Neuroprotective effect of naringenin-loaded solid lipid nanoparticles against streptozocin-induced neurotoxicity through autophagy blockage
Zeinab Nouri
Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Conceptualization, Investigation, Methodology, Writing - original draft, Writing - review & editing
Search for more papers by this authorSoraya Sajadimajd
Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
Contribution: Conceptualization, Methodology, Writing - review & editing
Search for more papers by this authorLeila Hoseinzadeh
Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Writing - review & editing
Search for more papers by this authorGholamreza Bahrami
Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Writing - review & editing
Search for more papers by this authorElham Arkan
Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Writing - review & editing
Search for more papers by this authorSajad Moradi
Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Investigation, Writing - review & editing
Search for more papers by this authorFereshteh Abdi
Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Investigation
Search for more papers by this authorCorresponding Author
Mohammad Hosein Farzaei
Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Correspondence
Mohammad Hosein Farzaei, Pharmaceutical Sciences Research Center, Kermanshah University of Medical, Sciences, Kermanshah, Iran.
Email: [email protected]
Contribution: Conceptualization, Supervision, Writing - review & editing
Search for more papers by this authorZeinab Nouri
Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Conceptualization, Investigation, Methodology, Writing - original draft, Writing - review & editing
Search for more papers by this authorSoraya Sajadimajd
Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
Contribution: Conceptualization, Methodology, Writing - review & editing
Search for more papers by this authorLeila Hoseinzadeh
Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Writing - review & editing
Search for more papers by this authorGholamreza Bahrami
Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Writing - review & editing
Search for more papers by this authorElham Arkan
Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Writing - review & editing
Search for more papers by this authorSajad Moradi
Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Investigation, Writing - review & editing
Search for more papers by this authorFereshteh Abdi
Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
Contribution: Investigation
Search for more papers by this authorCorresponding Author
Mohammad Hosein Farzaei
Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
Correspondence
Mohammad Hosein Farzaei, Pharmaceutical Sciences Research Center, Kermanshah University of Medical, Sciences, Kermanshah, Iran.
Email: [email protected]
Contribution: Conceptualization, Supervision, Writing - review & editing
Search for more papers by this authorZeinab Nouri and Soraya Sajadimajd contributed equally to this work.
Abstract
Autophagy is a pivotal contributing factor to modulate the progression of neurodegenerative diseases. Although naringenin (Nar) has shown beneficial effects against neurodegenerative diseases, its poor solubility and bioavailability have limited its application. The present research aimed to design a nanostructured formulation of Nar to achieve an enhanced therapeutic effect. Herein, Nar-loaded solid lipid nanoparticles (Nar-SLNs) were prepared and characterized. Then, PC12 cells were exposed to streptozocin (STZ) and/or Nar and Nar-SLNs in vitro to clarify the protective effect of Nar and its nanoformulation against STZ-stimulated neurotoxicity. The empty SLNs and Nar-SLNs indicated a narrow polydispersity index value with a negative zeta potential. As determined by the scanning electron microscopy images, the nanoparticles had a spherical shape and were less than 20 nm in size. FTIR results demonstrated the interaction between Nar and SLNs and supported the presence of Nar in the nanoparticle. The nanoformulation revealed an initial burst release followed by a sustained release manner. Treatment of PC12 cells with STZ resulted in mitochondrial dysfunction and increased autophagic markers, including LC3-II, Beclin1, Akt, ATG genes, and accumulation of miR-21 and miR-22. Both Nar and Nar-SLNs pre-treatment improved cell survival and augmented mitochondrial membrane potential, accompanied by reduced autophagic markers expression. However, Nar-SLNs were more effective than free Nar. As a result, our findings suggested that SLNs effectively enhance the neuroprotective effect of Nar, and Nar-SLNs may be a promising candidate to suppress or prevent STZ-elicited neurotoxicity.
Practical applications
According to the beneficial effect of Nar in the management of neurodegenerative diseases, we evaluated the protective effect of Nar and Nar-SLNs against STZ-stimulated neurotoxicity and analyzed the role of autophagy in STZ-stimulated neurotoxicity. Our results proposed that Nar-SLNs could be a promising option for neurological disorders prevention through autophagy suppression.
CONFLICT OF INTEREST
The authors declare that there is no conflict of interest regarding the publication of this paper.
Open Research
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the corresponding author upon reasonable request.
REFERENCES
- Ahmad, A., Fauzia, E., Kumar, M., Mishra, R. K., Kumar, A., Khan, M. A., Raza, S. S., & Khan, R. (2019). Gelatin-coated polycaprolactone nanoparticle-mediated naringenin delivery rescue human mesenchymal stem cells from oxygen glucose deprivation-induced inflammatory stress. ACS Biomaterials Science and Engineering, 5(2), 683–695. https://doi.org/10.1021/acsbiomaterials.8b01081
- Ahmad, N., Ahmad, R., Ahmad, F. J., Ahmad, W., Alam, M. A., Amir, M., & Ali, A. (2020). Poloxamer-chitosan-based naringenin nanoformulation used in brain targeting for the treatment of cerebral ischemia. Saudi Journal of Biological Sciences, 27(1), 500–517. https://doi.org/10.1016/j.sjbs.2019.11.008
- Ahmadifard, Z., Ahmeda, A., Rasekhian, M., Moradi, S., & Arkan, E. (2020). Chitosan-coated magnetic solid lipid nanoparticles for controlled release of letrozole. Journal of Drug Delivery Science and Technology, 57, 101621.
- Ahsan, A. U., Sharma, V. L., Wani, A., & Chopra, M. (2020). Naringenin upregulates AMPK-mediated autophagy to rescue neuronal cells from β-amyloid (1–42) evoked neurotoxicity. Molecular Neurobiology, 57(8), 3589–3602.
- Algul, D., Duman, G., Ozdemir, S., Acar, E. T., & Yener, G. (2018). Pre-formulation, characterization, and in vitro release studies of caffeine-loaded solid lipid nanoparticles. Journal of Cosmetic Science, 69, 165–173.
- Biswas, J., Goswami, P., Gupta, S., Joshi, N., Nath, C., & Singh, S. (2016). Streptozotocin induced neurotoxicity involves Alzheimer's related pathological markers: A study on N2A cells. Molecular Neurobiology, 53(5), 2794–2806.
- Bollimpelli, V. S., Kumar, P., Kumari, S., & Kondapi, A. K. (2016). Neuroprotective effect of curcumin-loaded lactoferrin nano particles against rotenone induced neurotoxicity. Neurochemistry International, 95, 37–45. https://doi.org/10.1016/j.neuint.2016.01.006
- Bussmann, A. J., Borghi, S. M., Zaninelli, T. H., Dos Santos, T., Guazelli, C. F., Fattori, V., Domiciano, T. P., Pinho-Ribeiro, F. A., Ruiz-Miyazawa, K. W., Casella, A. M. B., Vignoli, J. A., Camilios-Neto, D., Casagrande, R., & Verri, W. A., Jr. (2019). The citrus flavanone naringenin attenuates zymosan-induced mouse joint inflammation: Induction of Nrf2 expression in recruited CD45+ hematopoietic cells. Inflammopharmacology, 27(6), 1229–1242.
- Cao, Y., Cai, J., Li, X., Yuan, N., & Zhang, S. (2016). Autophagy governs erythroid differentiation both in vitro and in vivo. Hematology, 21(4), 225–233.
- Chen, J., Zhang, L., Zhou, H., Wang, W., Luo, Y., Yang, H., & Yi, H. (2018). Inhibition of autophagy promotes cisplatin-induced apoptotic cell death through Atg5 and Beclin1 in A549 human lung cancer cells. Molecular Medicine Reports, 17(5), 6859–6865.
- Chen, W., Sun, Y., Liu, K., & Sun, X. (2014). Autophagy: A double-edged sword for neuronal survival after cerebral ischemia. Neural Regeneration Research, 9(12), 1210–1216.
- Cordani, M., & Somoza, Á. (2019). Targeting autophagy using metallic nanoparticles: A promising strategy for cancer treatment. Cellular and Molecular Life Sciences, 76(7), 1215–1242.
- Corti, O., Blomgren, K., Poletti, A., & Beart, P. M. (2020). Autophagy in neurodegeneration: New insights underpinning therapy for neurological diseases. Journal of Neurochemistry, 154(4), 354–371.
- Danaei, M., Dehghankhold, M., Ataei, S., Hasanzadeh Davarani, F., Javanmard, R., Dokhani, A., Khorasani, S., & Mozafari, M. (2018). Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics, 10(2), 57.
- Eisenberg-Lerner, A., Bialik, S., Simon, H.-U., & Kimchi, A. (2009). Life and death partners: Apoptosis, autophagy and the cross-talk between them. Cell Death & Differentiation, 16(7), 966–975.
- Fahmy, A. M., & Labonté, P. (2017). The autophagy elongation complex (ATG5-12/16L1) positively regulates HCV replication and is required for wild-type membranous web formation. Scientific Reports, 7(1), 1–13.
- Gao, Y., Li, J., Wu, L., Zhou, C., Wang, Q., Li, X., Zhou, M., & Wang, H. (2016). Tetrahydro curcumin provides neuroprotection in rats after traumatic brain injury: Autophagy and the PI3K/AKT pathways as a potential mechanism. Journal of Surgical Research, 206(1), 67–76. https://doi.org/10.1016/j.jss.2016.07.014
- Ghofrani, S., Joghataei, M. T., Mohseni, S., Baluchnejadmojarad, T., Bagheri, M., Khamse, S., & Roghani, M. (2015). Naringenin improves learning and memory in an Alzheimer's disease rat model: Insights into the underlying mechanisms. European Journal of Pharmacology, 764, 195–201. https://doi.org/10.1016/j.ejphar.2015.07.001
- Gottlieb, E., Armour, S., Harris, M., & Thompson, C. (2003). Mitochondrial membrane potential regulates matrix configuration and cytochrome c release during apoptosis. Cell Death & Differentiation, 10(6), 709–717.
- Hidayat, A. F. A., Chan, C. K., Mohamad, J., & Kadir, H. A. (2018). Leptospermum flavescens Sm. Protect pancreatic β cell function from streptozotocin involving apoptosis and autophagy signaling pathway in in vitro and in vivo case study. Journal of Ethnopharmacology, 226, 120–131. https://doi.org/10.1016/j.jep.2018.08.020
- Honary, S., & Zahir, F. (2013). Effect of zeta potential on the properties of nano-drug delivery systems—A review (part 2). Tropical Journal of Pharmaceutical Research, 12(2), 265–273.
- Hu, F., Hong, Y., & Yuan, H. (2004). Preparation and characterization of solid lipid nanoparticles containing peptide. International Journal of Pharmaceutics, 273(1–2), 29–35.
- Ji, P., Yu, T., Liu, Y., Jiang, J., Xu, J., Zhao, Y., Hao, Y., Qiu, Y., Zhao, W., & Wu, C. (2016). Naringenin-loaded solid lipid nanoparticles: Preparation, controlled delivery, cellular uptake, and pulmonary pharmacokinetics. Drug Design, Development and Therapy, 10, 911.
- Kamat, P. K. (2015). Streptozotocin induced Alzheimer's disease like changes and the underlying neural degeneration and regeneration mechanism. Neural Regeneration Research, 10(7), 1050–1052.
- Kesh, S., Kannan, R. R., & Balakrishnan, A. (2021). Naringenin alleviates 6-hydroxydopamine induced Parkinsonism in SHSY5Y cells and zebrafish model. Comparative Biochemistry and Physiology Part C: Toxicology & Pharmacology, 239, 108893.
- Khan, M. B., Khan, M. M., Khan, A., Ahmed, M. E., Ishrat, T., Tabassum, R., Vaibhav, K., Ahmad, A., & Islam, F. (2012). Naringenin ameliorates Alzheimer's disease (AD)-type neurodegeneration with cognitive impairment (AD-TNDCI) caused by the intracerebroventricular-streptozotocin in rat model. Neurochemistry International, 61(7), 1081–1093.
- Kumar, R. P., & Abraham, A. (2016). PVP-coated naringenin nanoparticles for biomedical applications–in vivo toxicological evaluations. Chemico-Biological Interactions, 257, 110–118.
- Li, R., Zhang, L., Shi, Q., Guo, Y., Zhang, W., & Zhou, B. (2018). A protective role of autophagy in TDCIPP-induced developmental neurotoxicity in zebrafish larvae. Aquatic Toxicology, 199, 46–54.
- Li, X., Wu, Z., Zhang, Y., Xu, Y., Han, G., & Zhao, P. (2017). Activation of autophagy contributes to sevoflurane-induced neurotoxicity in fetal rats. Frontiers in Molecular Neuroscience, 10, 432.
- Li, Y., Cho, M. H., Lee, S. S., Lee, D. E., Cheong, H., & Choi, Y. (2020). Hydroxychloroquine-loaded hollow mesoporous silica nanoparticles for enhanced autophagy inhibition and radiation therapy. Journal of Controlled Release, 325, 100–110. https://doi.org/10.1016/j.jconrel.2020.06.025
- Lin, X., Wen, X., Wei, Z., Guo, K., Shi, F., Huang, T., Wang, W., & Zheng, J. (2021). Vitamin K2 protects against Aβ42-induced neurotoxicity by activating autophagy and improving mitochondrial function in drosophila. Neuroreport, 32(6), 431–437.
- Maity, S., Mukhopadhyay, P., Kundu, P. P., & Chakraborti, A. S. (2017). Alginate coated chitosan core-shell nanoparticles for efficient oral delivery of naringenin in diabetic animals—An in vitro and in vivo approach. Carbohydrate Polymers, 170, 124–132.
- Md, S., Alhakamy, N. A., Aldawsari, H. M., & Asfour, H. Z. (2019). Neuroprotective and antioxidant effect of naringenin-loaded nanoparticles for nose-to-brain delivery. Brain Sciences, 9(10), 275. https://doi.org/10.3390/brainsci9100275
- Md, S., Gan, S. Y., Haw, Y. H., Ho, C. L., Wong, S., & Choudhury, H. (2018). In vitro neuroprotective effects of naringenin nano-emulsion against β-amyloid toxicity through the regulation of amyloidogenesis and tau phosphorylation. International Journal of Biological Macromolecules, 118, 1211–1219.
- Meng, X.-M., Zhang, Y., Huang, X.-R., Ren, G.-L., Li, J., & Lan, H. Y. (2015). Treatment of renal fibrosis by rebalancing TGF-β/Smad signaling with the combination of asiatic acid and naringenin. Oncotarget, 6(35), 36984–36997.
- Millot, C., Millot, J.-M., Morjani, H., Desplaces, A., & Manfait, M. (1997). Characterization of acidic vesicles in multidrug-resistant and sensitive cancer cells by acridine orange staining and confocal microspectrofluorometry. Journal of Histochemistry & Cytochemistry, 45(9), 1255–1264.
- Mishra, V., Bansal, K. K., Verma, A., Yadav, N., Thakur, S., Sudhakar, K., & Rosenholm, J. M. (2018). Solid lipid nanoparticles: Emerging colloidal nano drug delivery systems. Pharmaceutics, 10(4), 191.
- Nouri, Z., Fakhri, S., El-Senduny, F. F., Sanadgol, N., Abd-Elghani, G. E., Farzaei, M. H., & Chen, J. T. (2019). On the neuroprotective effects of naringenin: Pharmacological targets, signaling pathways, molecular mechanisms, and clinical perspective. Biomolecules, 9(11), 690. https://doi.org/10.3390/biom9110690
- Nouri, Z., Hajialyani, M., Izadi, Z., Bahramsoltani, R., Farzaei, M. H., & Abdollahi, M. (2020). Nanophytomedicines for the prevention of metabolic syndrome: A pharmacological and biopharmaceutical review. Frontiers in Bioengineering and Biotechnology, 8, 425.
- Pantiya, P., Thonusin, C., Chattipakorn, N., & Chattipakorn, S. C. (2020). Mitochondrial abnormalities in neurodegenerative models and possible interventions: Focus on Alzheimer's disease, Parkinson's disease, Huntington's disease. Mitochondrion, 55, 14–47.
- Qizilbash, F. F., Ashhar, M. U., Zafar, A., Qamar, Z., Ali, J., Baboota, S., Ghoneim, M. M., Alshehri, S., & Ali, A. (2022). Thymoquinone-enriched naringenin-loaded nanostructured lipid carrier for brain delivery via nasal route: In vitro prospect and in vivo therapeutic efficacy for the treatment of depression. Pharmaceutics, 14(3), 656. https://doi.org/10.3390/pharmaceutics14030656
- Raeisi, S., Chavoshi, H., Mohammadi, M., Ghorbani, M., Sabzichi, M., & Ramezani, F. (2019). Naringenin-loaded nano-structured lipid carrier fortifies oxaliplatin-dependent apoptosis in HT-29 cell line. Process Biochemistry, 83, 168–175.
- Raza, S. S., Khan, M. M., Ahmad, A., Ashafaq, M., Islam, F., Wagner, A. P., & Safhi, M. M. (2013). Neuroprotective effect of naringenin is mediated through suppression of NF-κB signaling pathway in experimental stroke. Neuroscience, 230, 157–171. https://doi.org/10.1016/j.neuroscience.2012.10.041
- Rodius, S., De Klein, N., Jeanty, C., Sánchez-Iranzo, H., Crespo, I., Ibberson, M., Xenarios, I., Dittmar, G., Mercader, N., & Niclou, S. P. (2020). Fisetin protects against cardiac cell death through reduction of ROS production and caspases activity. Scientific Reports, 10(1), 1–12.
- Salehi, B., Fokou, P. V. T., Sharifi-Rad, M., Zucca, P., Pezzani, R., Martins, N., & Sharifi-Rad, J. (2019). The therapeutic potential of naringenin: A review of clinical trials. Pharmaceuticals, 12(1), 11.
- Seca, H., Lima, R. T., Lopes-Rodrigues, V., Guimarães, J. E., Almeida, G. M., & Vasconcelos, M. H. (2013). Targeting miR-21 induces autophagy and chemosensitivity of leukemia cells. Current Drug Targets, 14(10), 1135–1143. https://doi.org/10.2174/13894501113149990185
- Shah, S. Z. A., Zhao, D., Hussain, T., Sabir, N., & Yang, L. (2018). Regulation of microRNAs-mediated autophagic flux: A new regulatory avenue for neurodegenerative diseases with focus on prion diseases. Frontiers in Aging Neuroscience, 10, 139.
- Shi, L.-B., Tang, P.-F., Zhang, W., Zhao, Y.-P., Zhang, L.-C., & Zhang, H. (2016). Naringenin inhibits spinal cord injury-induced activation of neutrophils through miR-223. Gene, 592(1), 128–133.
- Thomas, K. T., Gross, C., & Bassell, G. J. (2018). MicroRNAs sculpt neuronal communication in a tight balance that is lost in neurological disease. Frontiers in Molecular Neuroscience, 11, 455.
- Tung, Y.-T., Wang, B.-J., Hu, M.-K., Hsu, W.-M., Lee, H., Huang, W.-P., & Liao, Y.-F. (2012). Autophagy: A double-edged sword in Alzheimer's disease. Journal of Biosciences, 37(1), 157–165.
- Ustuner, D., Kolac, U. K., Ustuner, M. C., Tanrikut, C., Ozdemir Koroglu, Z., Burukoglu Donmez, D., Ozen, H., & Ozden, H. (2020). Naringenin ameliorate carbon tetrachloride-induced hepatic damage through inhibition of endoplasmic reticulum stress and autophagy in rats. Journal of Medicinal Food, 23(11), 1192–1200. https://doi.org/10.1089/jmf.2019.0265
- Wang, X., Jiang, Y., Zhu, L., Cao, L., Xu, W., Rahman, S. U., Feng, S., Li, Y., & Wu, J. (2020). Autophagy protects PC12 cells against deoxynivalenol toxicity via the class III PI3K/beclin 1/Bcl-2 pathway. Journal of Cellular Physiology, 235(11), 7803–7815.
- Wang, Y., Wang, S., Firempong, C. K., Zhang, H., Wang, M., Zhang, Y., Zhu, Y., Yu, J., & Xu, X. (2017). Enhanced solubility and bioavailability of naringenin via liposomal nanoformulation: Preparation and in vitro and in vivo evaluations. AAPS PharmSciTech, 18(3), 586–594.
- Xu, L., Shen, J., Yu, L., Sun, J., McQuillan, P. M., Hu, Z., & Yan, M. (2018). Role of autophagy in sevoflurane-induced neurotoxicity in neonatal rat hippocampal cells. Brain Research Bulletin, 140, 291–298.
- Xu, Z., Han, X., Ou, D., Liu, T., Li, Z., Jiang, G., Liu, J., & Zhang, J. (2020). Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Applied Microbiology and Biotechnology, 104(2), 575–587.
- Zeng, Z., He, S., Lu, J., Liu, C., Lin, H., Xu, C., Xie, L., & Sun, S. (2018). MicroRNA-21 aggravates chronic obstructive pulmonary disease by promoting autophagy. Experimental Lung Research, 44(2), 89–97.
- Zhang, N., Hu, Z., Zhang, Z., Liu, G., Wang, Y., Ren, Y., Wu, X., & Geng, F. (2018). Protective role of naringenin against Aβ 25-35-caused damage via ER and PI3K/Akt-mediated pathways. Cellular and Molecular Neurobiology, 38(2), 549–557.
- Zhao, Q., Yang, H., Liu, F., Luo, J., Zhao, Q., Li, X., & Yang, Y. (2019). Naringenin exerts cardiovascular protective effect in a palmitate-induced human umbilical vein endothelial cell injury model via autophagy flux improvement. Molecular Nutrition & Food Research, 63(24), 1900601.