Next Article in Journal
Stereoselective Processes Based on σ-Hole Interactions
Next Article in Special Issue
Transcriptome Analysis of the Anti-Proliferative Effects of Ginsenoside Rh3 on HCT116 Colorectal Cancer Cells
Previous Article in Journal
Halogen-Doped Carbon Dots: Synthesis, Application, and Prospects
Previous Article in Special Issue
Antioxidant Interactions between S-allyl-L-cysteine and Polyphenols Using Interaction Index and Isobolographic Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Polyphenols from the Peels of Punica granatum L. and Their Bioactivity of Suppressing Lipopolysaccharide-Stimulated Inflammatory Cytokines and Mediators in RAW 264.7 Cells via Activating p38 MAPK and NF-κB Signaling Pathways

1
State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
2
Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(14), 4622; https://doi.org/10.3390/molecules27144622
Submission received: 1 July 2022 / Revised: 15 July 2022 / Accepted: 16 July 2022 / Published: 20 July 2022
(This article belongs to the Special Issue Functional and Bioactive Properties of Foods and Natural Products)

Abstract

:
Punica granatum L. (Punicaceae) is a popular fruit all over the world. Owning to its enriched polyphenols, P. granatum has been widely used in treating inflammation-related diseases, such as cardiovascular diseases and cancer. Twenty polyphenols, containing nine unreported ones, named punicagranins A–I (19), along with eleven known isolates (1020), were obtained from the peels. Their detailed structures were elucidated based on UV, IR, NMR, MS, optical rotation, ECD analyses and chemical evidence. The potential anti-inflammatory activities of all polyphenols were examined on a lipopolysaccharide (LPS)-induced inflammatory macrophages model, which indicated that enhancing nitric oxide (NO) production in response to inflammation stimulated in RAW 264.7 cells was controlled by compounds 1, 3, 58, 10, 11, 14 and 1620 in a concentration-dependent manner. The investigation of structure–activity relationships for tannins 68 and 1220 suggested that HHDP, flavogallonyl and/or gallagyl were key groups for NO production inhibitory activity. Western blotting indicated that compounds 68 could down-regulate the phosphorylation levels of proteins p38 MAPK, IKKα/β, IκBα and NF-κB p65 as well as inhibit the levels of inflammation-related cytokines and mediators, such as IL-6, TNF-α, iNOS and COX-2, at the concentration of 30 μM. In conclusion, polyphenols are proposed to be the potential anti-inflammatory active ingredients in P. granatum peels, and their molecular mechanism is likely related to the regulation of the p38 MAPK and NF-κB signaling pathways.

1. Introduction

Though moderate inflammation is a protective immune response against micro-organism invasion or damage, a pathological inflammation is the product of an uncontrolled of immune balance leading to chronic diseases, including inflammatory bowel disease, cardiovascular diseases, allergy, type 2 diabetes or even some kinds of cancer [1]. In the view of over-expression of pro-inflammatory cytokines and mediators lead to inflammatory response, this is a useful way to suppress their productions to avoid inflammation.
On the other hand, the pro-inflammatory cytokines and mediators are regulated by a large amount of signaling pathways, which indicates that selectively inhibiting the key target of corresponding signaling pathway may be a promising therapeutic strategy [2]. Macrophages, distributing throughout the body, can be activated by external stimuli, such as lipopolysaccharides (LPS—a major component of the outer membrane of Gram-negative bacteria).
As an endotoxin, LPS can activate the nuclear transcription factor kappa-B (NF-κB) pathway through LPS-binding protein (LBP), myeloid differentiation receptor (CD14) and Toll-like receptor 4 (TLR4) receptors, promote the release of inflammatory factors and induce a series of inflammatory reactions [3]. They will be recruited to inflammatory sites and will release cascades of inflammatory molecules, thereby, producing a variety of inflammatory cytokines, including interleukin (IL)-6 and tumor necrosis factor (TNF)-α, and inflammatory mediators, such as nitric oxide (NO), prostaglandin E2 (PGE2) and inducible cyclooxygenase-2 (COX-2), leading to severe damage at the inflamed sites by amplifying the inflammation response [4].
In recent years, greater attention has been paid to developing natural functional compounds or herbal plant extracts to prevent and treat inflammatory disorders or, at least, relieve its symptoms [5]. Polyphenols are usually found in vegetables, fruits, beverages cereals, etc. The long-term intake of diets rich in polyphenols can protect against gastrointestinal tract, arthritis, cancer and cardiovascular disease because of their anti-inflammatory properties to [2,6].
Punica granatum L. belongs to the Punicaceae family, is originated from central Asia and is now widely planted all around the world, such as India, Iran, America and the Mediterranean regions due to its strong adaptability to different climates and soil conditions [7]. P. granatum, famous for super-fruit, has been widely used for centuries in folk medicine for the treatment of various inflammation-related diseases, including cardiovascular diseases and cancer, owning to a high content in polyphenols, particularly ellagitannins [2,7].
The nonedible part, namely the peels, is an excellent source of polyphenols that are more abundant in specific medicinal and nutritional value compared with the edible part of the fruit [8], which was reported to have anti-oxidant, anti-inflammatory, anti-hyperglycemia; anti-hypertension, reduce body weight, anti-cancer and other activities [2,9,10,11]. However, its effective substances are not totally clear, and little is known about the anti-inflammatory mechanism.
Therefore, the phytochemistry investigation of polyphenols from P. granatum peels was developed. An LPS-treated RAW 264.7 macrophages model was established to evaluate the anti-inflammatory activity of polyphenols by measuring the NO level. Furthermore, the signaling pathways of compounds exerted anti-inflammatory effect were also studied through a western blot assay.

2. Results and Discussion

2.1. Structural Characterization

The 70% ethanol extraction of P. granatum peels were fractionated with various of materials for chromatographic separation, including D101 resin, silica gel, ODS and Sephadex LH-20 column chromatographies (CC). Then, the obtained fractions were purified by preparative high-performance liquid chromatography (pHPLC) to obtain polyphenols. The structures of gained polyphenols were identified by using kinds of spectroscopic methods, such as UV, IR, NMR, [α]D, MS, as well as electronic circular dichroism (ECD) spectra. The results were nine unreported polyphenols, named punicagranins A–I (19), together with eleven known isolates, brevifolin (10) [12], brevifolincarboxylic acid (11) [13], 4-O-α-l-rhamnopyranosyl ellagic acid (12) [14], 4-O-β-d-glucopyranosyl-3,3′-di-O-methylellagic acid (13) [15], (S)-flavogallonic acid (14) [16], valoneic acid dilactone (15) [17], corilagin (16) [18], 2,3-(S)-hexahydroxydiphenoyl-d-glucose (17) [19], punicalin (18) [20], punicalagin (19) [20] and punicacortein C (20) [21] (Figure 1) were obtained and identified.
Punicagranin A (1) was obtained as a white powder with molecular formula, C12H8O8 established on negative-ion HRESIMS analysis. Its IR spectrum exhibited characteristic absorptions accounting for hydroxyl (3171 cm−1), carbonyl (1790, 1715 cm−1) and aromatic ring (1608, 1551 cm−1). The 1H, 13C NMR (DMSO-d6) and HSQC spectra indicated the existences of one methyl [δ 2.14 (3H, s, H3-6)], one aromatic proton [δ 7.17 (1H, s, H-5′)] and ten quaternary carbons.
The presence of gallic acid fragment was speculated by the correlations from δH 7.17 (H-5′) to δC 108.0 (C-1′), 121.5 (C-6′), 134.5 (C-3′), 146.4 (C-4′), 167.7 (C-7′). In addition, the cross peaks from δH 2.14 (H3-6) to δC 108.0 (C-1′), 119.7 (C-4), 142.2 (C-3), 160.8 (C-5), 166.3 (C-2) were also observed in its HMBC spectrum (Figure 2). Consequently, the structure of punicagranin A (1) was elucidated to be 3-(2,3,4-trihydroxy-4-carboxyphenyl)-4-methylfuran-2,5-dione.
The HRESIMS determining result revealed the molecular formula of punicagranin B (2) was C13H10O8. Comparing its 1H and 13C NMR with compound 1, it was found that one more methoxyl [δH 3.70 (3H, s, 7′-COOCH3); δC 51.3 (7-COOCH3)] appeared in compound 2. The 13C NMR signal of C-7′ was decreased 0.8 [δC 167.7 (C-7′) for 1; 166.9 (C-7′) for 2]. In combination with the correlation from δH 3.70 (3H, s, 7′-COOCH3) to δC 166.9 (C-7′) (Figure 2), punicagranin B (2) was deduced to be punicagranin A 7′-methyl ester.
Punicagranin C (3) was obtained as a white powder with negative optical rotation ([α]D25 −7.0). HRESIMS spectrum revealed its molecular formula was C12H10O8. The 1H NMR signal at δ 6.21 (1H, br. s, H-5), 7.79 (1H, br. s, H-4) and 13C NMR signal at δ 71.6 (C-5), 127.4 (C-3), 153.3 (C-4), 171.2 (C-2), along with the HMBC correlations from δH 7.79 (H-4) to δC 71.6 (C-5), 171.2 (C-2); δH 6.21 (H-5) to δC 127.4 (C-3), 171.2 (C-2) confirmed the presence of α,β-unsaturated-γ-lactone. It was similar to compound 1, 1H, 13C NMR and 2D NMR spectra (Figure 2) suggested one 2-substituted gallic acid was present in compound 3. Moreover, the 1H NMR spectrum suggested the existence of one hydroxymethyl [δ 4.94, 5.02 (1H each, both d, J = 19.0 Hz, H2-6)].
Then, α,β-unsaturated-γ-lactone, 2-substituted gallic acid and hydroxymethyl were grouped together according to the correlations from δH 6.21 (H-5) to δC 115.6 (C-6′), 125.8 (C-1′), 139.9 (C-2′); δH 4.94, 5.01 (H2-6) to δC 127.4 (C-3), 153.3 (C-4), 171.2 (C-2). Finally, the absolute configuration of 5R was clarified by the positive Cotton effect at 223 nm displayed in its ECD spectrum [22].
Punicagranin D (4) was a pale yellow powder with molecular formula of C15H12O9 deduced from HRESIMS analysis. The thin layer chromatography (TLC) of it showed blue-purple fluorescent spots under the UV lamp of 365 nm before and after spraying with 10% aqueous H2SO4-EtOH. Additionally, its UV spectrum displayed the maximum absorption peaks at 274 and 344 nm. Then, punicagranin D (4) was speculated to be a coumarin.
Its 1H, 13C NMR and HSQC spectra suggested the presence of one ethyoxyl [δ 1.18 (3H, t, J = 7.0 Hz, H3-16), 4.09 (2H, q, J = 7.0 Hz, H2-15)], one methylene [δ 3.63 (2H, s, H2-11)], one oxygenated methine [δ 6.64 (1H, s, H-13)] and ten quaternary carbons. The long-range correlations from δH 7.42 (1H, s, H-6) to δC 109.6 (C-10), 109.8 (C-5), 139.6 (C-8), 149.7 (C-7), 161.2 (C-14) indicated the presence of galloyl.
Its planar structure was clarified by the cross peaks from δH 3.63 (H2-11) to δC 116.6 (C-3), 141.8 (C-4), 160.7 (C-2), 169.2 (C-12); δH 6.64 (1H, s, H-13) to δC 109.6 (C-10), 116.6 (C-3), 141.8 (C-4), 161.2 (C-14); δH 4.09 (H2-15) to δC 169.2 (C-12) observed in the HMBC spectrum (Figure 2). Its ECD spectrum displayed positive Cotton effects at 222 nm, which was similar to that of (S)-euphorhirtin H [23]. Then, its absolute configuration, 13R was revealed.
The molecular formula of punicagranin E (5) was determined to be C13H12O6 by HRESIMS. Its UV absorption (λmax: 266 and 328 nm) and the similarity phenomenon displayed in TLC indicated it was also a coumarin. Thirteen carbon signals appeared in its 13C NMR spectrum. Combining with its 1H NMR and HSQC spectra, the existence of two aromatic protons [δ 6.75, 6.84 (1H each, both d, J = 8.5 Hz, H-5 and 6)], one methyl [δ 2.01 (3H, s, H3-11)], one ethyoxyl [δ 1.35 (3H, t, J = 7.0 Hz, H3-14), 4.46 (2H, q, J = 7.0 Hz, H2-13)], two unsaturated carboxyl [δC 161.0 (C-2), 165.1 (C-12)], as well as six aromatic quaternary carbon were consolidated. In the HMBC spectrum, the correlations from δH 6.75 (H-5) to δC 108.7 (C-10), 142.3 (C-9), 142.5 (C-4), 149.0 (C-7); δH 6.84 (H-6) to δC 108.7 (C-10), 132.3 (C-8); δH 2.01 (H3-11) to δC 116.7 (C-3), 142.5 (C-4), 161.0 (C-2); δH 4.46 (H2-13) to δC 165.1 (C-12) were observed. Then, the structure of punicagranin E (5) was elucidated.
Punicagranin F (6) was measured on HRESIMS to give the molecular formular, C23H24O13. There was one ethyoxyl signals showed in its 1H NMR spectrum at δH 0.87 (3H, t, J = 7.0 Hz, 7″-OCH2CH3), 3.86 (2H, q, J = 7.0 Hz, 7″-OCH2CH3) and in its 13C NMR spectrum at δC 13.6 (7″-OCH2CH3), 59.2 (7″-OCH2CH3). Twenty-three carbon signals were observed in its 13C NMR spectrum, and twenty-one of them were in the field of 107–166 ppm. In particularly, carbonyl signal at δC 157.4 (C-7′), 159.2 (C-7), 165.9 (C-7″) suggested the presence of three gallic acid moieties. However, there were only two aromatic protons [δH 7.11 (1H, s, H-6″), 7.50 (1H, s, H-5)] displayed in its 1H NMR spectrum.
The above-mentioned information indicated that punicagranin F (6) was a tannin formed by the condensation of three molecules of gallic acid. The long-range correlations from δH 7.50 (1H, s, H-5) to δC 107.1 (C-6), 112.6 (C-1), 139.4 (C-3), 147.7 (C-4), 159.2 (C-7); δH 7.11 (1H, s, H-6″) to δC 117.9 (C-2″), 119.2 (C-6′), 125.3 (C-1″), 137.2 (C-4″), 143.9 (C-5″), 165.9 (C-7″), along with the comparison of its spectroscopic data with those of compound 14, the presence of flavogallonyl moiety was clarified. Moreover, according to the cross peak from δH 3.86 (7″-OCH2CH3) to δC 165.9 (C-7″), the substituted position of ethyoxyl was consolidated.
Finally, its Cotton effects displayed at 326 nm (positive), 298 nm (negative) and 255 nm (negative) were consistent with those of methyl (S)-heptamethylflavogallonate [24], suggesting the chirality of the flavogallonyl group was in the S-series. Moreover, its opposite optical rotation [([α]D25 −15.0, in MeOH)] with that of (R)-flavogallonic acid [([α]D25 +20.3, in MeOH)] [25] further clarified the conclusion. Then, the structure of punicagranin F (6) was elucidated to be ethyl (S)-flavogallonate. Though the plannar structure of it had been speculated by Abiodun et al. through LC-MS analysis [26], the absolute configuration of it was expounded here for the first time.
Punicagranin G (7) was obtained as yellow powder with negative optical rotation ([α]D25 −73.9, in H2O). Its molecular formular, C34H22O22, was established by HRESIMS. However, sixty-eight signals displayed in its 13C NMR spectrum. The 1H and 13C NMR spectra of 7 showed duplicated signals for the sugar and polyphenol moieties, indicating an equilibrium mixture of α and β anomeric forms with the ratio about 1:2 in solution. After hydrolysis with 1 M HCl, d-glucose was detected from its hydrolysate [27]. The cross-peaks between δH 2.46 (1H, dd, J = 3.6, 9.6 Hz, Glc-H-2α) and δH 4.81 (1H, d, J = 3.6 Hz, Glc-H-1α), 4.83 (1H, dd, J = 9.6, 10.0 Hz, Glc-H-3α); δH 1.75 (1H, dd, J = 10.0, 10.0 Hz, Glc-H-4α) and δH 3.55 (1H, m, Glc-H-5α), 4.83 (Glc-H-3α); δH 3.55 (Glc-H-5α) to δH [3.55 (1H, br. d, ca. J = 12 Hz), 4.67 (1H, dd, J = 3.2, 12.0 Hz), Glc-H2-6α] consolidated the α-d-glucopyranosyl.
The presence of β-d-glucopyranosyl was clarified by the correlations between δH 2.28 (1H, dd, J = 7.6, 9.2 Hz, Glc-H-2β) and δH 4.25 (1H, d, J = 7.6 Hz, Glc-H-1β), 4.57 (1H, dd, J = 9.2, 9.6 Hz, Glc-H-3β); δH 1.79 (1H, dd, J = 9.6, 9.6 Hz, Glc-H-4β) and δH 3.03 (1H, m, Glc-H-5β), 4.57 (Glc-H-3β); δH 3.03 (Glc-H-5β) and δH [3.53 (1H, br. d, ca. J = 12 Hz), 4.75 (1H, dd, J = 3.2, 12.4 Hz), Glc-H2-6β]. Except for the six carbon signals assigned to d-glucose, all of the twenty-eight duplicated carbon signals were in the low field.
The four pairs of carbonyl carbon signals at δC 160.0, 160.8 (lactone carbonyl-C-α), 169.1 (C-7′α), 170.7 (C-7″α); 159.9, 160.8 (lactone carbonyl-C-β), 169.0 (C-7′β), 170.7 (C-7″β) together with twenty-four duplicated aromatic carbon signals in the field of 108–161 ppm suggested the presence of four galloyl groups. The presence of (S,S)-gallagyl was confirmed by the comparison of its NMR data (in CD3OD), optical rotation ([α]D25 −73.9, in H2O) as well as ECD spectrum (Figure S50, Supplementary Materials) with those of punicalin (4,6-(S,S)-gallagyl-d-glucose (18) [20,28], whose 1H, 13NMR data, [α]D25 and ECD spectrum (Figure S69, Supplementary Materials) could be found in the supporting information.
Moreover, the correlations from δH 4.83 (Glc-H-3α) to δC 169.1 (C-7′α); δH 4.57 (Glc-H-3β) to δC 169.0 (C-7′β); δH 3.55, 4.67 (Glc-H2-6α) to δC 170.7 (C-7″α); δH 3.53, 4.75 (Glc-H2-6β) to δC 170.7 (C-7″β) indicated the two ester carbonyl groups in gallagyl were linked with C-3 and C-6 of d-glucosyl. Thus, the structure of punicagranin G (7) was determined to be 3,6-(S,S)-gallagyl-d-glucose.
Punicagranin H (8), [α]D25 −85.1 (in H2O). The HRESIMS revealed it had the same molecular formular, C34H22O22 as compound 7. d-fructose was yielded from compound 8 after acid hydrolysis [27]. Its 1H and 13C NMR spectra suggested that it also contained (S,S)-gallagyl [δC 160.3, 160.5 (lactone carbonyl-C-β); 160.2, 160.4 (lactone carbonyl-C-α), 169.3, 169.7 (ester carbonyl-C-β); 169.4, 170.0 (ester carbonyl-C-α)].
d-glucosyl disappeared, and d-fructofuranosyl appeared in compound 8, which was clarified by the cross peaks showed in the 1H 1H COSY spectrum, as well as the correlations from H2-1 to C-2, C-3 displayed in its HMBC spectrum (Figure 2). The correlations from H-4 to C-7′; H2-6 to C-7″ and the ECD similarity between it and compound 7 suggested the structure of punicagranin H (8) was 4,6-(S,S)-gallagyl-d-fructofuranose. It was a mixture of 4,6-(S,S)-gallagyl-α-d-fructofuranose and 4,6-(S,S)-gallagyl-β-d-fructofuranose with the ratio 1:3 nearly.
Punicagranin I (9) was obtained as a white powder. HRESIMS analysis showed its molecular formular was C22H24O12. Its IR spectrum displayed characteristic absorptions assigned to hydroxyl (3318 cm−1), α,β-unsaturated ketone (1718 cm−1), aromatic ring (1606, 1512, 1457 cm−1) and ether function (1076 cm−1). After hydrolyzing with 1 M HCl, the product of compound 9 was analyzed by HPLC combining with an optical rotation detector.
As a result, d-glucose was detected [27]. The 1H, 13C NMR and 2D NMR (1H 1H COSY, HSQC and HMBC) spectra indicated the existence of one AA′BB′ spin-coupled benzene ring [δ 6.90 (2H, d, J = 8.5 Hz, H-3′,5′), 7.98 (2H, d, J = 8.5 Hz, H-2′,6′)], one 1,2,3,5-tetra substituted benzene ring [δ 6.37, 6.65 (1H each, both d, J = 2.5 Hz, H-3 and 5)], one methylene [δ 3.60, 3.66 (1H each, both d, J = 16.5 Hz, H2-7)], two carboxyl [δC 166.0 (C-7′), 174.3 (C-8)], one methoxy group [δ 3.53 (3H, s, 8-COOCH3)] and one β-d-glucopyranosyl [δ 4.86 (1H, d, J = 7.5 Hz, H-1″)]. Finally, the long-range correlations were found from δH 3.60, 3.66 (H2-7) to δC 111.0 (C-1), 152.1 (C-2), 158.5 (C-6), 174.3 (C-8); δH 3.53 (8-COOCH3) to δC 174.3 (C-8); δH 7.98 (H-2′,6′) to δC 166.0 (C-7′); δH 4.86 (H-1″) to 158.5 (C-6) in its HMBC spectrum. Consequently, the structure of punicagranin I (9) was clarified.
The structures of the known compounds 1020 were identified by comparing the spectroscopic data with those reported in literature. Among all of the obtained compounds, 68 and 1220 were tannins.
To clarify whether compounds 2, 46 and 9 were artificial products, a 70% ethanol extract and 70% methanol extract of P. granatum peels, together with the reference compounds of 2, 46 and 9 were analyzed by LC-MS (Figure S65, Supplementary Materials). As compounds 6 and 9 could not be detected in the 70% MeOH and 70% EtOH extracts, respectively, they were elucidated to be artificial products. Compound 6 might be produced by refluxing with ethanol in the process of P. granatum peel extraction. Compound 9 might be the reaction product of the sample and methanol used as the mobile phase.

2.2. In Vitro Anti-Inflammatory Evaluation of Compounds 120

As an excess of NO can trigger severe damage at the inflamed sites by amplifying the inflammation response [4] and exploiting new strategies to treat the over production; thus, the release of NO is important and urgent. The LPS-induced RAW 264.7 macrophages model was established to evaluate the potential anti-inflammatory activities of the twenty polyphenols (120) by measuring the NO level as what has been reported [29].
In order to test the NO release inhibitory effects of all compounds at a safe concentration, a MTT assay was conducted first. The results showed that at the concentration of 30 μM, all compounds showed no cytotoxic on RAW 264.7 cell, except compound 19, which showed no toxic until 15 μM (Figure S72, Supplementary Materials). Thus, in the in vitro anti-inflammatory assay, the concentration for compounds 118, 20 and 19 were determined to be 30 μM and 15 μM, respectively. The results were compounds 1, 3, 58, 10, 11, 14 and 1620 displayed significant inhibitory effects on the NO release levels in LPS-activated RAW 264.7 cells (Table 1) at a non-toxic concentration.
It was found that compounds 1, 3, 58, 10, 11, 14, 1618 and 20 reduced NO production in a concentration-dependent manner at 3, 10 and 30 μM, while compound 19 showed a similar mode at 1, 5 and 15 μM (Figure 3).
By comparing their bioactivities, the structure–activity relationships (SARs) can be summarized:
(1)
Tannins containing gallagyl (68 and 1820) had good inhibitory activity on NO release production. Compound 19 displayed stronger activity than 17. Both of the results suggested gallagyl group may be a positive factor for NO inhibitory activity.
(2)
The HHDP group in tannins also played an important role in inhibiting NO release (18 vs. 19).
(3)
The activities of flavogallonic acid derivatives formed by the condensation of ellagic acid and gallic acid through carbon-carbon bonds were significantly stronger than those of valoneic acid derivatives produced by the condensation of ellagic acid and gallic acid through carbon-oxygen-carbon bond (6 and 14 vs. 15).
(4)
Moreover, it was found that ethyl or methyl esterification of carboxyl group could affect the activity slightly only (2 vs. 1 and 6 vs. 14).
(5)
Ellaganosides (12 and 13) showed no activity, which might be related to the introduction of sugar groups into ellagic acid (Table 1 and Figure 3).
This summary may provide more evidence for the structural modification of this type of polyphenols in the development of potential anti-inflammatory drugs.

2.3. Polyphenols from P. granatum Peels Exerted Anti-Inflammatory Effects through p38 MAPK and NF-κB Signaling Pathway

Moreover, the anti-inflammatory mechanism of compounds 68 at a concentration of 30 μM in LPS-induced RAW 264.7 macrophages model was investigated by western blot analyses. Comparing with the normal group, increased p38 mitogen-activated protein kinase (MAPK), IκB kinase (IKK)α/β, inhibitory nuclear factor κB-α (IκBα) and NF-κB p65 phosphorylation were observed in the model group.
Then, it was found that compounds 68 could down-regulate the phosphorylation levels of above-mentioned proteins. The expression levels of inflammation-related cytokines and mediators, such as IL-6, TNF-α, iNOS and COX-2 were down-regulated compared with the model group (Figure 4). The obtained findings also revealed that compound 8 owned the best anti-inflammatory activities, either in inhibiting NO production or iNOS expression, as well as down-regulating the IL-6 level in the LPS-treated RAW 264.7 cells.
p38 MAPK is one major member of MAPKs. As upstream modulators of inflammatory molecules, MAPKs have been considered to play critical role in the regulation of inflammatory mediators and cytokines, such as COX-2, inducible nitric oxide synthase (iNOS) in macrophage cells [30]. The p38 MAPK pathway, plays an important role in inflammation in particularly [31].
In addition, NF-κB, consists of subunits p50, p52, c-Rel, Rel A (p65) and Rel B, is normally interacted with IκBα proteins in the cytoplasm [32,33]. MAPKs, as one of the main regulatory factors of NF-κB, can up-regulate the expression of IKK, induce the phosphorylation and degradation of IκBα [34], thus, causing NF-κB p50 or p65 released to promote the transcription of pro-inflammatory genes, such as COX-2, iNOS, TNF-α, IL-6 and IL-1β [33,35,36]. Hence, p38 MAPK and NF-κB pathways have been regarded as potentially molecular targets for anti-inflammatory therapy.
According to the above-mentioned evidence, compounds 68 were confirmed to conduct anti-inflammatory activities by inhibiting p38 MAPK and NF-κB signaling pathways.

3. Conclusions

In summary, nine unreported polyphenols named punicagranins A–I (19), along with eleven known isolates (1020) were gained from P. granatum peels. New compounds 6 and 9 were elucidated to be artificial products by LC-MS analysis. The potential anti-inflammatory activities of all the polyphenols were examined on an LPS-induced inflammatory macrophages model. It was found that compounds 1, 3, 58, 10, 11, 14 and 1620 reduced the LPS-induced secretion of NO in a concentration-dependent manner. The SARs analysis for tannins 68 and 1220 suggested the introduction of HHDP, flavogallonyl and gallagyl played central roles in the NO inhibitory activity. Moreover, the new compounds 68 were found to suppress the expression of inflammatory cytokines and mediators, such as IL-6, TNF-α, iNOS and COX-2, through activating the p38 MAPK and NF-κB signaling pathways (Figure 5).
As described in the literature, corilagin (16) could restrict M1 macrophage activation by regulating the MAPK, NF-κB and interferon regulatory factor signaling pathways, resulting in a reduction in the expression of pro-inflammatory cytokines, such as IL-6, IL-12 and iNOS [37]. In addition, punicalin (18) reduced the release of inflammatory factors, including IL-1β and IL-18, via the ROS/NLRP3 pathway to exert anti-inflammatory function [38]. Punicalagin (19) could inhibit ERK/MAPK and JAK/SAT pathways in LPS-stimulated mouse primary peritoneal macrophages [39].
Combined with our investigation, polyphenols are proposed to be the potential anti-inflammatory active ingredients in P. granatum peels.

4. Experimental

4.1. Materials and Methods for Phytochemistry Research

4.1.1. General Experimental Procedures

NMR spectra were determined on a Varian 400 MR spectrometer and/or 500 MHz NMR (Bruker BioSpin AG Industriestrasse 26 CH-8117, Fällanden, Switzerland) spectrometer with tetramethylsilane (TMS) as an internal standard. Negative-ion or positive-ion mode ESI-Q-Orbitrap MS was acquired using a Thermo ESI-Q-Orbitrap MS mass spectrometer connected to the UltiMate 3000 UHPLC instrument via ESI interface (Thermo, Waltham, MA, USA). Optical rotations, UV and IR spectra were determined on a Rudolph Autopol® IV automatic polarimeter (l = 50 mm) (Rudolph Research Analytical, Hackettstown, NJ, USA), Varian Cary 50 UV-Vis (Varian, Inc., Hubbardston, MA, USA) and Varian 640-IR FT-IR spectrophotometer (Varian Australia Pty Ltd., Mulgrave, Australia), respectively.
CC were conducted on macroporous resin D101 (Haiguang Chemical Co., Ltd., Tianjin, China), silica gel (48–75 μm, Qingdao Haiyang Chemical Co., Ltd., Qingdao, China), ODS (40–75 µm, Chromatorex ODS MB, Fuji Silisia Chemical Co., Ltd., Tokyo, Japan) and Sephadex LH-20 (Ge Healthcare Bio-Sciences, Uppsala, Sweden). HPLC column: Cosmosil 5C18-MS-II column (4.6 mm i.d. × 250 mm, 5 µm, Nakalai Tesque, Inc., Tokyo, Japan) and Cosmosil 5C18-MS-II column (20 mm i.d. × 250 mm, 5 µm, Nakalai Tesque, Inc., Tokyo, Japan) were used to analysis and prepare the constituents, respectively. Precoated silica gel GF254 plates (10 cm × 20 cm, 0.25 mm thickness, Tianjin Silida Technology Co., Ltd., Tianjin, China) were used in TLC, and spots were detected under UV lights (254 and 365 nm) or by using the 10% sulfuric acid reagent.
Chloroform (CHCl3), methanol (MeOH), acetonitrile (CH3CN), and acetic acid (HAc) (chromatographically pure or analytical pure) were purchased from (Tianjin Concord Technology Co., Ltd., Tianjin, China).

4.1.2. Plant Material

The medicinal herbs of the dried P. granatum peels were purchased from Beijing Tong ren Tang drug store, whose origin is from Anqing city, Anhui province, China. Then, the medicinal herbs were identified by Professor Lin Ma according to their characters (School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine). The voucher specimen was deposited at the Academy of Traditional Chinese Medicine of Tianjin University of TCM.

4.1.3. Extraction and Isolation

The dried peels of P. granatum (9.0 kg) were extracted three times with 70% EtOH under reflux for 3, 2 and 2 h, successively. Evaporation of the solvent under reduced pressure provided the 70% EtOH extract (2450.0 g). Then, an aliquot (1.63 kg) of it was dissolved in water and subjected to D101 resin CC (H2O → 95% EtOH) to obtain H2O (1034.0 g) and 95% EtOH (525.0 g) eluates, respectively.
The 95% EtOH eluate (PGE, 400.0 g) was subjected to silica gel CC [CHCl3-MeOH (100:5, v/v) → CHCl3-MeOH-H2O (10:3:1 → 7:3:1 → 6:4:1, v/v/v, lower layer) → MeOH] to give PGE 1–PGE 10. PGE 3 (1.2 g) was prepared by pHPLC [MeOH-H2O (30:70 → 40:60 → 55:45 → 60:40 → 100:0, v/v)] to obtain PGE 3-1–PGE 3-15. PGE 3-11 (52.7 mg) was purified by pHPLC [CH3CN-1% HAc (25:75, v/v)] to gain punicagranin E (5, 5.3 mg).
PGE 6 (13.6 g) was separated by ODS CC [MeOH-H2O (20:80 → 30:70 → 40:60 → 50:50 → 60:40 → 70:30 → 80:20 → 100:0, v/v)] and PGE 6-1–PGE 6-13 were given. PGE 6-2 (1.0 g) was isolated by Sephadex LH-20 CC (MeOH) and pHPLC [CH3CN-1% HAc (7:93, v/v)] to yield punicagranins A (1, 16.4 mg), B (2, 17.5 mg) and C (3, 284.8 mg). PGE 6-4 (2.7 g) was fractionated by Sephadex LH-20 CC (MeOH) to obtain PGE 6-4-1–PGE 6-4-13.
PGE 6-4-5 (260.1 mg) was purified by pHPLC [CH3CN-H2O (18:82, v/v)] and [MeOH-1% HAc (32:68, v/v)] to produce punicagranin D (4, 7.0 mg). PGE 6-4-10 (143.4 mg) was prepared by pHPLC [CH3CN-1% HAc (16:84, v/v)] to gain punicagranin F (6, 35.0 mg). PGE 7 (12.8 g) was subjected to Sephadex LH-20 CC (MeOH), then PGE 7-1–PGE 7-9 were given. PGE 7-4 (1.7 g) was separated by pHPLC [MeOH-H2O (20:80 → 30:70 → 40:60 → 50:50 → 100:0, v/v)] and pHPLC [CH3CN-1% HAc (16:84, v/v)] to give punicagranin H (9, 11.0 mg).
PGE 7-6 (1.1 g) was subjected to pHPLC [MeOH-H2O (30:70 → 40:60 → 50:50 → 60:40 → 100:0, v/v)] to get PGE 7-6-1–PGE 7-6-19. PGE 7-6-10 (63.1 mg) was purified by pHPLC [CH3CN-1% HAc (16:84, v/v)] to obtain brevifolin (10, 7.5 mg). PGE 7-7 (2.9 g) was isolated by pHPLC [MeOH-H2O (30:70 → 40:60 → 50:50 → 60:40 → 100:0, v/v)], then PGE 7-7-1–PGE 7-7-20 were produced. PGE 7-7-5 (108.7 mg) was purified by pHPLC [CH3CN-H2O (12:88, v/v)] to yield corilagin (16, 37.3 mg). Using the same separation method, valoneic acid dilactone (15, 37.2 mg) was obtained from PGE 7-7-6 (112.7 mg).
PGE 7-7-10 (216.9 mg) was isolated by pHPLC [CH3CN-H2O (16:84, v/v)] to gain punicagranin F (6, 78.4 mg). PGE 8 (12.2 g) was prepared by Sephadex LH-20 CC (MeOH) to generate PGE 8-1–PGE 8-6. PGE 8-2 (2.6 g) was separated by pHPLC [MeOH-H2O (30:70 → 40:60 → 50:50 → 100:0, v/v)] to yield PGE 8-2-1–PGE 8-2-16. PGE 8-2-9 (93.0 mg) was purified by pHPLC [CH3CN-1% HAc (17:83, v/v)] and 4-O-β-d-glucopyranosyl-3,3′-di-O-methylellagic acid (13, 14.3 mg) was given.
PGE 8-2-10 (136.0 mg) was prepared by pHPLC [CH3CN-1% HAc (9:91, v/v)] to give 4-O-α-l-rhamnopyranosyl ellagic acid (12, 16.1 mg). PGE 8-5 (3.37 g) was isolated by pHPLC [MeOH-H2O (30:70 → 40:60 → 50:50 → 100:0, v/v)] and PGE 8-5-1–PGE 8-5-10 were yielded. PGE 8-5-2 (630.0 mg) was further purified by pHPLC [CH3CN-1% HAc (9:91, v/v)] to gain (S)-flavogallonic acid (14, 46.7 mg). PGE 8-5-4 (482.7 mg) was prepared by pHPLC [CH3CN-1% HAc (12:88, v/v)] to give corilagin (16, 234.7 mg).
H2O eluate (PGH, 20.0 g) was fractionated by ODS CC [MeOH-H2O (0:100 → 10:90 → 20:80 → 100:0, v/v)] to obtain PGH 1–PGH 5. PGH 2 (7.0 g) was pHPLC [MeOH-H2O (2:98 → 4:96 → 100:0, v/v)] to give PGH 2-1–PGH 2-13. PGH 2-4 (292.1 mg) was prepared by pHPLC [CH3CN-1% HAc (1:99, v/v)] to yield PGH 2-4-1–PGH 2-4-5. PGH 2-4-3 (37.3 mg) was subjected to Sephadex LH-20 CC (MeOH) to obtain punicagranin G (7, 14.4 mg).
PGH 2-4-4 (28.6mg) was purified by pHPLC [CH3CN-1% HAc (1:99, v/v) further, and 2,3-(S)-hexahydroxydiphenoyl-d-glucose (17, 13.9 mg) was produced. PGH 2-8 (1150.9 mg) was isolated by pHPLC [CH3CN-1% HAc (1:99, v/v)] to yield PGH 2-8-1–PGH 2-8-8. Among them, PGH 2-8-4 was identified as punicagranin H (8, 94.0 mg) and punicalin (18, 43.1 mg). PGH 3 (2.8 g) was fractionated by pHPLC [MeOH-H2O (8:92 → 10:90 → 100:0, v/v)] to give PGH 3-1–PGH 3-12.
PGH 3-6 (358.9 mg) was prepared by pHPLC [CH3CN-1% HAc (5:95, v/v)] to produce PGH 3-6-1–PGH 3-6-5. PGH 3-6-4 (75.3 mg) was further isolated by pHPLC [CH3CN-1% HAc (1:99, v/v)] to gain punicacortein C (20, 41.9 mg). PGH 3-11 (294.6 mg) was separated by pHPLC [MeOH-1% HAc (15:85, v/v)] and flavogallonic acid dilactone (14, 21.6 mg) as well as punicalagin (19, 42.7 mg) were yielded. PGH 4 (0.7 g) was purified by Sephadex LH-20 CC [MeOH-H2O (1:1, v/v)] and pHPLC [CH3CN-1% HAc (12:88, v/v)] in turn to give brevifolincarboxylic acid (11, 46.6 mg).
Punicagranin A (1): white powder; UV λmax (MeOH) nm (log ε): 264 (3.88), 338 (3.72); IR νmax (KBr) cm−1: 3171, 1790, 1715, 1608, 1551, 1513, 1339, 1240, 1201, 1108, 1024; 1H NMR (DMSO-d6, 500 MHz): δ 2.14 (3H, s, H3-6), 7.17 (1H, s, H-5′); 13C NMR (DMSO-d6, 125 MHz): δ 166.3 (C-2), 142.2 (C-3), 119.7 (C-4), 160.8 (C-5), 14.4 (C-6), 108.0 (C-1′), 143.0 (C-2′), 134.5 (C-3′), 146.4 (C-4′), 114.2 (C-5′), 121.5 (C-6′), 167.7 (C-7′); HRESIMS: m/z 279.0141 [M–H] (calcd for C12H7O8, 279.0146).
Punicagranin B (2): white powder; UV λmax (MeOH) nm (log ε): 261 (3.97), 334 (3.86); IR νmax (KBr) cm−1: 3163, 1800, 1716, 1610, 1552, 1515, 1436, 1351, 1242, 1119, 1024; 1H NMR (DMSO-d6, 500 MHz): δ 2.15 (3H, s, H3-6), 7.10 (1H, s, H-5′), 3.70 (3H, s, 7′-COOCH3); 13C NMR (DMSO-d6, 125 MHz): δ 166.5 (C-2), 142.5 (C-3), 120.1 (C-4), 160.7 (C-5), 14.4 (C-6), 107.9 (C-1′), 142.8 (C-2′), 135.0 (C-3′), 146.6 (C-4′), 114.3 (C-5′), 120.0 (C-6′), 166.9 (C-7′), 51.3 (7′-COOCH3); HRESIMS: m/z 293.0299 [M–H] (calcd for C13H9O8, 293.0303).
Punicagranin C (3): white powder; [α]D25 −7.0 (c 0.31, MeOH); UV λmax (MeOH) nm (log ε): 272 (3.78); CD (c 0.0022 M, MeOH) mdeg (λnm): +18.3 (223); IR νmax (KBr) cm−1: 3734, 3192, 2923, 1745, 1619, 1520, 1485, 1317, 1210, 1134, 1072; 1H NMR (DMSO-d6, 500 MHz): δ 7.79 (1H, br. s, H-4), 6.21 (1H, br. s, H-5), 4.94, 5.02 (1H each, both d, J = 19.0 Hz, H2-6), 6.81 (1H, s, H-5′); 13C NMR (DMSO-d6, 125 MHz): δ 171.2 (C-2), 127.4 (C-3), 153.3 (C-4), 71.6 (C-5), 70.6 (C-6), 125.8 (C-1′), 139.9 (C-2′), 140.1 (C-3′), 147.9 (C-4′), 102.0 (C-5′), 115.6 (C-6′), 170.0 (C-7′); HRESIMS: m/z 281.0297 [M–H] (calcd for C12H9O8, 281.0303).
Punicagranin D (4): pale yellow powder; [α]D25 −14.4 (c 0.14, MeOH); UV λmax (MeOH) nm (log ε): 274 (3.91), 280 (3.92, sh), 344 (3.91); CD (c 0.0018 M, MeOH) mdeg (λnm): +36.7 (222), +3.68 (278), +2.16 (343); IR νmax (KBr) cm−1: 3168, 2922, 2839, 1713, 1615, 1586, 1451, 1339, 1203, 1097, 1021; 1H NMR (DMSO-d6, 500 MHz): δ 7.42 (1H, s, H-6), 3.63 (2H, s, H2-11), 6.64 (1H, s, H-13), 4.09 (2H, q, J = 7.0 Hz, H2-15), 1.18 (3H, t, J = 7.0 Hz, H3-16); 13C NMR (DMSO-d6, 125 MHz): δ 160.7 (C-2), 116.6 (C-3), 141.8 (C-4), 109.8 (C-5), 112.1 (C-6), 149.7 (C-7), 139.6 (C-8), 139.8 (C-9), 109.6 (C-10), 31.5 (C-11), 169.2 (C-12), 92.6 (C-13), 161.2 (C-14), 60.5 (C-15), 13.9 (C-16); HRESIMS: m/z 335.0400 [M–H] (calcd for C15H11O9, 335.0409).
Punicagranin E (5): pale yellow powder; UV λmax (MeOH) nm (log ε): 266 (3.75), 328 (3.91); IR νmax (KBr) cm−1: 3732, 2923, 2861, 1716, 1587, 1514, 1470, 1373, 1255, 1208, 1094, 1048; 1H NMR (DMSO-d6, 500 MHz): δ 6.75 (1H, d, J = 8.5 Hz, H-5), 6.84 (1H, d, J = 8.5 Hz, H-6), 2.01 (3H, s, H3-11), 4.46 (2H, q, J = 7.0 Hz, H2-13), 1.35 (3H, t, J = 7.0 Hz, H3-14); 13C NMR (DMSO-d6, 125 MHz): δ 161.0 (C-2), 116.7 (C-3), 142.5 (C-4), 115.3 (C-5), 112.7 (C-6), 149.0 (C-7), 132.3 (C-8), 142.3 (C-9), 108.7 (C-10), 13.8 (C-11), 165.1 (C-12), 62.2 (C-13), 13.9 (C-14); HRESIMS: m/z 263.0557 [M–H] (calcd for C13H11O6, 263.0561).
Punicagranin F (6): pale yellow powder; [α]D25 −15.0 (c 0.10, MeOH); UV λmax (MeOH) nm (log ε): 256 (4.68), 372 (4.11); CD (c 0.00020 M, MeOH) mdeg (λnm): −0.80 (233), −1.52 (255), −0.06 (284), −0.07 (298), +0.05 (326). IR νmax (KBr) cm−1: 3246, 1720, 1600, 1521, 1442, 1336, 1235, 1181, 1058, 887, 767; 1H NMR (DMSO-d6, 500 MHz): δ 7.50 (1H, s, H-5), 7.11 (1H, s, H-6″), 3.86 (2H, q, J = 7.0 Hz, 7″-OCH2CH3), 0.87 (3H, t, J = 7.0 Hz, 7″-OCH2CH3); 13C NMR (DMSO-d6, 125 MHz): δ 112.6 (C-1), 136.3 (C-2)a, 139.4 (C-3), 147.7 (C-4), 109.9 (C-5), 107.1 (C-6), 159.2 (C-7), 107.2 (C-1′)b, 135.4 (C-2′)a, 138.5 (C-3′)a, 146.1 (C-4′)c, 111.5 (C-5′)b, 119.2 (C-6′)b, 157.4 (C-7′), 125.3 (C-1″), 117.9 (C-2″), 143.4 (C-3″)c, 137.2 (C-4″), 143.9 (C-5″), 109.1 (C-6″), 165.9 (C-7″), 59.2 (7″-OCH2CH3), 13.6 (7″-OCH2CH3) (a,b,cthe values with the same superscript markers may be interchangeable); HRESIMS: m/z 497.0358 [M–H] (calcd for C23H23O13, 497.0362).
Punicagranin G (7) (1α:1β ≈ 1:2): pale yellow powder; [α]D25 −73.9 (c 0.17, H2O); UV λmax (MeOH) nm (log ε): 254 (4.73), 372 (4.16); CD (c 0.00013 M, MeOH) mdeg (λnm): −4.20 (234), +1.36 (276), −0.21 (299), −0.07 (298), +1.29 (328), −1.87 (381); IR νmax (KBr) cm−1: 3290, 2921, 1698, 1601, 1548, 1440, 1339, 1223, 1058; 1H NMR (CD3OD, 400 MHz): δ 4.81 (1H, d, J = 3.6 Hz, Glc-H-1α), 2.46 (1H, dd, J = 3.6, 9.6 Hz, Glc-H-2α), 4.83 (1H, dd, J = 9.6, 10.0 Hz, Glc-H-3α), 1.75 (1H, dd, J = 10.0, 10.0 Hz, Glc-H-4α), 3.55 (1H, m, Glc-H-5α), [3.55 (1H, br. d, ca. J = 12 Hz), 4.67 (1H, dd, J = 3.2, 12.0 Hz), Glc-H2-6α], 7.33 (1H, s, H-5′α), 7.17 (1H, s, H-5″α); 4.25 (1H, d, J = 7.6 Hz, Glc-H-1β), 2.28 (1H, dd, J = 7.6, 9.2 Hz, Glc-H-2β), 4.57 (1H, dd, J = 9.2, 9.6 Hz, Glc-H-3β), 1.79 (1H, dd, J = 9.6, 9.6 Hz, Glc-H-4β), 3.03 (1H, m, Glc-H-5β), [3.53 (1H, br. d, ca. J = 12 Hz), 4.75 (1H, dd, J = 3.2, 12.4 Hz), Glc-H2-6β], 7.33 (1H, s, H-5′β), 7.13 (1H, s, H-5″β); 13C NMR (CD3OD, 100 MHz): δ 93.6 (Glc-C-1α), 71.8 (Glc-C-2α), 76.8 (Glc-C-3α), 69.1 (Glc-C-4α), 70.1 (Glc-C-5α), 62.5 (Glc-C-6α); 98.7 (Glc-C-1β), 74.2 (Glc-C-2β), 78.6 (Glc-C-3β), 68.8 (Glc-C-4β), 74.9 (Glc-C-5β), 62.1 (Glc-C-6β); the signals for gallagyl group linked to α-d-glucose: δ 108.7, 110.7, 112.0 (C-5″), 112.9 (C-5′), 115.0, 115.3, 118.3 (C-1″), 119.3 (C-1′), 120.9 (C-6′), 121.9 (C-6″), 124.9, 126.3, 137.3, 137.8, 138.9 (C-3″), 139.4 (C-3′), 140.5, 140.6, 144.6, 144.8, 145.8 (C-4′), 145.9 (C-4″), 147.2, 147.6, 160.0 (lactone carbonyl-C), 160.8 (lactone carbonyl-C), 169.1 (C-7′), 170.7 (C-7″); the signals for gallagyl group linked to β-d-glucose: δ 108.9, 110.7, 111.9 (C-5″), 113.0 (C-5′), 114.9, 115.2, 118.1 (C-1″), 119.3 (C-1′), 120.9 (C-6′), 121.7 (C-6″), 124.7, 125.9, 137.3, 137.9, 138.8 (C-3″), 139.5 (C-3′), 140.5, 140.6, 144.5, 144.8, 145.8 (C-4′), 145.9 (C-4″), 147.1, 147.7, 159.9 (lactone carbonyl-C), 160.8 (lactone carbonyl-C), 169.0 (C-7′), 170.7 (C-7″); HRESIMS: m/z 781.0530 [M–H] (calcd for C34H21O22, 781.0530).
Punicagranin H (8) (1α:1β ≈ 1:3): pale yellow powder; [α]D25 −85.1 (c = 0.54, H2O); UV λmax (MeOH), nm (log ε): 256 (4.66), 373 (4.06); CD (c 0.00014 M, MeOH) mdeg (λnm): −7.23 (232), +9.48 (276), +1.49 (306), +4.59 (335), −5.12 (385); IR νmax (KBr) cm−1: 3361, 2911, 2704, 1708, 1600, 1520, 1440, 1350, 1222, 1176, 1057, 877, 751; 1H NMR (CD3OD, 400 MHz): δ 3.44, 3.51 (1H each, both d, J = 11.6 Hz, Fruc-H2-1α), 3.91 (1H, s, Fruc-H-3α), 4.09 (1H, d, J = 2.8 Hz, Fruc-H-4α), 2.67 (1H, br. d, ca. J = 11 Hz, Fruc-H-5α), [3.12 (1H, br. d, ca. J = 10 Hz), 3.87 (1H, dd, J = 10.4, 10.8 Hz), Fruc-H2-6α], 6.92 (1H, s, H-5′α), 7.12 (1H, s, H-5″α); 3.23, 3.48 (1H each, both d, J = 11.6 Hz, Fruc-H2-1β), 3.93 (1H, d, J = 1.6 Hz, Fruc-H-3β), 4.38 (1H, t, J = 1.6 Hz, Fruc-H-4β), 2.36 (1H, br. d, ca. J = 11 Hz, Fruc-H-5β), [3.28 (1H, br. d, ca. J = 11 Hz), 4.03 (1H, dd, J = 11.2, 11.2 Hz), Fruc-H2-6β], 6.89 (1H, s, H-5′β), 7.15 (1H, s, H-5″β); 13C NMR (CD3OD, 100 MHz): δ 64.6 (Fruc-C-1α), 107.2 (Fruc-C-2α), 78.9 (Fruc-C-3α), 83.2 (Fruc-C-4α), 80.8 (Fruc-C-5α), 67.6 (Fruc-C-6α); 65.4 (Fruc-C-1β), 106.1 (Fruc-C-2β), 74.9 (Fruc-C-3β), 82.9 (Fruc-C-4β), 80.7 (Fruc-C-5β), 68.2 (Fruc-C-6β); the signals for gallagyl group linked to α-d-fructose: δ 109.9, 111.3, 111.4 (C-5′), 112.5 (C-5″), 115.2, 115.6, 117.5 (C-1′), 118.8 (C-1″), 121.8 (C-6″), 122.7, 123.5, 125.3, 137.1, 137.6, 138.5 (C-3′), 139.2 (C-3″), 140.1, 141.2, 144.6, 144.9, 145.9 (C-4′), 145.9 (C-4″), 147.9, 148.1, 160.2 (lactone carbonyl-C), 160.4 (lactone carbonyl-C), 169.4 (C-7″), 170.0 (C-7′); the signals for gallagyl group linked to β-d-fructose: δ 109.9, 110.8, 111.0 (C-5′), 112.6 (C-5″), 115.2, 115.4, 117.5 (C-1′), 118.7 (C-1″), 121.6 (C-6″), 122.9, 123.7, 125.5, 137.2, 138.1, 138.5 (C-3′), 139.2 (C-3″), 140.1, 141.2, 144.7, 145.0, 145.9 (C-4′), 145.9 (C-4″), 148.1, 148.2, 160.3 (lactone carbonyl-C), 160.5 (lactone carbonyl-C), 169.3 (C-7″), 169.7 (C-7′); HRESIMS: m/z 781.0519 [M–H] (calcd for C34H21O22, 781.0530).
Punicagranin H (9): white powder; UV λmax (MeOH) nm (log ε): 260 (4.10); IR νmax (KBr) cm−1: 3318, 2924, 1718, 1606, 1512, 1457, 1340, 1266, 1168, 1076, 851, 765; 1H NMR (CD3OD, 500 MHz): δ 6.37 (1H, d, J = 2.5 Hz, H-3), 6.65 (1H, d, J = 2.5 Hz, H-5), 3.60, 3.66 (1H each, both d, J = 16.5 Hz, H2-7), 3.53 (3H, s, 8-COOCH3), 7.98 (2H, d, J = 8.5 Hz, H-2′,6′), 6.90 (2H, d, J = 8.5 Hz, H-3′,5′), 4.86 (1H, d, J = 7.5 Hz, H-1″), 3.46 (1H, dd, J = 7.5, 9.0 Hz, H-2″), 3.47 (1H, dd, J = 9.0, 9.0 Hz, H-3″), 3.41 (1H, dd, J = 9.0, 9.0 Hz, H-4″), 3.42 (1H, m, H-5″), [3.72 (1H, dd, J = 4.5, 12.0 Hz), 3.91 (1H, br. d, ca. J = 12 Hz), H2-6″]; 13C NMR (CD3OD, 125 MHz): δ 111.0 (C-1), 152.1 (C-2), 105.4 (C-3), 159.0 (C-4), 102.4 (C-5), 158.5 (C-6), 30.1 (C-7), 174.3 (C-8), 52.4 (8-COOCH3), 121.1 (C-1′), 133.5 (C-2′,6′), 116.5 (C-3′,5′), 164.3 (C-4′), 166.0 (C-7′), 103.3 (C-1″), 74.3 (C-2″), 78.0 (C-3″), 71.3 (C-4″), 78.3 (C-5″), 62.6 (C-6″); HRESI-TOF-MS: m/z 479.1188 [M–H] (calcd for C22H23O12, 479.1195).

4.1.4. Acid Hydrolysis of 79

A solution of compounds 79 (each 1.5 mg) in 1 M HCl (1 mL) was heated under reflux, neutralized with Amberlite IRA-400 (OH form) and filtrated successively. Each aqueous layer was analyzed by HPLC: HPLC column, Kaseisorb LC NH2-60-5, 4.6 mm i.d. × 250 mm (Tokyo Kasei Co., Ltd., Tokyo, Japan); mobile phase, CH3CN-H2O [(75:25; flow rate, 1.0 mL/min)]. The results were d-glucose (tR: 8.1 min, positive optical rotation) for 7 and 9, d-fructose (tR: 7.5 min, negative) for 8 were identified by comparison of their retention times and optical rotations with those of the authentic sample.

4.2. Materials and Methods for Anti-Inflammatory Assay

4.2.1. Cell Culture

RAW 264.7 cells (Chinese Academy of Medical Science) were cultured in DMEM (Biological Industries, Beit HaEmek, Israel) supplemented with 10% heat-inactivated FBS (Biological Industries, Beit HaEmek, Israel), 100 U/mL penicillin (Waltham, MA, USA) and 100 µg/mL streptomycin (Waltham, MA, USA) in a 5% CO2 humidified incubator at 37 °C. When its confluency reached 80–90% approximately, the cells were seeded in 96-well plates.

4.2.2. Cytotoxicity Assay

RAW 264.7 macrophages-like cells were seeded in 96-well plates (2 × 105 cells/mL) and incubated for 24 h. After that, the supernatant was removed and RAW 264.7 cells were treated without or with test compounds (their primary concentration was 100 mM, and these solutions were then diluted to be 30 μM for compounds 118 and 20 and 15 μM for compound 19 as final concentration, respectively) for 18 h, respectively. Then, the supernatant was replaced with 0.5 mg/mL of MTT solution and incubated further. After 4 h, the supernatant was discarded, and the pellet at the bottom of the 96-well plate was dissolved with DMSO, measured at 490 nm using a BioTek Cytation five-cell imaging multi-mode reader (Winooski, VT, USA).

4.2.3. Determination of NO Production

The in vitro NO production inhibitory assay was conducted as what has been reported [29]. In the experiment, four groups, including the normal group, LPS group, the positive drug DEX group and the tested groups, were settled. RAW 264.7 cells were seeded at the 96-well plates of 2 × 106 cells/mL and incubated for 24 h to facilitate complete adherence to the well. The primary compound solution was the same as that in Section 4.2.2, with a concentration at 100 mM, and then, these solutions were diluted to 3, 10 and 30 μM for compounds 1, 3, 58, 10, 11, 14, 1618 and 20 and 1, 5 and 15 μM for compound 19, respectively.
Then, the media of the normal group was replaced with serum-containing medium, LPS (0.5 μg/mL) was administered to the LPS group, LPS (0.5 μg/mL) combined with positive drug DEX (1.5 μg/mL) was given to the DEX group, and the tested compounds together with LPS (0.5 μg/mL) were applied to the tested groups, followed by incubation 18 h. After, equal supernatant of the RAW 264.7 culture and the Griess reagent from Beyotime Biotechnology (Shanghai, China) were mixed and measured at 540 nm on a BioTek Cytation five-cell imaging multi-mode reader (Winooski, VT, USA). A standard sodium nitrite curve was used to calculate the amount of NO.

4.2.4. Western Blot Analysis

RAW 264.7 cells were seeded in the 6-cell plates for 24 h. Then, the supernatant of normal group, LPS group, the positive drug DEX group and the tested groups were replaced with serum-containing DMEM, LPS (0.5 μg/mL), LPS (0.5 μg/mL) as well as DEX (1.5 μg/mL) and LPS (0.5 μg/mL) combined with the tested compounds (final concentration was 30 μM), respectively. After 18 h, the RAW 264.7 cells were separated from the 6-cell plates (4 × 106 cells/mL) and then centrifuged at 4 °C and 12,000× g for 5 min. The cells were lysed by adding mixed solution of RIPA lysis buffer, protease and phosphatase inhibitor with the ratio of 100:1:1 for 30 min on ice.
Then, the supernatant was collected by centrifuging at 12,000× g for 5 min, and the total protein content was determined by using a BCA protein quantification kit (Thermo Fisher Scientific, Waltham, MA, USA). Next, the protein sample was mixed with 4 × sample buffer, incubated at 100 °C for 5 min, loaded onto 10% sodium dodecyl sulfate (SDS) polyacrylamide gel and electrophoresed. The electrophoretic SDS polyacrylamide gel was transferred onto polyvinylidene fluoride membranes (Merch/Millipore, Schwalbach, Germany).
They were probed with primary antibodies against NF-κB (1:1000, ab16502, Abcam, CBG, UK), phosphorylated NF-κB (p-NF-κB) (1:1000, ab16502, Abcam), p38 MAPK (1:1000, 8690, CST), phosphorylated p38 MAPK (1:1000, 4511, CST), IL-6 (1:1000, 21865-1-AP, Proteintech), iNOS (1:1000, ab3523, Abcam), COX-2 (1:1000, ab52237, Abcam), β-actin (1:1000, ab8227, Abcam), IκBα (1:1000, ab32518, Abcam), phosphorylated IκBα (1:1000, ab92700, Abcam), IKK (1:1000, 2682S, CST) and phosphorylated IKK (1:1000, 2697, CST) and incubated overnight at 4 °C.
Then, the membranes were washed three times, each for 10 min with Tris-buffered saline with 0.1% Tween 20 (TBST), followed by treatment with horseradish peroxidase (HRP)-conjugated goat anti-rabbit immunoglobulin G (IgG) (1:10,000, SR238; Beijing Solarbio Science & Technology, Beijing, China) at room temperature for 1 h and then washed with TBST buffer three times for 10 min. Finally, the immunoreactive protein bands were measured by Immobilon Western Chemilumescent HRP Substrate (Millipore, Massachusetts, USA), visualized using a ChemiDoc MP Imaging System (Bio-Rad Laboratories, Hercules, USA) and analyzed using Image Lab software (Version 1.0, National Institutes of Health, Bethesda, MD, USA).

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules27144622/s1, Supplementary data (the NMR and HRESIMS spectra of compounds 19, ECD spectra of compounds 3, 4, 68, 14, 1620, cell viability assay, the raw data for western blot assays, and the physical and chemical data of compounds 1020 are available in the Supporting Information.

Author Contributions

H.-M.L. contributed to the bioassay. O.K. and Y.Z. performed the isolation and structure elucidation of the compounds. D.-S.Y., Y.-Q.Z. and J.-Y.R., contributed to the interpretation of the spectra and also part of the preparation of the manuscript. L.-F.H. partially contributed to analyze the data, H.-M.L. and Y.Z. prepared the manuscript, Y.Z. and T.W. planned, designed and organized the whole research of this study. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the Program for National Natural Science Foundation of China (81673688, 82074118) and Tianjin Municipal Education Commission Scientific Research Program (2021KJ132).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not available.

References

  1. Olejnik, A.; Kowalska, K.; Olkowicz, M.; Rychlik, J.; Juzwa, W.; Myszka, K.; Dembczyński, R.; Wojciech Białas, W. Antiinflammatory effects of gastrointestinal digested Sambucus nigra L. fruit extract analysed in co-cultured intestinal epithelial cells and lipopolysaccharide-stimulated macrophages. J. Funct. Foods 2015, 19, 649–660. [Google Scholar] [CrossRef]
  2. Hollebeeck, S.; Winand, J.; Hérent, M.F.; During, A.; Leclercq, J.; Larondelle, Y.; Schneider, Y.J. Anti-inflammatory effects of pomegranate (Punica granatum L.) husk ellagitannins in Caco-2 cells, an in vitro model of human intestine. Food Funct. 2012, 3, 875–885. [Google Scholar] [CrossRef]
  3. Guo, Y.; Li, Y.; Liu, F.; Zhang, G. Review of active components of traditional Chinese medicine to reduce the lipopolysaccharide-induced inflammation. Huaxi Yaoxue Zazhi 2021, 36, 331–335. [Google Scholar]
  4. Hu, X.; Yu, Q.; Hou, K.; Ding, X.; Chen, Y.; Xie, J.; Nie, S.; Xie, M. Regulatory effects of Ganoderma atrum polysaccharides on LPS-induced inflammatory macrophages model and intestinal-like Caco-2/macrophages co-culture inflammation model. Food Chem. Toxicol. 2020, 140, 111321. [Google Scholar] [CrossRef] [PubMed]
  5. Aly, E.; López-Nicolás, R.; Darwish, A.A.; Ros-Berruezo, G.; Frontela-Saseta, C. In vitro effectiveness of recombinant human lactoferrin and its hydrolysate in alleviating LPS-induced inflammatory response. Food Res. Int. 2019, 118, 101–107. [Google Scholar] [CrossRef] [PubMed]
  6. Cao, Y.; Chen, J.; Ren, G.; Zhang, Y.; Tan, X.; Yang, L. Punicalagin prevents inflammation in LPS-induced RAW 264.7 macrophages by inhibiting FoxO3a/autophagy signaling pathway. Nutrients 2019, 11, 2794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Canuti, V.; Cecchi, L.; Khatib, M.; Guerrini, L.; Mulinacci, N.; Zanoni, B. A new extract from Pomegranate (Punica granatum L.) by-products as a potential oenological tannin: Preliminary characterization and comparison with existing commercial products. Molecules 2020, 25, 4460. [Google Scholar] [CrossRef]
  8. Nikfarjam, M.; Rashki Ghaleno, L.; Shahverdi, A.H.; Mirshahvalad, S.H.; Ghoreishi, S.M.; Alizadeh, A.R. Effects of dietary pomegranate peel on antioxidant gene expression and DJ-1 protein abundance in ram testes. Int. J. Fertil. Steril. 2021, 15, 258–262. [Google Scholar]
  9. Pan, Z.; Qu, W.; Ma, H.; Atungulu, G.G.; McHugh, T.H. Continuous and pulsed ultrasound-assisted extractions of antioxidants from pomegranate peel. Ultrason. Sonochem. 2012, 19, 365–372. [Google Scholar] [CrossRef]
  10. Laurindo, L.F.; Barbalho, S.M.; Marquess, A.R.; Grecco, A.I.S.; Goulart, R.A.; Tofano, R.J.; Bishayee, A. Pomegranate (Punica granatum L.) and metabolic syndrome risk factors and outcomes: A systematic review of clinical studies. Nutrients 2022, 14, 1665. [Google Scholar] [CrossRef]
  11. Eghbali, S.; Askari, S.F.; Avan, R.; Sahebkar, A. Therapeutic effects of Punica granatum (Pomegranate): An updated review of clinical trials. J. Nutr. Metab. 2021, 2021, 5297162. [Google Scholar] [CrossRef]
  12. Xu, W.D.; Lin, H.W.; Qiu, F.; Chen, W.S. Chemical constituents of Duchesnea indica Foche. Shenyang Yaokedaxue Xuebao 2007, 24, 402–406. [Google Scholar]
  13. Wang, Y.Q.; Si, J.Y.; Liu, X.M.; Tang, J.S.; Chang, Q. Isolation, identification and quantitative determination of ellagic acid and brevifolin carboxylic acid in Duchesnea indica by RP-HPLC. Tianran Chanwu Yanjiuyukaifa 2008, 20, 667–671. [Google Scholar]
  14. Zhen, G.M.; Wei, X.Y.; Xu, L.X.; Xie, H.H.; Wu, P. Chemical constituents from pericarp of longan fruits. Zhongcaoyao 2011, 42, 1485–1489. [Google Scholar]
  15. Zhao, Y.X.; Yang, D.; Ma, Q.Y.; Li, N.; Cheng, Z.Q.; Zhou, J. Chemical constituents from Osbeckia chinensis. Zhongcaoyao 2011, 42, 1061–1065. [Google Scholar]
  16. Pfundstein, B.; El Desouky, S.K.; Hull, W.E.; Haubner, R.; Erben, G.; Owen, R.W. Polyphenolic compounds in the fruits of Egyptian medicinal plants (Terminalia bellerica, Terminalia chebula and Terminalia horrida): Characterization, quantitation and determination of antioxidant capacities. Phytochemistry 2010, 71, 1132–1148. [Google Scholar] [CrossRef]
  17. Rocha, S.H.; Silva, C.C.M.; Caland, N.L.B.; Lopes, J.A.D.; Cito, A.M.; Chaves, M.H. Chemical constituents from bark of Cenostigma macrophyllum: Cholesterol occurrence. Quim. Nova 2007, 30, 1877–1881. [Google Scholar]
  18. Li, X.Y.; Wang, Y.H.; Wang, H.S.; Shi, Y.N.; Long, C.L. Phenolic derivatives from the leaves of Dipteronia dyeriana. Tianran Chanwu Yanjiuyukaifa 2010, 22, 5–10. [Google Scholar]
  19. Jossang, A.; Pousset, J.L.; Bodo, B. Comberglutinin, a hydrolysable tannin from Combertum glutinosum. J. Nat. Prod. 1994, 57, 732–737. [Google Scholar] [CrossRef]
  20. Ito, H.; Li, P.; Koreishi, M.; Nagatomo, A.; Nishida, N.; Yoshida, T. Ellagitannin oligomers and a neolignan from pomegranate arils and their inhibitory effects on the formation of advanced glycation end products. Food Chem. 2014, 152, 323–330. [Google Scholar] [CrossRef]
  21. Tanaka, T.; Nonaka, G.C.; Itsuo, N. Tannins and related compounds. XLI. Isolation and characterization of novel ellagitannins, punicacorteins A, B, C and D, and punigluconin from the bark of Punica granatum L. Chem. Pharm. Bull. 1986, 34, 656–663. [Google Scholar] [CrossRef]
  22. Chen, X.W.; Li, C.W.; Cui, C.B.; Hua, W.; Zhu, T.J.; Gu, Q.Q. Nine new and five known polyketides derived from a deep sea-sourced Aspergillus sp. 16-02-1. Mar. Drugs 2014, 12, 3116–3137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Yang, Z.N.; Su, B.J.; Wang, Y.Q.; Liao, H.B.; Chen, Z.F.; Liang, D. Isolation, absolute configuration, and biological activities of chebulic acid and brevifolincarboxylic acid derivatives from Euphorbia hirta. J. Nat. Prod. 2020, 83, 985–995. [Google Scholar] [CrossRef] [PubMed]
  24. Tanaka, T.; Nonaka, J.; Nishioka, I. Tannins and related compounds. XLII. Isolation and characterization of four new hydrolyzable tannins, terflavins A and B, tergallagin and tercatain from the leaves of Terminalia catappa L. Chem. Pharm. Bull. 1986, 34, 1039–1049. [Google Scholar] [CrossRef] [Green Version]
  25. Tanaka, T.; Tong, H.H.; Xu, Y.M.; Ishimaru, K.; Nonaka, J.; Nishioka, I. Tannins and related compounds. CXVII. Isolation and characterization of three new ellagitannins, lagerstannins A, B and C, having a gluconic acid core, from Lagerstroemia speciosa (L.) Pers. Chem. Pharm. Bull. 1992, 40, 2975–2980. [Google Scholar] [CrossRef] [Green Version]
  26. Abiodun, O.O.; Rodríguez-Nogales, A.; Algieri, F.; Gomez-Caravaca, A.M.; Segura-Carretero, A.; Utrilla, M.P.; Rodriguez-Cabezas, M.E.; Galvez, J. Antiinflammatory and immunomodulatory activity of an ethanolic extract from the stem bark of Terminalia catappa L. (Combretaceae): In vitro and in vivo evidences. J. Ethnopharmacol. 2016, 192, 309–319. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, Y.; Wu, C.; Guo, L.; Chen, Y.; Han, L.; Liu, E.; Gao, X.; Wang, T. Triglyceride accumulation inhibitory effects of phenylpropanoid glycosides from Boschniakia rossica Fedtsch et Flerov. Fitoterapia 2013, 85, 69–75. [Google Scholar] [CrossRef]
  28. Tanaka, T.; Nonaka, J.; Nishioka, I. Tannins and related compounds. XL. Revision of the structures of punicalin and punicalagin, and isolation and characterization of 2-O-galloylpunicalin from the bark of Punica granatum L. Chem. Pharm. Bull. 1986, 34, 650–655. [Google Scholar] [CrossRef] [Green Version]
  29. Hong, J.M.; Kim, J.E.; Min, S.K.; Kim, K.H.; Han, S.J.; Yim, J.H.; Park, H.; Kim, J.H.; Kim, I.C. Anti-inflammatory effects of antarctic lichen Umbilicaria antarctica methanol extract in lipopolysaccharide-stimulated RAW 264.7 macrophage cells and zebrafish model. Biomed. Res. Int. 2021, 2021, 8812090. [Google Scholar] [CrossRef]
  30. Jayakumar, T.; Yang, C.M.; Yen, T.L.; Hsu, C.Y.; Sheu, J.R.; Hsia, C.W.; Manubolu, M.; Huang, W.C.; Hsieh, C.Y.; Hsia, C.H. Anti-inflammatory mechanism of an alkaloid rutaecarpine in LTA-stimulated RAW 264.7 cells: Pivotal role on NF-κB and ERK/p38 signaling molecules. Int. J. Mol. Sci. 2022, 23, 5889. [Google Scholar] [CrossRef]
  31. Yong, H.Y.; Koh, M.S.; Moon, A. The p38 MAPK inhibitors for the treatment of inflammatory diseases and cancer. Expert. Opin. Investig. Drugs 2009, 18, 1893–1905. [Google Scholar] [CrossRef] [PubMed]
  32. Law, M.; Corsino, P.; Parker, N.T.; Law, B.K. Identification of a small molecule inhibitor of serine 276 phosphorylation of the p65 subunit of NF-kappaB using in silico molecular docking. Cancer Lett. 2010, 291, 217–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kim, K.M.; Kim, Y.S.; Lim, J.Y.; Min, S.J.; Ko, H.C.; Kim, S.J.; Kim, Y. Intestinal anti-inflammatory activity of Sasa quelpaertensis leaf extract by suppressing lipopolysaccharide-stimulated inflammatory mediators in intestinal epithelial Caco-2 cells co-cultured with RAW 264.7 macrophage cells. Nutr. Res. Pract. 2015, 9, 3–10. [Google Scholar] [CrossRef] [Green Version]
  34. Hsieh, I.N.; Chang, A.S.; Teng, C.M.; Chen, C.C.; Yang, C.R. Aciculatin inhibits lipopolysaccharide-mediated inducible nitric oxide synthase and cyclooxygenase-2 expression via suppressing NF-κB and JNK/p38 MAPK activation pathways. J. Biomed. Sci. 2011, 18, 28. [Google Scholar] [CrossRef] [Green Version]
  35. Oeckinghaus, A.; Ghosh, S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb Perspect. Biol. 2009, 1, a000034. [Google Scholar] [CrossRef]
  36. Atreya, I.; Atreya, R.; Neurath, M.F. NF-kappaB in inflammatory bowel disease. J. Intern. Med. 2008, 263, 591–596. [Google Scholar] [CrossRef]
  37. Yan, F.; Cheng, D.; Wang, H.; Gao, M.; Zhang, J.; Cheng, H.; Wang, C.; Zhang, H.; Xiong, H. Corilagin ameliorates con A-induced hepatic injury by restricting M1 macrophage polarization. Front. Immunol. 2022, 13, 807509. [Google Scholar] [CrossRef]
  38. Shen, R.; Yin, P.; Yao, H.; Chen, L.; Chang, X.; Li, H.; Hou, X. Punicalin ameliorates cell pyroptosis induced by LPS/ATP through suppression of ROS/NLRP3 pathway. J. Inflamm. Res. 2021, 14, 711–718. [Google Scholar] [CrossRef]
  39. Yan, H.; Gulimire, A.; Zhu, J.; Liang, H.; Gu, Z. Punicalagin suppresses LPS-induced inflammatory responses in murine macrophages via JAK/STAT signaling pathway and zymosan-induced mice paw edema. Indian. J. Pharm. Educ. Res. 2021, 55, 550–555. [Google Scholar] [CrossRef]
Figure 1. Structures of polyphenols obtained from the peels of P. granatum.
Figure 1. Structures of polyphenols obtained from the peels of P. granatum.
Molecules 27 04622 g001
Figure 2. The main 1H 1H COSY and HMBC correlations of 19.
Figure 2. The main 1H 1H COSY and HMBC correlations of 19.
Molecules 27 04622 g002
Figure 3. Concentration of compounds 1, 3, 58, 10, 11, 14 and 1620 on NO production in LPS-induced RAW 264.7 cells. N: normal group without LPS, DEX and tested samples; LPS: model group with 0.5 μg/mL LPS; DEX: positive drug group with 0.5 μg/mL LPS + 1.5 μg/mL DEX; tested compound groups were treated with 0.5 μg/mL LPS + compounds, and the final concentrations were 3, 10 and 30 μM for compounds 1, 3, 58, 10, 11, 14, 1618 and 20 and 1, 5 and 15 μM for compound 19, respectively. Values represent the mean ± SD of six determinations. * p < 0.05, ** p < 0.01, *** p < 0.001 (Differences between the compound-treated group and control group). ### p < 0.001 (Differences between the control group and normal group).
Figure 3. Concentration of compounds 1, 3, 58, 10, 11, 14 and 1620 on NO production in LPS-induced RAW 264.7 cells. N: normal group without LPS, DEX and tested samples; LPS: model group with 0.5 μg/mL LPS; DEX: positive drug group with 0.5 μg/mL LPS + 1.5 μg/mL DEX; tested compound groups were treated with 0.5 μg/mL LPS + compounds, and the final concentrations were 3, 10 and 30 μM for compounds 1, 3, 58, 10, 11, 14, 1618 and 20 and 1, 5 and 15 μM for compound 19, respectively. Values represent the mean ± SD of six determinations. * p < 0.05, ** p < 0.01, *** p < 0.001 (Differences between the compound-treated group and control group). ### p < 0.001 (Differences between the control group and normal group).
Molecules 27 04622 g003
Figure 4. Inhibitory effects of compounds 68 on the protein expression of TNF-α, IL-6, iNOS, COX-2, IKK-α/β, p-IKK-α/β, IκBα, p-IκBα, NF-κB/p65, p-NF-κB/p65, p38 MAPK and p-p38 MAPK in RAW 264.7 cells. N: normal group without LPS, DEX and tested samples; LPS: model group with 0.5 μg/mL LPS; DEX: positive drug group with 0.5 μg/mL LPS + 1.5 μg/mL DEX; tested compound groups were treated with 0.5 μg/mL LPS + 30 μM compounds 68, respectively. Values represent the mean ± SD of three determinations. * p < 0.05; ** p < 0.01; *** p < 0.001 (Differences between the compound-treated group and control group). ### p < 0.001 (Differences between the LPS-treated group and control group).
Figure 4. Inhibitory effects of compounds 68 on the protein expression of TNF-α, IL-6, iNOS, COX-2, IKK-α/β, p-IKK-α/β, IκBα, p-IκBα, NF-κB/p65, p-NF-κB/p65, p38 MAPK and p-p38 MAPK in RAW 264.7 cells. N: normal group without LPS, DEX and tested samples; LPS: model group with 0.5 μg/mL LPS; DEX: positive drug group with 0.5 μg/mL LPS + 1.5 μg/mL DEX; tested compound groups were treated with 0.5 μg/mL LPS + 30 μM compounds 68, respectively. Values represent the mean ± SD of three determinations. * p < 0.05; ** p < 0.01; *** p < 0.001 (Differences between the compound-treated group and control group). ### p < 0.001 (Differences between the LPS-treated group and control group).
Molecules 27 04622 g004
Figure 5. Summary graph of the investigation.
Figure 5. Summary graph of the investigation.
Molecules 27 04622 g005
Table 1. Inhibitory effects of compounds 120 on NO production in RAW 264.7 cells.
Table 1. Inhibitory effects of compounds 120 on NO production in RAW 264.7 cells.
No.NRC (%)No.NRC (%)No.NRC (%)
Normal4.0 ± 0.9685.9 ± 3.2 ***1488.2 ± 4.9 ***
Control100 ± 2.9784.1 ± 3.0 ***1599.2 ± 2.2
Dex83.2 ± 2.8 ***880.2 ± 3.6 ***1688.7 ± 1.8 ***
193.4 ± 3.7 *997.6 ± 3.01790.2 ± 1.4 ***
2101.4 ± 1.31081.9 ± 4.1 ***1874.8 ± 2.8 ***
394.4 ± 2.5 *1191.7 ± 1.8 ***1971.0 ± 1.3 ***
4100.1 ± 1.912100.6 ± 2.52071.1 ± 2.0 ***
591.3 ± 3.3 **13101.6 ± 2.7
Positive control: Dexamethasone (Dex). Nitrite relative concentration (NRC): percentage of control group (set as 100%). Values represent the mean ± SD of six determinations. * p < 0.05, ** p < 0.01, *** p < 0.001 (Differences between the compound-treated group and control group). Final concentrations were 30 μM for compounds 118 and 20, as well as 15 μM and 1.5 μg/mL for 19 and Dex, respectively.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Li, H.-M.; Kouye, O.; Yang, D.-S.; Zhang, Y.-Q.; Ruan, J.-Y.; Han, L.-F.; Zhang, Y.; Wang, T. Polyphenols from the Peels of Punica granatum L. and Their Bioactivity of Suppressing Lipopolysaccharide-Stimulated Inflammatory Cytokines and Mediators in RAW 264.7 Cells via Activating p38 MAPK and NF-κB Signaling Pathways. Molecules 2022, 27, 4622. https://doi.org/10.3390/molecules27144622

AMA Style

Li H-M, Kouye O, Yang D-S, Zhang Y-Q, Ruan J-Y, Han L-F, Zhang Y, Wang T. Polyphenols from the Peels of Punica granatum L. and Their Bioactivity of Suppressing Lipopolysaccharide-Stimulated Inflammatory Cytokines and Mediators in RAW 264.7 Cells via Activating p38 MAPK and NF-κB Signaling Pathways. Molecules. 2022; 27(14):4622. https://doi.org/10.3390/molecules27144622

Chicago/Turabian Style

Li, Hui-Min, Ongher Kouye, Ding-Shan Yang, Ya-Qi Zhang, Jing-Ya Ruan, Li-Feng Han, Yi Zhang, and Tao Wang. 2022. "Polyphenols from the Peels of Punica granatum L. and Their Bioactivity of Suppressing Lipopolysaccharide-Stimulated Inflammatory Cytokines and Mediators in RAW 264.7 Cells via Activating p38 MAPK and NF-κB Signaling Pathways" Molecules 27, no. 14: 4622. https://doi.org/10.3390/molecules27144622

Article Metrics

Back to TopTop