Biochemical and histopathological assessment of atorvastatin-loaded nanoemulsion effectiveness in Rats

Authors

  • Khadijah S. Balamash Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
  • Mona S. Al-ddyni Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia

DOI:

https://doi.org/10.18203/2319-2003.ijbcp20180009

Keywords:

Atorvastatin, Cholesterol, Fat, Kidney functions structure, Liver, Nanoparticles

Abstract

Background: Atorvastatin (ATV), a lipid lowering agent, has low solubility and poor dissolution affects its oral bioavailability. Nanoemulsion (NE) has been developed to improve the delivery of therapeutic agents. This study was aimed to assess the ability of the NE in enhancing ATV bioavailability and minimizing its side effects in hyperlipidemic rats.

Methods: Thirty-five rats divided into seven groups were utilized in this study. Hyperlipidemia was induced by feeding rats high fat diet (HFD) for 3 months. The antihyperlipidemic activity of 10 and 20 mg/kg of ATV loaded in two different delivery systems; nanoemulsion (10% and 20% ATV-LNE) or in water (10% and 20% ATV-sol), were investigated. At the end of the experiment, body weight, serum and plasma biochemical parameters (lipid profile, glucose, insulin, liver and kidney functions, oxidative stress markers were assessed. Liver and kidney were histopathologically examined. The physical characteristics of NE were determined by the Zetasizer (the z-average diameter and zeta potential).

Results: 20% ATV-LNE had the smallest nanoparticles (38.12±6.71nm) whereas it had the largest zeta negative potential of -26.8±4.16mV. The serum biochemical results and the histopathological examination revealed that treatment with 20% ATV-LNE improved the lipid profile by significantly increasing HDL and decreasing cholesterol and low-density lipoprotein. Both 10 and 20% ATV-LNE reduced serum glucose level compared to other used formulas.

Conclusions: NE formulas have the potential to improve the bioavailability and efficacy of ATV and reduce its side effects.

References

Mishra PR, Panda PK, Korla A, Panigrahi S. Evaluation of acute hypolipidemic activity of different plant extracts in triton Wr 1339 induced hyperlipidemia in albino rats. Pharmacologyonline. 2011;3:925-34.

Rodgers M. An Analysis of the Cause, Effect and Control of Hyperlipidemia from a Nursing Perspective. 2014.

Shattat GF. A Review Article on Hyperlipidemia: Types, Treatments and New Drug Targets. Biomedical and Pharmacology Journal. 2014;7:399-409.

Maji D, Shaikh S, Solanki D, Gaurav K. Safety of statins. Indian J Endocrinol Metab. 2014;17:636.

Meena A, Ratnam DV, Chandraiah G, Ankola D, Rao PR, Kumar MR. Oral nanoparticulate atorvastatin calcium is more efficient and safe in comparison to Lipicure® in treating hyperlipidemia. Lipids. 2008;43:231-41.

Shitara Y, Sugiyama Y. Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions. Pharmacol Therap. 2006;112:71-105.

Lea AP, Mctavish D. Atorvastatin. A review of its pharmacology and therapeutic potential in the. Drugs. 1997;53:828-47.

Wilczewska AZ, Niemirowicz K, Markiewicz KH, Car H. Nanoparticles as drug delivery systems. Pharmacological reports. 2012;64:1020-37.

Nevozhay D, Kańska U, Budzyńska R, Boratyński J. Current status of research on conjugates and related drug delivery systems in the treatment of cancer and other diseases. Postepy higieny i medycyny doswiadczalnej. 2006;61:350-60.

Shang L, Nienhaus K, Nienhaus GU. Engineered nanoparticles interacting with cells: size matters. J Nanobiotech. 2014;12:1.

Mason TG, Wilking J, Meleson K, Chang C, Graves S. Nanoemulsions: formation, structure, and physical properties. Journal of Physics: 2006;18:R635.

Liu P, Kurihara-Bergstrom T, Good WR. Cotransport of estradiol and ethanol through human skin in vitro: understanding the permeant/enhancer flux relationship. Pharmaceutical research. 1991;8:938-44.

Kadu PJ, Kushare SS, Thacker DD, Gattani SG. Enhancement of oral bioavailability of atorvastatin calcium by self-emulsifying drug delivery systems (SEDDS). Pharm Develop Technol. 2011;16:65-74.

Alkhatib MH, Albishi HM. In vitro evaluation of antitumor activity of doxorubicin-loaded nanoemulsion in MCF-7 human breast cancer cells. Journal of nanoparticle research. 2013;15:1-15.

Chouksey R, Pandey H, Jain A, Soni H, Saraogi G. Preparation and evaluation of the self-emulsifying drug delivery system containing atorvastatin HMG-CoA inhibiter. Int J Pharm Pharm Sci. 2011;3:147-52.

Alkhatib MH, Al-qaidi WA. Cytotoxicity effect of docetaxel-loadedmicroemulsion in A-549 non-small cell lung cancer and HCT116 colon cancer cells. Int J Pharm Bio Sci. 2014;5:300-15.

Basuny AM, Gaafar AM, Arafat SM. Tomato lycopene is a natural antioxidant and can alleviate hypercholesterolemia. African J Biotech. 2009;8.

Pedersen TR. Lipids: HDL, LDL, Role in Primary Prevention, the Message from Trials? Atherosclerosis: The 21st Century Epidemic. 2010;65.

Ahjel SW, Lupuleasa D. Enhancement of solubility and dissolution rate of different forms of atorvastatin calcium in direct compression tablet formulas. Farmacia. 2009;57:290-300.

Rohilla R, Garg T, Bariwal J, Goyal AK, Rath G. Development, optimization and characterization of glycyrrhetinic acid-chitosan nanoparticles of atorvastatin for liver targeting. Drug delivery. 2016;23:2290-7.

Kumar PP, Gayatri P, Sunil R, Jaganmohan S, Rao YM. Atorvastatin loaded solid lipid nanoparticles: formulation, optimization, and in vitro characterization. IOSR J Pharm. 2012;2:23-32.

Lovelyn C, Attama AA. Current state of nanoemulsions in drug delivery. J Biomat Nanobiotech. 2011;2:626.

Bathool A, Vishakante GD, Khan MS, Shivakumar H. Development and characterization of atorvastatin calcium loaded chitosan nanoparticles for sustain drug delivery. Adv Mat Lett. 2012;3:466-70.

Jahangiri A, Barzegar-Jalali M, Garjani A, Javadzadeh Y, Hamishehkar H, Afroozian A, et al. Pharmacological and histological examination of atorvastatin-PVP K30 solid dispersions. Powder Technology. 2015;286:538-45.

Rogers SL, Magliano DJ, Levison DB, Webb K, Clarke PJ, Grobler MP, et al. A dose-specific meta-analysis of lipid changes in randomized controlled trials of atorvastatin and simvastatin. Clini Therapeut. 2007;29:242-52.

Omar MA, Wilson JP, Cox TS. Rhabdomyolysis and HMG-CoA reductase inhibitors. Ann Pharmacotherap. 2001;35:1096-107.

Li X, Qi J, Xie Y, Zhang X, Hu S, Xu Y, et al. Nanoemulsions coated with alginate/chitosan as oral insulin delivery systems: preparation, characterization, and hypoglycemic effect in rats. Inte J Nanomed. 2013;8:23.

Smith CC, Bernstein LI, Davis RB, Rind DM, Shmerling RH. Screening for statin-related toxicity: the yield of transaminase and creatine kinase measurements in a primary care setting. Arch Internal Med. 2003;163:688-92.

Tolman K. Lovastatin and the liver. Am J Cardiol. 2002;89:1374-80.

Bader T. Liver tests are irrelevant when prescribing statins. The Lancet. 2010;376:1882-3.

Ayuob, NN. Can raisins ameliorate hypercholesterolemia-induced nephropathy? What is the evidence? The Egyptian Journal of Histology. 2014;37:677-88.

Aktas A, Tasdemir MS, Tuncer MC, Nergiz Y, Akkus M, Bagriyanik HA, et al. Atorvastatin has no effects on kidney tissues of wistar albino rats in the long-term Intake: an electron microscopic Study. Int J Morphol. 2011;29:144-50.

Downloads

Published

2018-01-23

How to Cite

Balamash, K. S., & Al-ddyni, M. S. (2018). Biochemical and histopathological assessment of atorvastatin-loaded nanoemulsion effectiveness in Rats. International Journal of Basic & Clinical Pharmacology, 7(2), 221–228. https://doi.org/10.18203/2319-2003.ijbcp20180009

Issue

Section

Original Research Articles