Drug Res (Stuttg) 2023; 73(03): 125-136
DOI: 10.1055/a-1960-3092
Original Article

Curcumin-Loaded Chitosan Nanoparticle Preparation and Its Protective Effect on Celecoxib-induced Toxicity in Rat isolated Cardiomyocytes and Mitochondria

Hossein Ali Ebrahimi
1   Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
,
Samira Esmaeli
1   Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
,
Saleh Khezri
2   Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences
3   Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
,
Ahmad Salimi
2   Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences
3   Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
› Author Affiliations
Funding Information This work was supported by Ardabil of Medical Sciences, Deputy of Research with ethics code IR.ARUMS.REC.1398.355.

Abstract

Curcumin has a wide range of pharmacological activities, including antioxidant, anti-inflammatory and tissue protective. In here we hypothesized that curcumin-loaded chitosan-coated solid lipid nanoparticles (CuCsSLN) are able to increase its overall bioavailability and hence its antioxidant and mitochondria;/lysosomal protective properties of curcumin. CuCsSLN were prepared using solvent diffusion technique for formation of solid lipid nanoparticles (SLNs) and electrostatic coating of positive-charged chitosan to negative surface of SLNs. CuCsSLN showed the encapsulation efficiency of 91.4±2.7%, the mean particle size of 208±9 nm, the polydispersity index of 0.34±0.07, and the zeta potential of+53.5±3.7 mV. The scanning electron microscope (SEM) images of nanoparticles verified their nanometric size and also spherical shape. Curcumin was released from CuCsSLN in a sustain release pattern up to 24 hours. Then isolated cardiomyocytes and mitochondria were simultaneously treated with (1) control (0.05% ethanol), (2) celecoxib (20 µg/ml) treatment, (3) celecoxib (20 µg/ml)+++CuCsSLN (1 µg/ml) treatment, (4) CuCsSLN (1 µg/ml) treatment, (5) celecoxib (20 µg/ml)+++curcumin (10 µM) treatment and (6) curcumin (10 µM) treatment for 4 h at 37°C. The results showed that celecoxib (20 µg/ml) induced a significant increase in cytotoxicity, reactive oxygen species (ROS) formation, mitochondria membrane potential (ΔΨm) collapse, lipid peroxidation, oxidative stress and mitochondrial swelling while CuCsSLN and curcumin reverted the above toxic effect of celecoxib. Our data indicated that the effect of CuCsSLN in a number of experiments, is significantly better than that of curcumin which shows the role of chitosan nanoparticles in increasing effect of curcumin.



Publication History

Received: 14 June 2022

Accepted: 12 October 2022

Article published online:
24 November 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Patra JK, Das G, Fraceto LF. et al. Nano based drug delivery systems: recent developments and future prospects. Journal of nanobiotechnology 2018; 16: 71
  • 2 Parveen S, Misra R, Sahoo SK. Nanoparticles: a boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine: Nanotechnology, Biology and Medicine 2012; 8: 147-166
  • 3 Muthu MS, Leong DT, Mei L. et al. Nanotheranostics-application and further development of nanomedicine strategies for advanced theranostics. Theranostics 2014; 4: 660
  • 4 Mohammed MA, Syeda J, Wasan KM. et al. An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics 2017; 9: 53
  • 5 Park JH, Saravanakumar G, Kim K. et al. Targeted delivery of low molecular drugs using chitosan and its derivatives. Advanced drug delivery reviews 2010; 62: 28-41
  • 6 Upadhyaya L, Singh J, Agarwal V. et al. The implications of recent advances in carboxymethyl chitosan based targeted drug delivery and tissue engineering applications. Journal of Controlled Release 2014; 186: 54-87
  • 7 Deng Y, Zhang X, Shen H. et al. Application of the Nano-Drug Delivery System in Treatment of Cardiovascular Diseases. Frontiers in Bioengineering and Biotechnology 2019; 7
  • 8 Swamy MK, Sinniah UR. Patchouli (Pogostemon cablin Benth.): botany, agrotechnology and biotechnological aspects. Industrial Crops and Products 2016; 87: 161-176
  • 9 Beutler JA. Natural products as a foundation for drug discovery. Current protocols in pharmacology 2019; 86: e67
  • 10 Bonifácio BV, da Silva PB, dos Santos Ramos MA. et al. Nanotechnology-based drug delivery systems and herbal medicines: a review. International journal of nanomedicine 2014; 9: 1
  • 11 Watkins R, Wu L, Zhang C. et al. Natural product-based nanomedicine: recent advances and issues. International journal of nanomedicine 2015; 10: 6055
  • 12 Jahangirian H, Lemraski EG, Webster TJ. et al. A review of drug delivery systems based on nanotechnology and green chemistry: green nanomedicine. International journal of nanomedicine 2017; 12: 2957
  • 13 Patra JK, Baek K-H. Green nanobiotechnology: factors affecting synthesis and characterization techniques. Journal of Nanomaterials 2014; 2014
  • 14 Arayne MS, Sultana N, Qureshi F. nanoparticles in delivery of cardiovascular drugs. Pakistan journal of pharmaceutical sciences 2007; 20: 340-348
  • 15 Kou L, Sun J, Zhai Y. et al. The endocytosis and intracellular fate of nanomedicines: Implication for rational design. Asian Journal of Pharmaceutical Sciences 2013; 8: 1-10
  • 16 Mosler C. Cardiovascular risk associated with NSAIDs and COX-2 inhibitors. US Pharm 2014; 39: 35-38
  • 17 Varga Z. rafay ali Sabzwari S, Vargova V. Cardiovascular risk of nonsteroidal anti-inflammatory drugs: an under-recognized public health issue. Cureus 2017; e1144 DOI: 10.7759/cureus.1144.
  • 18 Caldwell B, Aldington S, Weatherall M. et al. Risk of cardiovascular events and celecoxib: a systematic review and meta-analysis. Journal of the Royal Society of Medicine 2006; 99: 132-140
  • 19 Arber N, Eagle CJ, Spicak J. et al. Celecoxib for the prevention of colorectal adenomatous polyps. New England Journal of Medicine 2006; 355: 885-895
  • 20 Ghosh R, Alajbegovic A, Gomes AV. NSAIDs and cardiovascular diseases: role of reactive oxygen species. Oxidative medicine and cellular longevity 2015; 536962 DOI: 10.1155/2015/536962.
  • 21 Lal N, Kumar J, Erdahl WE. et al. Differential effects of non-steroidal anti-inflammatory drugs on mitochondrial dysfunction during oxidative stress. Archives of biochemistry and biophysics 2009; 490: 1-8
  • 22 Salimi A, Neshat MR, Naserzadeh P. et al. Mitochondrial permeability transition pore sealing agents and antioxidants protect oxidative stress and mitochondrial dysfunction induced by naproxen, diclofenac and celecoxib. Drug research 2019; 69: 598-605
  • 23 Shabalala S, Muller C, Louw J. et al. Polyphenols, autophagy and doxorubicin-induced cardiotoxicity. Life sciences 2017; 180: 160-170
  • 24 Nabofa WE, Alashe OO, Oyeyemi OT. et al. Cardioprotective effects of curcumin-nisin based poly lactic acid nanoparticle on myocardial infarction in guinea pigs. Scientific reports 2018; 8: 1-11
  • 25 Morin D, Barthélémy S, Zini R. et al. Curcumin induces the mitochondrial permeability transition pore mediated by membrane protein thiol oxidation. FEBS letters 2001; 495: 131-136
  • 26 Xu Q, Ming Z, Dart AM. et al. Optimizing dosage of ketamine and xylazine in murine echocardiography. Clinical and Experimental Pharmacology and Physiology 2007; 34: 499-507
  • 27 Naserzadeh P, Mehr SN, Sadabadi Z. et al. Curcumin protects mitochondria and cardiomyocytes from oxidative damage and apoptosis induced by hemiscorpius lepturus venom. Drug research 2018; 68: 113-120
  • 28 Huang P, Zhang YH, Zheng XW. et al. Phenylarsine oxide (PAO) induces apoptosis in HepG2 cells via ROS-mediated mitochondria and ER-stress dependent signaling pathways. Metallomics 2017; 9: 1756-1764
  • 29 Aghvami M, Ebrahimi F, Zarei MH. et al. Matrine Induction of ROS Mediated Apoptosis in Human ALL B-lymphocytes Via Mitochondrial Targeting. Asian Pacific journal of cancer prevention : APJCP 2018; 19: 555-560 DOI: 10.22034/apjcp.2018.19.2.555.
  • 30 Salimi A, Roudkenar MH, Sadeghi L. et al. Selective anticancer activity of acacetin against chronic lymphocytic leukemia using both in vivo and in vitro methods: key role of oxidative stress and cancerous mitochondria. Nutrition and cancer 2016; 68: 1404-1416
  • 31 Hissin PJ, Hilf R. A fluorometric method for determination of oxidized and reduced glutathione in tissues. Analytical biochemistry 1976; 74: 214-226
  • 32 Beach DC, Giroux E. Inhibition of lipid peroxidation promoted by iron (III) and ascorbate. Archives of biochemistry and biophysics 1992; 297: 258-264
  • 33 Huang L, Zhang K, Guo Y. et al. Honokiol protects against doxorubicin cardiotoxicity via improving mitochondrial function in mouse hearts. Scientific reports 2017; 7: 1-12
  • 34 Salimi A, Roudkenar MH, Seydi E. et al. Chrysin as an anti-cancer agent exerts selective toxicity by directly inhibiting mitochondrial complex II and V in CLL B-lymphocytes. Cancer investigation 2017; 35: 174-186
  • 35 Hosseini M-J, Naserzadeh P, Salimi A. et al. Toxicity of cigarette smoke on isolated lung, heart, and brain mitochondria: induction of oxidative stress and cytochrome c release. Toxicological & Environmental Chemistry 2013; 95: 1624-1637
  • 36 Escobales N, Nuñez RE, Jang S. et al. Mitochondria-targeted ROS scavenger improves post-ischemic recovery of cardiac function and attenuates mitochondrial abnormalities in aged rats. Journal of molecular and cellular cardiology 2014; 77: 136-146
  • 37 Salimi A, Nikoosiar Jahromi M, Pourahmad J. Maternal exposure causes mitochondrial dysfunction in brain, liver, and heart of mouse fetus: An explanation for perfluorooctanoic acid induced abortion and developmental toxicity. Environmental toxicology 2019; 34: 878-885 DOI: 10.1002/tox.22760.
  • 38 Xu X-Y, Meng X, Li S. et al. Bioactivity, health benefits, and related molecular mechanisms of curcumin: Current progress, challenges, and perspectives. Nutrients 2018; 10: 1553
  • 39 Salatin S, Yari Khosroushahi A. Overviews on the cellular uptake mechanism of polysaccharide colloidal nanoparticles. Journal of Cellular and Molecular Medicine 2017; 21: 1668-1686 DOI: 10.1111/jcmm.13110.
  • 40 Dutta PK, Dutta J, Tripathi V. Chitin and chitosan: Chemistry, properties and applications. 2004
  • 41 Zhou Y, Li J, Lu F. et al. A study on the hemocompatibility of dendronized chitosan derivatives in red blood cells. Drug design, development and therapy 2015; 9: 2635
  • 42 Wu J, Liu G, Qin Y-X. et al. Effect of low-intensity pulsed ultrasound on biocompatibility and cellular uptake of chitosan-tripolyphosphate nanoparticles. Biointerphases 2014; 9: 031016
  • 43 Huang M, Khor E, Lim L-Y. Uptake and cytotoxicity of chitosan molecules and nanoparticles: effects of molecular weight and degree of deacetylation. Pharmaceutical research 2004; 21: 344-353
  • 44 Zhao L, Yang G, Shi Y. et al. Co-delivery of Gefitinib and chloroquine by chitosan nanoparticles for overcoming the drug acquired resistance. Journal of Nanobiotechnology 2015; 13: 57
  • 45 Eguchi S, Takefuji M, Sakaguchi T. et al. Cardiomyocytes capture stem cell-derived, anti-apoptotic microRNA-214 via clathrin-mediated endocytosis in acute myocardial infarction. Journal of Biological Chemistry 2019; 294: 11665-11674
  • 46 Ma Z, Lim L-Y. Uptake of chitosan and associated insulin in Caco-2 cell monolayers: a comparison between chitosan molecules and chitosan nanoparticles. Pharmaceutical research 2003; 20: 1812-1819
  • 47 Zheng A-p, Liu H-x, Yuan L. et al. Comprehensive studies on the interactions between chitosan nanoparticles and some live cells. Journal of Nanoparticle Research 2011; 13: 4765-4776
  • 48 [Anonymous]. !!! INVALID CITATION !!! {}
  • 49 Montaigne D, Hurt C, Neviere R. Mitochondria death/survival signaling pathways in cardiotoxicity induced by anthracyclines and anticancer-targeted therapies. Biochemistry research international. 2012 2012.
  • 50 Varga ZV, Ferdinandy P, Liaudet L. et al. Drug-induced mitochondrial dysfunction and cardiotoxicity. American Journal of Physiology-Heart and Circulatory Physiology 2015; 309: H1453-H1467
  • 51 Tatematsu Y, Fujita H, Hayashi H. et al. Effects of the nonsteroidal anti-inflammatory drug celecoxib on mitochondrial function. Biological and Pharmaceutical Bulletin 2018; 41: 319-325
  • 52 Davis RE, Williams M. Mitochondrial function and dysfunction: an update. Journal of Pharmacology and Experimental Therapeutics 2012; 342: 598-607
  • 53 Camara AK, Bienengraeber M, Stowe DF. Mitochondrial approaches to protect against cardiac ischemia and reperfusion injury. Frontiers in physiology 2011; 2: 13
  • 54 Galluzzi L, Kepp O, Trojel-Hansen C. et al. Mitochondrial control of cellular life, stress, and death. Circulation research 2012; 111: 1198-1207
  • 55 Siasos G, Tsigkou V, Kosmopoulos M. et al. Mitochondria and cardiovascular diseases—from pathophysiology to treatment. Annals of translational medicine 2018; 6: 256 DOI: 10.21037/atm.2018.06.21.
  • 56 Sheu S-S, Nauduri D, Anders M. Targeting antioxidants to mitochondria: a new therapeutic direction. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 2006; 1762: 256-265
  • 57 He H, Luo Y, Qiao Y. et al. Curcumin attenuates doxorubicin-induced cardiotoxicity via suppressing oxidative stress and preventing mitochondrial dysfunction mediated by 14-3-3γ. Food & Function 2018; 9: 4404-4418
  • 58 Correa F, Buelna-Chontal M, Hernández-Reséndiz S. et al. Curcumin maintains cardiac and mitochondrial function in chronic kidney disease. Free Radical Biology and Medicine 2013; 61: 119-129