Journal List > Nutr Res Pract > v.5(2) > 1051123

Li and Wang: Anti-inflammatory effect of the water fraction from hawthorn fruit on LPS-stimulated RAW 264.7 cells

Abstract

The hawthorn fruit (Crataegus pinnatifida Bunge var. typica Schneider) is used as a traditional medicine in Korea. The objective of this study was to understand the mechanisms of the anti-inflammatory effects of the water fractionated portion of hawthorn fruit on a lipopolysaccharide (LPS)-stimulated RAW 264.7 cellular model. The level of nitric oxide (NO) production in the water fraction and LPS-treated RAW 264.7 cells were determined with an ELISA. The cytotoxicity of the water fraction and LPS was measured with an MTT assay. Expression of nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF)-α, interleukin 6 (IL-6), and interleukin 1β (IL-1β) mRNA were analyzed with a reverse transcription polymerase chain reaction (RT-PCR). The water fraction of hawthorn fruit was determined to be safe and significantly inhibited NO production in LPS-stimulated RAW 264.7 cells and suppressed COX-2, TNF-α, IL-1β, and IL-6 expression. The observed anti-inflammatory effects of the water fraction of hawthorn fruit might be attributed to the down-regulation of COX-2, TNF-α, IL-1β, and IL-6 expression in LPS-stimulated RAW 264.7 cells.

Introduction

Inflammation is a complex biological response to pathogens and damaged cells in the human body [1]. Chronic and uncontrolled inflammation can be common in various diseases, such as cardiovascular diseases, autoimmune rheumatoid arthritis, systemic lupus erythematosus, cancer, and Alzheimer's or Parkinson's diseases [2-5]. Many transcriptional factors, inflammatory cytokines, enzymes, and other mediators have been shown to be related to these effects [6]. Macrophages participate in the autoregulatory loop of the inflammatory process. Therefore, therapeutic interventions controlling various inflammatory diseases target macrophages and their products [7].
Lipopolysaccharide (LPS) is a major component of the cell walls of Gram-negative organisms. LPS activates immune cells to upregulate inflammatory responses and causes the production of pro-inflammatory cytokines [8]. Nitric oxide (NO) reflects the degree of LPS-induced inflammation in macrophages [9]. During the inflammatory process, NO is produced by nitric oxide synthase (iNOS), which plays a regulatory role in the expression of pro-inflammatory mediators [10,11]. NO is also reported to affect the activity of cyclooxygenase-2 (COX-2) [12]. Prostaglandins (PGs), the product of COX-2, are stimulated by LPS and cytokines and can contribute to the pain and swelling associated with inflammation [13]. Furthermore, LPS-stimulated inflammation can be identified by pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin 6 (IL-6), and interleukin 1β (IL-1β), which are related to the induction of insulin resistance and the development of Type 2 diabetes mellitus (T2DM) [14].
Hawthorn fruit is a traditional medicine used in treating the cardiovascular diseases, and angina pectoris of coronary heart disease [15,16]. Recent reports have shown that hawthorn fruit exhibits anti-oxidant, anti-atherosclerotic, and hypolipidemic activities [17-20]. We previously reported on the antioxidant and anti-inflammatory properties of the methanolic extract of hawthorn fruit [21,22]. In this study, our objective was to investigate the mechanisms behind the anti-inflammatory activities of the water fractionated portion of hawthorn fruit on RAW 264.7 cells.

Materials and Methods

We collected hawthorn fruits (Crataegus pinnatifida Bunge var. typica Schneider) from native trees in Chuncheon, Korea in autumn, 2008. The fresh fruits were air-dried and then ground into fine powder. Fifty grams of powder was extracted twice with one liter of 70% methanol. The extract was filtered with vacuum filtration (100-mm; Whatman, Maidstone, UK). The solvent was evaporated under reduced pressure using a vacuum rotary evaporator (CCA-1110; Eyela, Tokyo, Japan). Coarse extract was successively extracted with dichloromethane, ethyl acetate, n-butanol, and water in a continuous Soxhlet extractor. Among the extracted fractions, the water fraction exhibited significantly more anti-inflammatory activity than the others (data not shown), and so the water fraction of the hawthorn fruit was used for further analysis.

Cell line and cell culture

Murine macrophage RAW 264.7 cell lines were purchased from the Korean Cell Line Bank (Seoul, Korea) and grown in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum, 100 U/ml penicillin, and 100 µg/ml streptomycin. Cells were cultured in a humidified atmosphere and incubated at 37℃ in 5% CO2.

Treatment of cells with hawthorn fruit

Exponentially growing RAW 264.7 cells were plated at a density of 105 cells/well in 6- and 96-well microplates in culture medium and allowed to adhere for 16 h before treatment. Hawthorn fruit water fraction was added to the cultures at final concentrations of 200, 400, and 600 µg/ml with 2 µg/ml lipopolysaccharide (LPS) for 24 h.

MTT (1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan) assay

Cell viability was evaluated with a conventional MTT assay. At 24 h prior to culture termination, 20 µl of the MTT solution (4 mg/ml in a phosphate-buffered saline (PBS), pH 7.4) was added to each well, and the cells were continuously cultured for 4 h. Then 200 µl of dimethyl sulfoxide (DMSO) was added for solubilization. Absorbance was measured at 550 nm using an enzyme-linked immunosorbent assay (ELISA) plate reader (Bio-Tek, Winooski, VT, USA).

Nitric oxide (NO) determination

The level of NO production in cell culture supernatants was determined using a colorimetric assay based on the Griess reaction [23]. Aliquots of 100 µl of supernatants were mixed with 100 µl Griess reagent (50 µl of 1% sulfanilamide in 5% phosphoric acid and 50 µl of 0.1% naphthyl-ethylenediamine dihydrochloride). After 10 min, the absorbance was determined at 550 nm with an ELISA.

Total RNA extraction and reverse transcription polymerase chain reaction (RT-PCR)

The total RNA from the LPS-stimulated RAW264.7 cells was prepared with a TRIzol RNA isolation kit (Invitrogen, Carlsbad, CA, USA) and stored at -80℃ until required. One µg of the RNA was reverse-transcribed into cDNA and used as a template for RT-PCR amplification. The primers and the amplification conditions are listed in Table 1. PCR was performed with a DNA gene cycler, and the amplification was followed by denaturation at 94℃ for 30 s, annealation at 55℃ for 30 s, and primer extension a 72℃ for 45 s. PCR products were analyzed on 1% agarose gels, and bands were visualized with ethidium bromide staining and a Mini BIS image analysis system (DNR Bio-Imaging Systems Ltd., Kiryat Anavim, Israel). A densitometric analysis was preformed with image analysis software (Quantity one; Bio-Rad, Hercules, CA, USA).

Statistical analysis

All tests were carried out independently in triplicate (n = 3), and the data are expressed as the mean ± standard derivation (SD). Statistical significance was evaluated with analysis of variance (ANOVA) using SPSS 16.0 (SPSS Inc., Chicago, USA). Results were considered to be significant when P < 0.05.

Results

To investigate the cytotoxicity of the hawthorn fruit water fraction, we incubated RAW 264.7 cells with different dosages of the hawthorn fruit water fraction for 24 h. Cell viability did not seem to be affected for hawthorn fruit water fraction concentrations up to 600 µg/ml, as observed with an MTT assay (Fig. 1A). On the contrary, the hawthorn fruit water fraction protected the RAW 264.7 cells from LPS-induced apoptosis (Fig. 1B). 45.7% of cells were viable after stimulation with LPS for 24 h. However, after treatment with 200, 400, and 600 µg/ml of the hawthorn fruit water fraction, the viability of the RAW cells increased to 61.8, 72.7, and 83.4%, respectively.

Inhibition NO production in LPS-stimulated RAW 264.7 cells

The effect of the hawthorn fruit water fraction on NO inhibition was determined by treating the RAW264.7 cells with it in the presence or absence of LPS stimulation. The cells that were stimulated with LPS produced significant levels of NO in conditioned medium (Fig. 2). The level of NO production induced by LPS in RAW cells decreased significantly in a dose-dependent manner when treated with different concentrations of the hawthorn fruit water fraction.

Suppression of iNOS, COX-2, TNF-α, IL-1β, and IL-6 mRNA expression in LPS-stimulated RAW264.7

To determine if the above effect on NO production was related to the changes in the levels of iNOS, the expression of iNOS mRNA was measured with RT-PCR (Fig. 3A). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), the most commonly used housekeeping gene, was used to normalize gene expression in this study. The cells treated with hawthorn fruit water fraction exhibited stronger iNOS presentation than the control (Fig. 3B). However, cells treated with LPS along with the hawthorn fruit water fraction significantly inhibited COX-2 mRNA expression in a dose-dependent manner. Expression levels of COX-2 declined by 45.5, 80.2, and 85.7% when LPS-stimulated cells were treated with 200, 400, and 600 µg/ml of the hawthorn fruit water fraction, respectively (Fig. 3C).
We also examined the expression of several pro-inflammatory cytokines, such as TNF-α, IL-1β, and IL-6 mRNA. High concentrations of the hawthorn fruit water fraction inhibited TNF-α mRNA. Cells exhibited 60.2% of TNF-α mRNA when incubated with 600 µg/ml of the hawthorn fruit water fraction, as compared to the LPS-stimulated cells (Fig. 3D). A hawthorn fruit water fraction concentration of 200 µg/ml did not inhibit TNF-α mRNA expression.
The hawthorn fruit water fraction also attenuated the LPS-stimulated expression of IL-1β and IL-6 mRNA. At a dosing rate of 400 µg/ml of the water fraction of hawthorn fruit, control group cells showed reductions in expression of IL-1β and IL-6 by 44.8 and 34.4%, respectively, as compared to the LPS-stimulated group (Fig. 3E and Fig. 3F). When the dosage of hawthorn fruit water fraction was increased to 600 µg/ml, no further reduction in the expression of IL-16 was noted; contrastingly, the expression of IL-6 decreased sharply.

Discussion

Hawthorn fruit, used in traditional medicine, is considered to be safe in existing pharmacological and toxicological studies [15]. In previous studies, we showed that the methanolic extracts of hawthorn fruit possess anti-inflammatory properties, achieved through the inhibition of LPS-induced NO production in RAW 264.7 cells [22]. In the present study, we examined the potential anti-inflammatory effects and underlying molecular mechanisms of hawthorn fruit water fraction from 70% methanolic extract in an LPS-stimulated inflammatory model using RAW 264.7 cells.
NO is an important mediator and regulator of inflammatory responses [24]. In an inflammatory response, the overproduction of NO, a highly reactive molecule, reacts with superoxide, creating cytotoxicity and tissue damage in an organism [25]. NO-induced oxidative stress is associated with many diseases, such as septic shock, atherosclerosis, Alzheimer's disease, diabetes, and Parkinson's disease [26,27]. The down-regulation of NO has been used to treat such diseases. In the current study, hawthorn fruit water fraction was shown to inhibit the production of NO in LPS-stimulated RAW 264.7 cells in a dose-dependent manner. The hawthorn fruit water fraction also protected the RAW 264.7 cells against NO-induced toxicity and damage. In addition, the hawthorn fruit water fraction was quite safe at different concentrations. Thus, we considered the hawthorn fruit water fraction to have potential properties for preventing NO-related diseases.
Macrophages, important components in the human immune defense system, respond actively to inflammation by releasing pro-inflammatory cytokines, such as iNOS, COX-2, TNF-α, IL-12, IL-1β, and IL-6, and other inflammatory factors, such as NO and PGE2 [28]. These cytokines are playing a principal role in inflammatory diseases and processes [29]. Usually, activated inflammatory cells produce high quantities of NO, which in turn produce iNOS. In the present study, the water fraction from hawthorn fruit inhibited NO production but did not change the iNOS mRNA levels which produced by LPS stimulation. These unaffected levels of iNOS mRNA could be attributed to the state of macrophage differentiation, the complex of stimuli, and tissue location, which are dependent on iNOS expression [30]. Previously, our group reported that hawthorn fruit is a NO free radical scavenger [31]. The reduction of iNOS-catalyzed NO production could attribute to the NO free radical scavenging activity of the hawthorn fruit water fraction. NO is necessary for maintaining prolonged COX-2 gene expression, a central mediator in inflammation [32]. All of the steps that involve COX-2 gene expression are considered to be potential therapeutic targets [33]. In the present study, hawthorn fruit water fraction suppressed COX-2 mRNA expression, supporting its anti-inflammatory potential.
TNF-α is a key mediator in defense responses and the induction of apoptosis [34]. It can stimulate the production or expression of IL-6, IL-1β, PGE2, collagenase, and adhesion molecules, eliciting a number of physiological effects, including septic shock, inflammation, and cytotoxicity [35]. IL-6 is a well-known pro-inflammatory cytokine, regarded as an endogenous mediator of LPS-induced fever [36]. IL-1β is considered to be another pivotal pro-inflammatory cytokine, primarily released by macrophages, and plays a role in the pathophysiology of rheumatoid arthritis [37]. In this study, TNF-α, IL-1β, and IL-6 production in LPS-stimulated RAW 264.7 cells was significantly inhibited when the cells were incubated with increasing concentrations of hawthorn fruit water fraction. The decrease in production of TNF-α, IL-1β, and IL-6 seemed to attenuate cytokine-mediated host-destructive processes in inflammatory tissues.
In conclusion, our present findings indicate that hawthorn fruit water fraction is a potent inhibitor of LPS-induced NO production and COX-2, TNF-α, IL-1β, and IL-6 gene expression in RAW 264.7 cells. The hawthorn fruit water fraction did not inhibit iNOS mRNA expression, and it had relatively different mechanisms compared to its methanolic extract [22,38,39]. The difference may relate to the respective active mechanism of diverse compounds. Additional studies of the active compounds in hawthorn fruit water fraction are currently underway.

Figures and Tables

Fig. 1
Cell viability of murine macrophage RAW 264.7 cells. (A) Cell viability of RAW 264.7 cells after incubation in the presence of various concentrations of hawthorn fruit water fraction for 24 h. (B) Cell viability of RAW 264.7 cells co-treated with hawthorn fruit water fraction and 2 µg/ml of lipopolysaccharide (LPS) for 24 h. Cell viability was measured using an MTT assay. Data represent the mean ± SD of three independent experiments.
nrp-5-101-g001
Fig. 2
Inhibitory effect of hawthorn fruit water fraction on nitric oxide (NO) production in a culture medium of LPS-stimulated RAW 264.7 cells. RAW 264.7 cells were treated with 2 µg/ml of lipopolysaccharide (LPS) and hawthorn fruit water fraction for 24 h. The absorbance was determined at 550 nm with an ELISA. Data represent the mean ± SD of three independent experiments.
nrp-5-101-g002
Fig. 3
Effects of hawthorn fruit water fraction on nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumour necrosis factor (TNF)-α, interleukin 6 (IL-6), and interleukin 1β (IL-1β) mRNA production in LPS-stimulated RAW 264.7 cells. RAW 264.7 cells were treated with 2 µg/ml of lipopolysaccharide (LPS) and various concentrations of hawthorn fruit water fraction for 24 h. (A) Reverse transcription polymerase chain reaction (RT-PCR) analysis of the expression of iNOS, COX-2, TNF-α, IL-1β, and IL-6 mRNA. (B), (C), (D), (E), and (F) Quantification of the iNOS, COX-2, TNF-α, IL-1β, and IL-6 expression levels were achieved with densitometric measurement.
nrp-5-101-g003
Table 1
Primers and amplification conditions used for RT-PCR in RAW264.7 cells
nrp-5-101-i001

References

1. Ferrero-Miliani L, Nielsen OH, Andersen PS, Girardin SE. Chronic inflammation: importance of NOD2 and NALP3 in interleukin-1β generation. Clin Exp Immunol. 2007. 147:227–235.
crossref
2. Frostegård J, Ulfgren AK, Nyberg P, Hedin U, Swedenborg J, Andersson U, Hansson GK. Cytokine expression in advanced human atherosclerotic plaques: dominance of pro-inflammatory (Th1) and macrophage-stimulating cytokines. Atherosclerosis. 1999. 145:33–43.
crossref
3. Jara LJ, Medina G, Vera-Lastra O, Amigo MC. Accelerated atherosclerosis, immune response and autoimmune rheumatic diseases. Autoimmun Rev. 2006. 5:195–201.
crossref
4. Karin M, Lawrence T, Nizet V. Innate immunity gone awry: linking microbial infections to chronic inflammation and cancer. Cell. 2006. 124:823–835.
crossref
5. Sarkar D, Fisher PB. Molecular mechanisms of aging-associated inflammation. Cancer Lett. 2006. 236:13–23.
crossref
6. Walsh LJ. Mast cells and oral inflammation. Crit Rev Oral Biol Med. 2003. 14:188–198.
crossref
7. Fujiwara N, Kobayashi K. Macrophages in inflammation. Curr Drug Targets Inflamm Allergy. 2005. 4:281–286.
crossref
8. Nicholas C, Batra S, Vargo MA, Voss OH, Gavrilin MA, Wewers MD, Guttridge DC, Grotewold E, Doseff AI. Apigenin blocks lipopolysaccharide-induced lethality in vivo and proinflammatory cytokines expression by inactivating NF-κB through the suppression of p65 phosphorylation. J Immunol. 2007. 179:7121–7127.
crossref
9. Hortelano S, Zeini M, Boscá L. Nitric oxide and resolution of inflammation. Methods Enzymol. 2002. 359:459–465.
crossref
10. Sacco RE, Waters WR, Rudolph KM, Drew ML. Comparative nitric oxide production by LPS-stimulated monocyte-derived macrophages from Ovis canadensis and Ovis aries. Comp Immunol Microbiol Infect Dis. 2006. 29:1–11.
crossref
11. Farley KS, Wang LF, Razavi HM, Law C, Rohan M, McCormack DG, Mehta S. Effects of macrophage inducible nitric oxide synthase in murine septic lung injury. Am J Physiol Lung Cell Mol Physiol. 2006. 290:L1164–L1172.
crossref
12. Salvemini D, Misko TP, Masferrer JL, Seibert K, Currie MG, Needleman P. Nitric oxide activates cyclooxygenase enzymes. Proc Natl Acad Sci U S A. 1993. 90:7240–7244.
crossref
13. Yoon WJ, Ham YM, Kim KN, Park SY, Lee NH, Hyun CG, Lee WJ. Anti-inflammatory activity of brown alga Dictyota dichotoma in murine macrophage RAW 264.7 cells. J Med Plant Res. 2009. 3:1–8.
14. Neels JG, Olefsky JM. Inflamed fat: what starts the fire? J Clin Invest. 2006. 116:33–35.
crossref
15. Pryor WA. The antioxidant nutrients and disease prevention-what do we know and what do we need to find out? Am J Clin Nutr. 1991. 53:391S–393S.
crossref
16. Rigelsky JM, Sweet BV. Hawthorn: pharmacology and therapeutic uses. Am J Health Syst Pharm. 2002. 59:417–422.
crossref
17. Zhang Z, Ho WKK, Huang Y, James AE, Lam LW, Chen ZY. Hawthorn fruit is hypolipidemic in rabbits fed a high cholesterol diet. J Nutr. 2002. 132:5–10.
crossref
18. Froehlicher T, Hennebelle T, Martin-Nizard F, Cleenewerck P, Hilbert JL, Trotin F, Grec S. Phenolic profiles and antioxidative effects of hawthorn cell suspensions, fresh fruits, and medicinal dried parts. Food Chem. 2009. 115:897–903.
crossref
19. Kim KM, Choi JY, Yoo SE, Park MY, Lee BS, Ko TH, Sung SH, Shin HM, Park JE. HMCO5, herbal extract, inhibits NF-κB expression in lipopolysaccharide treated macrophages and reduces atherosclerotic lesions in cholesterol fed mice. J Ethnopharmacol. 2007. 114:316–324.
crossref
20. Ko SH, Choi SW, Ye SK, Yoo S, Kim HS, Chung MH. Comparison of anti-oxidant activities of seventy herbs that have been used in Korean traditional medicine. Nutr Res Pract. 2008. 2:143–151.
crossref
21. Park JH, Li C, Hu W, Wang MH. Antioxidant and free radical scavenging activity of different fractions from hawthorn fruit. J Food Sci Nutr. 2010. 15:44–50.
crossref
22. Li C, Son HJ, Huang C, Lee SK, Lohakare J, Wang MH. Comparison of Crataegus pinnatifida Bunge var. typica Schneider and C. pinnatifida Bunge fruits for antioxidant, anti-α-glucosidase, and anti-inflammatory activities. Food Sci Biotechnol. 2010. 19:769–775.
crossref
23. Sherman MP, Aeberhard EE, Wong VZ, Griscavage JM, Ignarro LJ. Pyrrolidine dithiocarbamate inhibits induction of nitric oxide synthase activity in rat alveolar macrophages. Biochem Biophys Res Commun. 1993. 191:1301–1308.
crossref
24. MacMicking J, Xie QW, Nathan C. Nitric oxide and macrophage function. Annu Rev Immunol. 1997. 15:323–350.
crossref
25. Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly. Am J Physiol. 1996. 271:C1424–C1437.
crossref
26. Li XA, Guo L, Asmis R, Nikolova-Karakashian M, Smart EJ. Scavenger receptor BI prevents nitric oxide-induced cytotoxicity and endotoxin-induced death. Circ Res. 2006. 98:e60–e65.
crossref
27. Jayakumar T, Thomas PA, Geraldine P. In-vitro antioxidant activities of an ethanolic extract of the oyster mushroom, Pleurotus ostreatus. IFSET. 2009. 10:228–234.
crossref
28. Boscá L, Zeini M, Través PG, Hortelano S. Nitric oxide and cell viability in inflammatory cells: a role for NO in macrophage function and fate. Toxicology. 2005. 208:249–258.
crossref
29. Sarkar D, Saha P, Gamre S, Bhattacharjee S, Hariharan C, Ganguly S, Sen R, Mandal G, Chattopadhyay S, Majumdar S, Chatterjee M. Anti-inflammatory effect of allylpyrocatechol in LPS-induced macrophages is mediated by suppression of iNOS and COX-2 via the NF-κB pathway. Int Immunopharmacol. 2008. 8:1264–1271.
crossref
30. Hori M, Kita M, Torihashi S, Miyamoto S, Won KJ, Sato K, Ozaki H, Karaki H. Upregulation of iNOS by COX-2 in muscularis resident macrophage of rat intestine stimulated with LPS. Am J Physiol Gastrointest Liver Physiol. 2001. 280:G930–G938.
crossref
31. Li C, Han W, Wang MH. Antioxidant activity of hawthorn fruit in vitro. J Appl Biol Chem. 2010. 53:8–12.
32. Perkins DJ, Kniss DA. Blockade of nitric oxide formation down-regulates cyclooxygenase-2 and decreases PGE2 biosynthesis in macrophages. J Leukoc Biol. 1999. 65:792–799.
crossref
33. Tsatsanis C, Androulidaki A, Venihaki M, Margioris AN. Signalling networks regulating cyclooxygenase-2. Int J Biochem Cell Biol. 2006. 38:1654–1661.
crossref
34. Wallach D, Varfolomeev EE, Malinin NL, Goltsev YV, Kovalenko AV, Boldin MP. Tumor necrosis factor receptor and Fas signaling mechanisms. Ann Rev Immunol. 1999. 17:331–367.
crossref
35. Aggarwal BB, Natarajan K. Tumor necrosis factors: developments during the last decade. Eur Cytokine Netw. 1996. 7:93–124.
36. Kim JY, Park SJ, Yun KJ, Cho YW, Park HJ, Lee KT. Isoliquiritigenin isolated from the roots of Glycyrrhiza uralensis inhibits LPS-induced iNOS and COX-2 expression via the attenuation of NF-kB in RAW264.7 macrophages. Eur J Pharmacol. 2008. 584:175–184.
crossref
37. Jung WK, Choi I, Lee DY, Yea SS, Choi YH, Kim MM, Park SG, Seo SK, Lee SW, Lee CM, Park YM, Choi IW. Caffeic acid phenethyl ester protects mice from lethal endotoxin shock and inhibits lipopolysaccharide-induced cyclooxygenase-2 and inducible nitric oxide synthase expression in RAW 264.7 macrophages via the p38/ERK and NF-kB pathways. Int J Biochem Cell Biol. 2008. 40:2572–2582.
crossref
38. Cheenpracha S, Park EJ, Yoshida WY, Barit C, Wall M, Pezzuto JM, Chang LC. Potential anti-inflammatory phenolic glycosides from the medicinal plant Moringa oleifera fruits. Bioorg Med Chem. 2010. 18:6598–6602.
crossref
39. Mueller M, Hobiger S, Jungbauer A. Anti-inflammatory activity of extracts from fruits, herbs and spices. Food Chem. 2010. 122:987–999.
crossref
TOOLS
Similar articles