We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Pharmaceutical characterization of probucol bile acid–lithocholic acid nanoparticles to prevent chronic hearing related and similar cellular oxidative stress pathologies

    Susbin R Wagle

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    ,
    Corina M Ionescu

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    ,
    Bozica Kovacevic

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    ,
    Melissa Jones

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    ,
    Thomas Foster

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    ,
    Patrick Lim

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    ,
    Michael Lewkowicz

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    ,
    Maja Ðanić

    Department of Pharmacology, Toxicology & Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad (Hajduk Veljkova 3, 21101), Serbia

    ,
    Momir Mikov

    Department of Pharmacology, Toxicology & Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad (Hajduk Veljkova 3, 21101), Serbia

    ,
    Armin Mooranian‡

    *Author for correspondence: Tel.: +61 892 661 318;

    E-mail Address: a.mooranian@curtin.edu.au

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    School of Pharmacy, University of Otago, Dunedin, 9016, Otago, New Zealand

    ‡Authors contributed equally

    Search for more papers by this author

    &
    Hani Al-Salami‡

    **Author for correspondence: Tel.: +61 892 669 816;

    E-mail Address: hani.al-salami@curtin.edu.au

    The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia

    Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia

    Medical School, University of Western Australia, Perth 6907, Western Australia, Australia

    ‡Authors contributed equally

    Search for more papers by this author

    Published Online:https://doi.org/10.2217/nnm-2023-0092

    Background: Sensorineural hearing loss has been associated with oxidative stress. However, an antioxidant that passes effectively through the ear remains elusive. Method: Probucol (PB)-based nanoparticles were formed using a spray-drying encapsulation technique, characterized and tested in vitro. Results: Uniform, spherical nanoparticles were produced. The addition of lithocholic acid to PB formulations did not affect drug content or production yield, but it did modify capsule size, surface tension, electrokinetic stability and drug release. Cell viability, bioenergetics and inflammatory profiles were improved when auditory cells were exposed to PB-based nanoparticles, which showed antioxidant properties (p < 0.05). Conclusion: PB-based nanoparticles can potentially protect the auditory cell line from oxidative stress and could be used in future in vivo studies as a potential new therapeutic agent for sensorineural hearing loss.

    Plain language summary

    Oxidative stress is an imbalance of cellular processes in which the production of free radicals outweighs the cellular defense mechanism. The association of oxidative stress with the pathophysiology of sensorineural hearing loss (SHL) is well established. SHL development is associated with chronic damage in the structure of the inner ear or auditory nerve. Therefore, potent antioxidants such as probucol could be one way to prevent or treat SHL. However, due to its isolated position, SHL is challenging to treat, imposing a desperate need for refining existing therapeutic methods; one way to do this is by optimizing the formulation using nanoparticles. We aimed to design a novel, stable formulation of PB using polymers and excipients to develop nanoparticles and examine the efficiency of these formulations on the HEI-OC1 stress cell line. We found that the prepared nanoparticle is robust and stable and protects HEI-OC1 from cellular toxicity and oxidative stress. It could be a novel therapeutic agent to treat or prevent SHL.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Henderson D, Bielefeld EC, Harris KC, Hu BH. The role of oxidative stress in noise-induced hearing loss. Ear Hear. 27(1), 1–19 (2006).
    • 2. Menardo J, Tang Y, Ladrech S et al. Oxidative stress, inflammation, and autophagic stress as the key mechanisms of premature age-related hearing loss in SAMP8 mouse cochlea. Antioxid. Redox Signal. 16(3), 263–274 (2012).
    • 3. Elias TGA, Monsanto RDC, do Amaral JB, Oyama LM, Maza PK, Penido NO. Evaluation of oxidative-stress pathway and recovery of sudden sensorineural hearing loss. Int. Arch. Otorhinolaryngol. 25(3), e428–e432 (2021).
    • 4. Böttger EC, Schacht J. The mitochondrion: a perpetrator of acquired hearing loss. Hear. Res. 303, 12–19 (2013).
    • 5. Nakae S, Tachibana M. The cochlea of the spontaneously diabetic mouse. Arch. Otorhinolaryngol. 243(5), 313–316 (1986).
    • 6. Raynor EM, Carrasco VN, Prazma J, Pillsbury HC. An assessment of cochlear hair-cell loss in insulin-dependent diabetes mellitus diabetic and noise-exposed rats. Arch. Otolaryngol. Head Neck Surg. 121(4), 452–456 (1995).
    • 7. Chester J, Johnston E, Walker D et al. A review on recent advancement on age-related hearing loss: the applications of nanotechnology, drug pharmacology, and biotechnology. Pharmaceutics 13(7), 1041 (2021). • Review which explains the importance of bile acid-based nanotechnology in drug-delivery systems.
    • 8. Zimetbaum P, Eder H, Frishman W. Probucol: pharmacology and clinical application. J. Clin. Pharmacol. 30(1), 3–9 (1990).
    • 9. Yamashita S, Matsuzawa Y. Where are we with probucol: a new life for an old drug? Atherosclerosis 207(1), 16–23 (2009).
    • 10. Bueno DC, Canto RFS, de Souza V et al. New probucol analogues inhibit ferroptosis, improve mitochondrial parameters, and induce glutathione peroxidase in HT22 cells. Mol. Neurobiol. 57(8), 3273–3290 (2020).
    • 11. Heel R, Brogden R, Speight T, Avery G. Probucol: a review of its pharmacological properties and therapeutic use in patients with hypercholesterolaemia. Drugs 15(6), 409–428 (1978).
    • 12. Mamo JC, Lam V, Brook E et al. Probucol prevents blood–brain barrier dysfunction and cognitive decline in mice maintained on pro-diabetic diet. Diab. Vasc. Dis. Res. 16(1), 87–97 (2019).
    • 13. Wagle SR, Kovacevic B, Walker D et al. Alginate-based drug oral targeting using bio-micro/nano encapsulation technologies. Expert Opin. Drug Deliv. 17(10), 1361–1376 (2020). • Review which explains that bile acid and nanotechnology could be one promising strategy to improve the therapeutic effectiveness of lipophilic drugs.
    • 14. Li F, Li L, Wang S et al. Improved dissolution and oral absorption by co-grinding active drug probucol and ternary stabilizers mixtures with planetary beads-milling method. Asian J. Pharm. Sci. 14(6), 649–657 (2019).
    • 15. Yehye WA, Rahman NA, Ariffin A et al. Understanding the chemistry behind the antioxidant activities of butylated hydroxytoluene (BHT): a review. Eur. J. Med. Chem. 101, 295–312 (2015).
    • 16. Mathavan S, Chen-Tan N, Arfuso F, Al-Salami H. A comprehensive study of novel microcapsules incorporating gliclazide and a permeation enhancing bile acid: hypoglycemic effect in an animal model of type-1 diabetes. Drug Deliv. 23(8), 2869–2880 (2016).
    • 17. Mikov M, Al-Salami H, Golocorbin-Kon S, Skrbic R, Raskovic A, Fawcett JP. The influence of 3α,7α-dihydroxy-12-keto-5β-cholanate on gliclazide pharmacokinetics and glucose levels in a rat model of diabetes. Eur. J. Drug Metab. Pharmacokinet. 33(3), 137–142 (2008).
    • 18. Mooranian A, Negrulj R, Mathavan S et al. An advanced microencapsulated system: a platform for optimized oral delivery of antidiabetic drug-bile acid formulations. Pharm. Dev. Technol. 20(6), 702–709 (2015).
    • 19. Stojančević M, Pavlović N, Goločorbin-Kon S, Mikov M. Application of bile acids in drug formulation and delivery. Front. Life Sci. 7(3–4), 112–122 (2013).
    • 20. Kovacevic B, Jones M, Ionescu C et al. The emerging role of bile acids as critical components in nanotechnology and bioengineering: pharmacology, formulation optimizers and hydrogel–biomaterial applications. Biomaterials 283, 121459 (2022). • Explores bile acids’ role and mechanism as permeation enhancers in drug delivery.
    • 21. Mooranian A, Negrulj R, Takechi R, Mamo J, Al-Sallami H, Al-Salami H. The biological effects of the hypolipidaemic drug probucol microcapsules fed daily for 4 weeks, to an insulin-resistant mouse model: potential hypoglycaemic and anti-inflammatory effects. Drug Deliv. Transl. Res. 8(3), 543–551 (2018).
    • 22. Mooranian A, Negrulj R, Chen-Tan N et al. Advanced bile acid-based multi-compartmental microencapsulated pancreatic β-cells integrating a polyelectrolyte-bile acid formulation, for diabetes treatment. Artif. Cells Nanomed. Biotechnol. 44(2), 588–595 (2016).
    • 23. Mooranian A, Negrulj R, Mikov M, Golocorbin-Kon S, Arfuso F, Al-Salami H. Novel chenodeoxycholic acid–sodium alginate matrix in the microencapsulation of the potential antidiabetic drug, probucol. An in vitro study. J. Microencapsul. 32(6), 589–597 (2015).
    • 24. Mooranian A, Raj Wagle S, Kovacevic B et al. Bile acid bio-nanoencapsulation improved drug targeted-delivery and pharmacological effects via cellular flux: 6-months diabetes preclinical study. Sci. Rep. 10(1), 106 (2020). •• Here we showed that probucol–bile acid microcapsules positively affected β-cells’ viability in the hyperglycemic state, and brought about hypoglycemic and anti-inflammatory effects on prediabetic mice.
    • 25. Wagle SR, Walker D, Kovacevic B et al. Micro-nano formulation of bile–gut delivery: rheological, stability and cell survival, basal and maximum respiration studies. Sci. Rep. 10(1), 7715 (2020).
    • 26. Yao B, He J, Yin X, Shi Y, Wan J, Tian Z. The protective effect of lithocholic acid on the intestinal epithelial barrier is mediated by the vitamin D receptor via a SIRT1/Nrf2 and NF-κB dependent mechanism in Caco-2 cells. Toxicol. Lett. 316, 109–118 (2019).
    • 27. Wagle SR, Kovacevic B, Ionescu CM et al. Pharmacological and biological study of microencapsulated probucol-secondary bile acid in a diseased mouse model. Pharmaceutics 13(8), 1223 (2021). •• Here we showed that probucol–lithocholic acid-based microcapsules may be a promising therapy for treating diabetes mellitus.
    • 28. Pavlovic N, Golocorbin-Kon S, Ethanic M et al. Bile acids and their derivatives as potential modifiers of drug release and pharmacokinetic profiles. Front. Pharmacol. 9, 1283 (2018).
    • 29. Sukmawati A, Utami W, Yuliani R, Da'i M, Nafarin A. Effect of Tween 80 on nanoparticle preparation of modified chitosan for targeted delivery of combination doxorubicin and curcumin analogue. IOP Conf. Ser. Mater. Sci. Eng. 311, 012024 (2018).
    • 30. Dumortier G, Grossiord JL, Agnely F, Chaumeil JC. A review of poloxamer 407 pharmaceutical and pharmacological characteristics. Pharm. Res. 23(12), 2709–2728 (2006).
    • 31. Lee CH, Singla A, Lee Y. Biomedical applications of collagen. Int. J. Pharm. 221(1–2), 1–22 (2001).
    • 32. Goh CH, Heng PWS, Chan LW. Alginates as a useful natural polymer for microencapsulation and therapeutic applications. Carbohydr. Polym. 88(1), 1–12 (2012).
    • 33. Tomeh MA, Hadianamrei R, Zhao X. Silk fibroin as a functional biomaterial for drug and gene delivery. Pharmaceutics 11(10), 494 (2019).
    • 34. Oliveira RN, Rouzé R, Quilty B et al. Mechanical properties and in vitro characterization of polyvinyl alcohol-nano-silver hydrogel wound dressings. Interface Focus 4(1), 20130049 (2014).
    • 35. Kumar R, Siril PF. Preparation and characterization of polyvinyl alcohol stabilized griseofulvin nanoparticles. Mater. Today Proc. 3(6), 2261–2267 (2016).
    • 36. Chakraborty G, Bhattarai A, De R. Polyelectrolyte–dye interactions: an overview. Polymers 14(3), 598 (2022).
    • 37. Panacek A, Prucek R, Hrbac J et al. Polyacrylate-assisted size control of silver nanoparticles and their catalytic activity. Chem. Mater. 26(3), 1332–1339 (2014).
    • 38. Almansour K, Ali R, Alheibshy F et al. Particle engineering by nano spray drying: optimization of process parameters with hydroethanolic versus aqueous solutions. Pharmaceutics 14(4), 800 (2022).
    • 39. Wagle SR, Kovacevic B, Walker D et al. Pharmacological and advanced cell respiration effects, enhanced by toxic human-bile nano-pharmaceuticals of probucol cell-targeting formulations. Pharmaceutics 12(8), 708 (2020).
    • 40. Mooranian A, Negrulj R, Al-Sallami HS et al. Probucol release from novel multicompartmental microcapsules for the oral targeted delivery in type 2 diabetes. AAPS PharmSciTech 16(1), 45–52 (2015).
    • 41. Salt AN, Hartsock JJ, Gill RM, Piu F, Plontke SK. Perilymph pharmacokinetics of markers and dexamethasone applied and sampled at the lateral semi-circular canal. J. Assoc. Res. Otolaryngol. 13(6), 771–783 (2012).
    • 42. Yagi N, Terashima Y, Kenmotsu H, Sekikawa H, Takada M. Dissolution behavior of probucol from solid dispersion systems of probucol-polyvinylpyrrolidone. Chem. Pharm. Bull. 44(1), 241–244 (1996).
    • 43. Uhljar LÉ, Kan SY, Radacsi N, Koutsos V, Szabó-Révész P, Ambrus R. In vitro drug release, permeability, and structural test of ciprofloxacin-loaded nanofibers. Pharmaceutics 13(4), 556 (2021).
    • 44. Kalinec G, Thein P, Park C, Kalinec F. HEI-OC1 cells as a model for investigating drug cytotoxicity. Hear. Res. 335, 105–117 (2016).
    • 45. Kalinec GM, Park C, Thein P, Kalinec F. Working with auditory HEI-OC1 cells. J. Vis. Exp. (115), 54425 (2016).
    • 46. Wu M, Neilson A, Swift AL et al. Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells. Am. J. Physiol. Cell Physiol. 292(1), C125–C136 (2007).
    • 47. Wolfe KL, Liu RH. Cellular antioxidant activity (CAA) assay for assessing antioxidants, foods, and dietary supplements. J. Agric. Food Chem. 55(22), 8896–8907 (2007).
    • 48. Liao W, Ning Z, Chen L et al. Intracellular antioxidant detoxifying effects of diosmetin on 2, 2-azobis (2-amidinopropane) dihydrochloride (AAPH)-induced oxidative stress through inhibition of reactive oxygen species generation. J. Agric. Food Chem. 62(34), 8648–8654 (2014).
    • 49. Kellett ME, Greenspan P, Pegg RB. Modification of the cellular antioxidant activity (CAA) assay to study phenolic antioxidants in a Caco-2 cell line. Food Chem. 244, 359–363 (2018).
    • 50. Xiao F, Xu T, Lu B, Liu R. Guidelines for antioxidant assays for food components. Food Front. 1(1), 60–69 (2020).
    • 51. Yarnpakdee S, Benjakul S, Kristinsson HG, Bakken HE. Preventive effect of Nile tilapia hydrolysate against oxidative damage of HepG2 cells and DNA mediated by H2O2 and AAPH. J. Food Sci. Technol. 52(10), 6194–6205 (2015).
    • 52. Ajun W, Yan S, Li G, Huili L. Preparation of aspirin and probucol in combination loaded chitosan nanoparticles and in vitro release study. Carbohydr. Polym. 75(4), 566–574 (2009).
    • 53. Lu P-J, Fu W-E, Huang S-C et al. Methodology for sample preparation and size measurement of commercial ZnO nanoparticles. J. Food Drug Anal. 26(2), 628–636 (2018).
    • 54. Malamatari M, Somavarapu S, Kachrimanis K, Buckton G, Taylor KMG. Preparation of respirable nanoparticle agglomerates of the low melting and ductile drug ibuprofen: impact of formulation parameters. Powder Technol. 308, 123–134 (2017).
    • 55. Malamatari M, Charisi A, Malamataris S, Kachrimanis K, Nikolakakis I. Spray drying for the preparation of nanoparticle-based drug formulations as dry powders for inhalation. Processes 8(7), 788 (2020).
    • 56. Danaei M, Dehghankhold M, Ataei S et al. Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics 10(2), 57 (2018).
    • 57. Mooranian A, Negrulj R, Arfuso F, Al-Salami H. The effect of a tertiary bile acid, taurocholic acid, on the morphology and physical characteristics of microencapsulated probucol: potential applications in diabetes: a characterization study. Drug Deliv. Transl. Res. 5(5), 511–522 (2015).
    • 58. Meier W. Polymer nanocapsules. Chem. Soc. Rev. 29(5), 295–303 (2000).
    • 59. Mooranian A, Negrulj R, Chen-Tan N et al. Microencapsulation as a novel delivery method for the potential antidiabetic drug, probucol. Drug Des. Devel. Ther. 8, 1221 (2014).
    • 60. Goycoolea MV. Clinical aspects of round window membrane permeability under normal and pathological conditions. Acta Otolaryngol. 121(4), 437–447 (2001).
    • 61. Draget KI, Taylor C. Chemical, physical and biological properties of alginates and their biomedical implications. Food Hydrocolloids 25(2), 251–256 (2011).
    • 62. Pamies R, Schmidt RR, del Carmen López Martínez M, de la Torre JG. The influence of mono and divalent cations on dilute and non-dilute aqueous solutions of sodium alginates. Carbohydr. Polym. 80(1), 248–253 (2010).
    • 63. Mooranian A, Negrulj R, Chen-Tan N, Watts GF, Arfuso F, Al-Salami H. An optimized probucol microencapsulated formulation integrating a secondary bile acid (deoxycholic acid) as a permeation enhancer. Drug Des. Devel. Ther. 8, 1673 (2014).
    • 64. Mooranian A, Negrulj R, Mathavan S et al. Stability and release kinetics of an advanced gliclazide-cholic acid formulation: the use of artificial-cell microencapsulation in slow release targeted oral delivery of antidiabetics. J. Pharm. Innov. 9, 150–157 (2014).
    • 65. Loebel C, Rodell CB, Chen MH, Burdick JA. Shear-thinning and self-healing hydrogels as injectable therapeutics and for 3D-printing. Nat. Protoc. 12(8), 1521–1541 (2017).
    • 66. Honary S, Zahir F. Effect of zeta potential on the properties of nano-drug delivery systems-a review (Part 2). Trop. J. Pharm. Res. 12(2), 265–273 (2013).
    • 67. Xie HG, Li XX, Lv GJ et al. Effect of surface wettability and charge on protein adsorption onto implantable alginate–chitosan–alginate microcapsule surfaces. J. Biomed. Mater. Res. A 92(4), 1357–1365 (2010).
    • 68. Madenci D, Egelhaaf S. Self-assembly in aqueous bile salt solutions. Curr. Opin. Colloid Interface Sci. 15(1–2), 109–115 (2010).
    • 69. Azarbayjani AF, Jouyban A, Chan SY. Impact of surface tension in pharmaceutical sciences. J. Pharm. Pharm. Sci. 12(2), 218–228 (2009).
    • 70. Mooranian A, Carey L, Ionescu CM et al. The effects of accelerated temperature-controlled stability systems on the release profile of primary bile acid-based delivery microcapsules. Pharmaceutics 13(10), 1667 (2021).
    • 71. Negrulj R, Mooranian A, Chen-Tan N et al. Swelling, mechanical strength, and release properties of probucol microcapsules with and without a bile acid, and their potential oral delivery in diabetes. Artif. Cells Nanomed. Biotechnol. 44(5), 1290–1297 (2016).
    • 72. Zou J, Saulnier P, Perrier T et al. Distribution of lipid nanocapsules in different cochlear cell populations after round window membrane permeation. J. Biomed. Mater. Res. B Appl. Biomater. 87B(1), 10–18 (2008).
    • 73. Huang X, Brazel CS. On the importance and mechanisms of burst release in matrix-controlled drug delivery systems. J. Control. Rel. 73(2), 121–136 (2001).
    • 74. Shively ML, Coonts BA, Renner WD, Southard JL, Bennett AT. Physico-chemical characterization of a polymeric injectable implant delivery system. J. Control. Rel. 33(2), 237–243 (1995).
    • 75. Jeong B, Bae YH, Kim SW. Drug release from biodegradable injectable thermosensitive hydrogel of PEG–PLGA–PEG triblock copolymers. J. Control. Rel. 63(1–2), 155–163 (2000).
    • 76. Valentini C, Szeto B, Kysar JW, Lalwani AK. Inner ear gene delivery: vectors and routes. Hear. Balance Commun. 18(4), 278–285 (2020).
    • 77. Fukui H, Raphael Y. Gene therapy for the inner ear. Hear. Res. 297, 99–105 (2013).
    • 78. Praetorius M, Brunner C, Lehnert B et al. Transsynaptic delivery of nanoparticles to the central auditory nervous system. Acta Otolaryngol. 127(5), 486–490 (2007).
    • 79. Pyykkö I, Zou J, Schrott-Fischer A, Glueckert R, Kinnunen P. An overview of nanoparticle based delivery for treatment of inner ear disorders. Methods Mol. Biol. 1427, 363–415 (2016).
    • 80. Szeto B, Chiang H, Valentini C, Yu M, Kysar JW, Lalwani AK. Inner ear delivery: challenges and opportunities. Laryngoscope Investig. Otolaryngol. 5(1), 122–131 (2019).
    • 81. Peluso I, Campolongo P, Valeri P, Romanelli L, Palmery M. Intestinal motility disorder induced by free radicals: a new model mimicking oxidative stress in gut. Pharmacol. Res. 46(6), 533–538 (2002).
    • 82. Moreira Gomes MD, Carvalho G, Casquilho NV et al. 2,2′-azobis (2-amidinopropane) dihydrochloride is a useful tool to impair lung function in rats. Front. Physiol. 7, 475 (2016).
    • 83. Zimowska W, Motyl T, Skierski J et al. Apoptosis and Bcl-2 protein changes in L1210 leukaemic cells exposed to oxidative stress. Apoptosis 2(6), 529–539 (1997).
    • 84. Cadenas E, Sies H. The lag phase. Free Radic. Res. 28(6), 601–609 (1998).
    • 85. Terao K, Niki E. Damage to biological tissues induced by radical initiator 2,2′-azobis (2-amidinopropane) dihydrochloride and its inhibition by chain-breaking antioxidants. J. Free Radic. Biol. Med. 2(3), 193–201 (1986).
    • 86. Ward JBJ, Lajczak NK, Kelly OB et al. Ursodeoxycholic acid and lithocholic acid exert anti-inflammatory actions in the colon. Am. J. Physiol. Gastrointest. Liver Physiol. 312(6), G550–G558 (2017).
    • 87. Brand MD, Nicholls DG. Assessing mitochondrial dysfunction in cells. Biochem. J. 435(2), 297–312 (2011).
    • 88. Joachims HZ, Segal J, Golz A, Netzer A, Goldenberg D. Antioxidants in treatment of idiopathic sudden hearing loss. Otol. Neurotol. 24(4), 572–575 (2003).
    • 89. Laurell G, Teixeira M, Sterkers O et al. Local administration of antioxidants to the inner ear: kinetics and distribution. Hear. Res. 173(1), 198–209 (2002).
    • 90. Fujimoto C, Yamasoba T. Mitochondria-targeted antioxidants for treatment of hearing loss: a systematic review. Antioxidants 8(4), 109 (2019).
    • 91. Lyu A-R, Kim T-H, Shin S-A et al. Hearing impairment in a mouse model of diabetes is associated with mitochondrial dysfunction, synaptopathy, and activation of the intrinsic apoptosis pathway. Int. J. Mol. Sci. 22(16), 8807 (2021).
    • 92. Fujioka M, Kanzaki S, Okano HJ, Masuda M, Ogawa K, Okano H. Proinflammatory cytokines expression in noise-induced damaged cochlea. J. Neurosci. Res. 83(4), 575–583 (2006).
    • 93. Sziklai I, Batta TJ, Karosi T. Otosclerosis: an organ-specific inflammatory disease with sensorineural hearing loss. Eur. Arch. Otorhinolaryngol. 266(11), 1711–1718 (2009).
    • 94. Moon SK, Woo JI, Lim DJ. Involvement of TNF-α and IFN-γ in inflammation-mediated cochlear injury. Ann. Otol. Rhinol. Laryngol. 128(Suppl. 6), S8–S15 (2019).
    • 95. Yang X, Merchant MS, Romero ME et al. Induction of caspase 8 by interferon γ renders some neuroblastoma (NB) cells sensitive to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) but reveals that a lack of membrane TR1/TR2 also contributes to TRAIL resistance in NB. Cancer Res. 63(5), 1122–1129 (2003).
    • 96. Young IS, Woodside JV. Antioxidants in health and disease. J. Clin. Pathol. 54(3), 176–186 (2001).