We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Probucol-bile acid based nanoparticles protect auditory cells from oxidative stress: an in vitro study

    Susbin Raj Wagle

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

    ,
    Corina Mihaela Ionescu

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

    ,
    Melissa Jones

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

    ,
    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 8 9266 1318;

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

    Authors contributed equally

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

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

    &
    Hani Al-Salami

    **Author for correspondence: Tel.: +61 8 9266 9816;

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

    Authors contributed equally

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

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

    Published Online:https://doi.org/10.4155/tde-2023-0099

    Aim: Excessive free radicals contribute to oxidative stress and mitochondrial dysfunction in sensorineural hearing loss (SNHL). The antioxidant probucol holds promise, but its limited bioavailability and inner ear barriers hinder effective SNHL treatment. Methodology: We addressed this by developing probucol-loaded nanoparticles with polymers and lithocholic acid and tested them on House Ear Institute-Organ of Corti cells. Results: Probucol-based nanoparticles effectively reduced oxidative stress-induced apoptosis, enhanced cellular viability, improved probucol uptake and promoted mitochondrial function. Additionally, they demonstrated the capacity to reduce reactive oxygen species through the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway. Conclusion: This innovative nanoparticle system holds the potential to prevent oxidative stress-related hearing impairment, providing an effective solution for SNHL.

    Plain language summary

    Hearing loss affects millions of people worldwide, and its prevalence is expected to double by 2050. Current treatments have limitations, pushing researchers to explore new options. Oxidative stress is a key player in hearing loss and is known to damage inner ear hair cells. While antioxidants, known for their protective effects, hold promise, delivering them effectively to the inner ear is challenging. Scientists have been testing nanoparticles loaded with the antioxidant probucol to fight hearing loss. In this study, these particles protected inner ear cells in cell studies, offering potential hope for preventing hearing problems. This research is a significant step toward finding better treatments for hearing loss.

    Graphical abstract

    References

    • 1. Chandrasekhar SS, Tsai Do BS, Schwartz SR et al. Clinical practice guideline: sudden hearing loss (update). Otolaryngol. Head Neck Surg. 161(Suppl. 1), S1–S45 (2019).
    • 2. Olusanya BO, Davis AC, Hoffman HJ. Hearing loss grades and the International classification of functioning, disability and health. Bull. World Health Organ. 97(10), 725 (2019).
    • 3. Olusanya BO, Neumann KJ, Saunders JE. The global burden of disabling hearing impairment: a call to action. Bull. World Health Organ. 92(5), 367–373 (2014).
    • 4. Pang J, Xiong H, Lin P et al. Activation of miR-34a impairs autophagic flux and promotes cochlear cell death via repressing ATG9A: implications for age-related hearing loss. Cell Death Dis. 8(10), e3079 (2017).
    • 5. Lee YY, Kim YJ, Gil ES, Kim H, Jang JH, Choung Y-H. Type 1 diabetes induces hearing loss: functional and histological findings in an Akita mouse model. Biomedicines 8(9), 343 (2020).
    • 6. Ohinata Y, Miller JM, Schacht J. Protection from noise-induced lipid peroxidation and hair cell loss in the cochlea. Brain Res. 966(2), 265–273 (2003).
    • 7. Benkafadar N, François F, Affortit C et al. ROS-induced activation of DNA damage responses drives senescence-like state in postmitotic cochlear cells: implication for hearing preservation. Mol. Neurobiol. 56(8), 5950–5969 (2019).
    • 8. Baker K, Staecker H. Low dose oxidative stress induces mitochondrial damage in hair cells. Anatom. Record 295(11), 1868–1876 (2012).
    • 9. Yamashita S, Matsuzawa Y. Where are we with probucol: a new life for an old drug? Atherosclerosis 207(1), 16–23 (2009).
    • 10. Tanous D, Hime N, Stocker R. Anti-atherosclerotic and anti-diabetic properties of probucol and related compounds. Redox. Rep. 13(2), 48–59 (2008).
    • 11. Yamamoto A. A uniqe antilipidemic drug–probucol. J. Atheroscler. Thromb. 15(6), 304–305 (2008).
    • 12. Endo K, Miyashita Y, Sasaki H et al. Probucol delays progression of diabetic nephropathy. Diabetes Res. Clin. Pract. 71(2), 156–163 (2006).
    • 13. Kondo S, Shimizu M, Urushihara M et al. Addition of the antioxidant probucol to angiotensin II Type I receptor antagonist arrests progressive mesangioproliferative glomerulonephritis in the rat. J. Am. Soc. Nephrol. 17(3), 783–794 (2006).
    • 14. Takechi R, Galloway S, Pallebage-Gamarallage MM, Lam V, Dhaliwal SS, Mamo JC. Probucol prevents blood-brain barrier dysfunction in wild-type mice induced by saturated fat or cholesterol feeding. Clin. Exp. Pharmacol. Physiol. 40(1), 45–52 (2013).
    • 15. Ribeiro RP, Moreira ELG, Santos DB et al. Probucol affords neuroprotection in a 6-OHDA mouse model of Parkinson's disease. Neurochem. Res. 38(3), 660–668 (2013).
    • 16. Santos DB, Peres KC, Ribeiro RP et al. Probucol, a lipid-lowering drug, prevents cognitive and hippocampal synaptic impairments induced by amyloid β peptide in mice. Exp. Neurol. 233(2), 767–775 (2012).
    • 17. 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).
    • 18. Wagle SR, Kovacevic B, Walker D et al. Alginate-based drug oral targeting using bio-micro/nano encapsulation technologies. Exp. Opin. Drug Deliv. 17(10), 1361–1376 (2020).
    • 19. 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. Scient. Rep. 10(1), 106 (2020).
    • 20. 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. Scient. Rep. 10(1), 7715 (2020).
    • 21. 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).
    • 22. 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).
    • 23. Mooranian A, Ionescu CM, Walker D et al. Single-cellular biological effects of cholesterol-catabolic bile acid-based nano/micro capsules as anti-inflammatory cell protective systems. Biomolecules 12(1) 19 (2022).
    • 24. 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 Design, Develop. Ther. 8, 1673 (2014).
    • 25. Mooranian A, Negrulj R, Mathavan S et al. An advanced microencapsulated system: a platform for optimized oral delivery of antidiabetic drug-bile acid formulations. Pharmaceut. Develop. Technol. 20(6), 702–709 (2015).
    • 26. 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. Translat. Res. 8(3), 543–551 (2018).
    • 27. Carey L, Walker D, Jones M et al. Bile acid permeation enhancement for inner ear cochlear drug pharmacological uptake: bio-nanotechnologies in chemotherapy-induced hearing loss. Therapeut. Deliv. 12(12), 807–819 (2021).
    • 28. 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).
    • 29. Kasai S, Mimura J, Ozaki T, Itoh K. Emerging regulatory role of Nrf2 in iron, heme, and hemoglobin metabolism in physiology and disease. Front. Vet. Sci. 5, 242 (2018).
    • 30. Zhou Z, Liu C, Chen S et al. Activation of the Nrf2/ARE signaling pathway by probucol contributes to inhibiting inflammation and neuronal apoptosis after spinal cord injury. Oncotarget 8(32), 52078–52093 (2017).
    • 31. Szeto B, Chiang H, Valentini C, Yu M, Kysar JW, Lalwani AK. Inner ear delivery: challenges and opportunities. Laryngosc. Investig. Otolaryngol. 5(1), 122–131 (2019).
    • 32. Yang K-J, Son J, Jung SY et al. Optimized phospholipid-based nanoparticles for inner ear drug delivery and therapy. Biomaterials 171, 133–143 (2018).
    • 33. Lajud SA, Nagda DA, Qiao P et al. A novel chitosan-hydrogel-based nanoparticle delivery system for local inner ear application. Otol. Neurotol. 36(2), 341 (2015).
    • 34. Pyykkö I, Zou J, Schrott-Fischer A, Glueckert R, Kinnunen P. An overview of nanoparticle based delivery for treatment of inner ear disorders. Audit. Vestib. Res. 1427, 363–415 (2016).
    • 35. Mondalek FG, Zhang YY, Kropp B et al. The permeability of SPION over an artificial three-layer membrane is enhanced by external magnetic field. J. Nanobiotechnol. 4(1), 1–9 (2006).
    • 36. Cai H, Wen X, Wen L et al. Enhanced local bioavailability of single or compound drugs delivery to the inner ear through application of PLGA nanoparticles via round window administration. Int. J. Nanomed. 9, 5591–5601 (2014).
    • 37. Wagle SR, Ionescu CM, Kovacevic B et al. Pharmaceutical characterization of probucol bile acid-lithocholic acid nanoparticles to prevent chronic hearing related and similar cellular oxidative stress pathologies. Nanomedicine 18(12), 923–940 (2023).
    • 38. Mooranian A, Jones M, Ionescu CM et al. Advancements in assessments of bio-tissue engineering and viable cell delivery matrices using bile acid-based pharmacological biotechnologies. Nanomaterials 11(7), 1861 (2021).
    • 39. Mooranian A, Foster T, Ionescu CM et al. Enhanced bilosomal properties resulted in optimum pharmacological effects by increased acidification pathways. Pharmaceutics 13(8), 1184 (2021).
    • 40. Kovacevic B, Ionescu CM, Wagle SR et al. Impact of novel teflon-DCA nanogel matrix on cellular bioactivity. J. Pharm. Sci. 112(3), 700–707 (2022).
    • 41. Kalinec GM, Park C, Thein P, Kalinec F. Working with auditory HEI-OC1 cells. J. Vis. Exp. doi: 10.3791/54425 (2016).
    • 42. Wu J, Ye J, Kong W, Zhang S, Zheng Y. Programmed cell death pathways in hearing loss: a review of apoptosis, autophagy and programmed necrosis. Cell Prolif. 53(11), e12915 (2020).
    • 43. Tan WJT, Song L. Hearing research special issue: mitochondrial function and dysfunction in the inner ear. Hear. Res. 434, 108783 (2023).
    • 44. He RR, Li Y, Li XD et al. A new oxidative stress model, 2,2-azobis(2-amidinopropane) dihydrochloride induces cardiovascular damages in chicken embryo. PLOS ONE 8(3), e57732 (2013).
    • 45. 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).
    • 46. Ehret G, Frankenreiter M. Quantitative analysis of cochlear structures in the house mouse in relation to mechanisms of acoustical information processing. J. Comp. Physiol. 122, 65–85 (1977).
    • 47. Fujimoto C, Yamasoba T. Oxidative stresses and mitochondrial dysfunction in age-related hearing loss. Oxid. Med. Cell. Longev. 2014, 582849 (2014).
    • 48. Li P, Li S, Wang L et al. Mitochondrial dysfunction in hearing loss: oxidative stress, autophagy and NLRP3 inflammasome. Front. Cell Developm. Biol. 11, 1119773 (2023).
    • 49. Circu ML, Aw TY. Glutathione and modulation of cell apoptosis. Biochim. et Biophys. Acta (BBA) – Molecular Cell Research 1823(10), 1767–1777 (2012).
    • 50. Pallepati P, Averill-Bates D. Mild thermotolerance induced at 40°C increases antioxidants and protects HeLa cells against mitochondrial apoptosis induced by hydrogen peroxide: role of p53. Arch. Biochem. Biophys. 495(2), 97–111 (2010).
    • 51. Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signaling pathways by reactive oxygen species. Biochim. Biophys. Acta 1863(12), 2977–2992 (2016).
    • 52. Twiddy D, Cain K. Caspase-9 cleavage, do you need it? Biochem. J. 405(1), e1–e2 (2007).
    • 53. Luna-Vargas MP, Chipuk JE. Physiological and pharmacological control of BAK, BAX, and beyond. Trend. Cell Biol. 26(12), 906–917 (2016).
    • 54. Martinou J-C, Youle Richard J. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Developmen. Cell 21(1), 92–101 (2011).
    • 55. Honkura Y, Matsuo H, Murakami S et al. NRF2 is a key target for prevention of noise-induced hearing loss by reducing oxidative damage of cochlea. Scient. Rep. 6(1), 19329 (2016).
    • 56. Honkura Y, Matsuo H, Murakami S et al. NRF2 is a key target for prevention of noise-induced hearing loss by reducing oxidative damage of cochlea. Scient. Rep. 6(1), 1–12 (2016).
    • 57. Cazanave S, Berson A, Haouzi D et al. High hepatic glutathione stores alleviate Fas-induced apoptosis in mice. J. Hepatol. 46(5), 858–868 (2007).
    • 58. Reinehr R, Becker S, Eberle A, Grether-Beck S, Häussinger D. Involvement of NADPH oxidase isoforms and Src family kinases in CD95-dependent hepatocyte apoptosis*. J. Biol. Chem. 280(29), 27179–27194 (2005).
    • 59. Matsumaru K, Ji C, Kaplowitz N. Mechanisms for sensitization to TNF-induced apoptosis by acute glutathione depletion in murine hepatocytes. Hepatology 37(6), 1425–1434 (2003).
    • 60. Takeya R, Ueno N, Kami K et al. Novel human homologues of p47phox and p67phox participate in activation of superoxide-producing NADPH oxidases*. J. Biol. Chem. 278(27), 25234–25246 (2003).
    • 61. Thompson JA, Franklin CC. Enhanced glutathione biosynthetic capacity promotes resistance to As3+-induced apoptosis. Toxicol. Lett. 193(1), 33–40 (2010).
    • 62. Lluis JM, Morales A, Blasco C et al. Critical role of mitochondrial glutathione in the survival of hepatocytes during hypoxia*. J. Biol. Chem. 280(5), 3224–3232 (2005).
    • 63. Yamasoba T, Nuttall AL, Harris C, Raphael Y, Miller JM. Role of glutathione in protection against noise-induced hearing loss. Brain Res. 784(1–2), 82–90 (1998).
    • 64. Hu BH, Zheng XY, McFadden SL, Kopke RD, Henderson D. R-phenylisopropyladenosine attenuates noise-induced hearing loss in the chinchilla. Hear Res. 113(1–2), 198–206 (1996).
    • 65. Vitale N, Kisslinger A, Paladino S et al. Resveratrol couples apoptosis with autophagy in UVB-irradiated HaCaT cells. PLOS ONE 8(11), e80728 (2013).
    • 66. Hosokawa K, Hosokawa S, Ishiyama G, Ishiyama A, Lopez IA. Immunohistochemical localization of Nrf2 in the human cochlea. Brain Res. 1700, 1–8 (2018).
    • 67. Zhang W, Xiong H, Pang J et al. Nrf2 activation protects auditory hair cells from cisplatin-induced ototoxicity independent on mitochondrial ROS production. Toxicol. Lett. 331, 1–10 (2020).
    • 68. Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J. Biol. Chem. 284(20), 13291–13295 (2009).
    • 69. Jiang T, Huang Z, Lin Y, Zhang Z, Fang D, Zhang DD. The protective role of Nrf2 in streptozotocin-induced diabetic nephropathy. Diabetes 59(4), 850–860 (2010).
    • 70. Kong L, Chen G-D, Zhou X, McGinnis JF, Li F, Cao W. Molecular mechanisms underlying cochlear degeneration in the tubby mouse and the therapeutic effect of sulforaphane. Neurochem. Int. 54(3), 172–179 (2009).
    • 71. Hoshino T, Tabuchi K, Nishimura B et al. Protective role of Nrf2 in age-related hearing loss and gentamicin ototoxicity. Biochem. Biophys. Res. Commun. 415(1), 94–98 (2011).
    • 72. Li D, Zhao H, Cui Z-K, Tian G. The role of Nrf2 in hearing loss. Front. Pharmacol. 12, 620921 (2021).
    • 73. Wang X, Zeng C, Lai Y et al. NRF2/HO-1 pathway activation by ATF3 in a noise-induced hearing loss murine model. Arch. Biochem. Biophys. 721, 109190 (2022).
    • 74. Huang J-L, Yu C, Su M et al. Probucol, a “non-statin” cholesterol-lowering drug, ameliorates D-galactose induced cognitive deficits by alleviating oxidative stress via Keap1/Nrf2 signaling pathway in mice. Aging 11(19), 8542 (2019).
    • 75. Zhongping C, Li C, Ai S, Zhou X. Effects of probucol on high glucose-induced expressions of SP1/Keap1/Nrf2/GCLC in the cultured human Müller cells. Investig. Ophthalmol. Vis. Sci. 60(9), 5368 (2019).
    • 76. Wang H, Zhang K, Ruan Z et al. Probucol enhances the therapeutic efficiency of mesenchymal stem cells in the treatment of erectile dysfunction in diabetic rats by prolonging their survival time via Nrf2 pathway. Stem Cell Res. Ther. 11(1), 302 (2020).
    • 77. Abe H, Arai H, Sakurai A, Yamashita K, Murayama T, Minami M. Probucol modulates Nrf2 function and ameliorates diabetic nephropathy in db/db mice. Med. Res. Archiv. 4(8), (2016).