Next Article in Journal
New Generation of Meso and Antiprogestins (SPRMs) into the Osteoporosis Approach
Next Article in Special Issue
Hybrid Quinoline-Thiosemicarbazone Therapeutics as a New Treatment Opportunity for Alzheimer’s Disease‒Synthesis, In Vitro Cholinesterase Inhibitory Potential and Computational Modeling Analysis
Previous Article in Journal
Revealing DNA Structure at Liquid/Solid Interfaces by AFM-Based High-Resolution Imaging and Molecular Spectroscopy
Previous Article in Special Issue
Stereoselective Synthesis of the Di-Spirooxindole Analogs Based Oxindole and Cyclohexanone Moieties as Potential Anticancer Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of the Inhibitory Effects of Pyridylpyrazole Derivatives on LPS-Induced PGE2 Productions and Nitric Oxide in Murine RAW 264.7 Macrophages

1
National Research Centre, Pharmaceutical and Drug Industries Research Division, Dokki-Giza 12622, Egypt
2
Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
3
Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
4
Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
5
Molecular Imaging & Therapeutic Medicine Research Center, Department of Nuclear Medicine, Jeonbuk National University Medical School and Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
6
Research Institute of Clinical Medicine, Jeonbuk National University-Biomedical Research Institute, Jeonbuk National University Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
7
Department of Pharmaceutical Biochemistry, Kyung Hee University, Seoul 02447, Korea
8
Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Seoul 02447, Korea
9
Center for Biomaterials, Korea Institute of Science and Technology, P.O. Box 131, Cheongryang, Seoul 130-650, Korea
*
Authors to whom correspondence should be addressed.
Submission received: 7 September 2021 / Revised: 7 October 2021 / Accepted: 26 October 2021 / Published: 27 October 2021

Abstract

:
A series of thirteen triarylpyrazole analogs were investigated as inhibitors of lipopolysaccharide (LPS)-induced prostaglandin E2 (PGE2) and nitric oxide (NO) production in RAW 264.7 macrophages. The target compounds 1am have first been assessed for cytotoxicity against RAW 264.7 macrophages to determine their non-cytotoxic concentration(s) for anti-inflammatory testing to make sure that the inhibition of PGE2 and NO production would not be caused by cytotoxicity. It was found that compounds 1f and 1m were the most potent PGE2 inhibitors with IC50 values of 7.1 and 1.1 μM, respectively. In addition, these compounds also showed inhibitory effects of 11.6% and 37.19% on LPS-induced NO production, respectively. The western blots analysis of COX-2 and iNOS showed that the PGE2 and NO inhibitory effect of compound 1m are attributed to inhibition of COX-2 and iNOS protein expression through inactivation of p38.

Graphical Abstract

1. Introduction

Inflammation is considered as a part of our body’s defense mechanisms against invasive organisms. It represents an attempt to get rid of such harmful organisms through releasing antibacterial or antiviral from cells close to it to help the body fight against infection [1]. In addition, it enhances injured tissue healing facilitating the return of the cells to their normal conditions. Despite these beneficial effects, it could have harmful effects triggering a list of disorders such as cardiovascular disorders [2], tumors [3], inflammatory bowel syndrome [4], arthritis [5], pulmonary disorders [6], Alzheimer’s [7], etc.
In order to treat inflammation, it is crucial to understand the role of inflammatory mediators that directly contribute to inflammatory responses. Inflammatory mediators arise from plasma proteins or some types of cells such as mast cells, platelets, neutrophils, monocytes, and macrophages. They are triggered by bacterial toxins or host cell proteins. The inflammatory mediators bind to particular receptors on the target cells and enhance vascular permeability and neutrophil chemotaxis, induce smooth muscle contraction, directly affect enzymatic activity, produce pain, or induce oxidative damage. The majority of these chemical mediators have short lives but produce harmful effects [1]. The inflammatory chemical mediators are exemplified by vasoactive amines (e.g., histamine and 5-HT), eicosanoids (e.g., prostaglandins and leukotrienes), and cytokines (e.g., tumor necrosis factor (TNF) and interleukin-1 (IL-1)).
Cyclooxygenase-2 (COX-2) converts arachidonic acid into PGE2, which is the mediator of inflammation [8]. Limiting PGE2 production via inhibition of COX-2 protein expression and/or enzymatic activity is another useful approach for the treatment of inflammation. Moreover, nitric oxide (NO) has another considerable contribution to inflammation development (although it could produce anti-inflammatory effect under other normal physiological conditions) [9,10,11]. On the other hand, it acts as a proinflammatory mediator to induce localized inflammatory response due to elevated secretion in cases of abnormal conditions. Inducible nitric oxide synthase (iNOS) enzyme forms NO in case of inflammation. NO produces localized vasodilation at the site of inflammation, leading to edema [12]. Therefore, Similar to PGE2 production inhibition, decreasing NO production via iNOS enzymatic activity inhibition, and/or iNOS protein expression inhibition could be a beneficial avenue for the management of inflammation.
Many substituted pyrazole derivatives have been recently reported to possess anti-inflammatory activity [13,14,15,16]. In our study, we evaluated a series of substituted pyrazole derivatives with a structural likeness to celecoxib, a pyrazole-based anti-inflammatory agent (Figure 1) as inhibitors of LPS-induced NO and PGE2 productions. Vicinal diarylheterocycles such as celecoxib have been reported as COX-2-inhibiting anti-inflammatory agents. The presence of vicinal diarylpyrazole scaffold in the structures of our target compounds encouraged us to investigate their anti-inflammatory activity. Our target compounds 1am were previously reported as antiproliferative agents [17]. Moreover, compound I (Figure 1) possessing triarylpyrazole nucleus has been reported as inhibitor of PGE2 and NO release [18].

2. Results and Discussion

2.1. Chemistry

The final compounds 1am were synthesized via the pathway demonstrated in Scheme 1. 2-Chloro-5-methylphenol (2) was reacted with dimethyl sulfate/potassium carbonate to obtain methoxy derivative. The methyl group was then oxidized to carboxylic acid by potassium permanganate to 4-chloro-3-methoxy-benzoic acid. Esterification of the resulting acid by methanol and acetyl chloride yielded the corresponding methyl ester 3. Compound 3 was activated using a strong base; lithium bis(trimethylsilyl)amide (LiHMDS) followed by slow addition of 4-picoline gave the pyridine ketide intermediate 4. The reaction of compound 4 with dimethylformamide dimethylacetal (DMF-DMA) produced compound 5. After that, adding hydrazine monohydrate yielded the pyrazolyl intermediate 6. Interaction of compound 6 with meta-iodonitrobenzene at 90 °C in dimethyl sulfoxide gave the meta-nitrophenyl intermediate 7. Reduction of the NO2 group of 7 using Pd/C and hydrogen gas produced amino compound 8. Interaction of the amino intermediate 8 with chloroacetyl chloride or chloropropionyl chloride produced the corresponding amide intermediates 9a,b, respectively. Interaction of the terminal alkyl halide group of compounds 9a,b with (substituted) alicyclic amines gave the target compounds 1am [17]. The detailed experimental procedures and the spectral analysis charts are shown in the Supplementary File. Structures of compounds 1am and their cell viability results against RAW 264.7 cells are shown in Table 1.

2.2. Biological Evaluation

Before screening the PGE2 and NO production inhibitory effects of the compounds, the compounds’ cytotoxicity was evaluated at 1 and 10 μM concentrations to make sure that the tested concentrations are safe enough and non-cytotoxic to avoid misleading results. All compounds were non-cytotoxic at 1 μM concentration, while by increasing concentration to 10 μM, all the target compounds except 1f, 1k, and 1m started showing cytotoxicity. These three compounds were found to be non-cytotoxic at 10 μM concentration (Figure 2). The three compounds possess N-benzylpiperazinyl, N-phenylpiperazinyl, and N-(4-fluorobenzyl)piperazinyl moieties, respectively. The piperidinyl and the morpholino moieties seem to be unfavorable to avoid cytotoxicity in this series of compounds.
Upon confirming the non-cytotoxicity of 10 μM concentration of these three derivatives against murine RAW 264.7 macrophages induced by LPS, compounds 1f, 1k, and 1m were tested for inhibitory effect against LPS-induced PGE2 production together with checking their cell viability. They have shown no cytotoxicity at these levels (2.5, 5, 10 μM) and good inhibition values against PGE2 production (Figure 2 and Figure 3). Among these selected derivatives, compounds 1f and 1m showed dose-dependent inhibition along with increasing its concentration (37.4 at 5 μM to 65.4% at 10 μM) for compound 1f, and 67% at 2.5 μM and 84.9% at 10 μM for compound 1m. Furthermore, the IC50 values of compounds 1f and 1m were 7.6 and 1.1 μM, respectively. This indicates that compound 1m with ethylene bridge was more active than compound 1f possessing methylene bridge. The fluorine atom of compound 1m might confer more lipophilicity that may result in more penetration inside the cell and hence better inhibition of PGE2 production. The fluorine atom can also add some more merits such as formation of an additional hydrogen bond with a hydrogen bond donor in the target protein and stronger hydrophobic interaction by fluorophenyl compared with unsubstituted phenyl. In addition, p-fluoro can prevent aromatic hydroxylation metabolic reaction and hence can elongate the duration of action [19]. Moreover, 1f and 1m were evaluated at 1 and 10 μM for inhibitory effects on LPS-induced NO production. It was found that compounds 1f and 1m showed inhibition values of 11.06% and 37.19%, respectively on LPS-induced NO production at 10 µM concentration (Table 2). Compound 1m is slightly more active than L-NIL at 10 µM.
Compounds 1f, 1k, and 1m were also tested for inhibitory effects against LPS-induced PGE2 production in addition to checking their cell viability. They have shown no cytotoxicity at these levels and good inhibition values against PGE2 production (Figure 2 and Figure 3). Among these selected derivatives, compounds 1f and 1m showed dose-dependent inhibition along with increasing its concentration (37.4% at 5 μM to 65.4% at 10 μM) for compound 1f, and 67% at 2.5 μM and 84.9% at 10 μM for compound 1m. Furthermore, the IC50 values of compounds 1f and 1m were 7.6 and 1.1 μM, respectively. This complies that compound 1m with ethylene bridge was more active than compound 1f possessing methylene bridge. The fluorine atom of compound 1m might confer more lipophilicity that may result in more penetration inside the cell and hence better inhibition of PGE2 production.
Furthermore, the most promising compound 1m was chosen for a more extensive investigation of its molecular mechanism(s) of action. It was tested for inhibitory effects on COX-2 and iNOS protein expressions (Figure 4). Compound 1m showed a concentration-dependent inhibitory effect against COX-2 and iNOS protein expression, especially at 10 µM concentration. Moreover, compound 1m markedly suppressed the phosphorylation of p38, a key molecule in regulating inflammation [20].

3. Conclusions

Our target compounds were tested for potential cytotoxicity. We then selected the safest compounds for further investigations as PGE2 and NO production inhibitors in LPS-induced murine RAW 264.7 macrophages. The two tested compounds (1f and 1m) act more against PGE2 production than over NO. We identified a couple of potential PGE2 production inhibitory compounds, namely 1f and 1m. The most potent compound, 1m, exerted a strong inhibitory effect on PGE2 production with IC50 value of 1.1 μM and NO production with 37% at 10 μM. It produces these effects due to inhibition of both COX-2 and iNOS protein expression through inactivation of p38. Further structural optimization is needed in order to optimize activity.

Supplementary Materials

The experimental procedures and the spectral analysis charts are available.

Author Contributions

Conceptualization, C.-H.O., K.-T.L., and H.-K.K.; methodology, M.M.G.E.-D., M.I.E.-G., S.-Y.K., H.-S.H., and S.-E.P.; data curation, Y.-D.K., writing-original draft preparation, M.M.G.E.-D. and M.I.E.-G.; writing-review and editing, M.M.G.E.-D., M.I.E.-G., C.-H.O., K.-T.L., and H.-K.K.; supervision, C.-H.O., K.-T.L., and H.-K.K.; funding acquisition, H.-K.K.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NRF-2021R1A2C1011204), and BK21FOUR 21st Century of Medical Science Creative Human Resource Development Center.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Experimental

The synthetic procedures and the protocols for the biological assay are provided in the Supplementary File.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are available from the authors upon request.

References

  1. Abdulkhaleq, L.A.; Assi, M.A.; Abdullah, R.; Zamri-Saad, M. The crucial roles of inflammatory mediators in inflammation: A review. Vet. World 2018, 11, 627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860. [Google Scholar] [CrossRef] [PubMed]
  3. Qui, H.; Johansson, A.S.; Sjostrom, M.; Wan, M.; Schroder, O.; Palmbald, J.; Haeggstrom, J.Z. Differential induction of BLT receptor expression on human endothelial cells by lipopolysaccharide, cytokines, and leukotriene B4. Proc. Natl. Acad. Sci. USA 2006, 103, 6913. [Google Scholar]
  4. Sung, B.; Prasad, S.; Yadav, V.R.; Lavasanifar, A.; Aggarwal, B.B. Cancer and diet: How are they related? Free Radical Res. 2011, 45, 864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lee, I.-A.; Bae, E.-A.; Hyun, Y.-J.; Kim, D.-H. Dextran sulfate sodium and 2,4,6-trinitrobenzene sulfonic acid induce lipid peroxidation by the proliferation of intestinal gram-negative bacteria in mice. J. Inflamm. 2010, 7, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hochberg, M.C. Semin. Changes in the incidence and prevalence of rheumatoid arthritis in England and Wales, 1970-1982. Arthritis Rheum. 1990, 19, 294. [Google Scholar] [CrossRef]
  7. Sastre, M.; Richardson, J.C.; Gentleman, S.M.; Brooks, D.J. Inflammatory risk factors and pathologies associated with Alzheimer’s disease. Curr. Alzheimer Res. 2011, 8, 132. [Google Scholar] [CrossRef] [PubMed]
  8. Hinz, B.; Brune, K.J. Cyclooxygenase-2–10 years later. Pharmacol. Exp. Ther. 2002, 300, 367. [Google Scholar] [CrossRef] [PubMed]
  9. Bhardwaj, A.; Batchu, S.N.; Kaur, J.; Huang, Z.; Seubert, J.M.; Knaus, E.E. Cardiovascular properties of a nitric oxide releasing rofecoxib analogue: beneficial anti-hypertensive activity and enhanced recovery in an ischemic reperfusion injury model. ChemMedChem 2012, 7, 1365. [Google Scholar] [CrossRef] [PubMed]
  10. Bhardwaj, A.; Huang, Z.; Kaur, J.; Knaus, E.E. Rofecoxib analogues possessing a nitric oxide donor sulfohydroxamic acid (SO2NHOH) cyclooxygenase-2 pharmacophore: synthesis, molecular modeling, and biological evaluation as anti-inflammatory agents. ChemMedChem 2012, 7, 62. [Google Scholar] [CrossRef] [PubMed]
  11. Kaur, J.; Bhardwaj, A.; Huang, Z.; Knaus, E.E. Aspirin analogues as dual cyclooxygenase-2/5-lipoxygenase inhibitors: synthesis, nitric oxide release, molecular modeling, and biological evaluation as anti-inflammatory agents. ChemMedChem 2011, 7, 144. [Google Scholar] [CrossRef] [PubMed]
  12. Yun, H.Y.; Dawson, V.L.; Dawson, T.M. Neurobiology of nitric oxide. Crit. Rev. Neurobiol. 1996, 10, 291. [Google Scholar] [CrossRef] [PubMed]
  13. Arora, R.K.; Kaur, N.; Bansal, Y.; Bansal, G. Novel coumarin-benzimidazole derivatives as antioxidants and safer anti-inflammatory agents. Acta Pharm. Sin. B. 2014, 4, 368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Azelmat, J.; Fiorito, S.; Taddeo, V.A.; Genovese, S.; Epifano, F.; Grenier, D. Synthesis and evaluation of antibacterial and anti-inflammatory properties of naturally occurring coumarins. Phytochem. Lett. 2015, 13, 399. [Google Scholar] [CrossRef]
  15. Wei, W.; Wu, X.-W.; Deng, G.-G.; Yang, X.-W. Anti-inflammatory coumarins with short- and long-chain hydrophobic groups from roots of Angelica dahurica cv. Hangbaizhi. Phytochemistry 2016, 123, 58. [Google Scholar] [CrossRef] [PubMed]
  16. Srivastava, P.; Vyas, V.K.; Variya, B.; Patel, P.; Qureshi, G.; Ghate, M. Synthesis, anti-inflammatory, analgesic, 5-lipoxygenase (5-LOX) inhibition activities, and molecular docking study of 7-substituted coumarin derivatives. Bioorg. Chem. 2016, 67, 130. [Google Scholar] [CrossRef] [PubMed]
  17. Gamal El-Din, M.M.; El-Gamal, M.I.; Abdel-Maksoud, M.S.; Yoo, K.H.; Oh, C.-H. Design, synthesis, in vitro potent antiproliferative activity, and kinase inhibitory effects of new triarylpyrazole derivatives possessing different heterocycle terminal moieties. J. Enz. Inhibit. Med. Chem. 2019, 34, 1534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. El-Gamal, M.I.; Abdel-Maksoud, M.S.; Gamal El-Din, M.M.; Shin, J.-S.; Lee, K.-T.; Yoo, K.H.; Oh, C.-H. Synthesis, in vitro Antiproliferative and Antiinflammatory Activities, and Kinase Inhibitory effects of New 1,3,4-triarylpyrazole Derivatives. Anti-Cancer Agents Med. Chem. 2017, 17, 75. [Google Scholar] [CrossRef]
  19. Shah, P.; Westwell, A.D. The role of fluorine in medicinal chemistry. J. Enz. Inhibit. Med. Chem. 2007, 22, 527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Madkour, M.M.; Anbar, H.S.; El-Gamal, M.I. Current status and future prospects of p38α/MAPK14 kinase and its inhibitors. Eur. J. Med. Chem. 2021, 213, 113216. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structures of the lead compound I [18], celecoxib, and the target compounds 1am.
Figure 1. Structures of the lead compound I [18], celecoxib, and the target compounds 1am.
Molecules 26 06489 g001
Scheme 1. Reagents and conditions: (a) (i) (CH3)2SO4, K2CO3, acetone, reflux, 1 h, 95%; (ii) KMnO4, C5H5N, H2O, 50 °C, 24 h, then rt, 13 h, 90%; (iii) acetyl chloride, CH3OH, rt, 15 h, 85%; (b) 4-picoline, LiHMDS, THF, rt, overnight, 45%; (c) (i) DMF-DMA, rt, 18 h; (d) hydrazine monohydrate, C2H5OH, rt, overnight, 81%; (e) 1-iodo-4-nitrobenzene, K2CO3, CuI, L-proline, DMSO, 90 °C, 8 h, 86%; (f) H2, Pd/C, THF, rt, 2 h, 86%; (g) chloroacetyl chloride, or chloropropionyl chloride, TEA, CH2Cl2, −10 °C, 15 min, 65%; (h) appropriate amine derivative, TEA, CH2Cl2, rt, 1 h, 46–71%.
Scheme 1. Reagents and conditions: (a) (i) (CH3)2SO4, K2CO3, acetone, reflux, 1 h, 95%; (ii) KMnO4, C5H5N, H2O, 50 °C, 24 h, then rt, 13 h, 90%; (iii) acetyl chloride, CH3OH, rt, 15 h, 85%; (b) 4-picoline, LiHMDS, THF, rt, overnight, 45%; (c) (i) DMF-DMA, rt, 18 h; (d) hydrazine monohydrate, C2H5OH, rt, overnight, 81%; (e) 1-iodo-4-nitrobenzene, K2CO3, CuI, L-proline, DMSO, 90 °C, 8 h, 86%; (f) H2, Pd/C, THF, rt, 2 h, 86%; (g) chloroacetyl chloride, or chloropropionyl chloride, TEA, CH2Cl2, −10 °C, 15 min, 65%; (h) appropriate amine derivative, TEA, CH2Cl2, rt, 1 h, 46–71%.
Molecules 26 06489 sch001
Figure 2. Effects of compounds 1f (a), 1k (b), 1m (c), NS398, and L-NIL on LPS-induced RAW 264.7 cell viability at various concentrations (2.5, 5, or 10 μM).
Figure 2. Effects of compounds 1f (a), 1k (b), 1m (c), NS398, and L-NIL on LPS-induced RAW 264.7 cell viability at various concentrations (2.5, 5, or 10 μM).
Molecules 26 06489 g002
Figure 3. Effects of compounds 1f (a), 1k (b), and 1m (c), and NS398 on LPS-stimulated PGE2 production in RAW 264.7 macrophages. # means significant difference from the negative control and *** means significant difference from the positive control.
Figure 3. Effects of compounds 1f (a), 1k (b), and 1m (c), and NS398 on LPS-stimulated PGE2 production in RAW 264.7 macrophages. # means significant difference from the negative control and *** means significant difference from the positive control.
Molecules 26 06489 g003
Figure 4. Effect of compound 1m on COX-2, iNOS, and p-p38 protein expression in LPS-induced RAW 264.7 macrophages.
Figure 4. Effect of compound 1m on COX-2, iNOS, and p-p38 protein expression in LPS-induced RAW 264.7 macrophages.
Molecules 26 06489 g004
Table 1. Structures of compounds 1am and their cell viability results at 1 and 10 μM concentrations against murine RAW 264.7 macrophages.
Table 1. Structures of compounds 1am and their cell viability results at 1 and 10 μM concentrations against murine RAW 264.7 macrophages.
Molecules 26 06489 i001
Compound No.nRCell Viability (%)
1 μM a10 μM a
1a1 Molecules 26 06489 i00299 ± 5.910 ± 1.2
1b1 Molecules 26 06489 i00392 ± 4.565 ± 2.8
1c1 Molecules 26 06489 i00478 ± 3.35 ± 0.8
1d1 Molecules 26 06489 i00575 ± 2.93 ± 0.4
1e1 Molecules 26 06489 i00687 ± 4.165 ± 4.1
1f1 Molecules 26 06489 i00766 ± 1.8107± 8.5
1g2 Molecules 26 06489 i00887 ± 3.85 ± 1.1
1h2 Molecules 26 06489 i00999 ± 2.875 ± 1.7
1i2 Molecules 26 06489 i01099 ± 3.45 ± 1.8
1j2 Molecules 26 06489 i01198 ± 3.15 ± 0.8
1k2 Molecules 26 06489 i01289 ± 2.8112 ± 7.6
1l2 Molecules 26 06489 i01386 ± 3.734 ± 1.1
1m2 Molecules 26 06489 i01490 ± 4.197 ± 3.9
a Data are presented as the means ± SDs of three independent experiments. Bold figures indicate non-cytotoxicity. * Indicates the site of connection to the main structure.
Table 2. Inhibitory effects of compounds 1f and 1m against NO production in LPS-induced RAW 264.7 cells.
Table 2. Inhibitory effects of compounds 1f and 1m against NO production in LPS-induced RAW 264.7 cells.
CompoundInhibition Rate (%)
1 (μM)10 (μM)
1f011.06 ± 1.5
1m037.19 ± 3.4
L-NIL3.9 ± 2.131.32 ± 2.9
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gamal El-Din, M.M.; El-Gamal, M.I.; Kwon, Y.-D.; Kim, S.-Y.; Han, H.-S.; Park, S.-E.; Oh, C.-H.; Lee, K.-T.; Kim, H.-K. Evaluation of the Inhibitory Effects of Pyridylpyrazole Derivatives on LPS-Induced PGE2 Productions and Nitric Oxide in Murine RAW 264.7 Macrophages. Molecules 2021, 26, 6489. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26216489

AMA Style

Gamal El-Din MM, El-Gamal MI, Kwon Y-D, Kim S-Y, Han H-S, Park S-E, Oh C-H, Lee K-T, Kim H-K. Evaluation of the Inhibitory Effects of Pyridylpyrazole Derivatives on LPS-Induced PGE2 Productions and Nitric Oxide in Murine RAW 264.7 Macrophages. Molecules. 2021; 26(21):6489. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26216489

Chicago/Turabian Style

Gamal El-Din, Mahmoud M., Mohammed I. El-Gamal, Young-Do Kwon, Su-Yeon Kim, Hee-Soo Han, Sang-Eun Park, Chang-Hyun Oh, Kyung-Tae Lee, and Hee-Kwon Kim. 2021. "Evaluation of the Inhibitory Effects of Pyridylpyrazole Derivatives on LPS-Induced PGE2 Productions and Nitric Oxide in Murine RAW 264.7 Macrophages" Molecules 26, no. 21: 6489. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26216489

Article Metrics

Back to TopTop