en Experimental Neurobiology

Cited by CrossRef (155)

  1. Kyung-Tai Kim, Da-Hee Kim, Bo-Kyung Kim, Ji-Seok Han, Han Young Eom, Mi-Jin Yang, Seung-Hyuk Shin, Doo-Wan Cho, Bo Ko Jang, Ki Duk Park, Young-Su Yang, Su-Cheol Han. Four-week repeated dose oral toxicity study of KDS2010, a novel selective monoamine oxidase B inhibitor, in Sprague Dawley rats. Regulatory Toxicology and Pharmacology 2020;117:104733
    https://doi.org/10.1016/j.yrtph.2020.104733
  2. Maria João Matos, Santiago Vilar, Verónica García‐Morales, Nicholas P. Tatonetti, Eugenio Uriarte, Lourdes Santana, Dolores Viña. Insight into the Functional and Structural Properties of 3‐Arylcoumarin as an Interesting Scaffold in Monoamine Oxidase B Inhibition. ChemMedChem 2014;9:1488
    https://doi.org/10.1002/cmdc.201300533
  3. Zhigang Liang, Zhuli Liu, Xuwen Sun, Manli Tao, Xiao Xiao, Guoping Yu, Xiaomin Wang. The Effect of Fucoidan on Cellular Oxidative Stress and the CatD-Bax Signaling Axis in MN9D Cells Damaged by 1-Methyl-4-Phenypyridinium. Front. Aging Neurosci. 2019;10
    https://doi.org/10.3389/fnagi.2018.00429
  4. Trisha Bhatt, Bhoomika M. Patel, Mayur M. Patel. Drug Delivery Devices and Therapeutic Systems. 2019.
    https://doi.org/10.1016/B978-0-12-819838-4.00028-6
  5. Hannah B. L. Jones, Rory M. Crean, Anna Mullen, Emanuele G. Kendrick, Steven D. Bull, Stephen A. Wells, David R. Carbery, Fraser MacMillan, Marc W. van der Kamp, Christopher R. Pudney. Exposing the Interplay Between Enzyme Turnover, Protein Dynamics, and the Membrane Environment in Monoamine Oxidase B. Biochemistry 2019;58:2362
    https://doi.org/10.1021/acs.biochem.9b00213
  6. Patrícia Coelho, Ana Cristina Carvalho Rego. Mitochondrial Physiology and Vegetal Molecules. 2019.
    https://doi.org/10.1016/B978-0-12-821562-3.00018-6
  7. Narayan D. Chaurasiya, Jacob Midiwo, Pankaj Pandey, Regina N. Bwire, Robert J. Doerksen, Ilias Muhammad, Babu L. Tekwani. Selective Interactions of O-Methylated Flavonoid Natural Products with Human Monoamine Oxidase-A and -B. Molecules 2020;25:5358
    https://doi.org/10.3390/molecules25225358
  8. Miguel Pinto, Carlos Fernandes, Eva Martins, Renata Silva, Sofia Benfeito, Fernando Cagide, Ricardo F. Mendes, Filipe A. Almeida Paz, Jorge Garrido, Fernando Remião, Fernanda Borges. Boosting Drug Discovery for Parkinson’s: Enhancement of the Delivery of a Monoamine Oxidase-B Inhibitor by Brain-Targeted PEGylated Polycaprolactone-Based Nanoparticles. Pharmaceutics 2019;11:331
    https://doi.org/10.3390/pharmaceutics11070331
  9. Samay Prakash, Wayne G. Carter. The Neuroprotective Effects of Cannabis-Derived Phytocannabinoids and Resveratrol in Parkinson’s Disease: A Systematic Literature Review of Pre-Clinical Studies. Brain Sciences 2021;11:1573
    https://doi.org/10.3390/brainsci11121573
  10. Fumito Naganuma, Tadaho Nakamura, Takeo Yoshikawa, Tomomitsu Iida, Yamato Miura, Anikó Kárpáti, Takuro Matsuzawa, Atushi Yanai, Asuka Mogi, Takatoshi Mochizuki, Nobuyuki Okamura, Kazuhiko Yanai. Histamine N-methyltransferase regulates aggression and the sleep-wake cycle. Sci Rep 2017;7
    https://doi.org/10.1038/s41598-017-16019-8
  11. Tamás Tábi, László Vécsei, Moussa B. Youdim, Peter Riederer, Éva Szökő. Selegiline: a molecule with innovative potential. J Neural Transm 2020;127:831
    https://doi.org/10.1007/s00702-019-02082-0
  12. Mohamed Mahmoud Ali, Raguia Aly Shoukri, Carol Yousry. Effect of Particle Size versus Surface Charge on the Brain Targeting Behaviour of Elastic Nanovesicles: In-Vitro Characterization, Comparison between I-optimal and D-optimal Statistical Optimization and In-vivo Pharmacokinetic Evaluation. Journal of Drug Delivery Science and Technology 2024:105693
    https://doi.org/10.1016/j.jddst.2024.105693
  13. Begum E. Aksoz, Erkan Aksoz. Vital Role of Monoamine Oxidases and Cholinesterases in Central Nervous System Drug Research: A Sharp Dissection of the Pathophysiology. CCHTS 2020;23:877
    https://doi.org/10.2174/1386207323666200220115154
  14. Dokyoung Kim, Yong Woong Jun, Kyo Han Ahn. Fluorescent Probes for Analysis and Imaging of Monoamine Oxidase Activity. Bulletin of the Korean Chemical Society 2014;35:1269
    https://doi.org/10.5012/bkcs.2014.35.5.1269
  15. Keliang Wang, Jie Luo, Shuyuan Yeh, Bosen You, Jialin Meng, Philip Chang, Yuanjie Niu, Gonghui Li, Changxue Lu, Yezi Zhu, Emmanuel S. Antonarakis, Jun Luo, Chi-Ping Huang, Wanhai Xu, Chawnshang Chang. The MAO inhibitors phenelzine and clorgyline revert enzalutamide resistance in castration resistant prostate cancer. Nat Commun 2020;11
    https://doi.org/10.1038/s41467-020-15396-5
  16. George Mpekoulis, Vassilina Tsopela, Anna Chalari, Katerina I. Kalliampakou, Georgios Panos, Efseveia Frakolaki, Raphaela S. Milona, Diamantis C. Sideris, Dido Vassilacopoulou, Niki Vassilaki. Dengue Virus Replication Is Associated with Catecholamine Biosynthesis and Metabolism in Hepatocytes. Viruses 2022;14:564
    https://doi.org/10.3390/v14030564
  17. Sarah J. Stednitz, Briana Freshner, Samantha Shelton, Tori Shen, Donovan Black, Ethan Gahtan. Selective toxicity of L-DOPA to dopamine transporter-expressing neurons and locomotor behavior in zebrafish larvae. Neurotoxicology and Teratology 2015;52:51
    https://doi.org/10.1016/j.ntt.2015.11.001
  18. Trang N. N. Vo, Karen Frei, Daniel D. Truong. NeuroPsychopharmacotherapy. 2015.
    https://doi.org/10.1007/978-3-030-62059-2_357
  19. Guadalupe Palencia, Esperanza Garcia, Laura Osorio-Rico, Cristina Trejo-Solís, Angel Escamilla-Ramírez, Julio Sotelo. Neuroprotective effect of thalidomide on MPTP-induced toxicity. NeuroToxicology 2015;47:82
    https://doi.org/10.1016/j.neuro.2015.02.004
  20. Narayan Chaurasiya, Jianping Zhao, Pankaj Pandey, Robert Doerksen, Ilias Muhammad, Babu Tekwani. Selective Inhibition of Human Monoamine Oxidase B by Acacetin 7-Methyl Ether Isolated from Turnera diffusa (Damiana). Molecules 2019;24:810
    https://doi.org/10.3390/molecules24040810
  21. Eun-Hye Lee, Sang-Mi Kim, Chun-Hyung Kim, Suvarna H. Pagire, Haushabhau S. Pagire, Hee Yong Chung, Jin Hee Ahn, Chang-Hwan Park. Dopamine neuron induction and the neuroprotective effects of thyroid hormone derivatives. Sci Rep 2019;9
    https://doi.org/10.1038/s41598-019-49876-6
  22. Trang N. N. Vo, Karen Frei, Daniel D. Truong. NeuroPsychopharmacotherapy. 2019.
    https://doi.org/10.1007/978-3-319-56015-1_357-1
  23. Meisi Liu, Qing Wang, Zihan Lu, Liwei Wang, Shibo Zhao, Selinda Shi, Yonglie Zhao. Plasma metabolomics and network pharmacology analyses combined identify the action of Xiongzhi Dilong decoction in migraine treatment. Journal of Traditional Chinese Medical Sciences 2023;10:427
    https://doi.org/10.1016/j.jtcms.2023.09.005
  24. Sarel J. Robinson, Jacobus P. Petzer, Anél Petzer, Jacobus J. Bergh, Anna C.U. Lourens. Selected furanochalcones as inhibitors of monoamine oxidase. Bioorganic & Medicinal Chemistry Letters 2013;23:4985
    https://doi.org/10.1016/j.bmcl.2013.06.050
  25. Thomas Müller. Pharmacokinetic/pharmacodynamic evaluation of rasagiline mesylate for Parkinson’s disease. Expert Opinion on Drug Metabolism & Toxicology 2014;10:1423
    https://doi.org/10.1517/17425255.2014.943182
  26. Marcin Cieślak, Tomasz Danel, Olga Krzysztyńska-Kuleta, Justyna Kalinowska-Tłuścik. Machine learning accelerates pharmacophore-based virtual screening of MAO inhibitors. Sci Rep 2024;14
    https://doi.org/10.1038/s41598-024-58122-7
  27. Spyros N Deftereos, Evie Dodou, Christos Andronis, Aris Persidis. From depression to neurodegeneration and heart failure: re-examining the potential of MAO inhibitors. Expert Review of Clinical Pharmacology 2012;5:413
    https://doi.org/10.1586/ecp.12.29
  28. Matthias D. Mertens, Sonja Hinz, Christa E. Müller, Michael Gütschow. Alkynyl–coumarinyl ethers as MAO-B inhibitors. Bioorganic & Medicinal Chemistry 2014;22:1916
    https://doi.org/10.1016/j.bmc.2014.01.046
  29. Orly Weinreb, Tamar Amit, Peter Riederer, Moussa B.H. Youdim, Silvia A. Mandel. Monoamine Oxidase and their Inhibitors. 2014.
    https://doi.org/10.1016/B978-0-12-386467-3.00007-8
  30. Thomas Müller. Pharmacokinetics and pharmacodynamics of levodopa/carbidopa cotherapies for Parkinson’s disease. Expert Opinion on Drug Metabolism & Toxicology 2020;16:403
    https://doi.org/10.1080/17425255.2020.1750596
  31. Liliana Bernardino. The Functional Roles of Histamine Receptors. 2020.
    https://doi.org/10.1007/7854_2021_235
  32. Ola A.M. Ghoneim, Raymond G. Booth. Burger's Medicinal Chemistry and Drug Discovery. 2020.
    https://doi.org/10.1002/0471266949.bmc105.pub3
  33. Muhammed Khairujjaman Mazumder, Rajib Paul, Banashree Chetia Phukan, Ankumoni Dutta, Jayasree Chakrabarty, Pallab Bhattacharya, Anupom Borah. Garcinol, an effective monoamine oxidase-B inhibitor for the treatment of Parkinson's disease. Medical Hypotheses 2018;117:54
    https://doi.org/10.1016/j.mehy.2018.06.009
  34. Peter Riederer, Thomas Müller. Use of monoamine oxidase inhibitors in chronic neurodegeneration. Expert Opinion on Drug Metabolism & Toxicology 2017;13:233
    https://doi.org/10.1080/17425255.2017.1273901
  35. Elisa Gómez de Lope, Rebecca Ting Jiin Loo, Armin Rauschenberger, Muhammad Ali, Lukas Pavelka, Tainá M. Marques, Clarissa P. C. Gomes, Rejko Krüger, Enrico Glaab, Geeta Acharya, Gloria Aguayo, Myriam Alexandre, Muhammad Ali, Wim Ammerlann, Giuseppe Arena, Rudi Balling, Michele Bassis, Roxane Batutu, Katy Beaumont, Regina Becker, Camille Bellora, Guy Berchem, Daniela Berg, Alexandre Bisdorff, Ibrahim Boussaad, David Bouvier, Kathrin Brockmann, Jessica Calmes, Lorieza Castillo, Gessica Contesotto, Nancy De Bremaeker, Nico Diederich, Rene Dondelinger, Nancy E. Ramia, Daniela Esteves, Guy Fagherazzi, Jean-Yves Ferrand, Katrin Frauenknecht, Manon Gantenbein, Thomas Gasser, Piotr Gawron, Soumyabrata Ghosh, Marijus Giraitis, Enrico Glaab, Martine Goergen, Elisa Gómez De Lope, Jérôme Graas, Mariella Graziano, Valentin Groues, Anne Grünewald, Wei Gu, Gaël Hammot, Anne-Marie Hanff, Linda Hansen, Michael Heneka, Estelle Henry, Sylvia Herbrink, Sascha Herzinger, Michael Heymann, Michele Hu, Alexander Hundt, Nadine Jacoby, Jacek Jaroslaw Lebioda, Yohan Jarosz, Sonja Jónsdóttir, Quentin Klopfenstein, Jochen Klucken, Rejko Krüger, Pauline Lambert, Zied Landoulsi, Roseline Lentz, Inga Liepelt, Robert Liszka, Laura Longhino, Victoria Lorentz, Paula Cristina Lupu, Tainá M. Marques, Clare Mackay, Walter Maetzler, Katrin Marcus, Guilherme Marques, Patricia Martins Conde, Patrick May, Deborah Mcintyre, Chouaib Mediouni, Francoise Meisch, Myriam Menster, Maura Minelli, Michel Mittelbronn, Brit Mollenhauer, Friedrich Mühlschlegel, Romain Nati, Ulf Nehrbass, Sarah Nickels, Beatrice Nicolai, Jean-Paul Nicolay, Fozia Noor, Marek Ostaszewski, Clarissa P. C. Gomes, Sinthuja Pachchek, Claire Pauly, Laure Pauly, Lukas Pavelka, Magali Perquin, Rosalina Ramos Lima, Armin Rauschenberger, Rajesh Rawal, Dheeraj Reddy Bobbili, Kirsten Roomp, Eduardo Rosales, Isabel Rosety, Estelle Sandt, Stefano Sapienza, Venkata Satagopam, Margaux Schmitt, Sabine Schmitz, Reinhard Schneider, Jens Schwamborn, Raquel Severino, Amir Sharify, Ekaterina Soboleva, Kate Sokolowska, Hermann Thien, Elodie Thiry, Rebecca Ting Jiin Loo, Christophe Trefois, Johanna Trouet, Olena Tsurkalenko, Michel Vaillant, Mesele Valenti, Gilles Van Cutsem, Carlos Vega, Liliana Vilas Boas, Maharshi Vyas, Richard Wade-Martins, Paul Wilmes, Evi Wollscheid-Lengeling, Gelani Zelimkhanov. Comprehensive blood metabolomics profiling of Parkinson’s disease reveals coordinated alterations in xanthine metabolism. npj Parkinsons Dis. 2024;10
    https://doi.org/10.1038/s41531-024-00671-9
  36. Giuseppe Di Giovanni, Dubravka Svob Strac, Montse Sole, Mercedes Unzeta, Keith F. Tipton, Dorotea Mück-Šeler, Irene Bolea, Laura Della Corte, Matea Nikolac Perkovic, Nela Pivac, Ilse J. Smolders, Anna Stasiak, Wieslawa A. Fogel, Philippe De Deurwaerdère. Monoaminergic and Histaminergic Strategies and Treatments in Brain Diseases. Front. Neurosci. 2016;10
    https://doi.org/10.3389/fnins.2016.00541
  37. Sertan AYTAÇ. Synthesis of Pyridine-Based Imine Compounds and Molecular Docking Studies Against Dopamine D2 Receptors. 2023;10:161
    https://doi.org/10.17350/HJSE19030000303
  38. Ankita Paul, Khushwant S. Yadav. Parkinson's disease: Current drug therapy and unraveling the prospects of nanoparticles. Journal of Drug Delivery Science and Technology 2020;58:101790
    https://doi.org/10.1016/j.jddst.2020.101790
  39. Manfred Gerlach, Heinz Reichmann, Peter Riederer. A critical review of evidence for preclinical differences between rasagiline and selegiline. Basal Ganglia 2012;2:S9
    https://doi.org/10.1016/j.baga.2012.04.032
  40. Jennifer N. K. Nyarko, Ryan M. Heistad, Paul R. Pennington, Darrell D. Mousseau. Monoamine Oxidase. 2012.
    https://doi.org/10.1007/978-1-0716-2643-6_10
  41. Makoto Naoi, Wakako Maruyama, Masayo Shamoto-Nagai. Type A and B monoamine oxidases distinctly modulate signal transduction pathway and gene expression to regulate brain function and survival of neurons. J Neural Transm 2018;125:1635
    https://doi.org/10.1007/s00702-017-1832-6
  42. Aslihan Ugun-Klusek, Theodosis S. Theodosi, Julia C. Fitzgerald, Florence Burté, Christoph Ufer, David J. Boocock, Patrick Yu-Wai-Man, Lynn Bedford, E. Ellen Billett. Monoamine oxidase-A promotes protective autophagy in human SH-SY5Y neuroblastoma cells through Bcl-2 phosphorylation. Redox Biology 2019;20:167
    https://doi.org/10.1016/j.redox.2018.10.003
  43. Mehar Sahu, Shrutikirti Vashishth, Neha Kukreti, Ashima Gulia, Ashish Russell, Rashmi K. Ambasta, Pravir Kumar. . 2019.
    https://doi.org/10.1016/bs.pmbts.2024.03.023
  44. Thomas Müller. Nondopaminergic therapy of motor and nonmotor symptoms in Parkinson's disease: a clinician's perspective. Neurodegenerative Disease Management 2016;6:385
    https://doi.org/10.2217/nmt-2016-0025
  45. Thomas Müller. Diagnosis and Management in Parkinson's Disease. 2016.
    https://doi.org/10.1016/B978-0-12-815946-0.00028-4
  46. Vasanthi Chinraj, Ramakkamma Aishwarya Reddy, Jubie Selvaraj, Raman Sureshkumar. Design, Synthesis and In Vitro Evaluation of Levodopa Stearic Acid Hydrazide Conjugate for the Management of Parkinson’s DiseaseNovel Conjugate for Parkinson’s Disease. Drug Res (Stuttg) 2024;74:60
    https://doi.org/10.1055/a-2234-9859
  47. Ranju Bansal, Ranjit Singh. Exploring the potential of natural and synthetic neuroprotective steroids against neurodegenerative disorders: A literature review. Medicinal Research Reviews 2018;38:1126
    https://doi.org/10.1002/med.21458
  48. Manish Chamoli, Shankar J. Chinta, Julie K. Andersen. An inducible MAO-B mouse model of Parkinson’s disease: a tool towards better understanding basic disease mechanisms and developing novel therapeutics. J Neural Transm 2018;125:1651
    https://doi.org/10.1007/s00702-018-1887-z
  49. Siew L. Cheong, Stephanie Federico, Giampiero Spalluto, Karl-Norbert Klotz, Giorgia Pastorin. The current status of pharmacotherapy for the treatment of Parkinson’s disease: transition from single-target to multitarget therapy. Drug Discovery Today 2019;24:1769
    https://doi.org/10.1016/j.drudis.2019.05.003
  50. Madalina-Andreea Robea, Ioana-Miruna Balmus, Alin Ciobica, Stefan Strungaru, Gabriel Plavan, Lucian Dragos Gorgan, Alexandra Savuca, Mircea Nicoara. Parkinson’s Disease-Induced Zebrafish Models: Focussing on Oxidative Stress Implications and Sleep Processes. Oxidative Medicine and Cellular Longevity 2020;2020:1
    https://doi.org/10.1155/2020/1370837
  51. Sagar Kansara, Akash Trivedi, Sheng Chen, Joseph Jankovic, Weidong Le. Early diagnosis and therapy of Parkinson’s disease: can disease progression be curbed?. J Neural Transm 2013;120:197
    https://doi.org/10.1007/s00702-012-0840-9
  52. Gavin R. Hoffman, Madeline G. Olson, Allen M. Schoffstall, Ryan F. Estévez, Vincent Van den Eynde, Peter K. Gillman, Maureen E. Stabio. Classics in Chemical Neuroscience: Selegiline, Isocarboxazid, Phenelzine, and Tranylcypromine. ACS Chem. Neurosci. 2023;14:4064
    https://doi.org/10.1021/acschemneuro.3c00591
  53. David Ramírez, Julio Caballero. Is It Reliable to Take the Molecular Docking Top Scoring Position as the Best Solution without Considering Available Structural Data?. Molecules 2018;23:1038
    https://doi.org/10.3390/molecules23051038
  54. Wakako Maruyama, Makoto Naoi. ‘‘70th Birthday Professor Riederer’’ Induction of glial cell line-derived and brain-derived neurotrophic factors by rasagiline and (−)deprenyl: a way to a disease-modifying therapy?. J Neural Transm 2013;120:83
    https://doi.org/10.1007/s00702-012-0876-x
  55. Eoin Mulroy, Kailash P. Bhatia. Non-Motor Parkinson's Disease. 2013.
    https://doi.org/10.1017/9781009039291.022
  56. Ibrahim Yildiz, Banu Sizirici Yildiz. Mechanistic study of L-6-hydroxynicotine oxidase by DFT and ONIOM methods. J Mol Model 2021;27
    https://doi.org/10.1007/s00894-020-04646-4
  57. Pedro Cruz-Vicente, Luís A. Passarinha, Samuel Silvestre, Eugenia Gallardo. Recent Developments in New Therapeutic Agents against Alzheimer and Parkinson Diseases: In-Silico Approaches. Molecules 2021;26:2193
    https://doi.org/10.3390/molecules26082193
  58. Mikhail V. Voronin, Ilya A. Kadnikov, Dmitry N. Voronkov, Sergey B. Seredenin. Chaperone Sigma1R mediates the neuroprotective action of afobazole in the 6-OHDA model of Parkinson’s disease. Sci Rep 2019;9
    https://doi.org/10.1038/s41598-019-53413-w
  59. David L. Eaton, Julia Cui. Patty's Toxicology. 2019.
    https://doi.org/10.1002/0471125474.tox122
  60. Werner J. Geldenhuys, Akiko Kochi, Li Lin, Vijaykumar Sutariya, Dean E. Dluzen, Cornelis J. Van der Schyf, Mi Hee Lim. Methyl Yellow: A Potential Drug Scaffold for Parkinson's Disease. ChemBioChem 2014;15:1591
    https://doi.org/10.1002/cbic.201300770
  61. Adjia Hamadjida, Stephen G. Nuara, Cynthia Kwan, Imane Frouni, Dominique Bédard, Jim C. Gourdon, Philippe Huot. Monoamine oxidase A inhibition with moclobemide enhances the anti-parkinsonian effect of L-DOPA in the MPTP-lesioned marmoset. Naunyn-Schmiedeberg's Arch Pharmacol 2020;393:2157
    https://doi.org/10.1007/s00210-020-01933-y
  62. Martin Klietz, Stephan Greten, Florian Wegner, Günter U. Höglinger. Safety and Tolerability of Pharmacotherapies for Parkinson’s Disease in Geriatric Patients. Drugs Aging 2019;36:511
    https://doi.org/10.1007/s40266-019-00654-z
  63. Cecilia Mattsson, Peder Svensson, Clas Sonesson. A novel series of 6-substituted 3-(pyrrolidin-1-ylmethyl)chromen-2-ones as selective monoamine oxidase (MAO) A inhibitors. European Journal of Medicinal Chemistry 2014;73:177
    https://doi.org/10.1016/j.ejmech.2013.11.035
  64. Marc Morissette, Thérèse Di Paolo. Non-human primate models of PD to test novel therapies. J Neural Transm 2018;125:291
    https://doi.org/10.1007/s00702-017-1722-y
  65. Stefanie Hagenow, Anna Affini, Elsa Y. Pioli, Sonja Hinz, Yan Zhao, Gregory Porras, Vigneshwaran Namasivayam, Christa E. Müller, Jian-Sheng Lin, Erwan Bezard, Holger Stark. Adenosine A2AR/A1R Antagonists Enabling Additional H3R Antagonism for the Treatment of Parkinson’s Disease. J. Med. Chem. 2021;64:8246
    https://doi.org/10.1021/acs.jmedchem.0c00914
  66. Arif Jamal Siddiqui, Sadaf Jahan, Maqsood Ahmed Siddiqui, Andleeb Khan, Mohammed Merae Alshahrani, Riadh Badraoui, Mohd Adnan. Targeting Monoamine Oxidase B for the Treatment of Alzheimer’s and Parkinson’s Diseases Using Novel Inhibitors Identified Using an Integrated Approach of Machine Learning and Computer-Aided Drug Design. Mathematics 2023;11:1464
    https://doi.org/10.3390/math11061464
  67. Bhupinder Kumar, Mohit Kumar, Ashish Ranjan Dwivedi, Vinod Kumar. Synthesis, Biological Evaluation and Molecular Modeling Studies of Propargyl‐Containing 2,4,6‐Trisubstituted Pyrimidine Derivatives as Potential Anti‐Parkinson Agents. ChemMedChem 2018;13:705
    https://doi.org/10.1002/cmdc.201700589
  68. Anupom Borah, Rajib Paul, Sabanum Choudhury, Amarendranath Choudhury, Bornalee Bhuyan, Anupam Das Talukdar, Manabendra Dutta Choudhury, Kochupurackal P Mohanakumar. Neuroprotective Potential of Silymarin against CNS Disorders: Insight into the Pathways and Molecular Mechanisms of Action. CNS Neurosci Ther 2013;19:847
    https://doi.org/10.1111/cns.12175
  69. Yan Song, Jian-Yu E, Tracy Guo, Rahul Sasane, Steve Arcona, Nirmal Keshava, Eric Wu. Treatment Patterns and Healthcare Resource Use in Medicare Beneficiaries with Parkinson’s Disease. CEOR 2023;Volume 15:631
    https://doi.org/10.2147/CEOR.S422023
  70. José-Rubén García-Montes, Alejandra Boronat-García, René Drucker-Colín. Pharmacological strategies for Parkinson’s disease. Health 2012;04:1153
    https://doi.org/10.4236/health.2012.431174
  71. Thomas Müller, Paul Foley. Clinical Pharmacokinetics and Pharmacodynamics of Safinamide. Clin Pharmacokinet 2017;56:251
    https://doi.org/10.1007/s40262-016-0449-5
  72. Annunziatina Laurino, Manuela Gencarelli, Laura Raimondi. The 3-iodothyronamine (T1AM) and the 3-iodothyroacetic acid (TA1) indicate a novel connection with the histamine system for neuroprotection. European Journal of Pharmacology 2021;912:174606
    https://doi.org/10.1016/j.ejphar.2021.174606
  73. Abha Harfouche, Wael Alata, Karine Leblanc, Bruno Figadère, Alexandre Maciuk. Label-Free LC/HRMS-Based Enzymatic Activity Assay for the Detection of DDC, MAO and COMT Inhibitors in Natural Extracts. SSRN Journal 2021
    https://doi.org/10.2139/ssrn.3866383
  74. Sam Dawbaa, Asaf Evrim Evren, Begüm Nurpelin Sağlik, Nalan Gundogdu-Karaburun, Ahmet Cagri Karaburun. Biological activity evaluation of novel monoamine oxidase inhibitory compounds targeting Parkinson disease. Future Medicinal Chemistry 2022;14:1663
    https://doi.org/10.4155/fmc-2022-0167
  75. Moussa B. H. Youdim. Monoamine oxidase inhibitors, and iron chelators in depressive illness and neurodegenerative diseases. J Neural Transm 2018;125:1719
    https://doi.org/10.1007/s00702-018-1942-9
  76. Peter Riederer, Dan Rujescu. Discoveries in Pharmacology. 2018.
    https://doi.org/10.1016/B978-0-323-85519-8.00008-0
  77. Subham Panigrahy, Anupama Sikder, Etikala Amulya, Saurabh Shah, Pooja Khairnar, Shailendra Saraf, Saurabh Srivastava. Deciphering the role of nanocarrier-based nucleic acid delivery to the brain for the management of neurodegenerative disorders. Journal of Drug Delivery Science and Technology 2024;92:105325
    https://doi.org/10.1016/j.jddst.2023.105325
  78. I. A. Kadnikov, D. N. Voronkov, M. V. Voronin, S. B. Seredenin. Analysis of Quinone Reductase 2 Implication in Mechanism of Antiparkinsonian Action of Afobazole. Neurochem. J. 2020;14:227
    https://doi.org/10.1134/S1819712420010110
  79. Nobutaka Hattori, Atsushi Takeda, Shinichi Takeda, Akira Nishimura, Tadayuki Kitagawa, Hideki Mochizuki, Masahiro Nagai, Ryosuke Takahashi. Rasagiline monotherapy in early Parkinson's disease: A phase 3, randomized study in Japan. Parkinsonism & Related Disorders 2019;60:146
    https://doi.org/10.1016/j.parkreldis.2018.08.024
  80. Dilipkumar Pal, Pooja Sahu. Importance of Indazole against Neurological Disorders. CTMC 2022;22:1136
    https://doi.org/10.2174/1568026622666220225152443
  81. Alireza Abdanipour, Iraj Jafari Anarkooli, Saeed Shokri, Mehrdad Ghorbanlou, Vahid Bayati, Reza Nejatbakhsh. Neuroprotective effects of selegiline on rat neural stem cells treated with hydrogen peroxide. biom rep 2017
    https://doi.org/10.3892/br.2017.1023
  82. Kyu Hwan Shim, Niti Sharma, Seong Soo A. An. Mechanistic Insights into the Neuroprotective Potential of Sacred Ficus Trees. Nutrients 2022;14:4731
    https://doi.org/10.3390/nu14224731
  83. Anupom Borah, Amarendranath Choudhury, Rajib Paul, Muhammed K. Mazumder, Swapnali Chetia. Neuroprotective Natural Products. 2022.
    https://doi.org/10.1002/9783527803781.ch5
  84. Kinga Krzysztoforska, Dagmara Mirowska-Guzel, Ewa Widy-Tyszkiewicz. Pharmacological effects of protocatechuic acid and its therapeutic potential in neurodegenerative diseases: Review on the basis of in vitro and in vivo studies in rodents and humans. Nutritional Neuroscience 2019;22:72
    https://doi.org/10.1080/1028415X.2017.1354543
  85. Yasir Hasan Siddique, Falaq Naz, Wasi Khan, Smita Jyoti, Braj Raj Singh, Alim Hussain Naqvi. Effect of pramipexole alginate nanodispersion (PAND) on the transgenic Drosophila expressing human alpha synuclein in the brain. J Appl Biomed 2018;16:111
    https://doi.org/10.1016/j.jab.2017.11.002
  86. Mohammad-Hadi Karimi-Harandi, Mehdi Shabani-Nooshabadi, Rozhin Darabi. Simultaneous determination of citalopram and selegiline using an efficient electrochemical sensor based on ZIF-8 decorated with RGO and g-C3N4 in real samples. Analytica Chimica Acta 2022;1203:339662
    https://doi.org/10.1016/j.aca.2022.339662
  87. Sinem Ilgın, Derya Osmaniye, Serkan Levent, Begüm Sağlık, Ulviye Acar Çevik, Betül Çavuşoğlu, Yusuf Özkay, Zafer Kaplancıklı. Design and Synthesis of New Benzothiazole Compounds as Selective hMAO-B Inhibitors. Molecules 2017;22:2187
    https://doi.org/10.3390/molecules22122187
  88. Gabriele Dalla Torre, Jon I. Mujika, Joanna Izabela Lachowicz, Maria J. Ramos, Xabier Lopez. The interaction of aluminum with catecholamine-based neurotransmitters: can the formation of these species be considered a potential risk factor for neurodegenerative diseases?. Dalton Trans. 2019;48:6003
    https://doi.org/10.1039/C8DT04216K
  89. Aaron D. Schwab, Mackenzie J. Thurston, Jatin Machhi, Katherine E. Olson, Krista L. Namminga, Howard E. Gendelman, R. Lee Mosley. Immunotherapy for Parkinson’s disease. Neurobiology of Disease 2020;137:104760
    https://doi.org/10.1016/j.nbd.2020.104760
  90. Patrick A. Randall, Christie A. Lee, Eric J. Nunes, Samantha E. Yohn, Victoria Nowak, Bilal Khan, Priya Shah, Saagar Pandit, V. Kiran Vemuri, Alex Makriyannis, Younis Baqi, Christa E. Müller, Merce Correa, John D. Salamone, Harriet de Wit. The VMAT-2 Inhibitor Tetrabenazine Affects Effort-Related Decision Making in a Progressive Ratio/Chow Feeding Choice Task: Reversal with Antidepressant Drugs. PLoS ONE 2014;9:e99320
    https://doi.org/10.1371/journal.pone.0099320
  91. Anjana Pathania, Rajnish Kumar, Rajat Sandhir. Hydroxytyrosol as anti-parkinsonian molecule: Assessment using in-silico and MPTP-induced Parkinson’s disease model. Biomedicine & Pharmacotherapy 2021;139:111525
    https://doi.org/10.1016/j.biopha.2021.111525
  92. Anastasia Bougea. Some Novel Therapies in Parkinson’s Disease: A Promising Path Forward or Not Yet? A Systematic Review of the Literature. Biomedicines 2024;12:549
    https://doi.org/10.3390/biomedicines12030549
  93. Dharmendra Kumar Khatri, Mamta Choudhary, Anika Sood, Shashi Bala Singh. Anxiety: An ignored aspect of Parkinson’s disease lacking attention. Biomedicine & Pharmacotherapy 2020;131:110776
    https://doi.org/10.1016/j.biopha.2020.110776
  94. Valentina Oliveri. Toward the discovery and development of effective modulators of α-synuclein amyloid aggregation. European Journal of Medicinal Chemistry 2019;167:10
    https://doi.org/10.1016/j.ejmech.2019.01.045
  95. Amritha Manoharan, Jong Min Oh, Feba Benny, Sunil Kumar, Mohamed A. Abdelgawad, Mohammed M. Ghoneim, Mohamed E. Shaker, Mohamed El-Sherbiny, Hailah M. Almohaimeed, Prashant Gahtori, Hoon Kim, Bijo Mathew. Assembling a Cinnamyl Pharmacophore in the C3-Position of Substituted Isatins via Microwave-Assisted Synthesis: Development of a New Class of Monoamine Oxidase-B Inhibitors for the Treatment of Parkinson’s Disease. Molecules 2023;28:6167
    https://doi.org/10.3390/molecules28166167
  96. A Mingorance‐Le Meur, P Ghisdal, B Mullier, P De Ron, P Downey, C Van Der Perren, V Declercq, S Cornelis, M Famelart, J Van Asperen, E Jnoff, J P Courade. Reversible inhibition of the glycine transporter GlyT2 circumvents acute toxicity while preserving efficacy in the treatment of pain. British J Pharmacology 2013;170:1053
    https://doi.org/10.1111/bph.12343
  97. Olivia J. Kalimon, Hemendra J. Vekaria, Greg A. Gerhardt, Patrick G. Sullivan. Inhibition of monoamine oxidase-a increases respiration in isolated mouse cortical mitochondria. Experimental Neurology 2023;363:114356
    https://doi.org/10.1016/j.expneurol.2023.114356
  98. Thomas Müller, Peter Riederer, Edna Grünblatt. Simultaneous determination of MAO-A and -B activity following first time intake of an irreversible MAO-B inhibitor in patients with Parkinson’s disease. J Neural Transm 2017;124:745
    https://doi.org/10.1007/s00702-017-1705-z
  99. Joachim Schuster, Jens Dreyhaupt, Karla Mönkemöller, Luc Dupuis, Stéphane Dieterlé, Jochen H. Weishaupt, Jan Kassubek, Susanne Petri, Thomas Meyer, Julian Grosskreutz, Berthold Schrank, Matthias Boentert, Alexander Emmer, Andreas Hermann, Daniel Zeller, Johannes Prudlo, Andrea S. Winkler, Torsten Grehl, Michael T. Heneka, Siw Johannesen, Bettina Göricke, Simon Witzel, Johannes Dorst, Albert C. Ludolph. In‐depth analysis of data from the RAS‐ALS study reveals new insights in rasagiline treatment for amyotrophic lateral sclerosis. Euro J of Neurology 2024;31
    https://doi.org/10.1111/ene.16204
  100. Vaibhav Walia, Ashish Gakkhar, Munish Garg. Handbook of Research on Critical Examinations of Neurodegenerative Disorders. 2024.
    https://doi.org/10.4018/978-1-5225-5282-6.ch012
  101. Zufika Qamar, Muhammad Usama Ashhar, Annu, Farheen Fatima Qizilibash, Pravat Kumar Sahoo, Asgar Ali, Javed Ali, Sanjula Baboota. Lipid nanocarrier of selegiline augmented anti-Parkinson’s effect via P-gp modulation using quercetin. International Journal of Pharmaceutics 2021;609:121131
    https://doi.org/10.1016/j.ijpharm.2021.121131
  102. Damir Varešlija, Keith F. Tipton, Gavin P. Davey, Andrew G. McDonald. 6-Hydroxydopamine: a far from simple neurotoxin. J Neural Transm 2020;127:213
    https://doi.org/10.1007/s00702-019-02133-6
  103. Éva Szökő, Tamás Tábi, Peter Riederer, László Vécsei, Kálmán Magyar. Pharmacological aspects of the neuroprotective effects of irreversible MAO-B inhibitors, selegiline and rasagiline, in Parkinson’s disease. J Neural Transm 2018;125:1735
    https://doi.org/10.1007/s00702-018-1853-9
  104. Hongjia Zhang, Rongsheng Tong, Lan Bai, Jianyou Shi, Liang Ouyang. Emerging targets and new small molecule therapies in Parkinson’s disease treatment. Bioorganic & Medicinal Chemistry 2016;24:1419
    https://doi.org/10.1016/j.bmc.2016.02.030
  105. Theresa A. Zesiewicz, Yarema Bezchlibnyk, Nicolas Dohse, Shaila D. Ghanekar. Management of Early Parkinson Disease. Clinics in Geriatric Medicine 2020;36:35
    https://doi.org/10.1016/j.cger.2019.09.001
  106. Sven Ulrich, Ute Lewitzka. Psychopharmakotherapie mit dem MAO-Hemmer Tranylcypromin Schwerpunkte und Trends aus Theorie und Praxis. Fortschr Neurol Psychiatr 2023
    https://doi.org/10.1055/a-2182-5365
  107. Jonas Drechsel, Christina Kyrousi, Silvia Cappello, Stephan A. Sieber. Tranylcypromine specificity for monoamine oxidase is limited by promiscuous protein labelling and lysosomal trapping. RSC Chem. Biol. 2020;1:209
    https://doi.org/10.1039/D0CB00048E
  108. Danielle N. Jones, Mary Ann Raghanti. The role of monoamine oxidase enzymes in the pathophysiology of neurological disorders. Journal of Chemical Neuroanatomy 2021;114:101957
    https://doi.org/10.1016/j.jchemneu.2021.101957
  109. Ling Yang, Chi-Lin Li, Tung-Hu Tsai. Preclinical Herb–Drug Pharmacokinetic Interaction of Panax ginseng Extract and Selegiline in Freely Moving Rats. ACS Omega 2020;5:4682
    https://doi.org/10.1021/acsomega.0c00123
  110. Dincer Erdag, Oguz Merhan, Baris Yildiz. Biogenic Amines. 2020.
    https://doi.org/10.5772/intechopen.81569
  111. Liliana Pacureanu, Alina Bora, Luminita Crisan. New Insights on the Activity and Selectivity of MAO-B Inhibitors through In Silico Methods. IJMS 2023;24:9583
    https://doi.org/10.3390/ijms24119583
  112. Daniel Chavarria, Carlos Fernandes, Vera Silva, Catia Silva, Eva Gil-Martins, Pedro Soares, Tiago Silva, Renata Silva, Fernando Remião, Paulo J. Oliveira, Fernanda Borges. Design of novel monoamine oxidase-B inhibitors based on piperine scaffold: Structure-activity-toxicity, drug-likeness and efflux transport studies. European Journal of Medicinal Chemistry 2020;185:111770
    https://doi.org/10.1016/j.ejmech.2019.111770
  113. Branden J. Stansley, Bryan K. Yamamoto. Chronic l-Dopa Decreases Serotonin Neurons in a Subregion of the Dorsal Raphe Nucleus. J Pharmacol Exp Ther 2014;351:440
    https://doi.org/10.1124/jpet.114.218966
  114. Michael Entzeroth, Anil K. Ratty. Monoamine Oxidase Inhibitors—Revisiting a Therapeutic Principle. OJD 2017;06:31
    https://doi.org/10.4236/ojd.2017.62004
  115. Nafsika Afentou, Johan Jarl, Ulf‐G Gerdtham, Sanjib Saha. Economic Evaluation of Interventions in Parkinson's Disease: A Systematic Literature Review. Movement Disord Clin Pract 2019;6:282
    https://doi.org/10.1002/mdc3.12755
  116. Neeraj Kumar, Anita Singh, Dinesh Kumar Sharma, Kamal Kishore. Novel Target Sites for Drug Screening: A Special Reference to Cancer, Rheumatoid Arthritis and Parkinson’s Disease. CST 2019;14:107
    https://doi.org/10.2174/1574362413666180320112810
  117. Thomas Müller, Jan-Dominique Möhr. Pharmacokinetics of monoamine oxidase B inhibitors in Parkinson’s disease: current status. Expert Opinion on Drug Metabolism & Toxicology 2019;15:429
    https://doi.org/10.1080/17425255.2019.1607292
  118. Masahiro Nagai, Nobutaka Hattori. Pharmacological properties and clinical efficacy of rasagiline mesylate (Azilect®). Folia Pharmacol. Jpn. 2020;155:187
    https://doi.org/10.1254/fpj.19146
  119. Raj V. Kavitha, J.R. Kumar, Chukwuebuka Egbuna, Jonathan C. Ifemeje. Phytochemicals as Lead Compounds for New Drug Discovery. 2020.
    https://doi.org/10.1016/B978-0-12-817890-4.00010-X
  120. A. Connor Whitfield, Ben T. Moore, R. Nathan Daniels. Classics in Chemical Neuroscience: Levodopa. ACS Chem. Neurosci. 2014;5:1192
    https://doi.org/10.1021/cn5001759
  121. Luiz Roberto Carraro Junior, Amália Gonçalves Alves, Taís da Silva Teixeira Rech, José Coan Campos Júnior, Geonir Machado Siqueira, Wilson Cunico, César Augusto Brüning, Cristiani Folharini Bortolatto. Three ‐(pyridin‐2‐yl)‐2‐(pyridin‐2‐ylimino)thiazolidin‐4‐one as a novel inhibitor of cerebral MAO‐B activity with antioxidant properties and low toxicity potential. J Biochem & Molecular Tox 2021;35
    https://doi.org/10.1002/jbt.22833
  122. Thomas Müller, Peter Riederer, Edna Grünblatt. Determination of Monoamine Oxidase A and B Activity in Long-Term Treated Patients With Parkinson Disease. Clin Neuropharm 2017;40:208
    https://doi.org/10.1097/WNF.0000000000000233
  123. Khalid Orayj, Tahani Almeleebia, Easwaran Vigneshwaran, Sultan Alshahrani, Sirajudeen. S. Alavudeen, Wael Alghamdi. Trend of recognizing depression symptoms and antidepressants use in newly diagnosed Parkinson's disease: Population‐based study. Brain and Behavior 2021;11
    https://doi.org/10.1002/brb3.2228
  124. Ali Moazzam, Farnaz Jafarpour. Chlorophyll-catalyzed photochemical regioselective coumarin C–H arylation with diazonium salts. New J. Chem. 2020;44:16692
    https://doi.org/10.1039/D0NJ02012E
  125. Deendyal Bhurta, Sandip B. Bharate. Styryl Group, a Friend or Foe in Medicinal Chemistry. ChemMedChem 2022;17
    https://doi.org/10.1002/cmdc.202100706
  126. Zhi Xin Chew, Chooi Ling Lim, Khuen Yen Ng, Soi Moi Chye, Anna Pick Kiong Ling, Rhun Yian Koh. The Role of Monoamine Oxidase B Inhibitors in the Treatment of Parkinson’s Disease - An Update. CNSNDDT 2023;22:329
    https://doi.org/10.2174/1871527321666211231100255
  127. Fabiola Kamecki, Damijan Knez, Diego Carvalho, Carolina Marcucci, Marina Rademacher, Josefina Higgs, Simon Žakelj, Alejandra Marcos, Felicitas de Tezanos Pinto, Juan Andrés Abin-Carriquiry, Stanislav Gobec, Natalia Colettis, Mariel Marder. Multitarget 2′-hydroxychalcones as potential drugs for the treatment of neurodegenerative disorders and their comorbidities. Neuropharmacology 2021;201:108837
    https://doi.org/10.1016/j.neuropharm.2021.108837
  128. Thomas Müller. Current and investigational non-dopaminergic agents for management of motor symptoms (including motor complications) in Parkinson’s disease. Expert Opinion on Pharmacotherapy 2017;18:1457
    https://doi.org/10.1080/14656566.2017.1373089
  129. Liyue Qin, Ziyu Chen, Liu Yang, Hailian Shi, Hui Wu, Beibei Zhang, Weiqi Zhang, Qi Xu, Fei Huang, Xiaojun Wu. Luteolin-7-O-glucoside protects dopaminergic neurons by activating estrogen-receptor-mediated signaling pathway in MPTP-induced mice. Toxicology 2019;426:152256
    https://doi.org/10.1016/j.tox.2019.152256
  130. Robert Otto, Robert Penzis, Friedemann Gaube, Oliver Adolph, Karl J. Föhr, Paul Warncke, Dina Robaa, Dorothea Appenroth, Christian Fleck, Christoph Enzensperger, Jochen Lehmann, Thomas Winckler. Evaluation of Homobivalent Carbolines as Designed Multiple Ligands for the Treatment of Neurodegenerative Disorders. J. Med. Chem. 2015;58:6710
    https://doi.org/10.1021/acs.jmedchem.5b00958
  131. Vikram S. Sawant, Hyeri Park, Soo Yoon Baek, Jieon Lee, Ji Won Choi, Ki Duk Park, Kyung Il Choi, Jihye Seong, Sanghee Lee, Hyunah Choo. Benzoxazoles as Selective Monoamine Oxidase B (MAO‐B) Inhibitors. Bulletin Korean Chem Soc 2019;40:457
    https://doi.org/10.1002/bkcs.11704
  132. Hayrettin O. Gülcan, Ilkay E. Orhan. The Main Targets Involved in Neuroprotection for the Treatment of Alzheimer’s Disease and Parkinson Disease. CPD 2020;26:509
    https://doi.org/10.2174/1381612826666200131103524
  133. Ibrahim Yildiz. Computational Analysis of the Nicotine Oxidoreductase Mechanism by the ONIOM Method. ACS Omega 2021;6:22422
    https://doi.org/10.1021/acsomega.1c03357
  134. Yu-Chen Wang, Xi Wang, Jiaji Yu, Feiyang Ma, Zhe Li, Yang Zhou, Samuel Zeng, Xiaoya Ma, Yan-Ruide Li, Adam Neal, Jie Huang, Angela To, Nicole Clarke, Sanaz Memarzadeh, Matteo Pellegrini, Lili Yang. Targeting monoamine oxidase A-regulated tumor-associated macrophage polarization for cancer immunotherapy. Nat Commun 2021;12
    https://doi.org/10.1038/s41467-021-23164-2
  135. Fabian Schumacher, Sudipta Chakraborty, Burkhard Kleuser, Erich Gulbins, Tanja Schwerdtle, Michael Aschner, Julia Bornhorst. Highly sensitive isotope-dilution liquid-chromatography–electrospray ionization–tandem-mass spectrometry approach to study the drug-mediated modulation of dopamine and serotonin levels in Caenorhabditis elegans. Talanta 2015;144:71
    https://doi.org/10.1016/j.talanta.2015.05.057
  136. John P.M. Finberg. Update on the pharmacology of selective inhibitors of MAO-A and MAO-B: Focus on modulation of CNS monoamine neurotransmitter release. Pharmacology & Therapeutics 2014;143:133
    https://doi.org/10.1016/j.pharmthera.2014.02.010
  137. Mariama Jaiteh, Alexey Zeifman, Marcus Saarinen, Per Svenningsson, Jose Bréa, Maria Isabel Loza, Jens Carlsson. Docking Screens for Dual Inhibitors of Disparate Drug Targets for Parkinson’s Disease. J. Med. Chem. 2018;61:5269
    https://doi.org/10.1021/acs.jmedchem.8b00204
  138. Ramakrishna Nirogi, Raghava Choudary Palacharla, Abdul Rasheed Mohammed, Arunkumar Manoharan, Ranjith kumar Ponnamaneni, Gopinadh Bhyrapuneni. Evaluation of metabolism dependent inhibition of CYP2B6 mediated bupropion hydroxylation in human liver microsomes by monoamine oxidase inhibitors and prediction of potential as perpetrators of drug interaction. Chemico-Biological Interactions 2015;230:9
    https://doi.org/10.1016/j.cbi.2015.01.028
  139. Masha G. Savelieff, Geewoo Nam, Juhye Kang, Hyuck Jin Lee, Misun Lee, Mi Hee Lim. Development of Multifunctional Molecules as Potential Therapeutic Candidates for Alzheimer’s Disease, Parkinson’s Disease, and Amyotrophic Lateral Sclerosis in the Last Decade. Chem. Rev. 2019;119:1221
    https://doi.org/10.1021/acs.chemrev.8b00138
  140. Rohit Bisht, Bhuwan Chandra Joshi, Ajudhiya Nath Kalia, Atish Prakash. Antioxidant-Rich Fraction of Urtica dioica Mediated Rescue of Striatal Mito-Oxidative Damage in MPTP-Induced Behavioral, Cellular, and Neurochemical Alterations in Rats. Mol Neurobiol 2017;54:5632
    https://doi.org/10.1007/s12035-016-0084-z
  141. Makoto Naoi, Wakako Maruyama, Hong Yi. Rasagiline prevents apoptosis induced by PK11195, a ligand of the outer membrane translocator protein (18 kDa), in SH-SY5Y cells through suppression of cytochrome c release from mitochondria. J Neural Transm 2013;120:1539
    https://doi.org/10.1007/s00702-013-1033-x
  142. Liping Huang, Minzhen Deng, Sheng Zhang, Shiyao Lu, Xuehong Gui, Yongqi Fang. β-asarone and levodopa coadministration increases striatal levels of dopamine and levodopa and improves behavioral competence in Parkinson's rat by enhancing dopa decarboxylase activity. Biomedicine & Pharmacotherapy 2017;94:666
    https://doi.org/10.1016/j.biopha.2017.07.125
  143. Mohammad Yasin Zamanian, Rosario Mireya Romero Parra, Afsaneh Soltani, Małgorzata Kujawska, Yasser Fakri Mustafa, Ghaidaa Raheem, Lateef Al-Awsi, Holya A. Lafta, Niloofar Taheri, Mahsa Heidari, Maryam Golmohammadi, Gholamreza Bazmandegan. Targeting Nrf2 signaling pathway and oxidative stress by resveratrol for Parkinson’s disease: an overview and update on new developments. Mol Biol Rep 2023;50:5455
    https://doi.org/10.1007/s11033-023-08409-1
  144. Satarupa Deb, Banashree Chetia Phukan, Muhammed Khairujjaman Mazumder, Ankumoni Dutta, Rajib Paul, Pallab Bhattacharya, Rajat Sandhir, Anupom Borah. Garcinol, a multifaceted sword for the treatment of Parkinson's disease. Neurochemistry International 2019;128:50
    https://doi.org/10.1016/j.neuint.2019.04.004
  145. Marco Bisaglia, Elisa Greggio, Mariano Beltramini, Luigi Bubacco. Dysfunction of dopamine homeostasis: clues in the hunt for novel Parkinson's disease therapies. FASEB j. 2013;27:2101
    https://doi.org/10.1096/fj.12-226852
  146. Jörg B. Schulz. Effects of selegiline and rasagiline on disease progression in Parkinson’s disease. Basal Ganglia 2012;2:S41
    https://doi.org/10.1016/j.baga.2012.08.003
  147. Makoto Naoi, Wakako Maruyama, Keiko Inaba-Hasegawa. Revelation in the neuroprotective functions of rasagiline and selegiline: the induction of distinct genes by different mechanisms. Expert Review of Neurotherapeutics 2013;13:671
    https://doi.org/10.1586/ern.13.60
  148. Zelan Wei, Tamara Satram-Maharaj, Bradley Chaharyn, Kelly Kuski, Paul R. Pennington, Xia Cao, Jennifer Chlan, Darrell D. Mousseau. Aspartic acid substitutions in monoamine oxidase-A reveal both catalytic-dependent and -independent influences on cell viability and proliferation. J Neural Transm 2012;119:1285
    https://doi.org/10.1007/s00702-012-0779-x
  149. Geunhyeong Jo, Seunghyun Ahn, Bong-Gyu Kim, Hye Ri Park, Young Hwa Kim, Hyun Ah Choo, Dongsoo Koh, Youhoon Chong, Joong-Hoon Ahn, Yoongho Lim. Chromenylchalcones with inhibitory effects on monoamine oxidase B. Bioorganic & Medicinal Chemistry 2013;21:7890
    https://doi.org/10.1016/j.bmc.2013.10.004
  150. Robin Fackrell, Camille B Carroll, Donald G Grosset, Biju Mohamed, Prashanth Reddy, Miriam Parry, Kallol Ray Chaudhuri, Tom Foltynie. Noninvasive options for ‘wearing-off’ in Parkinson's disease: a clinical consensus from a panel of UK Parkinson's disease specialists. Neurodegenerative Disease Management 2018;8:349
    https://doi.org/10.2217/nmt-2018-0020
  151. Lorenzo Antonio Justo, Rafael Durán, Miguel Alfonso, Daniel Fajardo, Lilian Rosana F. Faro. Effects and mechanism of action of isatin, a MAO inhibitor, on in vivo striatal dopamine release. Neurochemistry International 2016;99:147
    https://doi.org/10.1016/j.neuint.2016.06.012
  152. Narayan Chaurasiya, Francisco Leon, Ilias Muhammad, Babu Tekwani. Natural Products Inhibitors of Monoamine Oxidases—Potential New Drug Leads for Neuroprotection, Neurological Disorders, and Neuroblastoma. Molecules 2022;27:4297
    https://doi.org/10.3390/molecules27134297
  153. Yan Chen, Ying-yin Ni, Jie Liu, Jia-wei Lu, Fang Wang, Xiao-lin Wu, Ming-min Gu, Zhen-yu Lu, Zhu-gang Wang, Zhi-hua Ren. Dopamine receptor 3 might be an essential molecule in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity. BMC Neurosci 2013;14
    https://doi.org/10.1186/1471-2202-14-76
  154. John P. M. Finberg, Jose M. Rabey. Inhibitors of MAO-A and MAO-B in Psychiatry and Neurology. Front. Pharmacol. 2016;7
    https://doi.org/10.3389/fphar.2016.00340
  155. Ying Chang, Li-Bo Wang, Dan Li, Ke Lei, Song-Yan Liu. Efficacy of rasagiline for the treatment of Parkinson’s disease: an updated meta-analysis. Annals of Medicine 2017;49:421
    https://doi.org/10.1080/07853890.2017.1293285