Generic placeholder image

Current Drug Discovery Technologies

Editor-in-Chief

ISSN (Print): 1570-1638
ISSN (Online): 1875-6220

Research Article

One-Step Preparation of Luteolin Nanoemulsion and Evaluation of its Anti-inflammatory Effect in Animal Models

Author(s): Somayyeh Karami-Mohajeri, Narges Hashemi, Mehdi Ranjbar*, Mohammad Mohajeri and Fariba Sharififar*

Volume 20, Issue 3, 2023

Published on: 05 April, 2023

Article ID: e270323215003 Pages: 7

DOI: 10.2174/1570163820666230327133142

Price: $65

Abstract

Background: Nanoemulsions are promising drug delivery systems for topical application owing to the high transdermal penetration.

Objective: Due to the side effects of existing anti-inflammatory drugs, much attention has been paid to natural products such as flavonoids. The aim of this work was to formulate luteolin nanoemulsion (LNE) and to evaluate its anti-inflammatory effect.

Methods: LNE was prepared using the low-energy spontaneous emulsion method and characterized by transmission electron microscopy (TEM), Fourier transform infrared (FTIR) spectroscopy and dynamic light scattering (DLS). The anti-inflammatory effect of LNE was assessed in formalin and acetic acid-induced inflammation methods (Whittle test). Treatment with LNE (i.p, 4 consecutive days, 40 mg/kg) was compared with diclofenac 25 mg/kg and normal saline. In the formalin test, data were recorded at 1, 2 and 4 hours after formalin injection and in the Wittle test, the extent of Evans blue leakage in the peritoneal cavity was considered as vascular permeability.

Results: Formalin-induced edema decreased in the LNE group, but this reduction was not significant (p > 0.05), however, in Whittle test, both LNE and diclofenac significantly reduced Evans blue leakage compared with the group treated with acetic acid alone (p < 0.05).

Conclusion: Our results confirm the anti-inflammatory effect of LNE and give up a new platform for the design and development of bio-based carriers for more successful drug delivery.

Keywords: Luteolin, anti-inflammatory effect, nanoemulsion, formalin test, whilttle method, characterization.

Graphical Abstract
[1]
Chen L, Deng H, Cui H, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018; 9(6): 7204-18.
[http://dx.doi.org/10.18632/oncotarget.23208] [PMID: 29467962]
[2]
Galley HF, Webster NR. The immuno-inflammatory cascade. Br J Anaesth 1996; 77(1): 11-6.
[http://dx.doi.org/10.1093/bja/77.1.11] [PMID: 8703619]
[3]
Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: Positioning cells for host defense and immunity. Annu Rev Immunol 2014; 32(1): 659-702.
[http://dx.doi.org/10.1146/annurev-immunol-032713-120145] [PMID: 24655300]
[4]
Mandegary A, Pournamdari M, Sharififar F, Pournourmohammadi S, Fardiar R, Shooli S. Alkaloid and flavonoid rich fractions of fenugreek seeds (Trigonella foenum-graecum L.) with antinociceptive and anti-inflammatory effects. Food Chem Toxicol 2012; 50(7): 2503-7.
[http://dx.doi.org/10.1016/j.fct.2012.04.020] [PMID: 22542922]
[5]
Sharififar F, Dehghn-Nudeh G, Mirtajaldini M. Major flavonoids with antioxidant activity from Teucrium polium L. Food Chem 2009; 112(4): 885-8.
[http://dx.doi.org/10.1016/j.foodchem.2008.06.064]
[6]
Pournamdari M, Mandegary A, Sharififar F, et al. Anti-inflammatory subfractions separated from acidified chloroform fraction of fenugreek seeds (Trigonella foenum-graecum L.). J Diet Suppl 2018; 15(1): 98-107.
[http://dx.doi.org/10.1080/19390211.2017.1326431] [PMID: 28558255]
[7]
Pandurangan AK, Esa NM. Luteolin, a bioflavonoid inhibits colorectal cancer through modulation of multiple signaling pathways: A review. Asian Pac J Cancer Prev 2014; 15(14): 5501-8.
[http://dx.doi.org/10.7314/APJCP.2014.15.14.5501] [PMID: 25081655]
[8]
Wall C, Lim R, Poljak M, Lappas M. Dietary flavonoids as therapeutics for preterm birth: Luteolin and kaempferol suppress inflammation in human gestational tissues in vitro. Oxid Med Cell Longev 2013; 2013: 1-10.
[http://dx.doi.org/10.1155/2013/485201] [PMID: 23840918]
[9]
Kao TK, Ou YC, Lin SY, et al. Luteolin inhibits cytokine expression in endotoxin/cytokine-stimulated microglia. J Nutr Biochem 2011; 22(7): 612-24.
[http://dx.doi.org/10.1016/j.jnutbio.2010.01.011] [PMID: 21036586]
[10]
Chen CY, Peng WH, Tsai KD, Hsu SL. Luteolin suppresses inflammation-associated gene expression by blocking NF-κB and AP-1 activation pathway in mouse alveolar macrophages. Life Sci 2007; 81(23-24): 1602-14.
[http://dx.doi.org/10.1016/j.lfs.2007.09.028] [PMID: 17977562]
[11]
Hu C, Kitts DD. Luteolin and luteolin-7-O-glucoside from dandelion flower suppress iNOS and COX-2 in RAW264.7 cells. Mol Cell Biochem 2004; 265(1/2): 107-13.
[http://dx.doi.org/10.1023/B:MCBI.0000044364.73144.fe] [PMID: 15543940]
[12]
Wu W, Li D, Zong Y, et al. Luteolin inhibits inflammatory responses via p38/MK2/TTP-mediated mRNA stability. Molecules 2013; 18(7): 8083-94.
[http://dx.doi.org/10.3390/molecules18078083] [PMID: 23839113]
[13]
Mastrotto F, Salmaso S, Alexander C, Mantovani G, Caliceti P. Novel pH-responsive nanovectors for controlled release of ionisable drugs. J Mater Chem B 2013; 1(39): 5335-46.
[14]
Goldim MPS, Della Giustina A, Petronilho F. Using evans blue dye to determine blood‐brain barrier integrity in rodents. Curr Protoc Immunol 2019; 126(1): e83.
[http://dx.doi.org/10.1002/cpim.83] [PMID: 31483106]
[15]
Altamimi MA, Hussain A, Alshehri S, Imam SS, Alnemer UA. Development and evaluations of transdermally delivered luteolin loaded cationic nanoemulsion: In vitro and ex vivo evaluations. Pharmaceutics 2021; 13(8): 1218.
[http://dx.doi.org/10.3390/pharmaceutics13081218] [PMID: 34452179]
[16]
Tawornchat P, Pattarakankul T, Palaga T, Intasanta V, Wanichwecharungruang S. Polymerized luteolin nanoparticles: synthesis, structure elucidation, and anti-inflammatory activity. ACS Omega 2021; 6(4): 2846-55.
[http://dx.doi.org/10.1021/acsomega.0c05142] [PMID: 33553902]
[17]
Imam SS, Alshehri S, Altamimi MA, et al. Formulation and evaluation of luteolin-loaded nanovesicles: In vitro physicochemical characterization and viability assessment. ACS Omega 2022; 7(1): 1048-56.
[http://dx.doi.org/10.1021/acsomega.1c05628] [PMID: 35036768]
[18]
Sharififar F, Ahmadi S, Mandegary A, Soodi M. Anticholinesterase, antioxidant, and neuroprotective effects of Tripleurospermum disciforme and Dracocephalum multicaule. J Ayurveda Integr Med 2014; 5(3): 162-6.
[http://dx.doi.org/10.4103/0975-9476.140474] [PMID: 25336847]
[19]
Esmaeili M, Zohari F, Sadeghi H. Antioxidant and protective effects of major flavonoids from Teucrium polium on beta-cell destruction in a model of streptozotocin-induced diabetes. Planta Med 2009; 75(13): 1418-20.
[http://dx.doi.org/10.1055/s-0029-1185704] [PMID: 19452438]
[20]
Karami-Mohajeri S, Najafi A, Behnam B, et al. Protective effect of Zataria multiflora Boiss. and its main compound, rosmarinic acid, against malathion induced oxidative stress and apoptosis in HepG2 cells. J Environ Sci Health B 2021; 56(4): 297-306.
[http://dx.doi.org/10.1080/03601234.2021.1879595] [PMID: 33560903]
[21]
Aziz N, Kim MY, Cho JY. Anti-inflammatory effects of luteolin: A review of in vitro, in vivo, and in silico studies. J Ethnopharmacol 2018; 225: 342-58.
[http://dx.doi.org/10.1016/j.jep.2018.05.019] [PMID: 29801717]
[22]
Seelinger G, Merfort I, Schempp C. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med 2008; 74(14): 1667-77.
[http://dx.doi.org/10.1055/s-0028-1088314] [PMID: 18937165]
[23]
Lee JO, Jeong D, Kim MY, Cho JY. ATP-binding pocket-targeted suppression of Src and Syk by luteolin contributes to its anti-inflammatory action. Mediators Inflamm 2015; 2015: 1-12.
[http://dx.doi.org/10.1155/2015/967053] [PMID: 26236111]
[24]
Rooban BN, Sasikala V, Gayathri Devi V, Sahasranamam V, Abraham A. Prevention of selenite induced oxidative stress and cataractogenesis by luteolin isolated from Vitex negundo. Chem Biol Interact 2012; 196(1-2): 30-8.
[http://dx.doi.org/10.1016/j.cbi.2012.01.005] [PMID: 22342831]
[25]
Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta Mol Cell Res 2014; 1843(11): 2563-82.
[http://dx.doi.org/10.1016/j.bbamcr.2014.05.014] [PMID: 24892271]
[26]
Funaro A, Wu X, Song M, et al. Enhanced anti-inflammatory activities by the combination of luteolin and tangeretin. J Food Sci 2016; 81(5): H1320-7.
[http://dx.doi.org/10.1111/1750-3841.13300] [PMID: 27095513]
[27]
Ampadu FA, Boakye-Gyasi E, Osafo N, Benneh CK, Ekuadzi E, Woode E. Antipleuritic and vascular permeability inhibition of the ethyl acetate-petroleum ether stem bark extract of Maerua angolensis DC (Capparaceae) in murine. Int J Inflamm 2018; 2018: 1-12.
[http://dx.doi.org/10.1155/2018/6123094] [PMID: 30112161]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy