1. Morikawa, T., Yasuno, R., and Wada, H. Do mammalian cells synthesize lipoic acid? Identification of a mouse cDNA encoding a lipoic acid synthase located in mitochondria, FEBS Letters 2001, 498, 16-21, doi.org/10.1016/S0014-5793(01)02469-3.
2. Onder, N. T., Alcay, S., and Nur, Z. Effects of alpha‐lipoic acid on ram semen cryopreservation and post‐thaw life span, Andrologia 2022, 54, e14249, doi.org/10.1111/and.14249.
3. Salehi, B., Yılmaz, B., Antika, G., Boyunegmez, T. T., Fawzi, M., and Sharifi-Rad, J. Insights on the use of α-lipoic acid for therapeutic purposes, Biomolecules 2019, 9, 356, doi.org/10.3390/biom9080356.
4. Tibullo, D., Li Volti, G., Giallongo, C., Grasso, S., Tomassoni, D., and Bramanti, V. Biochemical and clinical relevance of alpha lipoic acid: antioxidant and anti-inflammatory activity, molecular pathways and therapeutic potential, Inflammation Research 2017, 66, 947–959, doi.org/10.1007/s00011-017-1079-6.
5. Nikolić, R. S., Krstić, N. S., Nikolić, G. M., Kocić, G. M., and Signelković, D. H. Molecular mechanisms of beneficial effects of lipoic acid in copper intoxicated rats assessment by FTIR and ESI-MS, Polyhedron 2014, 80, 223-227, doi.org/10.1016/j.poly.2014.04.033.
6. Baumgartner, M. R., Schmalle, H., and Dubler, E. The interaction of transition metals with the coenzyme α-lipoic acid: synthesis, structure and characterization of copper and zinc complexes, Inorganica Chim. Acta 1996, 252, 319-331, doi.org/10.1016/S0020-1693(96)05331-5.
7. Uchida, R., Okamoto, H., Ikuta, N., and Hirota, T. Enantioselective pharmacokinetics of α-lipoic acid in rats, Int. J. Mol. Sci. 2015, 16, 22781-22794, doi.org/10.3390/ijms160922781.
8. Devasagayam, T. P. A., Subramanian, M., and Sies, H. Prevention of singlet oxygen-induced DNA damage by lipoate, Chem. Biol. Interact. 1993, 86, 79-92, doi.org/10.1016/0009-2797(93)90113-D.
9. Tripathi, A. K., Ray, A. K., Mishra, S. K., Bishen, S. M., and Khurana, A. Molecular and therapeutic insights of alpha-lipoic acid as a potential molecule for disease prevention, Rev. Bras. Farmacogn. 2023, 33, 272–287, doi.org/10.1007/s43450-023-00370-1.
10. Rezaei, Z. S., Hasani, M., Morvaridzadeh, M., Beatriz, P. A., Heydari, H., and Heshmati, J. Effect of alpha-lipoic acid on oxidative stress parameters: a systematic review and meta-analysis, J. Funct. Foods 2021, 87, 104774, doi.org/10.1016/j.jff.2021.104774.
11. Monastra, G., De Grazia, S., and Unfer, V. Immunomodulatory activities of alpha lipoic acid with a special focus on its efficacy in preventing miscarriage, Expert Opin. Drug Deliv. 2016, 13, 1695-1708, doi.org/10.1080/17425247.2016.1200556.
12. Cremer, D. R., Rabeler, R., and Lynch, B. Safety evaluation of α-lipoic acid (ALA), Regul. Toxicol. Pharmacol. 2006, 46, 29-41, doi.org/10.1016/j.yrtph.2006.06.004.
13. Ou, P., Tritschler, H. J., and Wolff, S. P. Thioctic (lipoic) acid: a therapeutic metal-chelating antioxidant? Biochem. Pharmacol. 1995, 50, 123-126, doi.org/10.1016/0006-2952(95)00116-H.
14. Spector, A., Huang, R. R. C., and Wang, R. R. Thioredoxin fragment 31-36 is reduced by dihydrolipoamide and reduces oxidized protein, Biochem. Biophys. Res. Commun. 1988, 150, 156-162, doi.org/10.1016/0006-291X(88)90499-8.
15. Guseva, E. A., Pavlova, J. A., and Sergiev, P. V. Synthetic activators of autophagy, Biochemistry (Moscow) 2024, 89, 27–52, doi.org/10.1134/S0006297924010024.
16. Pavlova, J. A., Guseva, E. A., and Sergiev, P. V. Natural activators of autophagy, Biochemistry (Moscow) 2024, 89, 1–26, doi.org/10.1134/S0006297924010012.
17. Bossio, S., Perri, A., Gallo, R., and Aversa, A. Alpha-lipoic acid reduces cell growth, inhibits autophagy, and counteracts prostate cancer cell migration and invasion: evidence from in vitro studies, Int. J. Mol. Sci. 2023, 24, 17111, doi.org/10.3390/ijms242317111.
18. Peng, P., Zhang, X., Qi, T., Cheng, H., Kong, Q., and Ding, Z. Alpha‐lipoic acid inhibits lung cancer growth via mTOR‐mediated autophagy inhibition, FEBS Open Bio. 2020, 10, 607–618, doi.org/10.1002/2211-5463.12820.
19. Stoll, S., Hartmann, H., Cohen, S. A., and Müller, W. E. The potent free radical scavenger α-lipoic acid improves memory in aged mice: putative relationship to NMDA receptor deficits, Pharmacol. Biochem. Behav. 1993, 46, 799-805, doi.org/10.1016/0091-3057(93)90204-7.
20. Stoll, S., Rostock, A., Bartsch, R., Korn, E., and Müller, W. E. The potent free radical scavenger α‐lipoic acid improves cognition in rodents, Ann. N. Y. Acad. Sci. 1994, 717, 122-128, doi.org/10.1111/j.1749-6632.1994.tb12079.x.
21. Xu, C., Li, E., Liu, S., Huang, Z., and Chen, L. Effects of α-lipoic acid on growth performance, body composition, antioxidant status and lipid catabolism of juvenile Chinese mitten crab Eriocheir sinensis fed different lipid percentage, Aquaculture 2018, 484, 286-292, doi.org/10.1016/j.aquaculture.2017.09.036.
22. Lovell, M. A., Xie, C., Xiong, S., and Markesbery, W. R. Protection against amyloid beta peptide and iron/hydrogen peroxide toxicity by alpha lipoic acid, J. Alzheimer’s Dis. 2003, 5, 229-239, doi.org/10.3233/JAD-2003-5306.
23. Fiedler, S. E., Yadav, V., Kerns, A. R., Tsang, C., Markwardt, S., and Salinthone, S. Lipoic acid stimulates cAMP production in healthy control and secondary progressive MS subjects, Mol. Neurobiol. 2018, 55, 6037-6049, doi.org/10.1007/s12035-017-0813-y.
24. Scumpia, P. O., and Stevens, B. R. Alpha-lipoic acid effects on brain glial functions accompanying double-stranded RNA antiviral and inflammatory signaling, Neurochem. Int. 2014, 64, 55-63, doi.org/10.1016/j.neuint.2013.11.006.
25. Micili, S. C., Goker, A., Kuscu, K., and Fuso, A. α-lipoic acid vaginal administration contrasts inflammation and preterm delivery in rats, Reproductive Sci. 2019, 26, 128-138, doi.org/10.1177/1933719118766266.
26. Suh, J. H., Shenvi, S. V., and Hagen, T. M. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid, Proc. Natl. Acad. Sci. U. S. A. 2004, 101, 3381-3386, doi.org/10.1073/pnas.0400282101.
27. Kozlov, A. V., Gille, L., Staniek, K., and Nohl, H. Dihydrolipoic acid maintains ubiquinone in the antioxidant active form by two-electron reduction of ubiquinone and one-electron reduction of ubisemiquinone, Arch. Biochem. Biophys. 1999, 363, 148-154, doi.org/10.1006/abbi.1998.1064.
28. Chen, W. L., Kang, C. H., and Lee, H. M. α-lipoic acid regulates lipid metabolism through induction of Sirtuin 1 (SIRT1) and activation of AMP-activated protein kinase, Diabetologia 2012, 55, 1824-1835, doi.org/10.1007/s00125-012-2530-4.
29. Smith, A. R., Shenvi, S. V., Widlansky, M., and Hagen, T. M. Lipoic acid as a potential therapy for chronic diseases associated with oxidative stress, Curr. Med. Chem. 2012, 11, 1135-1146, doi.org/10.2174/0929867043365387.
30. Zhang, W. J., Bird, K. E., and Frei, B. Dietary α-lipoic acid supplementation inhibits atherosclerotic lesion development in apolipoprotein e-deficient and apolipoprotein e/low-density lipoprotein receptor-deficient mice, Circulation 2008, 117, 421-428, doi.org/10.1161/CIRCULATIONAHA.107.725275.
31. Packer, L., Witt, E. H., and Tritschler, H. J. Alpha-lipoic acid as a biological antioxidant, Free Radic. Biol. Med. 1995, 19, 227-250, doi.org/10.1016/0891-5849(95)00017-R.
32. Abdulghani, M., and Naser, A. Estimation of pharmacokinetic parameters of alpha-lipoic acid in the chicks model, Baghdad J. Biochem. Appl. Biol. Sci. 2022, 3, 122-132, doi.org/10.47419/bjbabs.v3i02.91.
33. Hagen, T. M., Ingersoll, R. T., and Ames, B. N. (R)‐α‐Lipoic acid‐supplemented old rats have improved mitochondrial function, decreased oxidative damage, and increased metabolic rate, The FASEB Journal 1999, 13, 411-418, doi.org/10.1096/fasebj.13.2.411.
34. Murali, P., George, S. K., and Dominic, G. Effect of dietary supplementation of l-carnitine on serum lipid profile and antioxidant status in broiler chicken fed with animal fat-rich diet, Appl. Biol. Res. 2020, 22, 118-122, doi.org/10.5958/0974-4510.2020.00018.9.
35. Banik, S., Halder, S., and Onoue, S. Self-emulsifying drug delivery system of (R)-α-lipoic acid to improve its stability and oral absorption, Biopharm. Drug Dispos. 2021, 42, 226-233, doi.org/10.1002/bdd.2277.
36. Brufani, M. α-lipoic acid: drug or dietary supplement? An overview on the pharmacokinetics, available for-mulations and clinical evidence in the diabetes complications, Progress in Nutrition 2014, 16, 62-74, doi.org/10.23751/pn.v19i1.6325.
37. Hermann, R., Mungo, J., and Ziegler, D. Enantiomer-selective pharmacokinetics, oral bioavailability, and sex effects of various alpha-lipoic acid dosage forms, Clin. Pharmacol. 2014, 6, 195–204, doi.org/10.2147/CPAA.S71574.
38. Bustamante, J., Lodge, J. K., Marcocci, L., Tritschler, H. J., and Rihn, B. H. α-lipoic acid in liver metabolism and disease, Free Radic. Biol. Med. 1998, 24, 1023-1039, doi.org/10.1016/S0891-5849(97)00371-7.
39. Wepierre, J., Corroller, M., and Dupuis, D. In vivo cutaneous distribution of linoleic acid following topical application in the hairless rat, J. Soc. Cosmet. Chem. Jpn. 1986, 37, 191-198, doi.org/10.1007/bf00455174.
40. Wepierre, J., Corroller, M., and Didry, J. R. Distribution and dissociation of benzoyl peroxide in cutaneous tissue after application on skin in the hairless rat, Int. J. Cosmet. Sci. 1986, 8, 97-104, doi.org/10.1111/j.1467-2494.1986.tb00437.x.
41. Podda, M., Rallis, M., Traber, M. G., and Maiboch, H. I. Kinetic study of cutaneous and subcutaneous distribution following topical application of [7,8-14C]rac-α-lipoic acid onto hairless mice, Biochem. Pharmacol. 1996, 52, 627-633, doi.org/10.1016/0006 2952(96)00337-1.
42. Teichert, J., Hermann, R., and Preiss, R. Plasma kinetics, metabolism, and urinary excretion of alpha-lipoic acid following oral administration in healthy volunteers, J. Clin. Pharmacol. 2003, 43, 1257-1267, doi.org/10.1177/0091270003258654.
43. Angelucci, L., and Mascitelli-Coriandoli, E. Anticholesterol activity of α-lipoic acid, Nature 1958, 182, 911-912, doi.org/10.1038/182396b0.
44. Brufani, M., and Figliola, R. (R)-α-lipoic acid oral liquid formulation: pharmacokinetic parameters and therapeutic efficacy, Acta Biomedica 2014, 85, 108-115, PMID: 25245645.
45. Teichert, J., Tuemmers, T., Achenbach, H., Preiss, C., and Preiss, R. Pharmacokinetics of alpha-lipoic acid in subjects with severe kidney damage and end-stage renal disease, J. Clin. Pharmacol. 2005, 45, 313-328, doi.org/10.1177/0091270004270792.
46. Akünal, T. C., and Yunusoğlu, O. Oleanolic acid suppresses pentylenetetrazole-induced seizure in vivo, Int. J. Environ. Health Res. 2023, 33, 529–540, doi.org/10.1080/09603123.2023.2167947.
47. Berköz, M., Yunusoğlu, O., and Bozkurt, A. Investigation of antiepileptic potentials of usnic acid and some lichen species on the behavioral and biochemical levels in pentylenetetrazole-induced kindling model of epilepsy, J. Res. Pharm. 2024, 28, 1378–1390, doi.org/10.29228/jrp.816.
48. Yunusoğlu, O., Ayaz, İ., & Dovankaya, E. H. Pharmacological, medicinal and biological properties of flavonoids: A comprehensive review. Journal of Research in Pharmacy 2025, 29(2), 561-584. doi.org/10.12991/jrespharm.1661054
49. Sevindik, M., Krupodorova, T., Sevindik, E., Koçer, O., Uysal, I., & Ünal, O. Elaeagnus angustifolia L.: A Comprehensive Review of Its Biological Activities, Phenolic and Chemical Constituents, and Applications. Applied Fruit Science 2025, 67(2), 70, doi.org/10.1007/s10341-025-01294-x.
50. Lasota, M., Jankowski, D., Wiśniewska, A., Szeleszczuk, Ł., Misterka-Kozaka, A., Kaczor-Kamińska, M., ... & Brzozowski, T. Interaction of avapritinib with congo red in pancreatic cancer cells: molecular modeling and biophysical studies. International Journal of Molecular Sciences 2025, 26(5), 1980. doi.org/10.3390/ijms26051980.
51. Shay, K. P., Moreau, R. F., Smith, E. J., and Hagen, T. M. Alpha-lipoic acid as a dietary supplement: molecular mechanisms and therapeutic potential, Biochim. Biophys. Acta Gen. Subj. 2009, 1790, 1149–1160, doi.org/10.1016/j.bbagen.2009.07.026.
52. Peng, P., Zhang, X., Qi, T., Cheng, H., Kong, Q., and Ding, Z. Alpha‐lipoic acid inhibits lung cancer growth via mTOR‐mediated autophagy inhibition, FEBS Open Bio. 2020, 10, 607–618, doi.org/10.1002/2211-5463.12820.
53. Hassan, A., Ibrahim, A., Mbodji, K., Coëffier, M., Ziegler, F., and Marion-Letellier, R. An α-linolenic acid-rich formula reduces oxidative stress and inflammation by regulating NF-κB in rats with TNBS-induced colitis, J. Nutr. 2010, 140, 1714-1721, doi.org/10.3945/jn.109.119768.
54. Kütter, M. T., Monserrat, J. M., Primel, E. G., and Tesser, M. B. Effects of dietary α-lipoic acid on growth, body composition and antioxidant status in the plata pompano Trachinotus Marginatus (Pisces, Carangidae), Aquaculture 2012, 368–369, 29–35, doi.org/10.1016/j.aquaculture.2012.09.010.
55. Cruz, L. C., Josende, M. E., Tavares, P. B., Wasielesky, W., and Maciel, F. E. Lipoic acid modulates energetic metabolism and antioxidant defense systems in Litopenaeus Vannamei under hypoxia/reoxygenation conditions, Aquaculture 2018, 497, 396–404, doi.org/10.1016/j.aquaculture.2018.08.020.
56. He, M., Wu, Y., Hong, M., Yun, Z., and Jiang, Y. α-Lipoic acid treatment alleviates postharvest pericarp browning of litchi fruit by regulating antioxidant ability and energy metabolism, Postharvest Biol. Technol. 2021, 180, 111629, doi.org/10.1016/j.postharvbio.2021.111629.
57. Turk, H., Erdal, S., Karayel, U., and Dumlupinar, R. Attenuation of lead toxicity by promotion of tolerance mechanism in wheat roots by lipoic acid, Cereal Res. Commun. 2018, 46, 424–435, doi.org/10.1556/0806.46.2018.020.
58. Monserrat, J. M., Garcia, M. L., Ventura-Lima, J., González, M., Ballesteros, M. L., and Wunderlin, D. A. Antioxidant, phase II and III responses induced by lipoic acid in the fish Jenynsia Multidentata (Anablapidae) and its influence on endolsulfan accumulation and toxicity, Pesticide Biochem. Physiol. 2014, 108, 8–15, doi.org/10.1016/j.pestbp.2013.10.009.
59. Zhao, G., Hu, C., and Xue, Y. In vitro evaluation of chitosan‐coated liposome containing both coenzyme Q10 and alpha‐lipoic acid: cytotoxicity, antioxidant activity, and antimicrobial activity, J. Cosmet. Dermatol. 2018, 17, 258–262, doi.org/10.1111/jocd.12369.
60. Piechota-Polanczyk, A., Zielińska, M., and Fichna, J. The influence of lipoic acid on caveolin-1-regulated antioxidative enzymes in the mouse model of acute ulcerative colitis, Biomed. Pharmacother. 2016, 84, 470-475, doi.org/10.1016/j.biopha.2016.09.066.
61. Bhattacharyya, A., Chattopadhyay, R., and Crowe, S. E. Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases, Physiol. Rev. 2014, 94, 329-354, doi.org/10.1152/physrev.00040.2012.
62. Packer, L., Kraemer, K., and Rimbach, G. Molecular aspects of lipoic acid in the prevention of diabetes complications, Nutrition 2001, 17, 888-895, doi.org/10.1016/S0899-9007(01)00658-X.
63. Sezgin, A., Altuntaş, C., Demiralay, M., and Terzi, R. Exogenous alpha lipoic acid can stimulate photosystem II activity and the gene expressions of carbon fixation and chlorophyll metabolism enzymes in maize seedlings under drought, J. Plant Physiol. 2019, 232, 65–73, doi.org/10.1016/j.jplph.2018.11.026.
64. Biewenga, G. P., Dorstijn, M. A., Verhagen, J. V., and Bast, A. Reduction of lipoic acid by lipoamide dehydrogenase, Biochem. Pharmacol. 1996, 51, 233-238, doi.org/10.1016/0006-2952(95)02124-8.
65. Tan, W., Zhang, J., Mi, Y., and Guo, Z. Synthesis and characterization of α-lipoic acid grafted chitosan derivatives with antioxidant activity, React. Funct. Polym. 2022, 172, 105205, doi.org/10.1016/j.reactfunctpolym.2022.105205.
66. El Basuini, M. F., Shahin, S. A., Eldenary, M. E., Elshora, S. M., and Mourad, M. M. Growth variables, feed efficacy, survival rate, and antioxidant capacity of european seabass (Dicentrarchus Labrax l.) larvae treated with coenzyme Q10 or lipoic acid, Aquac. Rep. 2022, 27, 101373, doi.org/10.1016/j.aqrep.2022.101373.
67. Huang, C., Sun, J., Ji, H., Oku, H., Chang, Z., and Xie, J. Influence of dietary alpha-lipoic acid and lipid level on the growth performance, food intake and gene expression of peripheral appetite regulating factors in juvenile grass carp (Ctenopharyngodon Idellus), Aquaculture 2019, 505, 412–422, doi.org/10.1016/j.aquaculture.2019.02.054.
68. Biewenga, G. P., and Bast, A. The pharmacology of the antioxidant: lipoic acid, Gen. Pharmacol. 1997, 29, 315-331, doi.org/10.1016/S0306-3623(96)00474-0.
69. Lopez-Maldonado, A., Pastoriza, S., and Rufián-Henares, J. Á. Assessing the antioxidant and metabolic effect of an alpha-lipoic acid and acetyl-L-carnitine nutraceutical, Curr. Res. Food Sci. 2021, 4, 336-344, doi.org/10.1016/j.crfs.2021.05.002.
70. Singer, A. J., and Clark, R. A. F. Cutaneous wound healing, N. Engl. J. Med. 1999, 341, 738–746, doi.org/10.1056/NEJM199909023411006.
71. Leu, J.G., Chen, S.A., Chen, H.M., Wu, W.M., Hung, C.F., and Liang, Y.J. The effects of gold nanoparticles in wound healing with antioxidant epigallocatechin gallate and α-lipoic acid, Nanomedicine 2012, 8, 767–775, doi.org/10.1016/j.nano.2011.08.013.
72. Alleva, R., Tomasetti, M., Sartini, D., Emanuelli, M., Nasole, E., and Neuzil, J. α-Lipoic acid modulates extracellular matrix and angiogenesis gene expression in non-healing wounds treated with hyperbaric oxygen therapy, Mol. Med. 2008, 14, 175-183, doi.org/10.2119/2007-00095.Alleva.
73. Salinthone, S., Schillace, R. V., Marracci, G. H., and Carr, D. W. Lipoic acid stimulates cAMP production via the EP2 and EP4 prostanoid receptors and inhibits IFN gamma synthesis and cellular cytotoxicity in NK cells, J. Neuroimmunol. 2008, 199, 46-55, doi.org/10.1016/j.jneuroim.2008.05.003.
74. Tanaka, Y., Kaibori, M., Miki, H., Nakatake, R., Tokuhara, K., and Kwon, A. H. Alpha-lipoic acid exerts a liver-protective effect in acute liver injury rats, J. Surg. Res. 2015, 193, 675-683, doi.org/10.1016/j.jss.2014.08.057.
75. Mor, G., Cardenas, I., Abrahams, V., and Guller, S. Inflammation and pregnancy: the role of the immune system at the implantation site, Ann. N. Y. Acad. Sci. 2011, 1221, 80-87, doi.org/10.1111/j.1749-6632.2010.05938.x.
76. Bao, P., Kodra, A., Tomic-Canic, M., Golinko, M. S., and Brem, H. The role of vascular endothelial growth factor in wound healing, J. Surg. Res. 2009, 153, 347-358, doi.org/10.1016/j.jss.2008.04.023.
77. Gora̧ca, A., Piechota, A., Kleniewska, P., and Skibska, B. Lipoic acid - biological activity and therapeutic potential, Pharmacol. Rep. 2011, 63, 849-858, doi.org/10.1016/S1734-1140(11)70600-4.
78. Collins, T., and Maniatis, T. Transcriptional regulation of endothelial cell adhesion molecules: NF-kappa B and cytokine-inducible enhancers, FASEB J. 1995, 9, 899-909, doi.org/10.1096/fasebj.9.10.7542214.
79. Zhang, W. J., and Frei, B. α‐lipoic acid inhibits TNF‐a‐induced NF‐κB activation and adhesion molecule expression in human aortic endothelial cells, FASEB J. 2001, 15, 2423-2432, doi.org/10.1096/fj.01-0260com.
80. Weber, C., Erl, W., Pietsch, A., and Weber, P. C. Aspirin inhibits nuclear factor-κB mobilization and monocyte adhesion in stimulated human endothelial cells, Circulation 1995, 91, 1914-1917, doi.org/10.1161/01.CIR.91.7.1914.
81. Albelda, S. M., Smith, C. W., and Ward, P. A. Adhesion molecules and inflammatory injury, FASEB J. 1994, 8, 504-512, doi.org/10.1096/fasebj.8.8.8181668.
82. Yadav, V., Marracci, G., Lovera, J., Woodward, W., Bogardus, K., and Bourdette, D. N. Lipoic acid in multiple sclerosis: a pilot study, Multiple Sclerosis 2005, 11, 159-165, doi.org/10.1191/1352458505ms1143oa.
83. Ziegler, Dan. Thioctic acid for patients with symptomatic diabetic polyneuropathy: a critical review, Treat. Endocrinol. 2004, 3, 173-189, doi.org/10.2165/00024677-200403030-00005.
84. Melhem, A., Stern, M., Shibolet, O., Israeli, E., Ackerman, Z., and Ilan, Y. Treatment of chronic hepatitis C virus infection via antioxidants, J. Clin. Gastroenterol. 2005, 39, 737–742, doi.org/10.1097/01.mcg.0000174023.73472.29.
85. Jesudason, E. P., Masilamoni, J. G., and Jayakumar, R. The protective role of DL- α -lipoic acid in the oxidative vulnerability triggered by A β -amyloid vaccination in mice, Mol. Cell. Biochem. 2005, 270, 29–37, doi.org/10.1007/s11010-005-3301-z.
86. Guha, M., and Mackman, N. The phosphatidylinositol 3-kinase-akt pathway limits lipopolysaccharide activation of signaling pathways and expression of inflammatory mediators in human monocytic cells, J. Biol. Chem. 2002, 277, 32124–32132, doi.org/10.1074/jbc.M203298200.
87. Fukao, T., and Koyasu, S. PI3K and negative regulation of TLR signaling, Trends Immunol. 2003, 24, 358–363, doi.org/10.1016/S1471-4906(03)00139-X.
88. Alhakamy, N. A., Al-Rabia, M. W., Asfour, H. Z., Alshehri, S., Alharbi, W. S., and Kotta, S. 2-Methoxy-estradiol loaded alpha lipoic acid nanoparticles augment cytotoxicity in MCF-7 breast cancer cells, Dose-Response 2021, 19, 15593258211055023, doi.org/10.1177/15593258211055023.
89. Campoccia, D., Montanaro, L., and Arciola, C. R. The significance of infection related to orthopedic devices and issues of antibiotic resistance, Biomaterials 2006, 27, 2331-2339¸ doi.org/10.1016/j.biomaterials.2005.11.044.
90. Choi, M. J., Kim, S. A., and Rhee, M. S. New decontamination method based on caprylic acid in combination with citric acid or vanillin for eliminating Cronobacter Sakazakii and Salmonella Enterica serovar typhimurium in reconstituted infant formula, Int. J. Food Microbiol. 2013, 166, 499-507, doi.org/10.1016/j.ijfoodmicro.2013.08.016.
91. Abdelmoneim, D., Porter, G., Duncan, W., Lim, K., Easingwood, R., and Coates, D. Three-dimensional evaluation of the cytotoxicity and antibacterial properties of alpha lipoic acid-capped silver nanoparticle constructs for oral applications, Nanomaterials 2023, 13, 705, doi.org/10.3390/nano13040705.
92. Iversen, C., Waddington, M., and Forsythe, S. Identification and phylogeny of Enterobacter Sakazakii relative to Enterobacter and Citrobacter species, J. Clin. Microbiol. 2004, 42, 5368-5370, doi.org/10.1128/JCM.42.11.5368-5370.2004.
93. Hajtuch, J., Wojcik, M., Tomczyk, E., Jaskiewicz, M., and Inkielewicz-Stepniak, I. Lipoic acid-coated silver nanoparticles: biosafety potential on the vascular microenvironment and antibacterial properties, Front. Pharmacol. 2022, 12, 733743, doi.org/10.3389/fphar.2021.733743.
94. Martínez, G. K. D., Zertuche, A. T., Iñiguez, E., and Kretzchmar, T. Radical scavenging, hemocompatibility, and antibacterial activity against MDR acinetobacter baumannii in alginate-based aerogels containing lipoic acid-capped silver nanoparticles, ACS Omega 2024, 9, 2350-2361, doi.org/10.1021/acsomega.3c06114.
95. Wright, E. J., Brew, B. J., and Wesselingh, S. L. Pathogenesis and diagnosis of viral infections of the nervous system, Neurol. Clin. 2008, 26, 617-633, doi.org/10.1016/j.ncl.2008.03.006.
96. Cure, E., and Cumhur Cure, M. Alpha-lipoic acid may protect patients with diabetes against COVID-19 infection, Med. Hypotheses 2020, 143, 110185, doi.org/10.1016/j.mehy.2020.110185.
97. Koufaki, M., Detsi, A., and Kiziridi, C. Multifunctional lipoic acid conjugates, Curr. Med. Chem. 2009, 16, 4728-4742, doi.org/10.2174/092986709789878274.
98. Spisakova, M., Cizek, Z., and Melkova, Z. Ethacrynic and α-lipoic acids inhibit vaccinia virus late gene expression, Antiviral Res. 2009, 81, 156–165, doi.org/10.1016/j.antiviral.2008.11.001.
99. Zhang, W., Chen, X., Yu, F., Li, F., Li, W., and Jia, K. α-lipoic acid exerts its antiviral effect against viral hemorrhagic septicemia virus (VHSV) by promoting upregulation of antiviral genes and suppressing VHSV-induced oxidative stress, Virol. Sin. 2021, 36, 1520–1531, doi.org/10.1007/s12250-021-00440-5.
100. Baur, A., Harrer, T., Peukert, M., Jahn, G., and Fleckenstein, B. Alpha-lipoic acid is an effective inhibitor of human immuno-deficiency virus (HIV-1) replication, Klin. Wochenschrift 1991, 69, 722-724, doi.org/10.1007/BF01649442.
101. Ray, P. D., Huang, B.W., and Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling, Cell. Signal. 2012, 24, 981–990, doi.org/10.1016/j.cellsig.2012.01.008.
102. Huang, W.J., Zhang, X., and Chen, W.W. Role of oxidative stress in Alzheimer’s disease, Biomed. Rep. 2016, 4, 519–522, doi.org/10.3892/br.2016.630.
103. Iqbal, K., and Grundke-Iqbal, I. Alzheimer neurofibrillary degeneration: significance, etiopathogenesis, therapeutics and prevention: Alzheimer review series, J. Cell. Mol. Med. 2008, 12, 38-55, doi.org/10.1111/j.1582-4934.2008.00225.x.
104. Rubio-Perez, J. M., and Morillas-Ruiz, J. M. (2012) A review: inflammatory process in Alzheimer’s disease, role of cytokines, Sci. World J. 2012, 756357, doi.org/10.1100/2012/756357.
105. Sano, M., Ernesto, C., Thomas, R. G., Klauber, M. R., Schafer, K., and Thal, L. J. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease, N. Engl. J. Med. 1997, 336, 1216-1222, doi.org/10.1056/nejm199704243361704.
106. Dinicola, S., Proietti, S., Cucina, A., Bizzarri, M., and Fuso, A. Alpha-lipoic acid downregulates IL-1β and IL-6 by DNA hypermethylation in SK-N-BE neuroblastoma cells, Antioxidants 2017, 6, 74, doi.org/10.3390/antiox6040074.
107. Jia, Z., Hallur, S., Zhu, H., Li, Y., and Misra, H. P. Potent upregulation of glutathione and NAD(P)H: quinone oxidoreductase 1 by alpha-lipoic acid in human neuroblastoma SH-SY5Y cells: protection against neurotoxicant-elicited cytotoxicity, Neurochem. Res. 2008, 33, 790-800, doi.org/10.1007/s11064-007-9496-5.
108. Cacciatore, I., Marinelli, L., Fornasari, E., Cerasa, L. S., and Eusepi, P. Novel NSAID-derived drugs for the potential treatment of Alzheimer’s disease, Int. J. Mol. Sci. 2016, 17, 1035, doi.org/10.3390/ijms17071035.
109. Allahverdiyev, O., Dzhafar, S., Berköz, M., and Yıldırım, M. Advances in current medication and new therapeutic approaches in epilepsy, Eastern J. Med. 2018, 23, 48–59, doi.org/10.5505/ejm.2018.62534.
110. Büget, B., Türkmen, A. Z., Allahverdiyev, O., and Enginar, N. Antimuscarinic-induced convulsions in fasted animals after food intake: evaluation of the effects of levetiracetam, topiramate and different doses of atropine, Naunyn-Schmiedebergs Arch. Pharmacol. 2016, 389, 57–62, doi.org/10.1007/s00210-015-1175-5.
111. Martinc, B., Grabnar, I., and Vovk, T. The role of reactive species in epileptogenesis and influence of antiepileptic drug therapy on oxidative stress, Curr. Neuropharmacol. 2012, 10, 328-343, doi.org/10.2174/157015912804143504.
112. Javaid, M. S., Antonic-Baker, A., Pitsillou, E., Liang, J., French, C., and Anderson, A. Alpha-lipoic acid analogues in the regulation of redox balance in epilepsy: a molecular docking and simulation study, J. Mol. Graph. Model. 2022, 112, 108116, doi.org/10.1016/j.jmgm.2021.108116.
113. Sagmanligil, H., Yunusoglu, O., and Catalkaya, E. Investigation of the pharmacological, behavioral and biochemical effects of boron on rats with rotenone-induced Parkinson’s disease, Cell. Mol. Biol. 2022, 68, 13–21, doi.org/10.14715/cmb/2022.68.8.3.
114. Kaygısız, M., & Gürer, E. S. Determination of the antimicrobial, antioxidant activities and effects on oxidative DNA damage of extracts from three different Salvia species grown in Turkey. Prospects in Pharmaceutical Sciences 2025, 23(1), 1-8. doi.org/10.56782/pps.300
115. Xicoy, H., Wieringa, B., and Martens, G. J. M. The role of lipids in Parkinson’s disease, Cells 2019, 8, 1–58, doi.org/10.3390/cells8010027.
116. Zheng, Q., Ma, P., Yang, P., Zhai, S., He, M., and Zhang, C. Alpha lipoic acid ameliorates motor deficits by inhibiting ferroptosis in Parkinson’s disease, Neurosci. Lett. 2023, 810, 137346, doi.org/10.1016/j.neulet.2023.137346.
117. Piersimoni, M. E., Teng, X., and Ying, L. Antioxidant lipoic acid ligand-shell gold nanoconjugates against oxidative stress caused by α-synuclein aggregates, Nanoscale Adv. 2020, 2, 5666–5681, doi.org/10.1039/D0NA00688B.
118. Tai, S., Zheng, Q., Zhai, S., Cai, T., Xu, L., and Zhang, C. Alpha-lipoic acid mediates clearance of iron accumulation by regulating iron metabolism in a Parkinson’s disease model induced by 6-OHDA, Front. Neurosci. 2020, 14, 612, doi.org/10.3389/fnins.2020.00612.
119. Li, X., Zou, Y., Xing, J., Wang, K. Y., and Zhai, X. Y. A-lipoic acid alleviates folic acid-induced renal damage through inhibition of ferroptosis, Front. Physiol. 2021, 12, 680544, doi.org/10.3389/fphys.2021.680544.
120. Abdin, A. A., and Sarhan, N. I. Intervention of mitochondrial dysfunction-oxidative stress-dependent apoptosis as a possible neuroprotective mechanism of α-lipoic acid against rotenone-induced parkinsonism and l-dopa toxicity, Neurosci. Res. 2011, 71, 387–39, doi.org/10.1016/j.neures.2011.08.008.
121. Pei, X., Hu, F., Hu, Z., Luo, F., Li, X., and Long, D. Neuroprotective effect of α-lipoic acid against Aβ25–35-induced damage in BV2 cells, Molecules 2023, 28, 1–13, doi.org/10.3390/molecules28031168.
122. Sahin, M., Sagdıc, G., Elmas, O., Akpınar, D., Derin, N., and Yargıcoğlu, P. Effect of chronic restraint stress and alpha-lipoic acid on lipid peroxidation and antioxidant enzyme activities in rat peripheral organs, Pharmacol. Res. 2006, 54, 247–252, doi.org/10.1016/j.phrs.2006.05.007.
123. Liu, J., Wang, X., and Mori, A. Immobilization stress-induced antioxidant defense changes in rat plasma: effect of treatment with reduced glutathione, Int. J. Biochem. 1994, 26, 511-517, doi.org/10.1016/0020-711X(94)90008-6.
124. Akotkar, L., Aswar, U., Patil, R., Kumar, D., Aswar, M., and Gurav, S. Antidepressant effect of alpha lipoic acid in rats exposed to chronic unpredictable mild stress: putative role of neurotransmitters and 5HT3 receptor, Future Pharmacol. 2023, 3, 407–425, doi.org/10.3390/futurepharmacol3020025.
125. Sampaio, L. R. L., Cysne Filho, F. M. S., and Vasconcelos, S. M. M. Advantages of the alpha-lipoic acid association with chlorpromazine in a model of schizophrenia induced by ketamine in rats: behavioral and oxidative stress evidences, Neuroscience 2018, 373, 72–81, doi.org/10.1016/j.neuroscience.2018.01.008.
126. Mansur, R. B., Cha, D. S., Asevedo, E., and Brietzke, E. Selfish brain and neuroprogression in bipolar disorder, Prog. Neuropsychopharmacol. Biol. Psychiatry 2013, 43, 66-71, doi.org/10.1016/j.pnpbp.2012.12.004.
127. Kim, M. S., Park, J. Y., Namkoong, C., Jang, P. G., Ryu, J. W., and Lee, K. U. Anti-obesity effects of α-lipoic acid mediated by suppression of hypothalamic AMP-activated protein kinase, Nat. Med. 2004, 10, 727-733, doi.org/10.1038/nm1061.
128. Kim, E., Park, D. W., Choi, S. H., and Cho, H. S. A preliminary investigation of α-lipoic acid treatment of antipsychotic drug-induced weight gain in patients with schizophrenia, J. Clin. Psychopharmacol. 2008, 28, 138-146, doi.org/10.1097/JCP.0b013e31816777f7.
129. Sampaio, L. R. L., Borges, L. T. N., Barbosa, T. M., Matos, N. C. B., Lima, R. de F., and Vasconcelos, S. M. M. de. Electroencephalographic study of chlorpromazine alone or combined with alpha-lipoic acid in a model of schizophrenia induced by ketamine in rats, J. Psychiatr. Res. 2017, 86, 73–82, doi.org/10.1016/j.jpsychires.2016.12.003.
130. Farr, S. A., Poon, H. F., Drake, J., Banks, W. A., and Morley, J. E. The antioxidants α‐lipoic acid and N ‐acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice, J. Neurochemi. 2003, 84, 1173–1183, doi.org/10.1046/j.1471-4159.2003.01580.x.
131. Memudu, A. E., and Adanike, R. P. Alpha lipoic acid reverses scopolamine-induced spatial memory loss and pyramidal cell neurodegeneration in the prefrontal cortex of wistar rats, IBRO Neurosci. Rep. 2022, 13, 1–8, doi.org/10.1016/j.ibneur.2022.05.005.
132. Liu, B., Ma, X., Guo, D., Guo, Y., and Bi, H. Neuroprotective effect of alpha-lipoic acid on hydrostatic pressure-induced damage of retinal ganglion cells in vitro, Neurosci. Lett. 2012, 526, 24–28, doi.org/10.1016/j.neulet.2012.08.016.
133. Han, D., Sen, C. K., Roy, S., Kobayashi, M. S., and Packer, L. Protection against glutamate-induced cytotoxicity in C6 glial cells by thiol antioxidants, Am. J. Physiol. Regul. Integr. Comp. Physiol. 1997, 273, 1771–1778, doi.org/10.1152/ajpregu.1997.273.5.R1771.
134. Sun, H., Yao, W., Tang, Y., Zhuang, W., Wu, D., and Sheng, H. Urinary exosomes as a novel biomarker for evaluation of α-lipoic acid’s protective effect in early diabetic nephropathy, J. Clin. Lab. Anal. 2017, 31, e22129, doi.org/10.1002/jcla.22129.
135. Lin, H., Ye, S., Xu, J., and Wang, W. The alpha-lipoic acid decreases urinary podocalyxin excretion in type 2 diabetics by inhibiting oxidative stress in vivo, J. Diabetes Complications 2015, 29, 64–67, doi.org/10.1016/j.jdiacomp.2014.09.011.
136. Wang, L., Tang, Y., Eisner, W., Sparks, M. A., and Spurney, R. F. Augmenting podocyte injury promotes advanced diabetic kidney disease in Akita mice, Biochem. Bioph Res. Commun. 2014, 444, 622-627, doi.org/10.1016/j.bbrc.2014.01.115.
137. Gui, D., Guo, Y., Wang, F., Liu, W., Chen, J., and Wang, N. Astragaloside IV, a novel antioxidant, prevents glucose-induced podocyte apoptosis in vitro and in vivo, PLoS One 2012, 7, e39824, doi.org/10.1371/journal.pone.0039824.
138. Jacob, S., Streeper, R. S., Fogt, D. L., Hokama, J. Y., Tritschler, H. J., and Henriksen, E. J. The antioxidant α-lipoic acid enhances insulin-stimulated glucose metabolism in insulin-resistant rat skeletal muscle, Diabetes 1996, 45, 1024-1029, doi.org/10.2337/diabetes.45.8.1024.
139. Khamaisi, M., Potashnik, R., Tirosh, A., Demshchak, E., Rudich, A., and Bashan, N. Lipoic acid reduces glycemia and increases muscle GLUT4 content in streptozotocin-diabetic rats, Metabolism 1997, 46, 763-768, doi.org/10.1016/S0026-0495(97)90120-7.
140. Nagamatsu, M., Nickander, K. K., Schmelzer, J. D., Raya, A., Wittrock, D. A., and Low, P. A. Lipoic acid improves nerve blood flow, reduces oxidative stress, and improves distal nerve conduction in experimental diabetic neuropathy, Diabetes Care 1995, 18, 1160-1167, doi.org/10.2337/diacare.18.8.1160.
141. Papanas, N., and Ziegler, D. Efficacy of α-lipoic acid in diabetic neuropathy, Expert Opin. Pharmacother. 2014, 15, 2721–2731, doi.org/10.1517/14656566.2014.972935.
142. Jain, S. K., and Lim, G. Lipoic acid decreases lipid peroxidation and protein glycosylation and increases (Na+ + K+) and Ca++ATPase activities in high glucose-treated human erythrocytes, Free Radic. Biol. Med. 2000, 29, 1122-1128, doi.org/10.1016/S0891-5849(00)00410-X.
143. Ziegler, D., Hanefeld, M., Ruhnau, K. J., Hasche, H., Lobisch, M., and Malessa, R. Treatment of symptomatic diabetic polyneuropathy with the antioxidant α-lipoic acid: a 7-month multicenter randomized controlled trial (ALADIN III study), Diabetes Care 1999, 22, 1296-1301, doi.org/10.2337/diacare.22.8.1296.
144. Mervaala, E., Finckenberg, P., Lapatto, R., Müller, D. N., Park, J. K., and Luft, F. C. Lipoic acid supplementation prevents angiotensin II-induced renal injury, Kidney Int. 2003, 64, 501-508, doi.org/10.1046/j.1523-1755.2003.00108.x.
145. Melhem, M. F., Craven, P. A., and DeRubertis, F. R. Effects of dietary supplementation of α-lipoic acid on early glomerular injury in diabetes mellitus, J. Am. Soc. Nephrol. 2001, 12, 124-133, doi.org/10.1681/asn.v121124.
146. Dinçer, Y., Telci, A., Kayali, R., Yilmaz, I. A., and Akçay, T. Effect of α-lipoic acid on lipid peroxidation and anti-oxidant enzyme activities in diabetic rats, Clin. Exp. Pharmacol. Physiol. 2002, 29, 281-284, doi.org/10.1046/j.1440-1681.2002.03642.x.
147.Maritim, A. C., Sanders, R. A., and Watkins, J. B. Effects of α-lipoic acid on biomarkers of oxidative stress in streptozotocin-induced diabetic rats, J. Nutr. Biochem. 2003, 14, 288-294, doi.org/10.1016/S0955-2863(03)00036-6.
148. Bhatti, F., Mankhey, R. W., Asico, L., Quinn, M. T., and Maric, C. Mechanisms of antioxidant and pro-oxidant effects of α-lipoic acid in the diabetic and nondiabetic kidney, Kidney Int. 2005, 67, 1371-1380, doi.org/10.1111/j.1523-1755.2005.00214.x.
149. Manaviat M., Rashidi M., and Shoja RM. Prevalence of dry eye syndrome and diabetic retinopathy in type 2 diabetic patients, BMC Ophthalmol. 2008, 8, 1-5, doi.org/10.1186/1471-2415-8-10.
150. Alvarez-Rivera F., Fernández-Villanueva D., and Concheiro A. α-Lipoic acid in Soluplus(®) polymeric nanomicelles for ocular treatment of diabetes-associated corneal diseases, J. Pharm. Sci. 2016, 105, 2855–2863, doi.org/10.1016/j.xphs.2016.03.006.
151. Pawankar, R. Allergic diseases and asthma: a global public health concern and a call to action, World Allergy Organization 2014, 7, 1-3, doi.org/10.1186/1939-4551-7-12.
152. Druce, H. M. Allergic Rhinitis, JAMA 1988, 259, 260, doi.org/10.1001/jama.1988.03720020062038.
153. Choi, Y. H., Chai, O. H., Han, E.H., Choi, S.Y., and Song, C. H. Lipoic acid suppresses compound 48/80-induced anaphylaxis-like reaction, Anat. Cell Biol. 2010, 43, 317, doi.org/10.5115/acb.2010.43.4.317.
154. Small, P., and Kim, H. Allergic rhinitis, Allergy, Asthma Clin. Immunol. 2011, 7, 1-8, doi.org/10.1186/1710-1492-7-S1-S3.
155. Van Nguyen, T., Piao, C. H., Fan, Y. J., Shin, D. U., Kim, S. Y., and Chai, O. H. Anti-allergic rhinitis activity of α-lipoic acid via balancing Th17/Treg expression and enhancing Nrf2/HO-1 pathway signaling, Sci. Rep. 2020, 10, 12528, doi.org/10.1038/s41598-020-69234-1.
156. Zhao, H., Zhao, X., Liu, L., Zhang, H., Xuan, M., and Liu, C. Neurochemical effects of the R form of α-lipoic acid and its neuroprotective mechanism in cellular models of Parkinson’s disease, Int. J. Biochem. Cell Biol. 2017, 87, 86–94, doi.org/10.1016/j.biocel.2017.04.002.
157. Sen, C. K., Roy, S., and Packer, L. Fas mediated apoptosis of human jurkat T-cells: intracellular events and potentiation by redox-active α-lipoic acid, Cell Death Differ. 1999, 6, 481-491, doi.org/10.1038/sj.cdd.4400514.
158. Wenzel, U., Nickel, A., and Daniel, H. α-lipoic acid induces apoptosis in human colon cancer cells by increasing mitochondrial respiration with a concomitant O2−generation, Apoptosis 2005, 10, 359–368, doi.org/10.1007/s10495-005-0810-x.
159. Schwartz, L., Abolhassani, M., Guais, A., Sanders, E., and Campion, F. A combination of alpha lipoic acid and calcium hydroxycitrate is efficient against mouse cancer models: preliminary results, Oncol. Rep. 2010, 23, 1407-1416, doi.org/10.3892/or_00000778.
160. Ahmadi, A., Hosseini, F., and Soukhtanloo, M. Anticancer effects of alpha-lipoic acid, a potent organosulfur compound by modulating matrix metalloproteinases and apoptotic markers in osteosarcoma MG-63 cells, J. Steroid Biochem. Mol. Biol. 2025, 247, 106664, doi.org/10.1016/j.jsbmb.2024.106664.
161. Jeon, M. J., Kim, W. G., Lim, S., Choi, H.J., Sim, S., and Kim, W. B. Alpha lipoic acid inhibits proliferation and epithelial mesenchymal transition of thyroid cancer cells, Mol. Cell. Endocrinol. 2016, 419, 113–123, doi.org/10.1016/j.mce.2015.10.005.
162.Na, M. H., Seo, E. Y., and Kim, W. K.Effects of α-lipoic acid on cell proliferation and apoptosis in MDA-MB-231 human breast cells, Nutr. Res. Pract. 2009, 3, 265, doi.org/10.4162/nrp.2009.3.4.265.
163. Kuban-Jankowska, A., Gorska-Ponikowska, M., and Wozniak, M. Lipoic acid decreases the viability of breast cancer cells and activity of PTP1B and SHP2, Anticancer Res. 2017, 37, 2893-2898, doi.org/10.21873/anticanres.11642.
164. Zhang, W. J., Wei, H., and Frei, B. α-Lipoic acid attenuates LPS-induced inflammatory responses by activating the phosphoinositide 3-kinase/Akt signaling pathway, Proc. Natl. Acad. Sci. 2007, 104, 4077–4082, doi.org/10.1073/pnas.0700305104.
165. Faverani, L. P., Polo, T. O. B., Ramalho-Ferreira, G., Momesso, G. A. C., Hassumi, J. S., Rossi, A. C., ... & Okamoto, R. Raloxifene but not alendronate can compensate the impaired osseointegration in osteoporotic rats. Clinical oral investigations 2018, 22, 255-265.
166. Koh, J. M., Lee, Y. S., Byun, C. H., Chang, E. J., Kim, H., and Kim, G. S. α-Lipoic acid suppresses osteoclastogenesis despite increasing the receptor activator of nuclear factor κB ligand/osteoprotegerin ratio in human bone marrow stromal cells, J. Endocrinol. 2005, 185, 401-413, doi.org/10.1677/joe.1.05995.
167. Lin, Z., Guichun, Z., Lifeng, L., Chen, C., and Jinfang, C. Protective effect of α-lipoic acid against antimycin a cytotoxicity in MC3T3-E1 osteoblastic cells, Cell Stress Chaperones 2017, 22, 5–13, doi.org/10.1007/s12192-016-0735-z.
168. Cheng, M., Wang, Q., Fan, Y., Liu, X., Wang, L., and Sun, W. A traditional Chinese herbal preparation, er-zhi-wan, prevent ovariectomy-induced osteoporosis in rats, J. Ethnopharmacol. 2011, 138, 279–285, doi.org/10.1016/j.jep.2011.09.030.
169. Vivanco, I., and Sawyers, C. L. The phosphatidylinositol 3-kinase–AKT pathway in human cancer, Nat. Rev. Cancer 2002, 2, 489–501, doi.org/10.1038/nrc839.
170. Fu, C., Xu, D., Wang, C. Y., Jin, Y., Liu, Q., and Liu, M. Z. Alpha-lipoic acid promotes osteoblastic formation in H2O2-treated MC3T3-E1 cells and prevents bone loss in ovariectomized rats, J. Cell. Physiol. 2015, 230, 2184-2201, doi.org/10.1002/jcp.24947.
171. Xiao, Y., Cui, J., and Le, G. Lipoic acid increases the expression of genes involved in bone formation in mice fed a high-fat diet, Nutrition Research 2011, 31, 309–317, doi.org/10.1016/j.nutres.2011.03.013.
172. Abdelhalim, M. A. K., Qaid, H. A. Y., and Ghannam, M. M. The protective roles of vitamin E and α-lipoic acid against nephrotoxicity, lipid peroxidation, and inflammatory damage induced by gold nanoparticles, Int. J. Nanomedicine 2020, 15, 729–734, doi.org/10.2147/IJN.S192740.
173. Ossipov, D. A. Bisphosphonate-modified biomaterials for drug delivery and bone tissue engineering, Expert Opin. Drug Deliv. 2015, 12, 1443–1458, doi.org/10.1517/17425247.2015.1021679.
174. Gao, W., Li, J. J., Shi, J., Lan, H., and Fu, D. Ångstrom-scale gold particles loaded with alendronate via alpha-lipoic acid alleviate bone loss in osteoporotic mice, J. Nanobiotechnology 2024, 22, 212, doi.org/10.1186/s12951-024-02466-9.
175. Lu, S.Y., Wang, C.Y., Jin, Y., Meng, Q., Liu, Q., and Liu, M.Z. The osteogenesis-promoting effects of alpha-lipoic acid against glucocorticoid-induced osteoporosis through the NOX4, NF-kappaB, JNK and PI3K/AKT pathways, Sci. Rep. 2017, 7, 3331, doi.org/10.1038/s41598-017-03187-w.
176. Wollin, S. D., Wang, Y., and Jones, P. J. H. Effects of a medium chain triglyceride oil mixture and α-lipoic acid diet on body composition, antioxidant status, and plasma lipid levels in the golden syrian hamster, J. Nutr. Biochem. 2004, 15, 402–410, doi.org/10.1016/j.jnutbio.2003.12.001.
177. Bai, J., Chen, C., Sun, Y., Li, S., He, R., and Han, Z. α-LA attenuates microcystin-LR-induced hepatocellular oxidative stress in mice through Nrf2-mediated antioxidant and detoxifying enzymes, Toxicon 2023, 235, 107313, doi.org/10.1016/j.toxicon.2023.107313.
178. Zhang, L., Zou, J., Chai, E., and Zhang, Y. Alpha-lipoic acid attenuates cardiac hypertrophy via downregulation of PARP-2 and subsequent activation of SIRT-1, Eur. J. Pharmacol. 2014, 744, 203–210, doi.org/10.1016/j.ejphar.2014.09.037.
179. Quinn, M. T., Parthasarathy, S., and Steinberg, D. Oxidatively modified low density lipoproteins: a potential role in recruitment and retention of monocyte/macrophages during atherogenesis, Proc. Natl. Acad. Sci. U. S. A. 1987, 84, 2995-2998, doi.org/10.1073/pnas.84.9.2995.
180. Meneses, L. N., Vasconcelos, G. S., Da Silva Medeiros, I., Silva, M. C. C., and Vasconcelos, S. M. M. Neuroprotective evidence of alpha-lipoic acid and desvenlafaxine on memory deficit in a neuroendocrine model of depression, Naunyn Schmiedebergs Arch. Pharmacol. 2018, 391, 803-817, doi.org/10.1007/s00210-018-1509-1.
181. Berköz, M., Aslan, A., Yunusoğlu, O., and Francik, R. Hepatoprotective potentials of Usnea Longissima Ach. and Xanthoparmelia Somloensis (gyelnik) hale extracts in ethanol-induced liver injury, Drug Chem. Toxicol. 2025, 48, 136–149, doi.org/10.1080/01480545.2024.2407867.
182. Ceylanlı, D., Şehirli, A. Ö., Gençosman, S., Teralı, K., Şah, H., and Sayıner, S. Protective effects of alpha-lipoic acid against 5-fluorouracil-induced gastrointestinal mucositis in rats, Antioxidants 2022, 11, 1930, doi.org/10.3390/antiox11101930.
183. Babu, S., Manoharan, S., and Perumal, E. Role of oxidative stress-mediated cell death and signaling pathways in experimental fluorosis, Chem. Biol. Interact. 2022, 365, 110106, doi.org/10.1016/j.cbi.2022.110106.
184. Elshazly, S. M., El-Moselhy, M. A., and Barakat, W. Insights in the mechanism underlying the protective effect of α-lipoic acid against acetaminophen-hepatotoxicity, Eur. J. Pharmacol. 2014, 726, 116–123, doi.org/10.1016/j.ejphar.2014.01.042.
185. Keith, D. J., Butler, J. A., Bemer, B., Dixon, B., Johnson, S., and Hagen, T. M. Age and gender dependent bioavailability of R- and S-α-lipoic acid: a pilot study, Pharmacol. Res. 2012, 66, 199-206, doi.org/10.1016/j.phrs.2012.05.002.
186. Guzel, E. E., Kaya, N., Ozan, G., Tektemur, A., and Ozan, I. E. The investigation of effect of alpha lipoic acid against damage on neonatal rat lung to maternal tobacco smoke exposure, Toxicol. Rep. 2018, 5, 714–722, doi.org/10.1016/j.toxrep.2018.05.014.
187. Yunusoglu, O., Türkmen, Ö., Berkoz, M., and Yalın, S. In vitro anti-obesity effect of aloe vera extract through transcription factors and lipolysis-associated genes, Eastern J. Med. 2022, 27, 519–528, doi.org/10.5505/ejm.2022.13285.
188. Romo-Hualde, A., Huerta, A. E., González-Navarro, C. J., Ramos-López, O., and Martínez, J. A. Untargeted metabolomic on urine samples after α-lipoic acid and/or eicosapentaenoic acid supplementation in healthy overweight/obese women, Lipids Health Dis. 2018, 17, 1-13, doi.org/10.1186/s12944-018-0750-4.
189. Namazi, N., Larijani, B., and Azadbakht, L. Alpha-lipoic acid supplement in obesity treatment: a systematic review and meta-analysis of clinical trials, Clinical Nutrition 2018, 37, 419–428, doi.org/10.1016/j.clnu.2017.06.002.
190. Dajnowicz-Brzezik, P., Żebrowska, E., Maciejczyk, M., and Chabowski, A. The effect of α-lipoic acid on oxidative stress in adipose tissue of rats with obesity-induced insulin resistance, Cell. Physiol. Biochem. 2022, 56, 239–253, doi.org/10.33594/000000528.
191. McNeilly, A. M., Davison, G. W., Murphy, M. H., Nadeem, N., Trinick, T., and McEneny, J. Effect of α-lipoic acid and exercise training on cardiovascular disease risk in obesity with impaired glucose tolerance, Lipids Health Dis. 2011, 10, 1-9, doi.org/10.1186/1476-511X-10-217.
192. Arivazhagan, P., Ramanathan, K., and Panneerselvam, C. Effect of DL-α-lipoic acid on the status of lipid peroxidation and antioxidants in mitochondria of aged rats, J. Nutr. Biochem. 2001, 12, 2–6, doi.org/10.1016/S0955-2863(00)00138-8.
193. Yawalkar, N. Drug hypersensitivity, Acta Clin. Belg. 2009, 64, 529–533, doi.org/10.1179/acb.2009.090.
194. Gomaa, A. M. S., Abd El-Mottaleb, N. A., and Aamer, H. A. Antioxidant and anti-inflammatory activities of alpha lipoic acid protect against indomethacin-induced gastric ulcer in rats, Biomed. Pharmacother. 2018, 101, 188–194, doi.org/10.1016/j.biopha.2018.02.070.
195. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications, Nature 2001, 414, 813-820, doi.org/10.1038/414813a.
196. Azevedo, Í. M., Lima, M. L., and Moreira, M. D. Effects of simvastatin on 5-fluorouracil-induced gastrointestinal mucositis in rats, Rev. Col. Bras. Cir. 2018, 45, e1968, doi.org/10.1590/0100-6991e-20181968.
197. Kleniewska, P., and Pawliczak, R. The influence of apocynin, lipoic acid and probiotics on antioxidant enzyme levels in the pulmonary tissues of obese asthmatic mice, Life Sci. 2019, 234, 116780, doi.org/10.1016/j.lfs.2019.116780.
198. Mims, J. W. Asthma: definitions and pathophysiology, Int. Forum Allergy Rhinol. 2015, 5, 2-6, doi.org/10.1002/alr.21609.
199. Sook Cho, Y., Lee, J., Lee, T.H., Young Lee, E., Lee, K.U., and Moon, H.B. α-Lipoic acid inhibits airway inflammation and hyperresponsiveness in a mouse model of asthma, J. Allergy Clin. Immunol. 2004, 114, 429–435, doi.org/10.1016/j.jaci.2004.04.004.
200. Dhar, D., Baglieri, J., Kisseleva, T., and Brenner, D. A. Mechanisms of liver fibrosis and its role in liver cancer, Exp. Biol. Med. 2020, 245, 96-108, doi.org/10.1177/1535370219898141.
201. El-Senousey, H. K., Chen, B., Wang, J. Y., Atta, A. M., and Nie, Q. H. Effects of dietary vitamin C, vitamin E, and alpha-lipoic acid supplementation on the antioxidant defense system and immune-related gene expression in broilers exposed to oxidative stress by dexamethasone, Poultry Sci. 2018, 97, 30–38, doi.org/10.3382/ps/pex298.
202. Morini, M., Roccatagliata, L., Dell’Eva, R., Pedemonte, E., Furlan, R., and Uccelli, A. α-Lipoic acid is effective in prevention and treatment of experimental autoimmune encephalomyelitis, J. Neuroimmunol. 2004, 148, 146–153, doi.org/10.1016/j.jneuroim.2003.11.021.
203. Li, Y., Ma, Q.G., Zhao, L.H., Wei, H., Duan, G.X., and Ji, C. Effects of lipoic acid on immune function, the antioxidant defense system, and inflammation-related genes expression of broiler chickens fed aflatoxin contaminated diets, Int. J. Mol. Sci. 2014, 15, 5649–5662, doi.org/10.3390/ijms15045649.
204. Prathima, P., Venkaiah, K., Reddy, M. H., and Sainath, S. B. Antioxidant effects of α-lipoic acid against epididymal oxidative damage in adult offspring rats exposed to maternal hypothyroidism stress, Reprod. Toxicol. 2024, 125, 108555, doi.org/10.1016/j.reprotox.2024.108555.
205. Sastre, J., Pallardó, F. V., Plá, R., Pellín, A., Juan, G., and Viña, J. Aging of the liver: age-associated mitochondrial damage in intact hepatocytes, Hepatology 1996, 24, 1199-1205, doi.org/10.1002/hep.510240536.
206. Jungwirth, A., Giwercman, A., Tournaye, H., Diemer, T., Kopa, Z., and Krausz, C. European association of urology guidelines on male infertility: the 2012 update, Eur. Urol. 2012, 62, 324–332, doi.org/10.1016/j.eururo.2012.04.048.
207. Gharagozloo, P., and Aitken, R. J. The role of sperm oxidative stress in male infertility and the significance of oral antioxidant therapy, Hum. Reprod. 2011, 26, 1628–1640, doi.org/10.1093/humrep/der132.
208. Ibrahim, S. F., Osman, K., Das, S., Othman, A. M., and Rahman, M. P. A. A Study of the antioxidant effect of alpha lipoic acids on sperm quality, Clinics 2008, 63, 545–550, doi.org/10.1590/S1807-59322008000400022.
209. Di Tucci, C., Galati, G., Mattei, G., Bonanni, V., Capri, O., and Benedetti Panici, P. The role of alpha lipoic acid in female and male infertility: a systematic review, Gynecol. Endocrinol. 2021, 37, 497–505, doi.org/10.1080/09513590.2020.1843619.
210. Asci, H., Erol, O., Ellidag, H. Y., Tola, E. N., and Ozmen, O. Pathology of cigarettes on the reproductive system and ameliorative effects of alpha lipoic acid: a rat model study, Toxicol. Ind. Health 2018, 34, 385–395, doi.org/10.1177/0748233718755160.
211. Genazzani, A. D., Prati, A., Marchini, F., Petrillo, T., and Simoncini, T. Differential insulin response to oral glucose tolerance test (OGTT) in overweight/obese polycystic ovary syndrome patients undergoing to myo-inositol (MYO), alpha lipoic acid (ALA), or combination of both, Gynecol. Endocrinol. 2019, 35, 1088–1093, doi.org/10.1080/09513590.2019.1640200.
212. Fruzzetti, F., Benelli, E., Fidecicchi, T., and Tonacchera, M. Clinical and metabolic effects of alpha-lipoic acid associated with two different doses of myo-inositol in women with polycystic ovary syndrome, Int. J. Endocrinol. 2020, 1–8, doi.org/10.1155/2020/2901393.
213. Fruzzetti, F., Capozzi, A., and Lello, S. Treatment with d-chiro-inositol and alpha lipoic acid in the management of polycystic ovary syndrome, Gynecol. Endocrinol. 2019, 35, 506–510, doi.org/10.1080/09513590.2018.1540573.
214. Emir, D. F., Ozturan, I. U., and Yilmaz, S. Alpha lipoic acid intoxicatıon: an adult, Am. J. Emerg. Med. 2018, 36, 1125-e3, doi.org/10.1016/j.ajem.2018.03.022.
215. Vidović, B., Milovanović, S., Stefanović, A., Takić, M., and Dordević, B. Effects of alpha-lipoic acid supplementation on plasma adiponectin levels and some metabolic risk factors in patients with schizophrenia, J. Med. Food 2017, 20, 79-85, doi.org/10.1089/jmf.2016.0070.
216. Schuff-Werner, P., Pönisch, W., and Kaiser, T. Transient Howell-Jolly-body-like cytoplasmic inclusions in neutrophils after severe intoxication with alpha-lipoic acid, Scand. J. Clin. Lab. Invest. 2021, 81, 8–11, doi.org/10.1080/00365513.2020.1855468.
217. Gatti, M., Ippoliti, I., Poluzzi, E., Antonazzo, I. C., Moro, P. A., and Raschi, Assessment of adverse reactions to α-lipoic acid containing dietary supplements through spontaneous reporting systems, Clin. Nutr. 2021, 40, 1176–1185, doi.org/10.1016/j.clnu.2020.07.028.