.0791 [M + H]+ (C41H27O26, 0.4)

2

1.21

1223.1460 [M + H]+ (C56H39O32, 2.9)

1240.1710 [M + H + NH3]+ (C56H42NO32, 1.6)

3

1.34

1207.1504 [M + H]+ (C56H39O31, 2.4)

1224.1741 [M + H+ NH3]+ (C56H42NO31, 0.0)

acutissimin A

Stark et al.32

4

1.53

937.0947 [M + H]+ (C41H29O26, 0.0)

617.0785 [M + H − H2O − HHDP]+ (C27H21O17, 1.0); 635.0845 [M + H − HHDP]+ (C27H23O18, 6.1); 785.0837 [M + H − galloyl group]+ (C34H25O22, 7.1)

casuarinin

Wu et al.10

5

1.64

937.0963 [M + H]+ (C41H29O26, 1.7)

919.0852 [M + H − H2O]+ (C41H27O25, 1.2); 617.0737 [M + H − H2O − HHDP]+ (C27H21O17, −0.6); 635.0885 [M + H − HHDP]+ (C27H23O18, 0.2); 785.0880 [M + H − galloyl group]+ (C34H25O22, 5.5)

casuarictin

Wu et al.10

6

2.38

303.0143 [M + H]+ (C14H7O8, 0.7)

605.0204 [2M + H]+ (C28H13O16, 0.7)

ellagic acid

Wu et al.10

7

2.69

396.9859 [M + H] (C15H9O11S, −1.8)

317.0297 [M + H − sulfuric acid] (C15H9O8, −1.9); 3-methyellagic acid713.0085 [2M + H − sulfuric acid]+ (C30H21O19S, 4-O-sulfate 2.2), 792.9653 [2M + H]+ (C30H17O22S2, 0.0)

first report in Myrciaria species

8

2.93

317.0297 [M + H]+ (C15H9O8, −3.2)

633.0517 [2M + H]+ (C30H17O16, −3.2)

first report in jaboticaba

9

2.96

411.0022 [M + H]+ (C16H11O11S, −2.2)

331.0454 [M + H − sulfuric acid]+ (C16H11O8, −3.0); 3,3′-dimethyellagic 741.0398 [2M + H − sulfuric acid]+ (C32H21O19S, acid-4-O-sulfate −1.6), 820.9966 [2M + H]+ (C32H21O11S2, 0.4)

first report in Myrciaria species; identification based on MS and NMR30

10

3.17

331.0445 [M + H]+ (C16H11O8, −2.7)

353.0273 [M + Na]+ (C16H10O8Na, −6.5); 661.0830 [2M + H]+ (C32H21O16, −0.8)

first report in jaboticaba

+

952.1057 [M + H + NH3]+ (C41H30NO26, 0.1); 917.0709 [M + H − H2O]+ (C41H25O25, 2.6); 633.0707 [M + H − HHDP]+ (C27H21O18, −3.3)

tentative identification

+

3-O-methylellagic acid

3,3′-dimethyellagic acid

Figure 6. Time course of bidirectional transcellular transport of jaboticabin from the apical to basolateral (A → B) side and from the basolateral to apical (B → A) side of the Caco-2 cell monolayers. Results are expressed as the mean cumulative amount transported ± standard deviation (SD) (n = 2 with triplicate Transwell inserts each).

Figure 5. IL-8 inhibition by 3,3′-dimethyellagic acid-4-O-sulfate (9) and jaboticabin (11) at 50 μM/mL in SAE cells untreated (open bars) and treated (bold bars) with CSE. Data are presented as mean values ± 95% confidence limits (n = 3). Open bars with the same lowercase letters (a) and bold bars with the same capital letters (A and B) are not significantly (p > 0.05) different.

compound atenolol, and it may play a role in the activities of the intestinal cell transporters, which awaits further investigation. Nevertheless, when the apical to basolateral absorption of jaboticabin was determined alone, which mimics the actual absorption in the intestine, there was 0.84% of the amount originally supplemented to the apical side of jaboticabin that crossed the monolayers of the target cell. Therefore, the compound would be anticipated to go in the bloodstream and arrive at the target cells after the intestinal absorption. While transport of jaboticabin is detected, it is not very efficient. Future studies are planned to look at jaboticabin analogues in the Caco-2 model to see if any are more efficiently transported. In conclusion, jaboticabin (present in the fruit) and compound 9 (present in wood) from jaboticaba (M. cauliflora) were both found to exhibit anti-inflammatory activities in CSE-exposed

monolayers. The apical side to basolateral side apparent permeability coefficient (Papp,A→B) of jaboticabin determined at 120 min was 1.45 ± 0.77 × 10−6 cm/s. The permeability value from the basolateral side to apical side (Papp,B→A) was 4.49 ± 1.54 × 10−6 cm/s (Figure 6). The jaboticabin transport is thus polarized, with the permeability from the basolateral side to apical side that was B → A surpassing the permeability from the apical side to basolateral side that was A → B by 3.09 efflux ratio. Jaboticabin could be regarded as a low-permeability compound when the Papp value was compared to the reference F

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry

Khoo, J. P.; Knowlton, L. M.; Kobusingye, O.; Koranteng, A.; Krishnamurthi, R.; Lipnick, M.; Lipshultz, S. E.; Ohno, S. L.; Mabweijano, J.; MacIntyre, M. F.; Mallinger, L.; March, L.; Marks, G. B.; Marks, R.; Matsumori, A.; Matzopoulos, R.; Mayosi, B. M.; McAnulty, J. H.; McDermott, M. M.; McGrath, J.; Mensah, G. A.; Merriman, T. R.; Michaud, C.; Miller, M.; Miller, T. R.; Mock, C.; Mocumbi, A. O.; Mokdad, A. A.; Moran, A.; Mulholland, K.; Nair, M. N.; Naldi, L.; Narayan, K. M.; Nasseri, K.; Norman, P.; O’Donnell, M.; Omer, S. B.; Ortblad, K.; Osborne, R.; Ozgediz, D.; Pahari, B.; Pandian, J. D.; Rivero, A. P.; Padilla, R. P.; Perez-Ruiz, F.; Perico, N.; Phillips, D.; Pierce, K.; Pope, C. A.; Porrini, E.; Pourmalek, F.; Raju, M.; Ranganathan, D.; Rehm, J. T.; Rein, D. B.; Remuzzi, G.; Rivara, F. P.; Roberts, T.; De León, F. R.; Rosenfeld, L. C.; Rushton, L.; Sacco, R. L.; Salomon, J. A.; Sampson, U.; Sanman, E.; Schwebel, D. C.; Segui-Gomez, M.; Shepard, D. S.; Singh, D.; Singleton, J.; Sliwa, K.; Smith, E.; Steer, A.; Taylor, J. A.; Thomas, B.; Tleyjeh, I. M.; Towbin, J. A.; Truelsen, T.; Undurraga, E. A.; Venketasubramanian, N.; Vijayakumar, L.; Vos, T.; Wagner, G. R.; Wang, M.; Wang, W.; Watt, K.; Weinstock, M. A.; Weintraub, R.; Wilkinson, J. D.; Woolf, A. D.; Wulf, S.; Yeh, P. H.; Yip, P.; Zabetian, A.; Zheng, Z. J.; Lopez, A. D.; Murray, C. J.; AlMazroa, M. A.; Memish, Z. A. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A systematic analysis for the global burden of disease study 2010. Lancet 2012, 380, 2095−2128. (3) Divo, M.; Cote, C.; de Torres, J. P.; Casanova, C.; Marin, J. M.; Pinto-Plata, V.; Zulueta, J.; Cabrera, C.; Zagaceta, J.; Hunninghake, G.; Celli, B. BODE Collaborative Group. Comorbidities and risk of mortality in patients with chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2012, 186, 155−161. (4) Jensen, A. B.; Moseley, P. L.; Oprea, T. I.; Ellesøe, S. G.; Eriksson, R.; Schmock, H.; Jensen, P. B.; Jensen, L. J.; Brunak, S. Temporal disease trajectories condensed from population-wide registry data covering 6.2 million patients. Nat. Commun. 2014, 5, 4022. (5) Divo, M. J.; Celli, B. R.; Poblador-Plou, B.; Calderón-Larrañaga, A.; de-Torres, J. P.; Gimeno-Feliu, L. A.; Bertó, J.; Zulueta, J. J.; Casanova, C.; Pinto-Plata, V. M.; Cabrera-Lopez, C.; Polverino, F.; Carmona Píréz, J.; Prados-Torres, A.; Marin, J. M. Chronic obstructive pulmonary disease (COPD) as a disease of early aging: Evidence from the epichron cohort. PLoS One 2018, 13, No. e0193143. (6) Boschetto, P.; Quintavalle, S.; Miotto, D.; Lo Cascio, N.; Zeni, E.; Mapp, C. E. Chronic obstructive pulmonary disease (COPD) and occupational exposures. J. Occup. Med. Toxicol. 2006, 1, 11. (7) Biswas, S.; Hwang, J. W.; Kirkham, P. A.; Rahman, I. Pharmacological and dietary antioxidant therapies for chronic obstructive pulmonary disease. Curr. Med. Chem. 2013, 20, 1496− 1530. (8) Santos, D.; Meireles, M. A. Jabuticaba as a source of functional pigments. Pharmacogn. Rev. 2009, 3, 137−142. (9) Wu, S. B.; Long, C.; Kennelly, E. J. Phytochemistry and health benefits of jaboticaba, an emerging fruit crop from Brazil. Food Res. Int. 2013, 54, 148−159. (10) Wu, S. B.; Dastmalchi, K.; Long, C.; Kennelly, E. J. Metabolite profiling of jaboticaba (Myrciaria cauliflora) and other dark-colored fruit juices. J. Agric. Food Chem. 2012, 60, 7513−7525. (11) Reynertson, K. A.; Wallace, A. M.; Adachi, S.; Gil, R. R.; Yang, H.; Basile, M. J.; D’Armiento, J.; Weinstein, I. B.; Kennelly, E. J. Bioactive depsides and anthocyanins from jaboticaba (Myrciaria cauliflora). J. Nat. Prod. 2006, 69, 1228−1230. (12) Neri-Numa, I. A.; Soriano Sancho, R. A.; Pereira, A. P. A.; Pastore, G. M. Small brazilian wild fruits: Nutrients, bioactive compounds, health-promotion properties and commercial interest. Food Res. Int. 2018, 103, 345−360. (13) Hsu, J. D.; Wu, C. C.; Hung, C. N.; Wang, C. J.; Huang, H. P. Myrciaria caulif lora extract improves diabetic nephropathy via suppression of oxidative stress and inflammation in streptozotocinnicotinamide mice. J. Food. Drug. Anal. 2016, 24, 730−737.

SAE cells, and thus, jaboticaba may be useful for the treatment of COPD. We are currently exploring the anti-inflammatory potential of jaboticabin in vivo.



ASSOCIATED CONTENT

S Supporting Information *

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jafc.8b05814. IL-8 inhibition by recombined jaboticaba wood fractions at 100 μM/mL in SAE cells untreated (open bars) and treated (bold bars) with CSE (Figure S1) (PDF)



AUTHOR INFORMATION

Corresponding Author

*Telephone: +1-718-960-1105. Fax: +1-718-960-8236. E-mail: [email protected]. ORCID

Edward J. Kennelly: 0000-0002-1682-2696 Author Contributions †

Da-Ke Zhao and Ya-Na Shi contributed equally to this work.

Funding

This work was supported by the National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Grant 5SC1HL096016. Notes

The authors declare no competing financial interest.



ACKNOWLEDGMENTS The authors thank Chris Rollins and Eric McDonough for collecting jaboticaba wood and fruits from the Fruit and Spice Park (Homestead, FL, U.S.A.). The authors also thank Dr. Lin Li and Eric C. W. Wong (from the Institute of Chinese Medicine, The Chinese University of Hong Kong) for their technical support on Caco-2 transport study.



ABBREVIATIONS USED COPD, chronic obstructive pulmonary disease; MRM, multiple reaction monitoring; SAE, small airway epithelial; CSE, cigarette smoke extract; TEER, transepithelial electrical resistance



REFERENCES

(1) Mannino, D. M.; Buist, A. S. Global burden of copd: Risk factors, prevalence, and future trends. Lancet 2007, 370, 765−773. (2) Lozano, R.; Naghavi, M.; Foreman, K.; Lim, S.; Shibuya, K.; Aboyans, V.; Abraham, J.; Adair, T.; Aggarwal, R.; Ahn, S. Y.; Alvarado, M.; Anderson, H. R.; Anderson, L. M.; Andrews, K. G.; Atkinson, C.; Baddour, L. M.; Barker-Collo, S.; Bartels, D. H.; Bell, M. L.; Benjamin, E. J.; Bennett, D.; Bhalla, K.; Bikbov, B.; Bin Abdulhak, A.; Birbeck, G.; Blyth, F.; Bolliger, I.; Boufous, S.; Bucello, C.; Burch, M.; Burney, P.; Carapetis, J.; Chen, H.; Chou, D.; Chugh, S. S.; Coffeng, L. E.; Colan, S. D.; Colquhoun, S.; Colson, K. E.; Condon, J.; Connor, M. D.; Cooper, L. T.; Corriere, M.; Cortinovis, M.; de Vaccaro, K. C.; Couser, W.; Cowie, B. C.; Criqui, M. H.; Cross, M.; Dabhadkar, K. C.; Dahodwala, N.; De Leo, D.; Degenhardt, L.; Delossantos, A.; Denenberg, J.; Des Jarlais, D. C.; Dharmaratne, S. D.; Dorsey, E. R.; Driscoll, T.; Duber, H.; Ebel, B.; Erwin, P. J.; Espindola, P.; Ezzati, M.; Feigin, V.; Flaxman, A. D.; Forouzanfar, M. H.; Fowkes, F. G.; Franklin, R.; Fransen, M.; Freeman, M. K.; Gabriel, S. E.; Gakidou, E.; Gaspari, F.; Gillum, R. F.; Gonzalez-Medina, D.; Halasa, Y. A.; Haring, D.; Harrison, J. E.; Havmoeller, R.; Hay, R. J.; Hoen, B.; Hotez, P. J.; Hoy, D.; Jacobsen, K. H.; James, S. L.; Jasrasaria, R.; Jayaraman, S.; Johns, N.; Karthikeyan, G.; Kassebaum, N.; Keren, A.; G

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX

Article

Journal of Agricultural and Food Chemistry (14) Lobo de Andrade, D. M.; Reis, C. F.; Castro, P. F. S.; Borges, L. L.; Amaral, N. O.; Torres, I. M. S.; Rezende, S. G.; Gil, E. S.; Cardoso da Conceiçaõ , E.; Pedrino, G. R.; Lavorenti Rocha, M. Vasorelaxant and hypotensive effects of jaboticaba fruit (Myrciaria cauliflora) extract in rats. Evidence-Based Complementary Altern. Med. 2015, 2015, 1−8. (15) Akter, M. S.; Oh, S.; Eun, J. B.; Ahmed, M. Nutritional compositions and health promoting phytochemicals of camu-camu (Myrciaria dubia) fruit: A review. Food Res. Int. 2011, 44, 1728−1732. (16) Bailão, E.; Devilla, I.; da Conceiçaõ , E.; Borges, L. Bioactive compounds found in Brazilian cerrado fruits. Int. J. Mol. Sci. 2015, 16, 23760. (17) Dastmalchi, K.; Flores, G.; Wu, S. B.; Ma, C.; Dabo, A. J.; Whalen, K.; Reynertson, K. A.; Foronjy, R. F.; D’Armiento, J. M.; Kennelly, E. J. Edible Myrciaria vexator fruits: Bioactive phenolics for potential COPD therapy. Bioorg. Med. Chem. 2012, 20, 4549−4555. (18) D’Armiento, J.; Reynertson, K.; Kennelly, E.; Wallace, A. Bioactive depside and anthocyanin compounds, compositions, and methods of use. U.S. Patent 20110110885 A1, May 12, 2011. (19) Wu, S.; Wu, J.; Yin, Z.; Zhang, J.; Long, C.; Kennelly, E.; Zheng, S. Bioactive and marker compounds from two edible darkcolored Myrciaria fruits and the synthesis of jaboticabin. J. Agric. Food Chem. 2013, 61, 4035−4043. (20) Wang, W. H.; Tyan, Y. C.; Chen, Z. S.; Lin, C.-G.; Yang, M. H.; Yuan, S. S.; Tsai, W.-C. Evaluation of the antioxidant activity and antiproliferative effect of the jaboticaba (Myrciaria cauliflora) seed extracts in oral carcinoma cells. BioMed Res. Int. 2014, 2014, 1−7. (21) Morales, P.; Barros, L.; Dias, M. I.; Santos-Buelga, C.; Ferreira, I. C. F. R.; Ramirez Asquieri, E.; Berrios, J. D. J. Non-fermented and fermented jabuticaba (Myrciaria cauliflora mart) pomaces as valuable sources of functional ingredients. Food Chem. 2016, 208, 220−227. (22) Wu, S. B.; Yue, G.; To, M.-H.; Keller, A.; Lau, C.; Kennelly, E. Transport in Caco-2 cell monolayers of antidiabetic cucurbitane triterpenoids from Momordica charantia fruits. Planta Med. 2014, 80, 907−911. (23) Artursson, P.; Karlsson, J. Correlation between oral drug absorption in humans and apparent drug permeability coefficients in human intestinal epithelial (Caco-2) cells. Biochem. Biophys. Res. Commun. 1991, 175, 880−885. (24) König, M.; Scholz, E.; Hartmann, R.; Lehmann, W.; Rimpler, H. Ellagitannins and complex tannins from quercus petraea bark. J. Nat. Prod. 1994, 57, 1411. (25) Jang, M.; Cai, L.; Udeani, G. O.; Slowing, K. V.; Thomas, C. F.; Beecher, C. W. W.; Fong, H. H. S.; Farnsworth, N. R.; Kinghorn, A. D.; Mehta, R. G.; Moon, R. C.; Pezzuto, J. M. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997, 275, 218−220. (26) Lenquiste, S. A.; Marineli, R. D. S.; Moraes, É .A.; Dionísio, A. P.; Brito, E. S. D.; Maróstica, M. R. Jaboticaba peel and jaboticaba peel aqueous extract shows in vitro and in vivo antioxidant properties in obesity model. Food Res. Int. 2015, 77, 162−170. (27) Plaza, M.; Batista, Â .G.; Cazarin, C. B. B.; Sandahl, M.; Turner, C.; Ö stman, E.; Maróstica Júnior, M. R. Characterization of antioxidant polyphenols from Myrciaria jaboticaba peel and their effects on glucose metabolism and antioxidant status: A pilot clinical study. Food Chem. 2016, 211, 185−197. (28) Borges, L. L.; Conceiçaõ , E. C.; Silveira, D. Active compounds and medicinal properties of Myrciaria genus. Food Chem. 2014, 153, 224−233. (29) Fracassetti, D.; Costa, C.; Moulay, L.; Tomás-Barberán, F. A. Ellagic acid derivatives, ellagitannins, proanthocyanidins and other phenolics, vitamin c and antioxidant capacity of two powder products from camu-camu fruit (Myrciaria dubia). Food Chem. 2013, 139, 578− 588. (30) Shi, X. H.; Xu, D. R.; Kong, L. Y. Chemical constituents in leaves of Euphorbia soongarica. Chin. Tradit. Herb. Drugs 2009, 40, 686−689.

(31) Koenig, M.; Scholz, E.; Hartmann, R.; Lehmann, W.; Rimpler, H. Ellagitannins and complex tannins from Quercus petraea bark. J. Nat. Prod. 1994, 57, 1411−1415. (32) Stark, T.; Wollmann, N.; Wenker, K.; Lösch, S.; Glabasnia, A.; Hofmann, T. Matrix-calibrated LC-MS/MS quantitation and sensory evaluation of oak ellagitannins and their transformation products in red wines. J. Agric. Food Chem. 2010, 58, 6360−6369.

H

DOI: 10.1021/acs.jafc.8b05814 J. Agric. Food Chem. XXXX, XXX, XXX−XXX