Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2024

Open Access 01-12-2024 | Post-COVID Syndrome | Review

Melatonin: a ferroptosis inhibitor with potential therapeutic efficacy for the post-COVID-19 trajectory of accelerated brain aging and neurodegeneration

Authors: Asmaa Yehia, Osama A. Abulseoud

Published in: Molecular Neurodegeneration | Issue 1/2024

Login to get access

Abstract

The unprecedented pandemic of COVID-19 swept millions of lives in a short period, yet its menace continues among its survivors in the form of post-COVID syndrome. An exponentially growing number of COVID-19 survivors suffer from cognitive impairment, with compelling evidence of a trajectory of accelerated aging and neurodegeneration. The novel and enigmatic nature of this yet-to-unfold pathology demands extensive research seeking answers for both the molecular underpinnings and potential therapeutic targets. Ferroptosis, an iron-dependent cell death, is a strongly proposed underlying mechanism in post-COVID-19 aging and neurodegeneration discourse. COVID-19 incites neuroinflammation, iron dysregulation, reactive oxygen species (ROS) accumulation, antioxidant system repression, renin-angiotensin system (RAS) disruption, and clock gene alteration. These events pave the way for ferroptosis, which shows its signature in COVID-19, premature aging, and neurodegenerative disorders. In the search for a treatment, melatonin shines as a promising ferroptosis inhibitor with its repeatedly reported safety and tolerability. According to various studies, melatonin has proven efficacy in attenuating the severity of certain COVID-19 manifestations, validating its reputation as an anti-viral compound. Melatonin has well-documented anti-aging properties and combating neurodegenerative-related pathologies. Melatonin can block the leading events of ferroptosis since it is an efficient anti-inflammatory, iron chelator, antioxidant, angiotensin II antagonist, and clock gene regulator. Therefore, we propose ferroptosis as the culprit behind the post-COVID-19 trajectory of aging and neurodegeneration and melatonin, a well-fitting ferroptosis inhibitor, as a potential treatment.
Literature
1.
go back to reference Marzouk M, et al. Modeling COVID-19 Effects on Sustainable Development Goals in Egypt Using System Dynamics. 2022.CrossRef Marzouk M, et al. Modeling COVID-19 Effects on Sustainable Development Goals in Egypt Using System Dynamics. 2022.CrossRef
2.
go back to reference Jennings G, et al. A systematic review of persistent symptoms and residual abnormal functioning following acute COVID-19: ongoing symptomatic phase vs. post-COVID-19 syndrome. J Clin Med. 2021;10(24):5913.PubMedCrossRefPubMedCentral Jennings G, et al. A systematic review of persistent symptoms and residual abnormal functioning following acute COVID-19: ongoing symptomatic phase vs. post-COVID-19 syndrome. J Clin Med. 2021;10(24):5913.PubMedCrossRefPubMedCentral
3.
go back to reference Carod-Artal FJ. Post-COVID-19 syndrome: epidemiology, diagnostic criteria and pathogenic mechanisms involved. Rev Neurol. 2021;72(11):384–96.PubMed Carod-Artal FJ. Post-COVID-19 syndrome: epidemiology, diagnostic criteria and pathogenic mechanisms involved. Rev Neurol. 2021;72(11):384–96.PubMed
5.
go back to reference Meo SA, et al. Magnetic Resonance Imaging (MRI) and neurological manifestations in SARS-CoV-2 patients. Eur Rev Med Pharmacol Sci. 2021;25(2):1101–8.PubMed Meo SA, et al. Magnetic Resonance Imaging (MRI) and neurological manifestations in SARS-CoV-2 patients. Eur Rev Med Pharmacol Sci. 2021;25(2):1101–8.PubMed
6.
go back to reference Anjana NKN, et al. Manifestations and risk factors of post COVID syndrome among COVID-19 patients presented with minimal symptoms - A study from Kerala, India. J Family Med Prim Care. 2021;10(11):4023–9.PubMedCrossRefPubMedCentral Anjana NKN, et al. Manifestations and risk factors of post COVID syndrome among COVID-19 patients presented with minimal symptoms - A study from Kerala, India. J Family Med Prim Care. 2021;10(11):4023–9.PubMedCrossRefPubMedCentral
7.
go back to reference Abdel-Gawad M. et al. Post-COVID-19 Syndrome Clinical Manifestations: A Systematic Review. Antiinflamm Antiallergy Agents Med Chem. 2022. Abdel-Gawad M. et al. Post-COVID-19 Syndrome Clinical Manifestations: A Systematic Review. Antiinflamm Antiallergy Agents Med Chem. 2022.
8.
go back to reference Taquet M, et al. 6-month neurological and psychiatric outcomes in 236 379 survivors of COVID-19: a retrospective cohort study using electronic health records. Lancet Psychiatry. 2021;8(5):416–27.PubMedCrossRefPubMedCentral Taquet M, et al. 6-month neurological and psychiatric outcomes in 236 379 survivors of COVID-19: a retrospective cohort study using electronic health records. Lancet Psychiatry. 2021;8(5):416–27.PubMedCrossRefPubMedCentral
9.
go back to reference Taquet M, et al. Bidirectional associations between COVID-19 and psychiatric disorder: retrospective cohort studies of 62 354 COVID-19 cases in the USA. Lancet Psychiatry. 2021;8(2):130–40.PubMedCrossRef Taquet M, et al. Bidirectional associations between COVID-19 and psychiatric disorder: retrospective cohort studies of 62 354 COVID-19 cases in the USA. Lancet Psychiatry. 2021;8(2):130–40.PubMedCrossRef
10.
go back to reference Taquet M, et al. Neurological and psychiatric risk trajectories after SARS-CoV-2 infection: an analysis of 2-year retrospective cohort studies including 1 284 437 patients. Lancet Psychiatry. 2022;9(10):815–27.PubMedCrossRefPubMedCentral Taquet M, et al. Neurological and psychiatric risk trajectories after SARS-CoV-2 infection: an analysis of 2-year retrospective cohort studies including 1 284 437 patients. Lancet Psychiatry. 2022;9(10):815–27.PubMedCrossRefPubMedCentral
11.
12.
go back to reference Crivelli L, et al. Changes in cognitive functioning after COVID-19: a systematic review and meta-analysis. Alzheimers Dement. 2022;18(5):1047–66.PubMedCrossRef Crivelli L, et al. Changes in cognitive functioning after COVID-19: a systematic review and meta-analysis. Alzheimers Dement. 2022;18(5):1047–66.PubMedCrossRef
13.
go back to reference Premraj L, et al. Mid and long-term neurological and neuropsychiatric manifestations of post-COVID-19 syndrome: a meta-analysis. J Neurol Sci. 2022;434:120162.PubMedCrossRefPubMedCentral Premraj L, et al. Mid and long-term neurological and neuropsychiatric manifestations of post-COVID-19 syndrome: a meta-analysis. J Neurol Sci. 2022;434:120162.PubMedCrossRefPubMedCentral
15.
go back to reference Dixon SJ, Stockwell BR. The hallmarks of ferroptosis. Ann Rev Cancer Biol. 2019;3:35–54.CrossRef Dixon SJ, Stockwell BR. The hallmarks of ferroptosis. Ann Rev Cancer Biol. 2019;3:35–54.CrossRef
18.
19.
go back to reference Sies H, et al. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat Rev Mol Cell Biol. 2022;23(7):499–515.PubMedCrossRef Sies H, et al. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat Rev Mol Cell Biol. 2022;23(7):499–515.PubMedCrossRef
21.
go back to reference Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995;82:969–74.PubMedCrossRef Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995;82:969–74.PubMedCrossRef
23.
go back to reference Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 2020;21(7):363–83.PubMedCrossRef Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 2020;21(7):363–83.PubMedCrossRef
24.
go back to reference Juan CA, et al. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int J Mol Sci. 2021;22(9):4642.PubMedCrossRefPubMedCentral Juan CA, et al. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int J Mol Sci. 2021;22(9):4642.PubMedCrossRefPubMedCentral
25.
go back to reference Wang W, et al. Chemistry and biology of ω-3 PUFA peroxidation-derived compounds. Prostaglandins Other Lipid Mediat. 2017;132:84–91.PubMedCrossRef Wang W, et al. Chemistry and biology of ω-3 PUFA peroxidation-derived compounds. Prostaglandins Other Lipid Mediat. 2017;132:84–91.PubMedCrossRef
26.
go back to reference Catalá A. Five decades with polyunsaturated fatty acids: chemical synthesis, enzymatic formation, lipid peroxidation and its biological effects. J Lipids. 2013;2013:710290.PubMedCrossRefPubMedCentral Catalá A. Five decades with polyunsaturated fatty acids: chemical synthesis, enzymatic formation, lipid peroxidation and its biological effects. J Lipids. 2013;2013:710290.PubMedCrossRefPubMedCentral
27.
go back to reference Wagner BA, Buettner GR, Burns CP. Free radical-mediated lipid peroxidation in cells: oxidizability is a function of cell lipid bis-allylic hydrogen content. Biochemistry. 1994;33(15):4449–53.PubMedCrossRef Wagner BA, Buettner GR, Burns CP. Free radical-mediated lipid peroxidation in cells: oxidizability is a function of cell lipid bis-allylic hydrogen content. Biochemistry. 1994;33(15):4449–53.PubMedCrossRef
28.
29.
go back to reference Yin H, Xu L, Porter NA. Free radical lipid peroxidation: mechanisms and analysis. Chem Rev. 2011;111(10):5944–72.PubMedCrossRef Yin H, Xu L, Porter NA. Free radical lipid peroxidation: mechanisms and analysis. Chem Rev. 2011;111(10):5944–72.PubMedCrossRef
30.
32.
go back to reference Halliwell B, Chirico S. Lipid peroxidation: its mechanism, measurement, and significance. Am J Clin Nutr. 1993;57(5):715S–725S.PubMedCrossRef Halliwell B, Chirico S. Lipid peroxidation: its mechanism, measurement, and significance. Am J Clin Nutr. 1993;57(5):715S–725S.PubMedCrossRef
33.
go back to reference Niki E, et al. Lipid peroxidation: mechanisms, inhibition, and biological effects. Biochem Biophys Res Commun. 2005;338(1):668–76.PubMedCrossRef Niki E, et al. Lipid peroxidation: mechanisms, inhibition, and biological effects. Biochem Biophys Res Commun. 2005;338(1):668–76.PubMedCrossRef
34.
go back to reference Kühn H, Borchert A. Regulation of enzymatic lipid peroxidation: the interplay of peroxidizing and peroxide reducing enzymes1 1This article is part of a series of reviews on “Regulatory and Cytoprotective Aspects of Lipid Hydroperoxide Metabolism.” The full list of papers may be found on the homepage of the journal. Free Rad Biol Med. 2002;33(2):154–72.PubMedCrossRef Kühn H, Borchert A. Regulation of enzymatic lipid peroxidation: the interplay of peroxidizing and peroxide reducing enzymes1 1This article is part of a series of reviews on “Regulatory and Cytoprotective Aspects of Lipid Hydroperoxide Metabolism.” The full list of papers may be found on the homepage of the journal. Free Rad Biol Med. 2002;33(2):154–72.PubMedCrossRef
37.
go back to reference Minotti G, Aust SD. The role of iron in oxygen radical mediated lipid peroxidation. Chem Biol Interact. 1989;71(1):1–19.PubMedCrossRef Minotti G, Aust SD. The role of iron in oxygen radical mediated lipid peroxidation. Chem Biol Interact. 1989;71(1):1–19.PubMedCrossRef
38.
go back to reference Cheng Z, Li Y. What is responsible for the initiating chemistry of iron-mediated lipid peroxidation: an update. Chem Rev. 2007;107(3):748–66.PubMedCrossRef Cheng Z, Li Y. What is responsible for the initiating chemistry of iron-mediated lipid peroxidation: an update. Chem Rev. 2007;107(3):748–66.PubMedCrossRef
39.
go back to reference Pamplona R. Membrane phospholipids, lipoxidative damage and molecular integrity: a causal role in aging and longevity. Biochim Biophys Acta. 2008;1777(10):1249–62.PubMedCrossRef Pamplona R. Membrane phospholipids, lipoxidative damage and molecular integrity: a causal role in aging and longevity. Biochim Biophys Acta. 2008;1777(10):1249–62.PubMedCrossRef
41.
go back to reference Taso OV, et al. Lipid peroxidation products and their role in neurodegenerative diseases. Ann Res Hosp. 2019;3(2):10.21037. Taso OV, et al. Lipid peroxidation products and their role in neurodegenerative diseases. Ann Res Hosp. 2019;3(2):10.21037.
42.
go back to reference Guéraud F, et al. Chemistry and biochemistry of lipid peroxidation products. Free Radical Res. 2010;44(10):1098–124.CrossRef Guéraud F, et al. Chemistry and biochemistry of lipid peroxidation products. Free Radical Res. 2010;44(10):1098–124.CrossRef
43.
go back to reference Miyamoto S, et al. Singlet molecular oxygen generated by biological hydroperoxides. J Photochem Photobiol, B. 2014;139:24–33.PubMedCrossRef Miyamoto S, et al. Singlet molecular oxygen generated by biological hydroperoxides. J Photochem Photobiol, B. 2014;139:24–33.PubMedCrossRef
45.
go back to reference Angeli JPF, Conrad M. Selenium and GPX4, a vital symbiosis. Free Radical Biol Med. 2018;127:153–9.CrossRef Angeli JPF, Conrad M. Selenium and GPX4, a vital symbiosis. Free Radical Biol Med. 2018;127:153–9.CrossRef
46.
go back to reference Weaver K, Skouta R. The selenoprotein glutathione peroxidase 4: from molecular mechanisms to novel therapeutic opportunities. Biomedicines. 2022;10(4):891.PubMedCrossRefPubMedCentral Weaver K, Skouta R. The selenoprotein glutathione peroxidase 4: from molecular mechanisms to novel therapeutic opportunities. Biomedicines. 2022;10(4):891.PubMedCrossRefPubMedCentral
48.
go back to reference Forman HJ, Zhang H, Rinna A. Glutathione: overview of its protective roles, measurement, and biosynthesis. Mol Aspects Med. 2009;30(1–2):1–12.PubMedCrossRef Forman HJ, Zhang H, Rinna A. Glutathione: overview of its protective roles, measurement, and biosynthesis. Mol Aspects Med. 2009;30(1–2):1–12.PubMedCrossRef
50.
go back to reference Gilbert HF. [2] Thiol/disulfide exchange equilibria and disulfidebond stability. Methods Enzymol. 1995;251:8–28.PubMedCrossRef Gilbert HF. [2] Thiol/disulfide exchange equilibria and disulfidebond stability. Methods Enzymol. 1995;251:8–28.PubMedCrossRef
51.
go back to reference Rossi R, et al. Oxidized forms of glutathione in peripheral blood as biomarkers of oxidative stress. Clin Chem. 2006;52(7):1406–14.PubMedCrossRef Rossi R, et al. Oxidized forms of glutathione in peripheral blood as biomarkers of oxidative stress. Clin Chem. 2006;52(7):1406–14.PubMedCrossRef
52.
go back to reference Li F-J, et al. System Xc−/GSH/GPX4 axis: An important antioxidant system for the ferroptosis in drug-resistant solid tumor therapy. Front Pharmacol. 2022;13:910292.PubMedCrossRefPubMedCentral Li F-J, et al. System Xc−/GSH/GPX4 axis: An important antioxidant system for the ferroptosis in drug-resistant solid tumor therapy. Front Pharmacol. 2022;13:910292.PubMedCrossRefPubMedCentral
53.
54.
go back to reference Lewerenz J, et al. The cystine/glutamate antiporter system xc− in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18(5):522–55.PubMedCrossRefPubMedCentral Lewerenz J, et al. The cystine/glutamate antiporter system xc− in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18(5):522–55.PubMedCrossRefPubMedCentral
55.
go back to reference Harvey C, et al. Nrf2-regulated glutathione recycling independent of biosynthesis is critical for cell survival during oxidative stress. Free Radical Biol Med. 2009;46(4):443–53.CrossRef Harvey C, et al. Nrf2-regulated glutathione recycling independent of biosynthesis is critical for cell survival during oxidative stress. Free Radical Biol Med. 2009;46(4):443–53.CrossRef
56.
go back to reference Shih AY, et al. Coordinate regulation of glutathione biosynthesis and release by Nrf2-expressing glia potently protects neurons from oxidative stress. J Neurosci. 2003;23(8):3394–406.PubMedCrossRefPubMedCentral Shih AY, et al. Coordinate regulation of glutathione biosynthesis and release by Nrf2-expressing glia potently protects neurons from oxidative stress. J Neurosci. 2003;23(8):3394–406.PubMedCrossRefPubMedCentral
58.
59.
60.
go back to reference Abbaspour N, Hurrell R, Kelishadi R. Review on iron and its importance for human health. J Res Med Sci. 2014;19(2):164.PubMedPubMedCentral Abbaspour N, Hurrell R, Kelishadi R. Review on iron and its importance for human health. J Res Med Sci. 2014;19(2):164.PubMedPubMedCentral
63.
go back to reference Katsarou A, Pantopoulos K. Basics and principles of cellular and systemic iron homeostasis. Mol Aspects Med. 2020;75:100866.PubMedCrossRef Katsarou A, Pantopoulos K. Basics and principles of cellular and systemic iron homeostasis. Mol Aspects Med. 2020;75:100866.PubMedCrossRef
66.
go back to reference McKie AT, et al. An iron-regulated ferric reductase associated with the absorption of dietary iron. Science. 2001;291(5509):1755–9.PubMedCrossRef McKie AT, et al. An iron-regulated ferric reductase associated with the absorption of dietary iron. Science. 2001;291(5509):1755–9.PubMedCrossRef
67.
go back to reference Fuqua BK, Vulpe CD, Anderson GJ. Intestinal iron absorption. J Trace Elem Med Biol. 2012;26(2):115–9.PubMedCrossRef Fuqua BK, Vulpe CD, Anderson GJ. Intestinal iron absorption. J Trace Elem Med Biol. 2012;26(2):115–9.PubMedCrossRef
68.
go back to reference Kawabata H. Transferrin and transferrin receptors update. Free Radical Biol Med. 2019;133:46–54.CrossRef Kawabata H. Transferrin and transferrin receptors update. Free Radical Biol Med. 2019;133:46–54.CrossRef
69.
go back to reference Yanatori I, Kishi F. DMT1 and iron transport. Free Radical Biol Med. 2019;133:55–63.CrossRef Yanatori I, Kishi F. DMT1 and iron transport. Free Radical Biol Med. 2019;133:55–63.CrossRef
70.
go back to reference Harrison PM, Arosio P. The ferritins: molecular properties, iron storage function and cellular regulation. Biochim Biophys Acta. 1996;1275(3):161–203.PubMedCrossRef Harrison PM, Arosio P. The ferritins: molecular properties, iron storage function and cellular regulation. Biochim Biophys Acta. 1996;1275(3):161–203.PubMedCrossRef
71.
go back to reference del QuilesRey M, Mancias JD. NCOA4-mediated ferritinophagy: a potential link to neurodegeneration. Front Neurosci. 2019;13:238.CrossRef del QuilesRey M, Mancias JD. NCOA4-mediated ferritinophagy: a potential link to neurodegeneration. Front Neurosci. 2019;13:238.CrossRef
74.
go back to reference Rui T, et al. Deletion of ferritin H in neurons counteracts the protective effect of melatonin against traumatic brain injury-induced ferroptosis. J Pineal Res. 2021;70(2):e12704.PubMedCrossRef Rui T, et al. Deletion of ferritin H in neurons counteracts the protective effect of melatonin against traumatic brain injury-induced ferroptosis. J Pineal Res. 2021;70(2):e12704.PubMedCrossRef
76.
go back to reference Nielsen P, et al. Serum ferritin iron in iron overload and liver damage: correlation to body iron stores and diagnostic relevance. J Lab Clin Med. 2000;135(5):413–8.PubMedCrossRef Nielsen P, et al. Serum ferritin iron in iron overload and liver damage: correlation to body iron stores and diagnostic relevance. J Lab Clin Med. 2000;135(5):413–8.PubMedCrossRef
78.
go back to reference Kruszewski M. Labile iron pool: the main determinant of cellular response to oxidative stress. Mutat Res. 2003;531(1–2):81–92.PubMedCrossRef Kruszewski M. Labile iron pool: the main determinant of cellular response to oxidative stress. Mutat Res. 2003;531(1–2):81–92.PubMedCrossRef
79.
go back to reference Muckenthaler MU, Galy B, Hentze MW. Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu Rev Nutr. 2008;28:197–213.PubMedCrossRef Muckenthaler MU, Galy B, Hentze MW. Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu Rev Nutr. 2008;28:197–213.PubMedCrossRef
80.
go back to reference Styś A, et al. Iron regulatory protein 1 outcompetes iron regulatory protein 2 in regulating cellular iron homeostasis in response to nitric oxide. J Biol Chem. 2011;286(26):22846–54.PubMedCrossRefPubMedCentral Styś A, et al. Iron regulatory protein 1 outcompetes iron regulatory protein 2 in regulating cellular iron homeostasis in response to nitric oxide. J Biol Chem. 2011;286(26):22846–54.PubMedCrossRefPubMedCentral
81.
go back to reference Recalcati S, et al. Molecular regulation of cellular iron balance. IUBMB Life. 2017;69(6):389–98.PubMedCrossRef Recalcati S, et al. Molecular regulation of cellular iron balance. IUBMB Life. 2017;69(6):389–98.PubMedCrossRef
82.
go back to reference Montesinos J, Guardia-Laguarta C, Area-Gomez E. The fat brain. Curr Opin Clin Nutr Metab Care. 2020;23(2):68–75.PubMedCrossRef Montesinos J, Guardia-Laguarta C, Area-Gomez E. The fat brain. Curr Opin Clin Nutr Metab Care. 2020;23(2):68–75.PubMedCrossRef
83.
go back to reference Friedman J. Why is the nervous system vulnerable to oxidative stress? Oxidative Stress Free Radical Damage Neurol. 2011:19–27. Friedman J. Why is the nervous system vulnerable to oxidative stress? Oxidative Stress Free Radical Damage Neurol. 2011:19–27.
84.
go back to reference Rodencal J, Dixon SJ. A tale of two lipids: Lipid unsaturation commands ferroptosis sensitivity. Proteomics. 2023;23(6):2100308.CrossRef Rodencal J, Dixon SJ. A tale of two lipids: Lipid unsaturation commands ferroptosis sensitivity. Proteomics. 2023;23(6):2100308.CrossRef
87.
go back to reference Kuwata H, Hara S. Role of acyl-CoA synthetase ACSL4 in arachidonic acid metabolism. Prostaglandins Other Lipid Mediat. 2019;144:106363.PubMedCrossRef Kuwata H, Hara S. Role of acyl-CoA synthetase ACSL4 in arachidonic acid metabolism. Prostaglandins Other Lipid Mediat. 2019;144:106363.PubMedCrossRef
88.
go back to reference Doll S, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13(1):91–8.PubMedCrossRef Doll S, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13(1):91–8.PubMedCrossRef
89.
go back to reference Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radical Biol Med. 2019;133:144–52.CrossRef Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radical Biol Med. 2019;133:144–52.CrossRef
90.
go back to reference Ursini F, Maiorino M. Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radical Biol Med. 2020;152:175–85.CrossRef Ursini F, Maiorino M. Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radical Biol Med. 2020;152:175–85.CrossRef
91.
go back to reference Forcina GC, Dixon SJ. GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 2019;19(18):1800311.CrossRef Forcina GC, Dixon SJ. GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 2019;19(18):1800311.CrossRef
93.
go back to reference Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12(8):599–620.PubMedCrossRef Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12(8):599–620.PubMedCrossRef
97.
go back to reference Song Y, et al. Human umbilical cord blood–derived MSCs exosome attenuate myocardial injury by inhibiting ferroptosis in acute myocardial infarction mice. Cell Biol Toxicol. 2021;37:51–64.PubMedCrossRef Song Y, et al. Human umbilical cord blood–derived MSCs exosome attenuate myocardial injury by inhibiting ferroptosis in acute myocardial infarction mice. Cell Biol Toxicol. 2021;37:51–64.PubMedCrossRef
98.
go back to reference Yao F, et al. Iron regulatory protein 1 promotes ferroptosis by sustaining cellular iron homeostasis in melanoma. Oncol Lett. 2021;22(3):1–12.CrossRef Yao F, et al. Iron regulatory protein 1 promotes ferroptosis by sustaining cellular iron homeostasis in melanoma. Oncol Lett. 2021;22(3):1–12.CrossRef
99.
go back to reference Wen J, et al. Sulforaphane triggers iron overload-mediated ferroptosis in gastric carcinoma cells by activating the PI3K/IRP2/DMT1 pathway. Hum Exp Toxicol. 2023;42:09603271231177295.CrossRef Wen J, et al. Sulforaphane triggers iron overload-mediated ferroptosis in gastric carcinoma cells by activating the PI3K/IRP2/DMT1 pathway. Hum Exp Toxicol. 2023;42:09603271231177295.CrossRef
100.
go back to reference Geng N, et al. Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharmacol Sci. 2018;22(12):3826–36.PubMed Geng N, et al. Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharmacol Sci. 2018;22(12):3826–36.PubMed
101.
go back to reference Bao W-D, et al. Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer’s disease. Cell Death Differ. 2021;28(5):1548–62.PubMedCrossRefPubMedCentral Bao W-D, et al. Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer’s disease. Cell Death Differ. 2021;28(5):1548–62.PubMedCrossRefPubMedCentral
102.
go back to reference Zhang H. et al. Hepcidin promoted ferroptosis through iron metabolism which is associated with DMT1 signaling activation in early brain injury following subarachnoid hemorrhage. Oxidative Med Cell Longevity. 2021;2021. Zhang H. et al. Hepcidin promoted ferroptosis through iron metabolism which is associated with DMT1 signaling activation in early brain injury following subarachnoid hemorrhage. Oxidative Med Cell Longevity. 2021;2021.
103.
104.
go back to reference Liu N, et al. The role of ferroptosis mediated by NRF2/ERK-regulated ferritinophagy in CdTe QDs-induced inflammation in macrophage. J Hazard Mater. 2022;436:129043.PubMedCrossRef Liu N, et al. The role of ferroptosis mediated by NRF2/ERK-regulated ferritinophagy in CdTe QDs-induced inflammation in macrophage. J Hazard Mater. 2022;436:129043.PubMedCrossRef
105.
go back to reference Cheng Y, et al. TrkB agonist N-acetyl serotonin promotes functional recovery after traumatic brain injury by suppressing ferroptosis via the PI3K/Akt/Nrf2/Ferritin H pathway. Free Radical Biol Med. 2023;194:184–98.CrossRef Cheng Y, et al. TrkB agonist N-acetyl serotonin promotes functional recovery after traumatic brain injury by suppressing ferroptosis via the PI3K/Akt/Nrf2/Ferritin H pathway. Free Radical Biol Med. 2023;194:184–98.CrossRef
106.
go back to reference Yang, M., et al., Clockophagy is a novel selective autophagy process favoring ferroptosis. Science advances, 2019. 5(7): p. eaaw2238. Yang, M., et al., Clockophagy is a novel selective autophagy process favoring ferroptosis. Science advances, 2019. 5(7): p. eaaw2238.
107.
go back to reference Rozenfeld Y, et al. A model of disparities: risk factors associated with COVID-19 infection. Int J Equity Health. 2020;19(1):1–10.CrossRef Rozenfeld Y, et al. A model of disparities: risk factors associated with COVID-19 infection. Int J Equity Health. 2020;19(1):1–10.CrossRef
108.
go back to reference Pang L, et al. Influence of aging on deterioration of patients with COVID-19. Aging (Albany NY). 2020;12(24):26248.PubMedCrossRef Pang L, et al. Influence of aging on deterioration of patients with COVID-19. Aging (Albany NY). 2020;12(24):26248.PubMedCrossRef
109.
go back to reference Aguado J. et al. Senolytic therapy alleviates physiological human brain aging and COVID-19 neuropathology. bioRxiv, 2023: p. 2023.01. 17.524329. Aguado J. et al. Senolytic therapy alleviates physiological human brain aging and COVID-19 neuropathology. bioRxiv, 2023: p. 2023.01. 17.524329.
110.
go back to reference Sargiacomo C, Sotgia F, Lisanti MP. COVID-19 and chronological aging: senolytics and other anti-aging drugs for the treatment or prevention of corona virus infection? Aging (Albany NY). 2020;12(8):6511.PubMedCrossRef Sargiacomo C, Sotgia F, Lisanti MP. COVID-19 and chronological aging: senolytics and other anti-aging drugs for the treatment or prevention of corona virus infection? Aging (Albany NY). 2020;12(8):6511.PubMedCrossRef
111.
go back to reference Tripathi U, et al. SARS-CoV-2 causes senescence in human cells and exacerbates the senescence-associated secretory phenotype through TLR-3. Aging (albany NY). 2021;13(18):21838.PubMedCrossRef Tripathi U, et al. SARS-CoV-2 causes senescence in human cells and exacerbates the senescence-associated secretory phenotype through TLR-3. Aging (albany NY). 2021;13(18):21838.PubMedCrossRef
112.
115.
go back to reference Ineichen C, et al. Worsened Parkinson’s disease progression: impact of the COVID-19 pandemic. J Parkinsons Dis. 2021;11(4):1579–83.PubMedCrossRef Ineichen C, et al. Worsened Parkinson’s disease progression: impact of the COVID-19 pandemic. J Parkinsons Dis. 2021;11(4):1579–83.PubMedCrossRef
116.
go back to reference Fedeli U, et al. Parkinson’s disease related mortality: Long-term trends and impact of COVID-19 pandemic waves. Parkinsonism Relat Disord. 2022;98:75–7.PubMedCrossRefPubMedCentral Fedeli U, et al. Parkinson’s disease related mortality: Long-term trends and impact of COVID-19 pandemic waves. Parkinsonism Relat Disord. 2022;98:75–7.PubMedCrossRefPubMedCentral
117.
go back to reference Parihar R, et al. Outcome of Hospitalized Parkinson’s Disease Patients with and without COVID-19. Movement Disorders Clinical Practice. 2021;8(6):859–67.PubMedCrossRefPubMedCentral Parihar R, et al. Outcome of Hospitalized Parkinson’s Disease Patients with and without COVID-19. Movement Disorders Clinical Practice. 2021;8(6):859–67.PubMedCrossRefPubMedCentral
118.
go back to reference Zenesini C, et al. Risk of SARS-CoV-2 infection, hospitalization, and death for COVID-19 in people with Parkinson disease or parkinsonism over a 15-month period: A cohort study. Eur J Neurol. 2022;29(11):3205–17.CrossRef Zenesini C, et al. Risk of SARS-CoV-2 infection, hospitalization, and death for COVID-19 in people with Parkinson disease or parkinsonism over a 15-month period: A cohort study. Eur J Neurol. 2022;29(11):3205–17.CrossRef
119.
go back to reference Wang L. et al. Association of COVID-19 with new-onset Alzheimer’s disease. J Alzheimer's Dis. 2022(Preprint):1–4. Wang L. et al. Association of COVID-19 with new-onset Alzheimer’s disease. J Alzheimer's Dis. 2022(Preprint):1–4.
120.
go back to reference Chung SJ. et al. Association of Alzheimer’s disease with COVID-19 susceptibility and severe complications: a nationwide cohort study. J Alzheimer's Dis. 2022(Preprint):1–10. Chung SJ. et al. Association of Alzheimer’s disease with COVID-19 susceptibility and severe complications: a nationwide cohort study. J Alzheimer's Dis. 2022(Preprint):1–10.
121.
go back to reference Rudnicka-Drożak E, et al. Links between COVID-19 and Alzheimer’s disease—what do we already know? Int J Environ Res Public Health. 2023;20(3):2146.PubMedCrossRefPubMedCentral Rudnicka-Drożak E, et al. Links between COVID-19 and Alzheimer’s disease—what do we already know? Int J Environ Res Public Health. 2023;20(3):2146.PubMedCrossRefPubMedCentral
122.
go back to reference Frontera JA, et al. Comparison of serum neurodegenerative biomarkers among hospitalized COVID-19 patients versus non-COVID subjects with normal cognition, mild cognitive impairment, or Alzheimer’s dementia. Alzheimers Dement. 2022;18(5):899–910.PubMedCrossRef Frontera JA, et al. Comparison of serum neurodegenerative biomarkers among hospitalized COVID-19 patients versus non-COVID subjects with normal cognition, mild cognitive impairment, or Alzheimer’s dementia. Alzheimers Dement. 2022;18(5):899–910.PubMedCrossRef
123.
go back to reference Idrees D, Kumar V. SARS-CoV-2 spike protein interactions with amyloidogenic proteins: Potential clues to neurodegeneration. Biochem Biophys Res Commun. 2021;554:94–8.PubMedCrossRefPubMedCentral Idrees D, Kumar V. SARS-CoV-2 spike protein interactions with amyloidogenic proteins: Potential clues to neurodegeneration. Biochem Biophys Res Commun. 2021;554:94–8.PubMedCrossRefPubMedCentral
124.
125.
go back to reference Matias-Guiu JA, et al. Development of criteria for cognitive dysfunction in post-COVID syndrome: the IC-CoDi-COVID approach. Psychiatry Res. 2023;319:115006.PubMedCrossRef Matias-Guiu JA, et al. Development of criteria for cognitive dysfunction in post-COVID syndrome: the IC-CoDi-COVID approach. Psychiatry Res. 2023;319:115006.PubMedCrossRef
127.
128.
go back to reference Graham EL, et al. Persistent neurologic symptoms and cognitive dysfunction in non-hospitalized Covid-19 “long haulers.” Ann Clin Transl Neurol. 2021;8(5):1073–85.PubMedCrossRefPubMedCentral Graham EL, et al. Persistent neurologic symptoms and cognitive dysfunction in non-hospitalized Covid-19 “long haulers.” Ann Clin Transl Neurol. 2021;8(5):1073–85.PubMedCrossRefPubMedCentral
129.
go back to reference Ferrando SJ, et al. Neuropsychological, Medical, and Psychiatric Findings After Recovery From Acute COVID-19: A Cross-sectional Study. J Acad Consult Liaison Psychiatry. 2022;63(5):474–84.PubMedCrossRefPubMedCentral Ferrando SJ, et al. Neuropsychological, Medical, and Psychiatric Findings After Recovery From Acute COVID-19: A Cross-sectional Study. J Acad Consult Liaison Psychiatry. 2022;63(5):474–84.PubMedCrossRefPubMedCentral
130.
go back to reference Schild AK, et al. Multidomain cognitive impairment in non-hospitalized patients with the post-COVID-19 syndrome: results from a prospective monocentric cohort. J Neurol. 2023;270(3):1215–23.PubMedCrossRef Schild AK, et al. Multidomain cognitive impairment in non-hospitalized patients with the post-COVID-19 syndrome: results from a prospective monocentric cohort. J Neurol. 2023;270(3):1215–23.PubMedCrossRef
132.
133.
go back to reference Ariza M. et al. COVID-19 severity is related to poor executive function in people with post-COVID conditions. J Neurol. 2023:1–17. Ariza M. et al. COVID-19 severity is related to poor executive function in people with post-COVID conditions. J Neurol. 2023:1–17.
134.
go back to reference Lerner AB, et al. Isolation of melatonin, the pineal gland factor that lightens melanocyteS1. J Am Chem Soc. 1958;80(10):2587–2587.CrossRef Lerner AB, et al. Isolation of melatonin, the pineal gland factor that lightens melanocyteS1. J Am Chem Soc. 1958;80(10):2587–2587.CrossRef
136.
go back to reference Claustrat B, Brun J, Chazot G. The basic physiology and pathophysiology of melatonin. Sleep Med Rev. 2005;9(1):11–24.PubMedCrossRef Claustrat B, Brun J, Chazot G. The basic physiology and pathophysiology of melatonin. Sleep Med Rev. 2005;9(1):11–24.PubMedCrossRef
137.
go back to reference Reiter RJ. Melatonin: the chemical expression of darkness. Mol Cell Endocrinol. 1991;79(1–3):C153–8.PubMedCrossRef Reiter RJ. Melatonin: the chemical expression of darkness. Mol Cell Endocrinol. 1991;79(1–3):C153–8.PubMedCrossRef
139.
140.
go back to reference Cagnacci A, Elliott J, Yen S. Melatonin: a major regulator of the circadian rhythm of core temperature in humans. J Clin Endocrinol Metab. 1992;75(2):447–52.PubMed Cagnacci A, Elliott J, Yen S. Melatonin: a major regulator of the circadian rhythm of core temperature in humans. J Clin Endocrinol Metab. 1992;75(2):447–52.PubMed
141.
go back to reference Arendt J. Melatonin and the pineal gland: influence on mammalian seasonal and circadian physiology. Rev Reprod. 1998;3:13–22.PubMedCrossRef Arendt J. Melatonin and the pineal gland: influence on mammalian seasonal and circadian physiology. Rev Reprod. 1998;3:13–22.PubMedCrossRef
142.
go back to reference Axelrod J, Wurtman RJ. Photic and Neural Control of Indoleamine Metabolism in the Rat Pineal Gland, in Advances in Pharmacology, S. Garattini and P.A. Shore, Editors. 1968, Academic Press. p. 157–166. Axelrod J, Wurtman RJ. Photic and Neural Control of Indoleamine Metabolism in the Rat Pineal Gland, in Advances in Pharmacology, S. Garattini and P.A. Shore, Editors. 1968, Academic Press. p. 157–166.
143.
go back to reference Axelrod J. The Pineal Gland: a neurochemical transducer: chemical signals from nerves regulate synthesis of melatonin and convey information about internal clocks. Science. 1974;184(4144):1341–8.PubMedCrossRef Axelrod J. The Pineal Gland: a neurochemical transducer: chemical signals from nerves regulate synthesis of melatonin and convey information about internal clocks. Science. 1974;184(4144):1341–8.PubMedCrossRef
144.
go back to reference Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002;295(5557):1070–3.PubMedCrossRef Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002;295(5557):1070–3.PubMedCrossRef
145.
go back to reference Pevet P, Challet E. Melatonin: both master clock output and internal time-giver in the circadian clocks network. Journal of Physiology-Paris. 2011;105(4–6):170–82.PubMedCrossRef Pevet P, Challet E. Melatonin: both master clock output and internal time-giver in the circadian clocks network. Journal of Physiology-Paris. 2011;105(4–6):170–82.PubMedCrossRef
146.
go back to reference Krause DN, Dubocovich ML. Regulatory sites in the melatonin system of mammals. Trends Neurosci. 1990;13(11):464–70.PubMedCrossRef Krause DN, Dubocovich ML. Regulatory sites in the melatonin system of mammals. Trends Neurosci. 1990;13(11):464–70.PubMedCrossRef
147.
go back to reference Klein DC, Moore RY. Pineal N-acetyltransferase and hydroxyindole-O-methyl-transferase: control by the retinohypothalamic tract and the suprachiasmatic nucleus. Brain Res. 1979;174(2):245–62.PubMedCrossRef Klein DC, Moore RY. Pineal N-acetyltransferase and hydroxyindole-O-methyl-transferase: control by the retinohypothalamic tract and the suprachiasmatic nucleus. Brain Res. 1979;174(2):245–62.PubMedCrossRef
148.
go back to reference Zimmermann R, et al. Effects of acute tryptophan depletion on nocturnal melatonin secretion in humans. J Clin Endocrinol Metab. 1993;76(5):1160–4.PubMed Zimmermann R, et al. Effects of acute tryptophan depletion on nocturnal melatonin secretion in humans. J Clin Endocrinol Metab. 1993;76(5):1160–4.PubMed
149.
go back to reference Ruddick JP, et al. Tryptophan metabolism in the central nervous system: medical implications. Expert Rev Mol Med. 2006;8(20):1–27.PubMedCrossRef Ruddick JP, et al. Tryptophan metabolism in the central nervous system: medical implications. Expert Rev Mol Med. 2006;8(20):1–27.PubMedCrossRef
150.
go back to reference King TS, Richardson BA, Reiter RJ. Regulation of rat pineal melatonin synthesis: effect of monoamine oxidase inhibition. Mol Cell Endocrinol. 1982;25(3):327–38.PubMedCrossRef King TS, Richardson BA, Reiter RJ. Regulation of rat pineal melatonin synthesis: effect of monoamine oxidase inhibition. Mol Cell Endocrinol. 1982;25(3):327–38.PubMedCrossRef
151.
go back to reference Acuña-Castroviejo D, et al. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol Life Sci. 2014;71(16):2997–3025.PubMedCrossRef Acuña-Castroviejo D, et al. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol Life Sci. 2014;71(16):2997–3025.PubMedCrossRef
152.
153.
go back to reference Costa EJ, Lopes RH, Lamy-Freund MT. Permeability of pure lipid bilayers to melatonin. J Pineal Res. 1995;19(3):123–6.PubMedCrossRef Costa EJ, Lopes RH, Lamy-Freund MT. Permeability of pure lipid bilayers to melatonin. J Pineal Res. 1995;19(3):123–6.PubMedCrossRef
155.
go back to reference Reppert SM, Weaver DR, Ebisawa T. Cloning and characterization of a mammalian melatonin receptor that mediates reproductive and circadian responses. Neuron. 1994;13(5):1177–85.PubMedCrossRef Reppert SM, Weaver DR, Ebisawa T. Cloning and characterization of a mammalian melatonin receptor that mediates reproductive and circadian responses. Neuron. 1994;13(5):1177–85.PubMedCrossRef
156.
go back to reference Reppert SM, et al. Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc Natl Acad Sci. 1995;92(19):8734–8.PubMedCrossRefPubMedCentral Reppert SM, et al. Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc Natl Acad Sci. 1995;92(19):8734–8.PubMedCrossRefPubMedCentral
157.
go back to reference Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol. 2007;213(3):589–602.PubMedCrossRef Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol. 2007;213(3):589–602.PubMedCrossRef
158.
go back to reference Cecon E, Oishi A, Jockers R. Melatonin receptors: molecular pharmacology and signalling in the context of system bias. Br J Pharmacol. 2018;175(16):3263–80.PubMedCrossRef Cecon E, Oishi A, Jockers R. Melatonin receptors: molecular pharmacology and signalling in the context of system bias. Br J Pharmacol. 2018;175(16):3263–80.PubMedCrossRef
159.
160.
go back to reference Nosjean O, et al. Identification of the Melatonin-binding SiteMT 3 as the Quinone Reductase 2. J Biol Chem. 2000;275(40):31311–7.PubMedCrossRef Nosjean O, et al. Identification of the Melatonin-binding SiteMT 3 as the Quinone Reductase 2. J Biol Chem. 2000;275(40):31311–7.PubMedCrossRef
161.
go back to reference Boutin JA. Quinone reductase 2 as a promising target of melatonin therapeutic actions. Expert Opin Ther Targets. 2016;20(3):303–17.PubMedCrossRef Boutin JA. Quinone reductase 2 as a promising target of melatonin therapeutic actions. Expert Opin Ther Targets. 2016;20(3):303–17.PubMedCrossRef
162.
go back to reference Becker-André M, et al. Pineal gland hormone melatonin binds and activates an orphan of the nuclear receptor superfamily. J Biol Chem. 1994;269(46):28531–4.PubMedCrossRef Becker-André M, et al. Pineal gland hormone melatonin binds and activates an orphan of the nuclear receptor superfamily. J Biol Chem. 1994;269(46):28531–4.PubMedCrossRef
163.
go back to reference Kojetin DJ, Burris TP. REV-ERB and ROR nuclear receptors as drug targets. Nat Rev Drug Discovery. 2014;13(3):197–216.PubMedCrossRef Kojetin DJ, Burris TP. REV-ERB and ROR nuclear receptors as drug targets. Nat Rev Drug Discovery. 2014;13(3):197–216.PubMedCrossRef
164.
go back to reference Wurtman RJ, Axelrod J, Chu EW. Melatonin, a pineal substance: effect on the rat ovary. Science. 1963;141(3577):277–8.PubMedCrossRef Wurtman RJ, Axelrod J, Chu EW. Melatonin, a pineal substance: effect on the rat ovary. Science. 1963;141(3577):277–8.PubMedCrossRef
165.
go back to reference Reiter, R., et al. Melatonin: reproductive effects. in The Pineal Gland: Proceedings of the International Symposium, Jerusalem, November 14–17, 1977. 1978. Springer. Reiter, R., et al. Melatonin: reproductive effects. in The Pineal Gland: Proceedings of the International Symposium, Jerusalem, November 14–17, 1977. 1978. Springer.
166.
go back to reference Fiske V, Bryant G, Putnam J. Effect of light on the weight of the pineal in the rat. 1960, ENDOCRINE SOC 4350 EAST WEST HIGHWAY SUITE 500, BETHESDA, MD 20814-4110. p. 489-491. Fiske V, Bryant G, Putnam J. Effect of light on the weight of the pineal in the rat. 1960, ENDOCRINE SOC 4350 EAST WEST HIGHWAY SUITE 500, BETHESDA, MD 20814-4110. p. 489-491.
167.
go back to reference Wurtman RJ, et al. Interactions of the pineal and exposure to continuous light on organ weights of female rats. Eur J Endocrinol. 1961;36(4):617–24.CrossRef Wurtman RJ, et al. Interactions of the pineal and exposure to continuous light on organ weights of female rats. Eur J Endocrinol. 1961;36(4):617–24.CrossRef
168.
go back to reference Tamarkin L, et al. Effect of melatonin on the reproductive systems of male and female Syrian hamsters: a diurnal rhythm in sensitivity to melatonin. Endocrinology. 1976;99(6):1534–41.PubMedCrossRef Tamarkin L, et al. Effect of melatonin on the reproductive systems of male and female Syrian hamsters: a diurnal rhythm in sensitivity to melatonin. Endocrinology. 1976;99(6):1534–41.PubMedCrossRef
169.
go back to reference Lewy AJ, et al. Melatonin shifts human orcadian rhythms according to a phase-response curve. Chronobiol Int. 1992;9(5):380–92.PubMedCrossRef Lewy AJ, et al. Melatonin shifts human orcadian rhythms according to a phase-response curve. Chronobiol Int. 1992;9(5):380–92.PubMedCrossRef
170.
go back to reference Strassman RJ, et al. Elevated rectal temperature produced by all-night bright light is reversed by melatonin infusion in men. J Appl Physiol. 1991;71(6):2178–82.PubMedCrossRef Strassman RJ, et al. Elevated rectal temperature produced by all-night bright light is reversed by melatonin infusion in men. J Appl Physiol. 1991;71(6):2178–82.PubMedCrossRef
171.
go back to reference Poeggeler B, et al. Melatonin—a highly potent endogenous radical scavenger and electron donor: new aspects of the oxidation chemistry of this indole accessed in vitro a. Ann N Y Acad Sci. 1994;738(1):419–20.PubMedCrossRef Poeggeler B, et al. Melatonin—a highly potent endogenous radical scavenger and electron donor: new aspects of the oxidation chemistry of this indole accessed in vitro a. Ann N Y Acad Sci. 1994;738(1):419–20.PubMedCrossRef
172.
go back to reference Reiter RJ, et al. Melatonin and its relation to the immune system and inflammation. Ann N Y Acad Sci. 2000;917(1):376–86.PubMedCrossRef Reiter RJ, et al. Melatonin and its relation to the immune system and inflammation. Ann N Y Acad Sci. 2000;917(1):376–86.PubMedCrossRef
173.
go back to reference Morgan L, et al. Effects of the endogenous clock and sleep time on melatonin, insulin, glucose and lipid metabolism. J Endocrinol. 1998;157(3):443–52.PubMedCrossRef Morgan L, et al. Effects of the endogenous clock and sleep time on melatonin, insulin, glucose and lipid metabolism. J Endocrinol. 1998;157(3):443–52.PubMedCrossRef
174.
go back to reference Arendt J. Melatonin and the mammalian pineal gland. 1994: Springer Science & Business Media. Arendt J. Melatonin and the mammalian pineal gland. 1994: Springer Science & Business Media.
175.
go back to reference Brainard GC, et al. Action spectrum for melatonin regulation in humans: evidence for a novel circadian photoreceptor. J Neurosci. 2001;21(16):6405–12.PubMedCrossRefPubMedCentral Brainard GC, et al. Action spectrum for melatonin regulation in humans: evidence for a novel circadian photoreceptor. J Neurosci. 2001;21(16):6405–12.PubMedCrossRefPubMedCentral
176.
177.
go back to reference Arendt J et al. Some effects of melatonin and the control of its secretion in humans. in Ciba Foundation Symposium 117‐Photoperiodism, Melatonin and the Pineal. 1985. Wiley Online Library. Arendt J et al. Some effects of melatonin and the control of its secretion in humans. in Ciba Foundation Symposium 117‐Photoperiodism, Melatonin and the Pineal. 1985. Wiley Online Library.
178.
go back to reference Liu C, et al. Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron. 1997;19(1):91–102.PubMedCrossRef Liu C, et al. Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron. 1997;19(1):91–102.PubMedCrossRef
179.
go back to reference Albrecht U. The mammalian circadian clock: a network of gene expression. Front Biosci. 2004;9(48–55):8. Albrecht U. The mammalian circadian clock: a network of gene expression. Front Biosci. 2004;9(48–55):8.
180.
go back to reference Vriend J, Reiter RJ. Melatonin feedback on clock genes: a theory involving the proteasome. J Pineal Res. 2015;58(1):1–11.PubMedCrossRef Vriend J, Reiter RJ. Melatonin feedback on clock genes: a theory involving the proteasome. J Pineal Res. 2015;58(1):1–11.PubMedCrossRef
181.
go back to reference Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. 2017;18(3):164–79.PubMedCrossRef Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. 2017;18(3):164–79.PubMedCrossRef
182.
go back to reference Hirayama J, Sassone-Corsi P. Structural and functional features of transcription factors controlling the circadian clock. Curr Opin Genet Dev. 2005;15(5):548–56.PubMedCrossRef Hirayama J, Sassone-Corsi P. Structural and functional features of transcription factors controlling the circadian clock. Curr Opin Genet Dev. 2005;15(5):548–56.PubMedCrossRef
184.
go back to reference Duez H, Staels B. The nuclear receptors Rev-erbs and RORs integrate circadian rhythms and metabolism. Diab Vasc Dis Res. 2008;5(2):82–8.PubMedCrossRef Duez H, Staels B. The nuclear receptors Rev-erbs and RORs integrate circadian rhythms and metabolism. Diab Vasc Dis Res. 2008;5(2):82–8.PubMedCrossRef
185.
186.
go back to reference Roy D, Belsham DD. Melatonin receptor activation regulates GnRH Gene expression and secretion in GT1–7 GnRH neurons: signal transduction mechanisms. J Biol Chem. 2002;277(1):251–8.PubMedCrossRef Roy D, Belsham DD. Melatonin receptor activation regulates GnRH Gene expression and secretion in GT1–7 GnRH neurons: signal transduction mechanisms. J Biol Chem. 2002;277(1):251–8.PubMedCrossRef
188.
go back to reference Waldhauser F, et al. Fall in nocturnal serum melatonin during prepuberty and pubescence. Lancet. 1984;323(8373):362–5.CrossRef Waldhauser F, et al. Fall in nocturnal serum melatonin during prepuberty and pubescence. Lancet. 1984;323(8373):362–5.CrossRef
189.
go back to reference Wójtowicz M, Jakiel G. Melatonin and its role in human reproduction. Ginekol Pol. 2002;73(12):1231–7.PubMed Wójtowicz M, Jakiel G. Melatonin and its role in human reproduction. Ginekol Pol. 2002;73(12):1231–7.PubMed
190.
go back to reference Olcese JM. Melatonin and female reproduction: an expanding universe. Front Endocrinol. 2020;11:85.CrossRef Olcese JM. Melatonin and female reproduction: an expanding universe. Front Endocrinol. 2020;11:85.CrossRef
191.
go back to reference Sun T-C, et al. Protective effects of melatonin on male fertility preservation and reproductive system. Cryobiology. 2020;95:1–8.PubMedCrossRef Sun T-C, et al. Protective effects of melatonin on male fertility preservation and reproductive system. Cryobiology. 2020;95:1–8.PubMedCrossRef
192.
go back to reference Roth JA, et al. Melatonin promotes osteoblast differentiation and bone formation*. J Biol Chem. 1999;274(31):22041–7.PubMedCrossRef Roth JA, et al. Melatonin promotes osteoblast differentiation and bone formation*. J Biol Chem. 1999;274(31):22041–7.PubMedCrossRef
193.
go back to reference Zhang L, et al. Melatonin inhibits adipogenesis and enhances osteogenesis of human mesenchymal stem cells by suppressing PPARγ expression and enhancing Runx2 expression. J Pineal Res. 2010;49(4):364–72.PubMedCrossRef Zhang L, et al. Melatonin inhibits adipogenesis and enhances osteogenesis of human mesenchymal stem cells by suppressing PPARγ expression and enhancing Runx2 expression. J Pineal Res. 2010;49(4):364–72.PubMedCrossRef
194.
go back to reference Arangino S, et al. Effects of melatonin on vascular reactivity, catecholamine levels, and blood pressure in healthy men. Am J Cardiol. 1999;83(9):1417–9.PubMedCrossRef Arangino S, et al. Effects of melatonin on vascular reactivity, catecholamine levels, and blood pressure in healthy men. Am J Cardiol. 1999;83(9):1417–9.PubMedCrossRef
196.
go back to reference Régrigny O, et al. Melatonin improves cerebral circulation security margin in rats. Am J Physiology-Heart Circulatory Physiol. 1998;275(1):H139–44.CrossRef Régrigny O, et al. Melatonin improves cerebral circulation security margin in rats. Am J Physiology-Heart Circulatory Physiol. 1998;275(1):H139–44.CrossRef
197.
go back to reference Yasmin F, et al. Gut melatonin: a potent candidate in the diversified journey of melatonin research. Gen Comp Endocrinol. 2021;303:113693.PubMedCrossRef Yasmin F, et al. Gut melatonin: a potent candidate in the diversified journey of melatonin research. Gen Comp Endocrinol. 2021;303:113693.PubMedCrossRef
198.
go back to reference Mazzucchelli C, et al. The melatonin receptor in the human brain: cloning experiments and distribution studies. Mol Brain Res. 1996;39(1–2):117–26.PubMedCrossRef Mazzucchelli C, et al. The melatonin receptor in the human brain: cloning experiments and distribution studies. Mol Brain Res. 1996;39(1–2):117–26.PubMedCrossRef
199.
go back to reference Pandi-Perumal SR, et al. Physiological effects of melatonin: role of melatonin receptors and signal transduction pathways. Prog Neurobiol. 2008;85(3):335–53.PubMedCrossRef Pandi-Perumal SR, et al. Physiological effects of melatonin: role of melatonin receptors and signal transduction pathways. Prog Neurobiol. 2008;85(3):335–53.PubMedCrossRef
200.
go back to reference Reiter RJ, et al. Delivery of pineal melatonin to the brain and SCN: role of canaliculi, cerebrospinal fluid, tanycytes and Virchow-Robin perivascular spaces. Brain Struct Funct. 2014;219:1873–87.PubMedCrossRef Reiter RJ, et al. Delivery of pineal melatonin to the brain and SCN: role of canaliculi, cerebrospinal fluid, tanycytes and Virchow-Robin perivascular spaces. Brain Struct Funct. 2014;219:1873–87.PubMedCrossRef
201.
go back to reference Ng KY, et al. Melatonin receptors: distribution in mammalian brain and their respective putative functions. Brain Struct Funct. 2017;222:2921–39.PubMedCrossRef Ng KY, et al. Melatonin receptors: distribution in mammalian brain and their respective putative functions. Brain Struct Funct. 2017;222:2921–39.PubMedCrossRef
202.
go back to reference Nir I, et al. Changes in the electrical activity of the brain following pinealectomy. Neuroendocrinology. 1969;4(2):122–7.PubMedCrossRef Nir I, et al. Changes in the electrical activity of the brain following pinealectomy. Neuroendocrinology. 1969;4(2):122–7.PubMedCrossRef
203.
go back to reference Bindoni M, Rizzo R. Effects of electrolytic lesions of the pineal gland on the electric activity of some brain structures in the rabbit. Bollettino Della Societa Italiana di Biologia Sperimentale. 1964;40(24): p. Suppl: 2010–3. Bindoni M, Rizzo R. Effects of electrolytic lesions of the pineal gland on the electric activity of some brain structures in the rabbit. Bollettino Della Societa Italiana di Biologia Sperimentale. 1964;40(24): p. Suppl: 2010–3.
204.
go back to reference Fariello RG, Bubenik GA. Melatonin-induced changes in the sensory activation of acute epileptic foci. Neurosci Lett. 1976;3(3):151–5.PubMedCrossRef Fariello RG, Bubenik GA. Melatonin-induced changes in the sensory activation of acute epileptic foci. Neurosci Lett. 1976;3(3):151–5.PubMedCrossRef
205.
go back to reference Stankov B, et al. Melatonin signal transduction and mechanism of action in the central nervous system: using the rabbit cortex as a model. Endocrinology. 1992;130(4):2152–9.PubMed Stankov B, et al. Melatonin signal transduction and mechanism of action in the central nervous system: using the rabbit cortex as a model. Endocrinology. 1992;130(4):2152–9.PubMed
206.
go back to reference Xu Z, et al. Melatonin alleviates PTSD-like behaviors and restores serum GABA and cortisol levels in mice. Psychopharmacology. 2023;240(2):259–69.PubMedCrossRef Xu Z, et al. Melatonin alleviates PTSD-like behaviors and restores serum GABA and cortisol levels in mice. Psychopharmacology. 2023;240(2):259–69.PubMedCrossRef
207.
go back to reference Pierrefiche G, Zerbib R, Laborit H. Anxiolytic activity of melatonin in mice: involvement of benzodiazepine receptors. Res Commun Chem Pathol Pharmacol. 1993;82(2):131–42.PubMed Pierrefiche G, Zerbib R, Laborit H. Anxiolytic activity of melatonin in mice: involvement of benzodiazepine receptors. Res Commun Chem Pathol Pharmacol. 1993;82(2):131–42.PubMed
208.
go back to reference Raghavendra V, Kulkarni SK. Reversal of morphine tolerance and dependence by melatonin: possible role of central and peripheral benzodiazepine receptors. Brain Res. 1999;834(1–2):178–81.PubMedCrossRef Raghavendra V, Kulkarni SK. Reversal of morphine tolerance and dependence by melatonin: possible role of central and peripheral benzodiazepine receptors. Brain Res. 1999;834(1–2):178–81.PubMedCrossRef
209.
go back to reference Raghavendra V, Kaur G, Kulkarni SK. Anti-depressant action of melatonin in chronic forced swimming-induced behavioral despair in mice, role of peripheral benzodiazepine receptor modulation. Eur Neuropsychopharmacol. 2000;10(6):473–81.PubMedCrossRef Raghavendra V, Kaur G, Kulkarni SK. Anti-depressant action of melatonin in chronic forced swimming-induced behavioral despair in mice, role of peripheral benzodiazepine receptor modulation. Eur Neuropsychopharmacol. 2000;10(6):473–81.PubMedCrossRef
210.
go back to reference Acufla-Castroviejo D, et al. Minireview: Cell protective role of melatonin in the brain. J Pineal Res. 1995;19(2):57–63.CrossRef Acufla-Castroviejo D, et al. Minireview: Cell protective role of melatonin in the brain. J Pineal Res. 1995;19(2):57–63.CrossRef
211.
go back to reference Vishnoi S, Raisuddin S, Parvez S. Glutamate excitotoxicity and oxidative stress in epilepsy: modulatory role of melatonin. J Environ Pathol Toxicol Oncol. 2016. 35(4). Vishnoi S, Raisuddin S, Parvez S. Glutamate excitotoxicity and oxidative stress in epilepsy: modulatory role of melatonin. J Environ Pathol Toxicol Oncol. 2016. 35(4).
212.
go back to reference Reiter RJ. Oxidative damage in the central nervous system: protection by melatonin. Prog Neurobiol. 1998;56(3):359–84.PubMedCrossRef Reiter RJ. Oxidative damage in the central nervous system: protection by melatonin. Prog Neurobiol. 1998;56(3):359–84.PubMedCrossRef
213.
go back to reference Leon J, et al. Modification of nitric oxide synthase activity and neuronal response in rat striatum by melatonin and kynurenine derivatives. J Neuroendocrinol. 1998;10(4):297–302.PubMedCrossRef Leon J, et al. Modification of nitric oxide synthase activity and neuronal response in rat striatum by melatonin and kynurenine derivatives. J Neuroendocrinol. 1998;10(4):297–302.PubMedCrossRef
214.
go back to reference León J, et al. Structure-related inhibition of calmodulin-dependent neuronal nitric-oxide synthase activity by melatonin and synthetic kynurenines. Mol Pharmacol. 2000;58(5):967–75.PubMedCrossRef León J, et al. Structure-related inhibition of calmodulin-dependent neuronal nitric-oxide synthase activity by melatonin and synthetic kynurenines. Mol Pharmacol. 2000;58(5):967–75.PubMedCrossRef
215.
go back to reference Larson J, et al. Impaired hippocampal long-term potentiation in melatonin MT2 receptor-deficient mice. Neurosci Lett. 2006;393(1):23–6.PubMedCrossRef Larson J, et al. Impaired hippocampal long-term potentiation in melatonin MT2 receptor-deficient mice. Neurosci Lett. 2006;393(1):23–6.PubMedCrossRef
217.
go back to reference Cipolla-Neto J, Amaral FGD. Melatonin as a hormone: new physiological and clinical insights. Endocrine Rev. 2018;39(6):990–1028.CrossRef Cipolla-Neto J, Amaral FGD. Melatonin as a hormone: new physiological and clinical insights. Endocrine Rev. 2018;39(6):990–1028.CrossRef
218.
219.
go back to reference Besag FM, Vasey MJ. Adverse events in long-term studies of exogenous melatonin. Expert Opin Drug Saf. 2022;21(12):1469–81.PubMedCrossRef Besag FM, Vasey MJ. Adverse events in long-term studies of exogenous melatonin. Expert Opin Drug Saf. 2022;21(12):1469–81.PubMedCrossRef
220.
go back to reference Papagiannidou E, Skene DJ, Ioannides C. Potential drug interactions with melatonin. Physiol Behav. 2014;131:17–24.PubMedCrossRef Papagiannidou E, Skene DJ, Ioannides C. Potential drug interactions with melatonin. Physiol Behav. 2014;131:17–24.PubMedCrossRef
221.
go back to reference Ashy NI, Shroff KV, Ashy N. Evaluation of the potential drug interaction of melatonin and warfarin: a case series. Life Sci J. 2016;13(6):124. Ashy NI, Shroff KV, Ashy N. Evaluation of the potential drug interaction of melatonin and warfarin: a case series. Life Sci J. 2016;13(6):124.
222.
go back to reference Hosseinzadeh A, et al. Melatonin effect on platelets and coagulation: Implications for a prophylactic indication in COVID-19. Life Sci. 2022;307:120866.PubMedCrossRefPubMedCentral Hosseinzadeh A, et al. Melatonin effect on platelets and coagulation: Implications for a prophylactic indication in COVID-19. Life Sci. 2022;307:120866.PubMedCrossRefPubMedCentral
223.
224.
go back to reference Herxheimer A, Petrie KJ, C.C.M.D. Group. Melatonin for the prevention and treatment of jet lag. Cochrane Database Syst Rev. 1996;2010(1). Herxheimer A, Petrie KJ, C.C.M.D. Group. Melatonin for the prevention and treatment of jet lag. Cochrane Database Syst Rev. 1996;2010(1).
225.
go back to reference Waldhauser F, et al. Serum melatonin in central precocious puberty is lower than in age-matched prepubertal children. J Clin Endocrinol Metab. 1991;73(4):793–6.PubMedCrossRef Waldhauser F, et al. Serum melatonin in central precocious puberty is lower than in age-matched prepubertal children. J Clin Endocrinol Metab. 1991;73(4):793–6.PubMedCrossRef
226.
go back to reference Tamura H, et al. Melatonin and the ovary: physiological and pathophysiological implications. Fertil Steril. 2009;92(1):328–43.PubMedCrossRef Tamura H, et al. Melatonin and the ovary: physiological and pathophysiological implications. Fertil Steril. 2009;92(1):328–43.PubMedCrossRef
227.
go back to reference Brugger P, Marktl W, Herold M. Impaired nocturnal secretion of melatonin in coronary heart disease. Lancet. 1995;345(8962):1408.PubMedCrossRef Brugger P, Marktl W, Herold M. Impaired nocturnal secretion of melatonin in coronary heart disease. Lancet. 1995;345(8962):1408.PubMedCrossRef
228.
229.
go back to reference Moradkhani F, et al. Immunoregulatory role of melatonin in cancer. J Cell Physiol. 2020;235(2):745–57.PubMedCrossRef Moradkhani F, et al. Immunoregulatory role of melatonin in cancer. J Cell Physiol. 2020;235(2):745–57.PubMedCrossRef
230.
go back to reference Patel R. et al. Diabetes mellitus and melatonin: where are we? Biochimie. 2022. Patel R. et al. Diabetes mellitus and melatonin: where are we? Biochimie. 2022.
231.
go back to reference Cipolla-Neto J, et al. Melatonin, energy metabolism, and obesity: a review. J Pineal Res. 2014;56(4):371–81.PubMedCrossRef Cipolla-Neto J, et al. Melatonin, energy metabolism, and obesity: a review. J Pineal Res. 2014;56(4):371–81.PubMedCrossRef
232.
go back to reference Potes Y, et al. Benefits of the neurogenic potential of melatonin for treating neurological and neuropsychiatric disorders. Int J Mol Sci. 2023;24(5):4803.PubMedCrossRefPubMedCentral Potes Y, et al. Benefits of the neurogenic potential of melatonin for treating neurological and neuropsychiatric disorders. Int J Mol Sci. 2023;24(5):4803.PubMedCrossRefPubMedCentral
233.
go back to reference Touitou Y, et al. Age-and mental health-related circadian rhythms of plasma levels of melatonin, prolactin, luteinizing hormone and follicle-stimulating hormone in man. J Endocrinol. 1981;91(3):467–75.PubMedCrossRef Touitou Y, et al. Age-and mental health-related circadian rhythms of plasma levels of melatonin, prolactin, luteinizing hormone and follicle-stimulating hormone in man. J Endocrinol. 1981;91(3):467–75.PubMedCrossRef
234.
go back to reference Iguchi H, Kato K-I, Ibayashi H. Age-dependent reduction in serum melatonin concentrations in healthy human subjects. J Clin Endocrinol Metab. 1982;55(1):27–9.CrossRef Iguchi H, Kato K-I, Ibayashi H. Age-dependent reduction in serum melatonin concentrations in healthy human subjects. J Clin Endocrinol Metab. 1982;55(1):27–9.CrossRef
235.
go back to reference Touitou Y, et al. Age-related changes in both circadian and seasonal rhythms of rectal temperature with special reference to senile dementia of Alzheimer type. Gerontology. 1986;32(2):110–8.PubMedCrossRef Touitou Y, et al. Age-related changes in both circadian and seasonal rhythms of rectal temperature with special reference to senile dementia of Alzheimer type. Gerontology. 1986;32(2):110–8.PubMedCrossRef
236.
go back to reference Magri F, et al. Changes in endocrine orcadian rhythms as markers of physiological and pathological brain aging. Chronobiol Int. 1997;14(4):385–96.PubMedCrossRef Magri F, et al. Changes in endocrine orcadian rhythms as markers of physiological and pathological brain aging. Chronobiol Int. 1997;14(4):385–96.PubMedCrossRef
237.
go back to reference Tresguerres IF, et al. Melatonin dietary supplement as an anti-aging therapy for age-related bone loss. Rejuvenation Res. 2014;17(4):341–6.PubMedCrossRef Tresguerres IF, et al. Melatonin dietary supplement as an anti-aging therapy for age-related bone loss. Rejuvenation Res. 2014;17(4):341–6.PubMedCrossRef
238.
go back to reference Lesnikov VA, Pierpaoli W. Pineal cross-transplantation (old-to-young and vice versa) as evidence for an endogenous" aging clock". Ann N Y Acad Sci. 1994;719:456–60.PubMedCrossRef Lesnikov VA, Pierpaoli W. Pineal cross-transplantation (old-to-young and vice versa) as evidence for an endogenous" aging clock". Ann N Y Acad Sci. 1994;719:456–60.PubMedCrossRef
239.
go back to reference Caroleo MC, Doria G, Nistico G. Melatonin restores immunodepression in aged and cyclophosphamide-treated mice. Ann N Y Acad Sci. 1994;719:343–52.PubMedCrossRef Caroleo MC, Doria G, Nistico G. Melatonin restores immunodepression in aged and cyclophosphamide-treated mice. Ann N Y Acad Sci. 1994;719:343–52.PubMedCrossRef
240.
go back to reference Yoo DY, et al. Melatonin improves D-galactose-induced aging effects on behavior, neurogenesis, and lipid peroxidation in the mouse dentate gyrus via increasing pCREB expression. J Pineal Res. 2012;52(1):21–8.PubMedCrossRef Yoo DY, et al. Melatonin improves D-galactose-induced aging effects on behavior, neurogenesis, and lipid peroxidation in the mouse dentate gyrus via increasing pCREB expression. J Pineal Res. 2012;52(1):21–8.PubMedCrossRef
241.
go back to reference Cachán-Vega C, et al. Chronic treatment with melatonin improves hippocampal neurogenesis in the aged brain and under neurodegeneration. Molecules. 2022;27(17):5543.PubMedCrossRefPubMedCentral Cachán-Vega C, et al. Chronic treatment with melatonin improves hippocampal neurogenesis in the aged brain and under neurodegeneration. Molecules. 2022;27(17):5543.PubMedCrossRefPubMedCentral
242.
243.
go back to reference Keskin-Aktan A et al. SIRT2 and FOXO3a expressions in the cerebral cortex and hippocampus of young and aged male rats: antioxidant and anti-apoptotic effects of melatonin. Biologia Futura. 2022:1–15. Keskin-Aktan A et al. SIRT2 and FOXO3a expressions in the cerebral cortex and hippocampus of young and aged male rats: antioxidant and anti-apoptotic effects of melatonin. Biologia Futura. 2022:1–15.
244.
go back to reference Lemoine P, et al. Prolonged-release melatonin improves sleep quality and morning alertness in insomnia patients aged 55 years and older and has no withdrawal effects. J Sleep Res. 2007;16(4):372–80.PubMedCrossRef Lemoine P, et al. Prolonged-release melatonin improves sleep quality and morning alertness in insomnia patients aged 55 years and older and has no withdrawal effects. J Sleep Res. 2007;16(4):372–80.PubMedCrossRef
245.
go back to reference Fan Y, et al. The effect of melatonin on early postoperative cognitive decline in elderly patients undergoing hip arthroplasty: a randomized controlled trial. J Clin Anesth. 2017;39:77–81.PubMedCrossRef Fan Y, et al. The effect of melatonin on early postoperative cognitive decline in elderly patients undergoing hip arthroplasty: a randomized controlled trial. J Clin Anesth. 2017;39:77–81.PubMedCrossRef
246.
go back to reference Furio AM, Brusco LI, Cardinali DP. Possible therapeutic value of melatonin in mild cognitive impairment: a retrospective study. J Pineal Res. 2007;43(4):404–9.PubMedCrossRef Furio AM, Brusco LI, Cardinali DP. Possible therapeutic value of melatonin in mild cognitive impairment: a retrospective study. J Pineal Res. 2007;43(4):404–9.PubMedCrossRef
247.
go back to reference O’Neal-Moffitt G, et al. Prophylactic melatonin significantly reduces Alzheimer’s neuropathology and associated cognitive deficits independent of antioxidant pathways in AβPPswe/PS1 mice. Mol Neurodegener. 2015;10(1):1–21.CrossRef O’Neal-Moffitt G, et al. Prophylactic melatonin significantly reduces Alzheimer’s neuropathology and associated cognitive deficits independent of antioxidant pathways in AβPPswe/PS1 mice. Mol Neurodegener. 2015;10(1):1–21.CrossRef
248.
go back to reference Labban S, et al. Effects of melatonin and resveratrol on recognition memory and passive avoidance performance in a mouse model of Alzheimer’s disease. Behav Brain Res. 2021;402:113100.PubMedCrossRef Labban S, et al. Effects of melatonin and resveratrol on recognition memory and passive avoidance performance in a mouse model of Alzheimer’s disease. Behav Brain Res. 2021;402:113100.PubMedCrossRef
249.
go back to reference Nie L, et al. Melatonin ameliorates anxiety and depression-like behaviors and modulates proteomic changes in triple transgenic mice of Alzheimer’s disease. BioFactors. 2017;43(4):593–611.PubMedCrossRef Nie L, et al. Melatonin ameliorates anxiety and depression-like behaviors and modulates proteomic changes in triple transgenic mice of Alzheimer’s disease. BioFactors. 2017;43(4):593–611.PubMedCrossRef
250.
go back to reference Patki G, Lau Y-S. Melatonin protects against neurobehavioral and mitochondrial deficits in a chronic mouse model of Parkinson’s disease. Pharmacol Biochem Behav. 2011;99(4):704–11.PubMedCrossRefPubMedCentral Patki G, Lau Y-S. Melatonin protects against neurobehavioral and mitochondrial deficits in a chronic mouse model of Parkinson’s disease. Pharmacol Biochem Behav. 2011;99(4):704–11.PubMedCrossRefPubMedCentral
251.
go back to reference Antolı́n, I., et al., Protective effect of melatonin in a chronic experimental model of Parkinson’s disease. Brain research, 2002. 943(2): p. 163–173. Antolı́n, I., et al., Protective effect of melatonin in a chronic experimental model of Parkinson’s disease. Brain research, 2002. 943(2): p. 163–173.
252.
go back to reference Naskar A, et al. Melatonin enhances L-DOPA therapeutic effects, helps to reduce its dose, and protects dopaminergic neurons in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced Parkinsonism in mice. J Pineal Res. 2015;58(3):262–74.PubMedCrossRef Naskar A, et al. Melatonin enhances L-DOPA therapeutic effects, helps to reduce its dose, and protects dopaminergic neurons in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced Parkinsonism in mice. J Pineal Res. 2015;58(3):262–74.PubMedCrossRef
253.
go back to reference Jand Y, et al. Melatonin ameliorates disease severity in a mouse model of multiple sclerosis by modulating the kynurenine pathway. Sci Rep. 2022;12(1):15963.PubMedCrossRefPubMedCentral Jand Y, et al. Melatonin ameliorates disease severity in a mouse model of multiple sclerosis by modulating the kynurenine pathway. Sci Rep. 2022;12(1):15963.PubMedCrossRefPubMedCentral
254.
go back to reference Shen X, et al. Melatonin induces autophagy in amyotrophic lateral sclerosis mice via upregulation of SIRT1. Mol Neurobiol. 2022;59(8):4747–60.PubMedCrossRef Shen X, et al. Melatonin induces autophagy in amyotrophic lateral sclerosis mice via upregulation of SIRT1. Mol Neurobiol. 2022;59(8):4747–60.PubMedCrossRef
255.
go back to reference Tseng P-T, et al. The dose and duration-dependent association between melatonin treatment and overall cognition in Alzheimer’s dementia: a network meta-analysis of randomized placebo-controlled trials. Curr Neuropharmacol. 2022;20(10):1816.PubMedCrossRefPubMedCentral Tseng P-T, et al. The dose and duration-dependent association between melatonin treatment and overall cognition in Alzheimer’s dementia: a network meta-analysis of randomized placebo-controlled trials. Curr Neuropharmacol. 2022;20(10):1816.PubMedCrossRefPubMedCentral
256.
go back to reference Alagiakrishnan K. Melatonin based therapies for delirium and dementia. Discov Med. 2016;21(117):363–71.PubMed Alagiakrishnan K. Melatonin based therapies for delirium and dementia. Discov Med. 2016;21(117):363–71.PubMed
257.
go back to reference Nous A, et al. Serum daytime melatonin levels reflect cerebrospinal fluid melatonin levels in Alzheimer’s disease but are not correlated with cognitive decline. J Alzheimers Dis. 2021;83(2):693–704.PubMedCrossRefPubMedCentral Nous A, et al. Serum daytime melatonin levels reflect cerebrospinal fluid melatonin levels in Alzheimer’s disease but are not correlated with cognitive decline. J Alzheimers Dis. 2021;83(2):693–704.PubMedCrossRefPubMedCentral
258.
go back to reference Lin CH, Chiu CC, Lane HY. Trough Melatonin Levels Differ between Early and Late Phases of Alzheimer Disease. Clin Psychopharmacol Neurosci. 2021;19(1):135–44.PubMedCrossRefPubMedCentral Lin CH, Chiu CC, Lane HY. Trough Melatonin Levels Differ between Early and Late Phases of Alzheimer Disease. Clin Psychopharmacol Neurosci. 2021;19(1):135–44.PubMedCrossRefPubMedCentral
259.
go back to reference Obayashi K, et al. Physiological levels of melatonin relate to cognitive function and depressive symptoms: the HEIJO-KYO cohort. J Clin Endocrinol Metab. 2015;100(8):3090–6.PubMedCrossRef Obayashi K, et al. Physiological levels of melatonin relate to cognitive function and depressive symptoms: the HEIJO-KYO cohort. J Clin Endocrinol Metab. 2015;100(8):3090–6.PubMedCrossRef
260.
go back to reference Daneshvar Kakhaki R, et al. Melatonin supplementation and the effects on clinical and metabolic status in Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin Neurol Neurosurg. 2020;195:105878.PubMedCrossRef Daneshvar Kakhaki R, et al. Melatonin supplementation and the effects on clinical and metabolic status in Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin Neurol Neurosurg. 2020;195:105878.PubMedCrossRef
261.
go back to reference Jallouli S et al. Effect of melatonin intake on postural balance, functional mobility and fall risk in persons with multiple sclerosis: a pilot study. Int J Neurosci. 2022:1–11. Jallouli S et al. Effect of melatonin intake on postural balance, functional mobility and fall risk in persons with multiple sclerosis: a pilot study. Int J Neurosci. 2022:1–11.
262.
go back to reference Bald EM, Nance CS, Schultz JL. Melatonin may slow disease progression in amyotrophic lateral sclerosis: findings from the Pooled Resource Open-Access ALS Clinic Trials database. Muscle Nerve. 2021;63(4):572–6.PubMedCrossRefPubMedCentral Bald EM, Nance CS, Schultz JL. Melatonin may slow disease progression in amyotrophic lateral sclerosis: findings from the Pooled Resource Open-Access ALS Clinic Trials database. Muscle Nerve. 2021;63(4):572–6.PubMedCrossRefPubMedCentral
263.
go back to reference Boga JA, et al. Beneficial actions of melatonin in the management of viral infections: a new use for this “molecular handyman”? Rev Med Virol. 2012;22(5):323–38.PubMedCrossRefPubMedCentral Boga JA, et al. Beneficial actions of melatonin in the management of viral infections: a new use for this “molecular handyman”? Rev Med Virol. 2012;22(5):323–38.PubMedCrossRefPubMedCentral
264.
go back to reference Bonilla E, et al. Melatonin protects mice infected with Venezuelan equine encephalomyelitis virus. Cell Mol Life Sci CMLS. 1997;53:430–4.PubMedCrossRef Bonilla E, et al. Melatonin protects mice infected with Venezuelan equine encephalomyelitis virus. Cell Mol Life Sci CMLS. 1997;53:430–4.PubMedCrossRef
266.
go back to reference Shiu SY, et al. Urgent search for safe and effective treatments of severe acute respiratory syndrome: is melatonin a promising candidate drug? J Pineal Res. 2003;35(1):69.PubMedCrossRefPubMedCentral Shiu SY, et al. Urgent search for safe and effective treatments of severe acute respiratory syndrome: is melatonin a promising candidate drug? J Pineal Res. 2003;35(1):69.PubMedCrossRefPubMedCentral
267.
go back to reference Tan D-X, et al. Ebola virus disease: potential use of melatonin as a treatment. J Pineal Res. 2014;57(4):381–4.PubMedCrossRef Tan D-X, et al. Ebola virus disease: potential use of melatonin as a treatment. J Pineal Res. 2014;57(4):381–4.PubMedCrossRef
268.
go back to reference Yılmaz S, Öner P. Melatonin levels are low in COVID-19 positive patients and these levels are associated with depression, death anxiety and insomnia. Scand J Clin Lab Invest. 2023;83(2):86–94.PubMedCrossRef Yılmaz S, Öner P. Melatonin levels are low in COVID-19 positive patients and these levels are associated with depression, death anxiety and insomnia. Scand J Clin Lab Invest. 2023;83(2):86–94.PubMedCrossRef
269.
go back to reference Eroğlu İ, Eroğlu BÇ, Güven GS. Altered tryptophan absorption and metabolism could underlie long-term symptoms in survivors of coronavirus disease 2019 (COVID-19). Nutrition. 2021;90: 111308.PubMedCrossRefPubMedCentral Eroğlu İ, Eroğlu BÇ, Güven GS. Altered tryptophan absorption and metabolism could underlie long-term symptoms in survivors of coronavirus disease 2019 (COVID-19). Nutrition. 2021;90: 111308.PubMedCrossRefPubMedCentral
270.
go back to reference Farnoosh G, et al. Efficacy of a low dose of melatonin as an adjunctive therapy in hospitalized patients with COVID-19: a randomized, double-blind clinical trial. Arch Med Res. 2022;53(1):79–85.PubMedCrossRef Farnoosh G, et al. Efficacy of a low dose of melatonin as an adjunctive therapy in hospitalized patients with COVID-19: a randomized, double-blind clinical trial. Arch Med Res. 2022;53(1):79–85.PubMedCrossRef
271.
go back to reference Fogleman C, et al. A pilot of a randomized control trial of melatonin and vitamin C for mild-to-moderate COVID-19. J Am Board Fam Med. 2022;35(4):695–707.PubMedCrossRef Fogleman C, et al. A pilot of a randomized control trial of melatonin and vitamin C for mild-to-moderate COVID-19. J Am Board Fam Med. 2022;35(4):695–707.PubMedCrossRef
272.
go back to reference Ameri A, et al. Efficacy and safety of oral melatonin in patients with severe COVID-19: a randomized controlled trial. Inflammopharmacology. 2023;31(1):265–74.PubMedCrossRef Ameri A, et al. Efficacy and safety of oral melatonin in patients with severe COVID-19: a randomized controlled trial. Inflammopharmacology. 2023;31(1):265–74.PubMedCrossRef
273.
go back to reference Hasan ZT, Atrakji D, Mehuaiden DAK. The effect of melatonin on thrombosis, sepsis and mortality rate in COVID-19 patients. Int J Infect Dis. 2022;114:79–84.PubMedCrossRef Hasan ZT, Atrakji D, Mehuaiden DAK. The effect of melatonin on thrombosis, sepsis and mortality rate in COVID-19 patients. Int J Infect Dis. 2022;114:79–84.PubMedCrossRef
274.
go back to reference Darban M, et al. Efficacy of high dose vitamin C, melatonin and zinc in Iranian patients with acute respiratory syndrome due to coronavirus infection: a pilot randomized trial. J Cell Mol Anesth. 2021;6:164–7. Darban M, et al. Efficacy of high dose vitamin C, melatonin and zinc in Iranian patients with acute respiratory syndrome due to coronavirus infection: a pilot randomized trial. J Cell Mol Anesth. 2021;6:164–7.
275.
go back to reference Sánchez-Rico M, et al. Melatonin does not reduce mortality in adult hospitalized patients with COVID-19: a multicenter retrospective observational study. J Travel Med. 2022;29(3):taab195.PubMedCrossRef Sánchez-Rico M, et al. Melatonin does not reduce mortality in adult hospitalized patients with COVID-19: a multicenter retrospective observational study. J Travel Med. 2022;29(3):taab195.PubMedCrossRef
276.
go back to reference Sahu N, et al. Retrospective review of melatonin in patients with COVID-19. Chest. 2021;160(4):A560.CrossRef Sahu N, et al. Retrospective review of melatonin in patients with COVID-19. Chest. 2021;160(4):A560.CrossRef
277.
go back to reference Faridzadeh A et al. The role of melatonin as an adjuvant in the treatment of COVID-19: a systematic review. Heliyon. 2022. Faridzadeh A et al. The role of melatonin as an adjuvant in the treatment of COVID-19: a systematic review. Heliyon. 2022.
278.
go back to reference Cecon E, et al. Melatonin drugs inhibit SARS-CoV-2 entry into the brain and virus-induced damage of cerebral small vessels. Cell Mol Life Sci. 2022;79(7):361.PubMedCrossRefPubMedCentral Cecon E, et al. Melatonin drugs inhibit SARS-CoV-2 entry into the brain and virus-induced damage of cerebral small vessels. Cell Mol Life Sci. 2022;79(7):361.PubMedCrossRefPubMedCentral
279.
go back to reference Yadalam PK, et al. Assessing the therapeutic potential of agomelatine, ramelteon, and melatonin against SARS-CoV-2. Saudi J Biol Sci. 2022;29(5):3140–50.PubMedCrossRefPubMedCentral Yadalam PK, et al. Assessing the therapeutic potential of agomelatine, ramelteon, and melatonin against SARS-CoV-2. Saudi J Biol Sci. 2022;29(5):3140–50.PubMedCrossRefPubMedCentral
280.
go back to reference Cecon E, et al. Therapeutic potential of melatonin and melatonergic drugs on K18-hACE2 mice infected with SARS-CoV-2. J Pineal Res. 2022;72(1):e12772.PubMedCrossRef Cecon E, et al. Therapeutic potential of melatonin and melatonergic drugs on K18-hACE2 mice infected with SARS-CoV-2. J Pineal Res. 2022;72(1):e12772.PubMedCrossRef
281.
go back to reference Kumar P, et al. Severe glutathione deficiency, oxidative stress and oxidant damage in adults hospitalized with COVID-19: implications for GlyNAC (glycine and N-acetylcysteine) supplementation. Antioxidants. 2022;11(1):50.CrossRef Kumar P, et al. Severe glutathione deficiency, oxidative stress and oxidant damage in adults hospitalized with COVID-19: implications for GlyNAC (glycine and N-acetylcysteine) supplementation. Antioxidants. 2022;11(1):50.CrossRef
282.
go back to reference Muhammad Y, et al. Deficiency of antioxidants and increased oxidative stress in COVID-19 patients: a cross-sectional comparative study in Jigawa, Northwestern Nigeria. SAGE Open Med. 2021;9:2050312121991246.PubMedCrossRefPubMedCentral Muhammad Y, et al. Deficiency of antioxidants and increased oxidative stress in COVID-19 patients: a cross-sectional comparative study in Jigawa, Northwestern Nigeria. SAGE Open Med. 2021;9:2050312121991246.PubMedCrossRefPubMedCentral
283.
go back to reference Yang M, Lai CL. SARS-CoV-2 infection: can ferroptosis be a potential treatment target for multiple organ involvement? Cell Death Discovery. 2020;6(1):130.PubMedCrossRefPubMedCentral Yang M, Lai CL. SARS-CoV-2 infection: can ferroptosis be a potential treatment target for multiple organ involvement? Cell Death Discovery. 2020;6(1):130.PubMedCrossRefPubMedCentral
284.
go back to reference Cui Y, et al. ACSL4 exacerbates ischemic stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav Immun. 2021;93:312–21.PubMedCrossRef Cui Y, et al. ACSL4 exacerbates ischemic stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav Immun. 2021;93:312–21.PubMedCrossRef
285.
go back to reference Abulseoud OA, et al. Attenuated initial serum ferritin concentration in critically ill coronavirus disease 2019 geriatric patients with comorbid psychiatric conditions. Front Psychiatry. 2022;13:1035986.PubMedCrossRefPubMedCentral Abulseoud OA, et al. Attenuated initial serum ferritin concentration in critically ill coronavirus disease 2019 geriatric patients with comorbid psychiatric conditions. Front Psychiatry. 2022;13:1035986.PubMedCrossRefPubMedCentral
286.
go back to reference Hortová-Kohoutková M, et al. Hepcidin and ferritin levels as markers of immune cell activation during septic shock, severe COVID-19 and sterile inflammation. Front Immunol. 2023;14:1110540.PubMedCrossRefPubMedCentral Hortová-Kohoutková M, et al. Hepcidin and ferritin levels as markers of immune cell activation during septic shock, severe COVID-19 and sterile inflammation. Front Immunol. 2023;14:1110540.PubMedCrossRefPubMedCentral
287.
go back to reference Gaiatto ACM, et al. COVID-19 compromises iron homeostasis: Transferrin as a target of investigation. J Trace Elem Med Biol. 2023;76:127109.PubMedCrossRef Gaiatto ACM, et al. COVID-19 compromises iron homeostasis: Transferrin as a target of investigation. J Trace Elem Med Biol. 2023;76:127109.PubMedCrossRef
288.
go back to reference Claise C, et al. Low transferrin levels predict heightened inflammation in patients with COVID-19: New insights. Int J Infect Dis. 2022;116:74–9.PubMedCrossRef Claise C, et al. Low transferrin levels predict heightened inflammation in patients with COVID-19: New insights. Int J Infect Dis. 2022;116:74–9.PubMedCrossRef
289.
go back to reference Kaushal K, et al. Serum ferritin as a predictive biomarker in COVID-19. A systematic review, meta-analysis and meta-regression analysis. J Crit Care. 2022;67:172–81.PubMedCrossRef Kaushal K, et al. Serum ferritin as a predictive biomarker in COVID-19. A systematic review, meta-analysis and meta-regression analysis. J Crit Care. 2022;67:172–81.PubMedCrossRef
290.
go back to reference Zhou C, et al. Increased serum levels of hepcidin and ferritin are associated with severity of COVID-19. Med Sci Monitor. 2020;26:e926178–81.CrossRef Zhou C, et al. Increased serum levels of hepcidin and ferritin are associated with severity of COVID-19. Med Sci Monitor. 2020;26:e926178–81.CrossRef
291.
go back to reference Suriawinata E, Mehta KJ. Iron and iron-related proteins in COVID-19. Clin Exp Med. 2022:1–23. Suriawinata E, Mehta KJ. Iron and iron-related proteins in COVID-19. Clin Exp Med. 2022:1–23.
292.
go back to reference Kempuraj D, et al. COVID-19, mast cells, cytokine storm, psychological stress, and neuroinflammation. Neuroscientist. 2020;26(5–6):402–14.PubMedCrossRef Kempuraj D, et al. COVID-19, mast cells, cytokine storm, psychological stress, and neuroinflammation. Neuroscientist. 2020;26(5–6):402–14.PubMedCrossRef
294.
go back to reference Savla SR, Prabhavalkar KS, Bhatt LK. Cytokine storm associated coagulation complications in COVID-19 patients: pathogenesis and management. Expert Rev Anti Infect Ther. 2021;19(11):1397–413.PubMedCrossRef Savla SR, Prabhavalkar KS, Bhatt LK. Cytokine storm associated coagulation complications in COVID-19 patients: pathogenesis and management. Expert Rev Anti Infect Ther. 2021;19(11):1397–413.PubMedCrossRef
295.
go back to reference Lino K et al. Serum ferritin at admission in hospitalized COVID-19 patients as a predictor of mortality. Brazilian J Infect Dis. 2021;25. Lino K et al. Serum ferritin at admission in hospitalized COVID-19 patients as a predictor of mortality. Brazilian J Infect Dis. 2021;25.
296.
297.
go back to reference Almutairi MM, et al. Neuroinflammation and Its Impact on the Pathogenesis of COVID-19. Front Med. 2021;8:745789.CrossRef Almutairi MM, et al. Neuroinflammation and Its Impact on the Pathogenesis of COVID-19. Front Med. 2021;8:745789.CrossRef
300.
go back to reference Mu Q, et al. The role of iron homeostasis in remodeling immune function and regulating inflammatory disease. Science Bulletin. 2021;66(17):1806–16.PubMedCrossRef Mu Q, et al. The role of iron homeostasis in remodeling immune function and regulating inflammatory disease. Science Bulletin. 2021;66(17):1806–16.PubMedCrossRef
301.
go back to reference Rodriguez R, et al. Hepcidin induction by pathogens and pathogen-derived molecules is strongly dependent on interleukin-6. Infect Immun. 2014;82(2):745–52.PubMedCrossRefPubMedCentral Rodriguez R, et al. Hepcidin induction by pathogens and pathogen-derived molecules is strongly dependent on interleukin-6. Infect Immun. 2014;82(2):745–52.PubMedCrossRefPubMedCentral
302.
go back to reference Zhang F-L, et al. Impairment of hepcidin upregulation by lipopolysaccharide in the interleukin-6 knockout mouse brain. Front Mol Neurosci. 2017;10:367.PubMedCrossRefPubMedCentral Zhang F-L, et al. Impairment of hepcidin upregulation by lipopolysaccharide in the interleukin-6 knockout mouse brain. Front Mol Neurosci. 2017;10:367.PubMedCrossRefPubMedCentral
303.
go back to reference Urrutia P, et al. Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells. J Neurochem. 2013;126(4):541–9.PubMedCrossRef Urrutia P, et al. Inflammation alters the expression of DMT1, FPN1 and hepcidin, and it causes iron accumulation in central nervous system cells. J Neurochem. 2013;126(4):541–9.PubMedCrossRef
306.
go back to reference Kroner A, et al. TNF and increased intracellular iron alter macrophage polarization to a detrimental M1 phenotype in the injured spinal cord. Neuron. 2014;83(5):1098–116.PubMedCrossRef Kroner A, et al. TNF and increased intracellular iron alter macrophage polarization to a detrimental M1 phenotype in the injured spinal cord. Neuron. 2014;83(5):1098–116.PubMedCrossRef
307.
go back to reference Zhang X, et al. Cellular iron status influences the functional relationship between microglia and oligodendrocytes. Glia. 2006;54(8):795–804.PubMedCrossRef Zhang X, et al. Cellular iron status influences the functional relationship between microglia and oligodendrocytes. Glia. 2006;54(8):795–804.PubMedCrossRef
310.
go back to reference Wang M, et al. Revisiting the intersection of microglial activation and neuroinflammation in Alzheimer’s disease from the perspective of ferroptosis. Chem Biol Interact. 2023;375:110387.PubMedCrossRef Wang M, et al. Revisiting the intersection of microglial activation and neuroinflammation in Alzheimer’s disease from the perspective of ferroptosis. Chem Biol Interact. 2023;375:110387.PubMedCrossRef
312.
go back to reference Kapralov AA, et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat Chem Biol. 2020;16(3):278–90.PubMedCrossRefPubMedCentral Kapralov AA, et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat Chem Biol. 2020;16(3):278–90.PubMedCrossRefPubMedCentral
313.
go back to reference Ponnappan S, Ponnappan U. Aging and immune function: molecular mechanisms to interventions. 2011. Ponnappan S, Ponnappan U. Aging and immune function: molecular mechanisms to interventions. 2011.
314.
go back to reference Liguori I et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018:757–772. Liguori I et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018:757–772.
315.
go back to reference Xia S et al. An update on inflamm-aging: mechanisms, prevention, and treatment. J Immunol Res. 2016;2016. Xia S et al. An update on inflamm-aging: mechanisms, prevention, and treatment. J Immunol Res. 2016;2016.
317.
go back to reference Salimi S, Hamlyn JM. COVID-19 and crosstalk with the hallmarks of aging. J Gerontol Series A. 2020;75(9):e34–41.CrossRef Salimi S, Hamlyn JM. COVID-19 and crosstalk with the hallmarks of aging. J Gerontol Series A. 2020;75(9):e34–41.CrossRef
318.
go back to reference Oronsky B, et al. A review of persistent post-COVID syndrome (PPCS). Clin Rev Allergy Immunol. 2023;64(1):66–74.PubMedCrossRef Oronsky B, et al. A review of persistent post-COVID syndrome (PPCS). Clin Rev Allergy Immunol. 2023;64(1):66–74.PubMedCrossRef
319.
go back to reference Jing X, et al. Association between inflammatory cytokines and anti-SARS-CoV-2 antibodies in hospitalized patients with COVID-19. Immunity Ageing. 2022;19(1):12.PubMedCrossRefPubMedCentral Jing X, et al. Association between inflammatory cytokines and anti-SARS-CoV-2 antibodies in hospitalized patients with COVID-19. Immunity Ageing. 2022;19(1):12.PubMedCrossRefPubMedCentral
320.
go back to reference Normandin E, et al. Intrathecal inflammatory responses in the absence of SARS-CoV-2 nucleic acid in the CSF of COVID-19 hospitalized patients. J Neurol Sci. 2021;430:120023.PubMedCrossRefPubMedCentral Normandin E, et al. Intrathecal inflammatory responses in the absence of SARS-CoV-2 nucleic acid in the CSF of COVID-19 hospitalized patients. J Neurol Sci. 2021;430:120023.PubMedCrossRefPubMedCentral
322.
324.
go back to reference Bao WD, et al. Targeting miR-124/Ferroportin signaling ameliorated neuronal cell death through inhibiting apoptosis and ferroptosis in aged intracerebral hemorrhage murine model. Aging Cell. 2020;19(11):e13235.PubMedCrossRefPubMedCentral Bao WD, et al. Targeting miR-124/Ferroportin signaling ameliorated neuronal cell death through inhibiting apoptosis and ferroptosis in aged intracerebral hemorrhage murine model. Aging Cell. 2020;19(11):e13235.PubMedCrossRefPubMedCentral
326.
go back to reference Habib HM, et al. The role of iron in the pathogenesis of COVID-19 and possible treatment with lactoferrin and other iron chelators. Biomed Pharmacother. 2021;136:111228.PubMedCrossRefPubMedCentral Habib HM, et al. The role of iron in the pathogenesis of COVID-19 and possible treatment with lactoferrin and other iron chelators. Biomed Pharmacother. 2021;136:111228.PubMedCrossRefPubMedCentral
327.
go back to reference Sfera A, et al. Ferrosenescence: the iron age of neurodegeneration? Mech Ageing Dev. 2018;174:63–75.PubMedCrossRef Sfera A, et al. Ferrosenescence: the iron age of neurodegeneration? Mech Ageing Dev. 2018;174:63–75.PubMedCrossRef
328.
go back to reference Lara PC, Macías-Verde D, Burgos-Burgos J. Age-induced NLRP3 inflammasome over-activation increases lethality of SARS-CoV-2 pneumonia in elderly patients. Aging Dis. 2020;11(4):756.PubMedCrossRefPubMedCentral Lara PC, Macías-Verde D, Burgos-Burgos J. Age-induced NLRP3 inflammasome over-activation increases lethality of SARS-CoV-2 pneumonia in elderly patients. Aging Dis. 2020;11(4):756.PubMedCrossRefPubMedCentral
329.
330.
go back to reference Mezzanotte M, et al. Activation of the Hepcidin-Ferroportin1 pathway in the brain and astrocytic–neuronal crosstalk to counteract iron dyshomeostasis during aging. Sci Rep. 2022;12(1):11724.PubMedCrossRefPubMedCentral Mezzanotte M, et al. Activation of the Hepcidin-Ferroportin1 pathway in the brain and astrocytic–neuronal crosstalk to counteract iron dyshomeostasis during aging. Sci Rep. 2022;12(1):11724.PubMedCrossRefPubMedCentral
332.
go back to reference Cameron B, Landreth GE. Inflammation, microglia, and Alzheimer’s disease. Neurobiol Dis. 2010;37(3):503–9.PubMedCrossRef Cameron B, Landreth GE. Inflammation, microglia, and Alzheimer’s disease. Neurobiol Dis. 2010;37(3):503–9.PubMedCrossRef
334.
go back to reference Ayton S, et al. Brain iron is associated with accelerated cognitive decline in people with Alzheimer pathology. Mol Psychiatry. 2020;25(11):2932–41.PubMedCrossRef Ayton S, et al. Brain iron is associated with accelerated cognitive decline in people with Alzheimer pathology. Mol Psychiatry. 2020;25(11):2932–41.PubMedCrossRef
336.
go back to reference Zhang R et al. COVID-19-related brain injury: the potential role of ferroptosis. J Inflammation Res. 2022: 2181–2198. Zhang R et al. COVID-19-related brain injury: the potential role of ferroptosis. J Inflammation Res. 2022: 2181–2198.
337.
339.
go back to reference Tu H, et al. Insights into the novel function of system Xc-in regulated cell death. Eur Rev Med Pharmacol Sci. 2021;25(3):1650–62.PubMed Tu H, et al. Insights into the novel function of system Xc-in regulated cell death. Eur Rev Med Pharmacol Sci. 2021;25(3):1650–62.PubMed
340.
341.
go back to reference Chen W, et al. Ferritin reduction is essential for cerebral ischemia-induced hippocampal neuronal death through p53/SLC7A11-mediated ferroptosis. Brain Res. 2021;1752:147216.PubMedCrossRef Chen W, et al. Ferritin reduction is essential for cerebral ischemia-induced hippocampal neuronal death through p53/SLC7A11-mediated ferroptosis. Brain Res. 2021;1752:147216.PubMedCrossRef
342.
go back to reference Cozzi A, et al. Stem cell modeling of neuroferritinopathy reveals iron as a determinant of senescence and ferroptosis during neuronal aging. Stem Cell Reports. 2019;13(5):832–46.PubMedCrossRefPubMedCentral Cozzi A, et al. Stem cell modeling of neuroferritinopathy reveals iron as a determinant of senescence and ferroptosis during neuronal aging. Stem Cell Reports. 2019;13(5):832–46.PubMedCrossRefPubMedCentral
345.
go back to reference Han K, et al. Nrf2 knockout altered brain iron deposition and mitigated age-related motor dysfunction in aging mice. Free Radical Biol Med. 2021;162:592–602.CrossRef Han K, et al. Nrf2 knockout altered brain iron deposition and mitigated age-related motor dysfunction in aging mice. Free Radical Biol Med. 2021;162:592–602.CrossRef
347.
go back to reference Olagnier D, et al. SARS-CoV2-mediated suppression of NRF2-signaling reveals potent antiviral and anti-inflammatory activity of 4-octyl-itaconate and dimethyl fumarate. Nat Commun. 2020;11(1):4938.PubMedCrossRefPubMedCentral Olagnier D, et al. SARS-CoV2-mediated suppression of NRF2-signaling reveals potent antiviral and anti-inflammatory activity of 4-octyl-itaconate and dimethyl fumarate. Nat Commun. 2020;11(1):4938.PubMedCrossRefPubMedCentral
348.
go back to reference Kyriakopoulos AM, et al. Mitogen Activated Protein Kinase (MAPK) Activation, p53, and Autophagy Inhibition Characterize the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Protein Induced Neurotoxicity. Cureus. 2022;14(12):e32361.PubMedPubMedCentral Kyriakopoulos AM, et al. Mitogen Activated Protein Kinase (MAPK) Activation, p53, and Autophagy Inhibition Characterize the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Protein Induced Neurotoxicity. Cureus. 2022;14(12):e32361.PubMedPubMedCentral
349.
go back to reference Zhang Y et al. SARS-COV-2 spike protein promotes RPE cell senescence via the ROS/P53/P21 pathway. Biogerontology. 2023. Zhang Y et al. SARS-COV-2 spike protein promotes RPE cell senescence via the ROS/P53/P21 pathway. Biogerontology. 2023.
350.
go back to reference Cecchini R, Cecchini AL. SARS-CoV-2 infection pathogenesis is related to oxidative stress as a response to aggression. Med Hypotheses. 2020;143:110102.PubMedCrossRefPubMedCentral Cecchini R, Cecchini AL. SARS-CoV-2 infection pathogenesis is related to oxidative stress as a response to aggression. Med Hypotheses. 2020;143:110102.PubMedCrossRefPubMedCentral
351.
go back to reference Lovell MA, et al. Decreased thioredoxin and increased thioredoxin reductase levels in Alzheimer’s disease brain. Free Radical Biol Med. 2000;28(3):418–27.CrossRef Lovell MA, et al. Decreased thioredoxin and increased thioredoxin reductase levels in Alzheimer’s disease brain. Free Radical Biol Med. 2000;28(3):418–27.CrossRef
352.
go back to reference Chiang GC, et al. Relationships among cortical glutathione levels, brain amyloidosis, and memory in healthy older adults investigated in vivo with 1H-MRS and Pittsburgh compound-B PET. Am J Neuroradiol. 2017;38(6):1130–7.PubMedCrossRefPubMedCentral Chiang GC, et al. Relationships among cortical glutathione levels, brain amyloidosis, and memory in healthy older adults investigated in vivo with 1H-MRS and Pittsburgh compound-B PET. Am J Neuroradiol. 2017;38(6):1130–7.PubMedCrossRefPubMedCentral
353.
go back to reference Chen L, et al. Enhanced defense against ferroptosis ameliorates cognitive impairment and reduces neurodegeneration in 5xFAD mice. Free Radical Biol Med. 2022;180:1–12.CrossRef Chen L, et al. Enhanced defense against ferroptosis ameliorates cognitive impairment and reduces neurodegeneration in 5xFAD mice. Free Radical Biol Med. 2022;180:1–12.CrossRef
354.
go back to reference Labandeira-Garcia JL, et al. Brain renin-angiotensin system and microglial polarization: implications for aging and neurodegeneration. Front Aging Neurosci. 2017;9:129.PubMedCrossRefPubMedCentral Labandeira-Garcia JL, et al. Brain renin-angiotensin system and microglial polarization: implications for aging and neurodegeneration. Front Aging Neurosci. 2017;9:129.PubMedCrossRefPubMedCentral
355.
go back to reference Dang R, et al. Activation of angiotensin-converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling. Aging Cell. 2021;20(10):e13480.PubMedCrossRefPubMedCentral Dang R, et al. Activation of angiotensin-converting enzyme 2/angiotensin (1–7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling. Aging Cell. 2021;20(10):e13480.PubMedCrossRefPubMedCentral
356.
go back to reference Min L-J, et al. Signaling mechanisms of angiotensin II in regulating vascular senescence. Ageing Res Rev. 2009;8(2):113–21.PubMedCrossRef Min L-J, et al. Signaling mechanisms of angiotensin II in regulating vascular senescence. Ageing Res Rev. 2009;8(2):113–21.PubMedCrossRef
357.
go back to reference Liu C, et al. Angiotensin II-induced vascular endothelial cells ferroptosis via P53-ALOX12 signal axis. Clin Exp Hypertens. 2023;45(1):2180019.PubMedCrossRef Liu C, et al. Angiotensin II-induced vascular endothelial cells ferroptosis via P53-ALOX12 signal axis. Clin Exp Hypertens. 2023;45(1):2180019.PubMedCrossRef
358.
go back to reference Benicky J, et al. Anti-Inflammatory Effects of Angiotensin Receptor Blockers in the Brain and the Periphery. Cell Mol Neurobiol. 2009;29(6):781–92.PubMedCrossRef Benicky J, et al. Anti-Inflammatory Effects of Angiotensin Receptor Blockers in the Brain and the Periphery. Cell Mol Neurobiol. 2009;29(6):781–92.PubMedCrossRef
359.
go back to reference Basso N, et al. Protective effect of the inhibition of the renin–angiotensin system on aging. Regul Pept. 2005;128(3):247–52.PubMedCrossRef Basso N, et al. Protective effect of the inhibition of the renin–angiotensin system on aging. Regul Pept. 2005;128(3):247–52.PubMedCrossRef
360.
go back to reference Garrido-Gil P, et al. Brain angiotensin regulates iron homeostasis in dopaminergic neurons and microglial cells. Exp Neurol. 2013;250:384–96.PubMedCrossRef Garrido-Gil P, et al. Brain angiotensin regulates iron homeostasis in dopaminergic neurons and microglial cells. Exp Neurol. 2013;250:384–96.PubMedCrossRef
361.
go back to reference Feng X, Wang L, Li Y. Change of telomere length in angiotensin II-induced human glomerular mesangial cell senescence and the protective role of losartan. Mol Med Rep. 2011;4(2):255–60.PubMed Feng X, Wang L, Li Y. Change of telomere length in angiotensin II-induced human glomerular mesangial cell senescence and the protective role of losartan. Mol Med Rep. 2011;4(2):255–60.PubMed
362.
go back to reference Duan R, et al. ACE2 activator diminazene aceturate ameliorates Alzheimer’s disease-like neuropathology and rescues cognitive impairment in SAMP8 mice. Aging (Albany NY). 2020;12(14):14819–29.PubMedCrossRef Duan R, et al. ACE2 activator diminazene aceturate ameliorates Alzheimer’s disease-like neuropathology and rescues cognitive impairment in SAMP8 mice. Aging (Albany NY). 2020;12(14):14819–29.PubMedCrossRef
363.
go back to reference Rocha NP, et al. Cerebrospinal fluid levels of angiotensin-converting enzyme are associated with amyloid-β 42 burden in Alzheimer’s disease. J Alzheimers Dis. 2018;64(4):1085–90.PubMedCrossRef Rocha NP, et al. Cerebrospinal fluid levels of angiotensin-converting enzyme are associated with amyloid-β 42 burden in Alzheimer’s disease. J Alzheimers Dis. 2018;64(4):1085–90.PubMedCrossRef
364.
go back to reference Evans CE, et al. ACE2 activation protects against cognitive decline and reduces amyloid pathology in the Tg2576 mouse model of Alzheimer’s disease. Acta Neuropathol. 2020;139(3):485–502.PubMedCrossRefPubMedCentral Evans CE, et al. ACE2 activation protects against cognitive decline and reduces amyloid pathology in the Tg2576 mouse model of Alzheimer’s disease. Acta Neuropathol. 2020;139(3):485–502.PubMedCrossRefPubMedCentral
365.
go back to reference Kehoe PG, et al. Angiotensin-converting enzyme 2 is reduced in Alzheimer’s disease in association with increasing amyloid-β and tau pathology. Alzheimer’s Res Ther. 2016;8(1):50.CrossRef Kehoe PG, et al. Angiotensin-converting enzyme 2 is reduced in Alzheimer’s disease in association with increasing amyloid-β and tau pathology. Alzheimer’s Res Ther. 2016;8(1):50.CrossRef
366.
go back to reference Wang X-L, et al. Deficiency of angiotensin-converting enzyme 2 causes deterioration of cognitive function. npj Aging Mechanisms Dis. 2016;2(1):16024.CrossRef Wang X-L, et al. Deficiency of angiotensin-converting enzyme 2 causes deterioration of cognitive function. npj Aging Mechanisms Dis. 2016;2(1):16024.CrossRef
368.
go back to reference Bakhtazad A, et al. Paying attention to circadian rhythms in the treatment of COVID-19. Basic Clin Neurosci J. 2020;11(2):233–46.CrossRef Bakhtazad A, et al. Paying attention to circadian rhythms in the treatment of COVID-19. Basic Clin Neurosci J. 2020;11(2):233–46.CrossRef
369.
go back to reference Tomar R, Raghav A. Association of circadian clock and severe acute respiratory syndrome coronavirus 2 infection. Chronobiol Med. 2021;3(2):60–3.CrossRef Tomar R, Raghav A. Association of circadian clock and severe acute respiratory syndrome coronavirus 2 infection. Chronobiol Med. 2021;3(2):60–3.CrossRef
370.
go back to reference Zhuang X, et al. The circadian clock component BMAL1 regulates SARS-CoV-2 entry and replication in lung epithelial cells. IScience. 2021;24(10):103144.PubMedCrossRefPubMedCentral Zhuang X, et al. The circadian clock component BMAL1 regulates SARS-CoV-2 entry and replication in lung epithelial cells. IScience. 2021;24(10):103144.PubMedCrossRefPubMedCentral
372.
go back to reference Borrmann H, McKeating JA, Zhuang X. The circadian clock and viral infections. J Biol Rhythms. 2021;36(1):9–22.PubMedCrossRef Borrmann H, McKeating JA, Zhuang X. The circadian clock and viral infections. J Biol Rhythms. 2021;36(1):9–22.PubMedCrossRef
373.
375.
go back to reference Bitto A, et al. p62/SQSTM1 at the interface of aging, autophagy, and disease. Age. 2014;36(3):1123–37.CrossRef Bitto A, et al. p62/SQSTM1 at the interface of aging, autophagy, and disease. Age. 2014;36(3):1123–37.CrossRef
376.
377.
go back to reference Liu J et al. Iron accumulation with age alters metabolic pattern and circadian clock gene expression through the reduction of AMP-modulated histone methylation. J Biol Chem. 2022;298(6). Liu J et al. Iron accumulation with age alters metabolic pattern and circadian clock gene expression through the reduction of AMP-modulated histone methylation. J Biol Chem. 2022;298(6).
379.
go back to reference Amidfar M, Garcez ML, Kim Y-K. The shared molecular mechanisms underlying aging of the brain, major depressive disorder, and Alzheimer’s disease: The role of circadian rhythm disturbances. Prog Neuropsychopharmacol Biol Psychiatry. 2023;123: 110721.PubMedCrossRef Amidfar M, Garcez ML, Kim Y-K. The shared molecular mechanisms underlying aging of the brain, major depressive disorder, and Alzheimer’s disease: The role of circadian rhythm disturbances. Prog Neuropsychopharmacol Biol Psychiatry. 2023;123: 110721.PubMedCrossRef
380.
go back to reference Kondratov RV, et al. Early aging and age-related pathologies in mice deficient in BMAL1, the core componentof the circadian clock. Genes Dev. 2006;20(14):1868–73.PubMedCrossRefPubMedCentral Kondratov RV, et al. Early aging and age-related pathologies in mice deficient in BMAL1, the core componentof the circadian clock. Genes Dev. 2006;20(14):1868–73.PubMedCrossRefPubMedCentral
381.
go back to reference Ali AA, et al. Premature aging of the hippocampal neurogenic niche in adult Bmal1-deficient mice. Aging (Albany NY). 2015;7(6):435.PubMedCrossRef Ali AA, et al. Premature aging of the hippocampal neurogenic niche in adult Bmal1-deficient mice. Aging (Albany NY). 2015;7(6):435.PubMedCrossRef
382.
go back to reference Musiek ES. Circadian clock disruption in neurodegenerative diseases: cause and effect? Front Pharmacol. 2015;6. Musiek ES. Circadian clock disruption in neurodegenerative diseases: cause and effect? Front Pharmacol. 2015;6.
383.
go back to reference Liu W-W, et al. BMAL1 regulation of microglia-mediated neuroinflammation in MPTP-induced Parkinson’s disease mouse model. FASEB J. 2020;34(5):6570–81.PubMedCrossRef Liu W-W, et al. BMAL1 regulation of microglia-mediated neuroinflammation in MPTP-induced Parkinson’s disease mouse model. FASEB J. 2020;34(5):6570–81.PubMedCrossRef
384.
go back to reference Ni J, et al. An impaired intrinsic microglial clock system induces neuroinflammatory alterations in the early stage of amyloid precursor protein knock-in mouse brain. J Neuroinflammation. 2019;16(1):173.PubMedCrossRefPubMedCentral Ni J, et al. An impaired intrinsic microglial clock system induces neuroinflammatory alterations in the early stage of amyloid precursor protein knock-in mouse brain. J Neuroinflammation. 2019;16(1):173.PubMedCrossRefPubMedCentral
386.
go back to reference Hu L, et al. Melatonin decreases M1 polarization via attenuating mitochondrial oxidative damage depending on UCP2 pathway in prorenin-treated microglia. PLoS ONE. 2019;14(2):e0212138.PubMedCrossRefPubMedCentral Hu L, et al. Melatonin decreases M1 polarization via attenuating mitochondrial oxidative damage depending on UCP2 pathway in prorenin-treated microglia. PLoS ONE. 2019;14(2):e0212138.PubMedCrossRefPubMedCentral
387.
go back to reference Zhou Q, et al. Melatonin reduces neuroinflammation and improves axonal hypomyelination by modulating M1/M2 microglia polarization via JAK2-STAT3-telomerase pathway in postnatal rats exposed to lipopolysaccharide. Mol Neurobiol. 2021;58(12):6552–76.PubMedCrossRefPubMedCentral Zhou Q, et al. Melatonin reduces neuroinflammation and improves axonal hypomyelination by modulating M1/M2 microglia polarization via JAK2-STAT3-telomerase pathway in postnatal rats exposed to lipopolysaccharide. Mol Neurobiol. 2021;58(12):6552–76.PubMedCrossRefPubMedCentral
389.
go back to reference Li J, et al. Melatonin ameliorates Parkinson’s disease via regulating microglia polarization in a RORα-depend4ent pathway. npj Parkinson’s Dis. 2022;8(1):90.CrossRef Li J, et al. Melatonin ameliorates Parkinson’s disease via regulating microglia polarization in a RORα-depend4ent pathway. npj Parkinson’s Dis. 2022;8(1):90.CrossRef
390.
go back to reference Ali T, et al. Melatonin attenuates D-galactose-induced memory impairment, neuroinflammation and neurodegeneration via RAGE/NF-KB/JNK signaling pathway in aging mouse model. J Pineal Res. 2015;58(1):71–85.PubMedCrossRef Ali T, et al. Melatonin attenuates D-galactose-induced memory impairment, neuroinflammation and neurodegeneration via RAGE/NF-KB/JNK signaling pathway in aging mouse model. J Pineal Res. 2015;58(1):71–85.PubMedCrossRef
391.
go back to reference Dou X et al. Therapeutic potential of melatonin in the intervertebral disc degeneration through inhibiting the ferroptosis of nucleus pulpous cells. J Cell Mol Med. 2023. Dou X et al. Therapeutic potential of melatonin in the intervertebral disc degeneration through inhibiting the ferroptosis of nucleus pulpous cells. J Cell Mol Med. 2023.
392.
go back to reference Qu W, et al. Targeting iNOS alleviates early brain injury after experimental subarachnoid hemorrhage via promoting ferroptosis of M1 microglia and reducing neuroinflammation. Mol Neurobiol. 2022;59(5):3124–39.PubMedCrossRef Qu W, et al. Targeting iNOS alleviates early brain injury after experimental subarachnoid hemorrhage via promoting ferroptosis of M1 microglia and reducing neuroinflammation. Mol Neurobiol. 2022;59(5):3124–39.PubMedCrossRef
393.
go back to reference Gulcin İ, Buyukokuroglu ME, Kufrevioglu OI. Metal chelating and hydrogen peroxide scavenging effects of melatonin. J Pineal Res. 2003;34(4):278–81.PubMedCrossRef Gulcin İ, Buyukokuroglu ME, Kufrevioglu OI. Metal chelating and hydrogen peroxide scavenging effects of melatonin. J Pineal Res. 2003;34(4):278–81.PubMedCrossRef
394.
go back to reference Limson J, Nyokong T, Daya S. The interaction of melatonin and its precursors with aluminium, cadmium, copper, iron, lead, and zinc: an adsorptive voltammetric study. J Pineal Res. 1998;24(1):15–21.PubMedCrossRef Limson J, Nyokong T, Daya S. The interaction of melatonin and its precursors with aluminium, cadmium, copper, iron, lead, and zinc: an adsorptive voltammetric study. J Pineal Res. 1998;24(1):15–21.PubMedCrossRef
395.
go back to reference Romero A, et al. A review of metal-catalyzed molecular damage: protection by melatonin. J Pineal Res. 2014;56(4):343–70.PubMedCrossRef Romero A, et al. A review of metal-catalyzed molecular damage: protection by melatonin. J Pineal Res. 2014;56(4):343–70.PubMedCrossRef
396.
go back to reference Hayter CL, Bishop GM, Robinson SR. Pharmacological but not physiological concentrations of melatonin reduce iron-induced neuronal death in rat cerebral cortex. Neurosci Lett. 2004;362(3):182–4.PubMedCrossRef Hayter CL, Bishop GM, Robinson SR. Pharmacological but not physiological concentrations of melatonin reduce iron-induced neuronal death in rat cerebral cortex. Neurosci Lett. 2004;362(3):182–4.PubMedCrossRef
397.
go back to reference Maharaj DS, et al. Melatonin and 6-hydroxymelatonin protect against iron-induced neurotoxicity. J Neurochem. 2006;96(1):78–81.PubMedCrossRef Maharaj DS, et al. Melatonin and 6-hydroxymelatonin protect against iron-induced neurotoxicity. J Neurochem. 2006;96(1):78–81.PubMedCrossRef
398.
go back to reference Kaptanoglu E, et al. Different responsiveness of central nervous system tissues to oxidative conditions and to the antioxidant effect of melatonin. J Pineal Res. 2003;34(1):32–5.PubMedCrossRef Kaptanoglu E, et al. Different responsiveness of central nervous system tissues to oxidative conditions and to the antioxidant effect of melatonin. J Pineal Res. 2003;34(1):32–5.PubMedCrossRef
399.
go back to reference Lin AM-Y, Ho L-T. Melatonin suppresses iron-induced neurodegeneration in rat brain. Free Radical Biol Med. 2000;28(6):904–11.CrossRef Lin AM-Y, Ho L-T. Melatonin suppresses iron-induced neurodegeneration in rat brain. Free Radical Biol Med. 2000;28(6):904–11.CrossRef
400.
go back to reference Ortega-Gutiérrez S, et al. Melatonin improves deferoxamine antioxidant activity in protecting against lipid peroxidation caused by hydrogen peroxide in rat brain homogenates. Neurosci Lett. 2002;323(1):55–9.PubMedCrossRef Ortega-Gutiérrez S, et al. Melatonin improves deferoxamine antioxidant activity in protecting against lipid peroxidation caused by hydrogen peroxide in rat brain homogenates. Neurosci Lett. 2002;323(1):55–9.PubMedCrossRef
401.
go back to reference Mi Y, et al. Melatonin inhibits ferroptosis and delays age-related cataract by regulating SIRT6/p-Nrf2/GPX4 and SIRT6/NCOA4/FTH1 pathways. Biomed Pharmacother. 2023;157:114048.PubMedCrossRef Mi Y, et al. Melatonin inhibits ferroptosis and delays age-related cataract by regulating SIRT6/p-Nrf2/GPX4 and SIRT6/NCOA4/FTH1 pathways. Biomed Pharmacother. 2023;157:114048.PubMedCrossRef
402.
go back to reference Gao Y, et al. Melatonin ameliorates neurological deficits through MT2/IL-33/ferritin H signaling-mediated inhibition of neuroinflammation and ferroptosis after traumatic brain injury. Free Radical Biol Med. 2023;199:97–112.CrossRef Gao Y, et al. Melatonin ameliorates neurological deficits through MT2/IL-33/ferritin H signaling-mediated inhibition of neuroinflammation and ferroptosis after traumatic brain injury. Free Radical Biol Med. 2023;199:97–112.CrossRef
403.
go back to reference Wang X, et al. Melatonin alleviates acute sleep deprivation-induced memory loss in mice by suppressing hippocampal ferroptosis. Front Pharmacol. 2021;12:708645.PubMedCrossRefPubMedCentral Wang X, et al. Melatonin alleviates acute sleep deprivation-induced memory loss in mice by suppressing hippocampal ferroptosis. Front Pharmacol. 2021;12:708645.PubMedCrossRefPubMedCentral
404.
go back to reference Yang J, Tang Q, Zeng Y. Melatonin: potential avenue for treating iron overload disorders. Ageing Res Rev. 2022;81:101717.PubMedCrossRef Yang J, Tang Q, Zeng Y. Melatonin: potential avenue for treating iron overload disorders. Ageing Res Rev. 2022;81:101717.PubMedCrossRef
405.
go back to reference Rodriguez C, et al. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res. 2004;36(1):1–9.PubMedCrossRef Rodriguez C, et al. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res. 2004;36(1):1–9.PubMedCrossRef
406.
go back to reference Reiter RJ, et al. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016;61(3):253–78.PubMedCrossRef Reiter RJ, et al. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016;61(3):253–78.PubMedCrossRef
407.
go back to reference Ahmadi Z, Ashrafizadeh M. Melatonin as a potential modulator of Nrf2. Fundam Clin Pharmacol. 2020;34(1):11–9.PubMedCrossRef Ahmadi Z, Ashrafizadeh M. Melatonin as a potential modulator of Nrf2. Fundam Clin Pharmacol. 2020;34(1):11–9.PubMedCrossRef
408.
go back to reference Ma S-J, et al. Melatonin alleviates early brain injury by inhibiting the NRF2-mediated ferroptosis pathway after subarachnoid hemorrhage. Free Radical Biol Med. 2023;208:555–70.CrossRef Ma S-J, et al. Melatonin alleviates early brain injury by inhibiting the NRF2-mediated ferroptosis pathway after subarachnoid hemorrhage. Free Radical Biol Med. 2023;208:555–70.CrossRef
409.
go back to reference Ren C, et al. Melatonin reduces radiation-induced ferroptosis in hippocampal neurons by activating the PKM2/NRF2/GPX4 signaling pathway. Prog Neuropsychopharmacol Biol Psychiatry. 2023;126:110777.PubMedCrossRef Ren C, et al. Melatonin reduces radiation-induced ferroptosis in hippocampal neurons by activating the PKM2/NRF2/GPX4 signaling pathway. Prog Neuropsychopharmacol Biol Psychiatry. 2023;126:110777.PubMedCrossRef
410.
go back to reference Gou Z, et al. Melatonin improves hypoxic-ischemic brain damage through the Akt/Nrf2/Gpx4 signaling pathway. Brain Res Bull. 2020;163:40–8.PubMedCrossRef Gou Z, et al. Melatonin improves hypoxic-ischemic brain damage through the Akt/Nrf2/Gpx4 signaling pathway. Brain Res Bull. 2020;163:40–8.PubMedCrossRef
411.
go back to reference Huang Y-B et al. Melatonin alleviates acute kidney injury by inhibiting NRF2/Slc7a11 axis-mediated ferroptosis. Oxidative Med Cell Longevity. 2022;2022. Huang Y-B et al. Melatonin alleviates acute kidney injury by inhibiting NRF2/Slc7a11 axis-mediated ferroptosis. Oxidative Med Cell Longevity. 2022;2022.
412.
go back to reference Guohua F, et al. Melatonin protects against PM2. 5-induced lung injury by inhibiting ferroptosis of lung epithelial cells in a Nrf2-dependent manner. Ecotoxicol Environ Safety. 2021;223:112588.PubMedCrossRef Guohua F, et al. Melatonin protects against PM2. 5-induced lung injury by inhibiting ferroptosis of lung epithelial cells in a Nrf2-dependent manner. Ecotoxicol Environ Safety. 2021;223:112588.PubMedCrossRef
413.
go back to reference Ma H et al. Melatonin suppresses ferroptosis induced by high glucose via activation of the Nrf2/HO-1 signaling pathway in type 2 diabetic osteoporosis. Oxidative Med Cell Longevity. 2020;2020. Ma H et al. Melatonin suppresses ferroptosis induced by high glucose via activation of the Nrf2/HO-1 signaling pathway in type 2 diabetic osteoporosis. Oxidative Med Cell Longevity. 2020;2020.
414.
415.
go back to reference Li N et al. PM2. 5 contributed to pulmonary epithelial senescence and ferroptosis by regulating USP3-SIRT3-P53 axis. Free Radical Biol Med. 2023. Li N et al. PM2. 5 contributed to pulmonary epithelial senescence and ferroptosis by regulating USP3-SIRT3-P53 axis. Free Radical Biol Med. 2023.
416.
go back to reference Li W, et al. Exogenous melatonin ameliorates steroid-induced osteonecrosis of the femoral head by modulating ferroptosis through GDF15-mediated signaling. Stem Cell Res Ther. 2023;14(1):171.PubMedCrossRefPubMedCentral Li W, et al. Exogenous melatonin ameliorates steroid-induced osteonecrosis of the femoral head by modulating ferroptosis through GDF15-mediated signaling. Stem Cell Res Ther. 2023;14(1):171.PubMedCrossRefPubMedCentral
417.
go back to reference Sun X, et al. Melatonin alleviates doxorubicin-induced mitochondrial oxidative damage and ferroptosis in cardiomyocytes by regulating YAP expression. Toxicol Appl Pharmacol. 2022;437:115902.PubMedCrossRef Sun X, et al. Melatonin alleviates doxorubicin-induced mitochondrial oxidative damage and ferroptosis in cardiomyocytes by regulating YAP expression. Toxicol Appl Pharmacol. 2022;437:115902.PubMedCrossRef
418.
go back to reference Wu C, et al. A novel mechanism linking ferroptosis and endoplasmic reticulum stress via the circPtpn14/miR-351-5p/5-LOX signaling in melatonin-mediated treatment of traumatic brain injury. Free Radical Biol Med. 2022;178:271–94.CrossRef Wu C, et al. A novel mechanism linking ferroptosis and endoplasmic reticulum stress via the circPtpn14/miR-351-5p/5-LOX signaling in melatonin-mediated treatment of traumatic brain injury. Free Radical Biol Med. 2022;178:271–94.CrossRef
419.
go back to reference Li H, et al. Alterations in the time course of expression of the Nox family in the brain in a rat experimental cerebral ischemia and reperfusion model: effects of melatonin. J Pineal Res. 2014;57(1):110–9.PubMedCrossRef Li H, et al. Alterations in the time course of expression of the Nox family in the brain in a rat experimental cerebral ischemia and reperfusion model: effects of melatonin. J Pineal Res. 2014;57(1):110–9.PubMedCrossRef
420.
go back to reference Patiño P, et al. Melatonin protects against oxygen and glucose deprivation by decreasing extracellular glutamate and Nox-derived ROS in rat hippocampal slices. Neurotoxicology. 2016;57:61–8.PubMedCrossRef Patiño P, et al. Melatonin protects against oxygen and glucose deprivation by decreasing extracellular glutamate and Nox-derived ROS in rat hippocampal slices. Neurotoxicology. 2016;57:61–8.PubMedCrossRef
421.
go back to reference Jumnongprakhon P, et al. Inhibitory effect of melatonin on cerebral endothelial cells dysfunction induced by methamphetamine via NADPH oxidase-2. Brain Res. 2016;1650:84–92.PubMedCrossRef Jumnongprakhon P, et al. Inhibitory effect of melatonin on cerebral endothelial cells dysfunction induced by methamphetamine via NADPH oxidase-2. Brain Res. 2016;1650:84–92.PubMedCrossRef
422.
go back to reference Najafi M, et al. Melatonin modulates regulation of NOX2 and NOX4 following irradiation in the lung. Curr Clin Pharmacol. 2019;14(3):224–31.PubMedCrossRef Najafi M, et al. Melatonin modulates regulation of NOX2 and NOX4 following irradiation in the lung. Curr Clin Pharmacol. 2019;14(3):224–31.PubMedCrossRef
423.
go back to reference Yılmaz S, Öner P. Melatonin levels are low in COVID-19 positive patients and these levels are associated with depression, death anxiety and insomnia. Scandinavian J Clin Lab Investig. 2023: 1–9. Yılmaz S, Öner P. Melatonin levels are low in COVID-19 positive patients and these levels are associated with depression, death anxiety and insomnia. Scandinavian J Clin Lab Investig. 2023: 1–9.
424.
go back to reference Li S, et al. Ferrostatin-1 alleviates angiotensin II (Ang II)-induced inflammation and ferroptosis in astrocytes. Int Immunopharmacol. 2021;90:107179.PubMedCrossRef Li S, et al. Ferrostatin-1 alleviates angiotensin II (Ang II)-induced inflammation and ferroptosis in astrocytes. Int Immunopharmacol. 2021;90:107179.PubMedCrossRef
425.
go back to reference Liu C et al. Paeonol improves angiotensin II-induced cardiac hypertrophy by suppressing ferroptosis. Heliyon. 2023;9(9). Liu C et al. Paeonol improves angiotensin II-induced cardiac hypertrophy by suppressing ferroptosis. Heliyon. 2023;9(9).
426.
go back to reference Ji ZZ, Xu YC. Melatonin protects podocytes from angiotensin II-induced injury in an in vitro diabetic nephropathy model. Mol Med Rep. 2016;14(1):920–6.PubMedCrossRef Ji ZZ, Xu YC. Melatonin protects podocytes from angiotensin II-induced injury in an in vitro diabetic nephropathy model. Mol Med Rep. 2016;14(1):920–6.PubMedCrossRef
427.
go back to reference Su H, et al. Melatonin attenuates angiotensin II-induced cardiomyocyte hypertrophy through the CyPA/CD147 signaling pathway. Mol Cell Biochem. 2016;422:85–95.PubMedCrossRef Su H, et al. Melatonin attenuates angiotensin II-induced cardiomyocyte hypertrophy through the CyPA/CD147 signaling pathway. Mol Cell Biochem. 2016;422:85–95.PubMedCrossRef
428.
go back to reference Kong J, et al. Melatonin attenuates angiotensin II-induced abdominal aortic aneurysm through the down-regulation of matrix metalloproteinases. Oncotarget. 2017;8(9):14283.PubMedCrossRefPubMedCentral Kong J, et al. Melatonin attenuates angiotensin II-induced abdominal aortic aneurysm through the down-regulation of matrix metalloproteinases. Oncotarget. 2017;8(9):14283.PubMedCrossRefPubMedCentral
429.
go back to reference Yang Y, et al. Melatonin alleviates angiotensin-II-induced cardiac hypertrophy via activating MICU1 pathway. Aging (Albany NY). 2021;13(1):493.CrossRef Yang Y, et al. Melatonin alleviates angiotensin-II-induced cardiac hypertrophy via activating MICU1 pathway. Aging (Albany NY). 2021;13(1):493.CrossRef
430.
go back to reference Yu L-M, et al. Activation of PKG-CREB-KLF15 by melatonin attenuates Angiotensin II-induced vulnerability to atrial fibrillation via enhancing branched-chain amino acids catabolism. Free Radical Biol Med. 2022;178:202–14.CrossRef Yu L-M, et al. Activation of PKG-CREB-KLF15 by melatonin attenuates Angiotensin II-induced vulnerability to atrial fibrillation via enhancing branched-chain amino acids catabolism. Free Radical Biol Med. 2022;178:202–14.CrossRef
431.
go back to reference Mahalanobish S, et al. Melatonin counteracts necroptosis and pulmonary edema in cadmium-induced chronic lung injury through the inhibition of angiotensin II. J Biochem Mol Toxicol. 2022;36(10):e23163.PubMedCrossRef Mahalanobish S, et al. Melatonin counteracts necroptosis and pulmonary edema in cadmium-induced chronic lung injury through the inhibition of angiotensin II. J Biochem Mol Toxicol. 2022;36(10):e23163.PubMedCrossRef
432.
go back to reference Zhao Y, et al. Melatonin prevents against ethanol-induced liver injury by mitigating ferroptosis via targeting brain and muscle ARNT-like 1 in mice liver and HepG2 cells. J Agric Food Chem. 2022;70(40):12953–67.PubMedCrossRef Zhao Y, et al. Melatonin prevents against ethanol-induced liver injury by mitigating ferroptosis via targeting brain and muscle ARNT-like 1 in mice liver and HepG2 cells. J Agric Food Chem. 2022;70(40):12953–67.PubMedCrossRef
433.
go back to reference Fan L et al. Melatonin ameliorates the progression of Alzheimer's disease by inducing TFEB nuclear translocation, promoting mitophagy, and regulating NLRP3 inflammasome activity. BioMed Res Int. 2022. 2022. Fan L et al. Melatonin ameliorates the progression of Alzheimer's disease by inducing TFEB nuclear translocation, promoting mitophagy, and regulating NLRP3 inflammasome activity. BioMed Res Int. 2022. 2022.
434.
go back to reference Li T et al. Peripheral clock system abnormalities in patients with Parkinson’s disease. Front Aging Neurosci. 2021;13. Li T et al. Peripheral clock system abnormalities in patients with Parkinson’s disease. Front Aging Neurosci. 2021;13.
435.
go back to reference Delgado-Lara D, et al. Effect of melatonin administration on the PER1 and BMAL1 clock genes in patients with Parkinson’s disease. Biomed Pharmacother. 2020;129:110485.PubMedCrossRef Delgado-Lara D, et al. Effect of melatonin administration on the PER1 and BMAL1 clock genes in patients with Parkinson’s disease. Biomed Pharmacother. 2020;129:110485.PubMedCrossRef
436.
go back to reference Mattam U, Jagota A. Differential role of melatonin in restoration of age-induced alterations in daily rhythms of expression of various clock genes in suprachiasmatic nucleus of male Wistar rats. Biogerontology. 2014;15:257–68.PubMedCrossRef Mattam U, Jagota A. Differential role of melatonin in restoration of age-induced alterations in daily rhythms of expression of various clock genes in suprachiasmatic nucleus of male Wistar rats. Biogerontology. 2014;15:257–68.PubMedCrossRef
Metadata
Title
Melatonin: a ferroptosis inhibitor with potential therapeutic efficacy for the post-COVID-19 trajectory of accelerated brain aging and neurodegeneration
Authors
Asmaa Yehia
Osama A. Abulseoud
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2024
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-024-00728-6

Other articles of this Issue 1/2024

Molecular Neurodegeneration 1/2024 Go to the issue