en Experimental Neurobiology

Cited by CrossRef (82)

  1. Rémi Moirand, Filipe Galvao, Clément Dondé. Pramipexole and Selegiline Combination Therapy in a Case of Treatment-Resistant Depression. J Clin Psychopharmacol 2019;39:684
    https://doi.org/10.1097/JCP.0000000000001139
  2. Bijo Mathew, Della G. T. Parambi, Githa E. Mathew, Md. Sahab Uddin, Sini T. Inasu, Hoon Kim, Akash Marathakam, Mazhuvancherry Kesavan Unnikrishnan, Simone Carradori. Emerging therapeutic potentials of dual‐acting MAO and AChE inhibitors in Alzheimer's and Parkinson's diseases. Archiv der Pharmazie 2019;352
    https://doi.org/10.1002/ardp.201900177
  3. Lieven D. Declercq, Rik Vandenberghe, Koen Van Laere, Alfons Verbruggen, Guy Bormans. Drug Development in Alzheimer’s Disease: The Contribution of PET and SPECT. Front. Pharmacol. 2016;7
    https://doi.org/10.3389/fphar.2016.00088
  4. D. P. Devanand, R. Fremont. Tasman’s Psychiatry. 2016.
    https://doi.org/10.1007/978-3-030-42825-9_139-1
  5. Fabio Rizzo-Aguiar, Carlos A.D. Sousa, Xerardo Garcia-Mera, José E. Rodríguez-Borges. Synthesis and characterization of 1-pyrindane derivatives as rasagiline analogues. Chemical Data Collections 2016;5-6:21
    https://doi.org/10.1016/j.cdc.2016.10.001
  6. Jing Xu, Feifei Ji, Xiaocong Sun, Xiangrong Cao, Shen Li, Yasushi Ohizumi, Yuanqiang Guo. Characterization and Biological Evaluation of Diterpenoids from Casearia graveolens. J. Nat. Prod. 2015;78:2648
    https://doi.org/10.1021/acs.jnatprod.5b00583
  7. Gerard Esteban, Jennifer Allan, Abdelouahid Samadi, Andrea Mattevi, Mercedes Unzeta, José Marco-Contelles, Claudia Binda, Rona R. Ramsay. Kinetic and structural analysis of the irreversible inhibition of human monoamine oxidases by ASS234, a multi-target compound designed for use in Alzheimer's disease. Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics 2014;1844:1104
    https://doi.org/10.1016/j.bbapap.2014.03.006
  8. Yanier Nuñez-Figueredo, Jeney Ramírez-Sánchez, Gisele Hansel, Elisa Nicoloso Simões Pires, Nelson Merino, Odalys Valdes, René Delgado-Hernández, Alicia Lagarto Parra, Estael Ochoa-Rodríguez, Yamila Verdecia-Reyes, Christianne Salbego, Silvia L. Costa, Diogo O. Souza, Gilberto L. Pardo-Andreu. A novel multi-target ligand (JM-20) protects mitochondrial integrity, inhibits brain excitatory amino acid release and reduces cerebral ischemia injury in vitro and in vivo. Neuropharmacology 2014;85:517
    https://doi.org/10.1016/j.neuropharm.2014.06.009
  9. 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
  10. Wei Liu, Ming Lang, Moussa B.H. Youdim, Tamar Amit, Yewei Sun, Zaijun Zhang, Yuqiang Wang, Orly Weinreb. Design, synthesis and evaluation of novel dual monoamine-cholinesterase inhibitors as potential treatment for Alzheimer’s disease. Neuropharmacology 2016;109:376
    https://doi.org/10.1016/j.neuropharm.2016.06.013
  11. Avinash C. Tripathi, Savita Upadhyay, Sarvesh Paliwal, Shailendra K. Saraf. Privileged scaffolds as MAO inhibitors: Retrospect and prospects. European Journal of Medicinal Chemistry 2018;145:445
    https://doi.org/10.1016/j.ejmech.2018.01.003
  12. Zheng Liu, Wei Cai, Ming Lang, Ruizuo Yan, Zhenshen Li, Gaoxiao Zhang, Pei Yu, Yuqiang Wang, Yewei Sun, Zaijun Zhang. Neuroprotective Effects and Mechanisms of Action of Multifunctional Agents Targeting Free Radicals, Monoamine Oxidase B and Cholinesterase in Parkinson’s Disease Model. J Mol Neurosci 2017;61:498
    https://doi.org/10.1007/s12031-017-0891-3
  13. Young-Cheol Lim, Hyeongi Kim, Sang Moo Lim, Jin Su Kim. Genetic analysis of a novel antioxidant multi-target iron chelator, M30 protecting against chemotherapy-induced alopecia in mice. BMC Cancer 2019;19
    https://doi.org/10.1186/s12885-019-5323-z
  14. Pankaj Mishra, Amit Kumar, Gautam Panda. Anti-cholinesterase hybrids as multi-target-directed ligands against Alzheimer’s disease (1998–2018). Bioorganic & Medicinal Chemistry 2019;27:895
    https://doi.org/10.1016/j.bmc.2019.01.025
  15. Rona R. Ramsay. Molecular aspects of monoamine oxidase B. Progress in Neuro-Psychopharmacology and Biological Psychiatry 2016;69:81
    https://doi.org/10.1016/j.pnpbp.2016.02.005
  16. Aitor Carneiro, Eugenio Uriarte, Fernanda Borges, Maria João Matos. Propargylamine: an important moiety in drug discovery. Future Medicinal Chemistry 2023;15:211
    https://doi.org/10.4155/fmc-2022-0243
  17. Ali Ulvi Uca, Hasan Hüseyin Kozak. A case of rasagiline-induced spontaneous orgasms in a female patient. Parkinsonism & Related Disorders 2014;20:929
    https://doi.org/10.1016/j.parkreldis.2014.04.004
  18. Sonali S. Bharate, Serge Mignani, Ram A. Vishwakarma. Why Are the Majority of Active Compounds in the CNS Domain Natural Products? A Critical Analysis. J. Med. Chem. 2018;61:10345
    https://doi.org/10.1021/acs.jmedchem.7b01922
  19. Maria L Bolognesi, Michele Rossi. Encyclopedia of Life Sciences. 2018.
    https://doi.org/10.1002/9780470015902.a0028845
  20. Nikolay T. Tzvetkov, Sonja Hinz, Petra Küppers, Marcus Gastreich, Christa E. Müller. Indazole- and Indole-5-carboxamides: Selective and Reversible Monoamine Oxidase B Inhibitors with Subnanomolar Potency. J. Med. Chem. 2014;57:6679
    https://doi.org/10.1021/jm500729a
  21. Md. Sahab Uddin, Md. Tanvir Kabir, Md. Habibur Rahman, Md. Abdul Alim, Md. Motiar Rahman, Anurag Khatkar, Abdullah Al Mamun, Abdur Rauf, Bijo Mathew, Ghulam Md. Ashraf. Exploring the Multifunctional Neuroprotective Promise of Rasagiline Derivatives for Multi-Dysfunctional Alzheimer’s Disease. CPD 2020;26:4690
    https://doi.org/10.2174/1381612826666200406075044
  22. Sarah Chajkowski-Scarry, John M Rimoldi. Monoamine oxidase A and B substrates: probing the pathway for drug development. Future Medicinal Chemistry 2014;6:697
    https://doi.org/10.4155/fmc.14.23
  23. Ramón Cacabelos, Pablo Cacabelos, Clara Torrellas, Iván Tellado, Juan C. Carril. Pharmacogenomics in Drug Discovery and Development. 2014.
    https://doi.org/10.1007/978-1-4939-0956-8_13
  24. İsmail Okan Ateş, Asaf Evrim Evren, Begüm Nurpelin Sağlik, Leyla Yurttaş. New indane derivatives containing 2-hydrazinothiazole as potential acetylcholinesterase and monoamine oxidase-B inhibitors. 2021;76:417
    https://doi.org/10.1515/znc-2021-0058
  25. N. André Sasaki, Pascal Sonnet. A novel multi-target strategy to attenuate the progression of Parkinson's disease by diamine hybrid AGE/ALE inhibitor. Future Medicinal Chemistry 2021;13:2185
    https://doi.org/10.4155/fmc-2021-0217
  26. Elahe Zarini-Gakiye, Javad Amini, Nima Sanadgol, Gholamhassan Vaezi, Kazem Parivar. Recent Updates in the Alzheimer’s Disease Etiopathology and Possible Treatment Approaches: A Narrative Review of Current Clinical Trials. CMP 2020;13:273
    https://doi.org/10.2174/1874467213666200422090135
  27. Rona R. Ramsay, Magdalena Majekova, Milagros Medina, Massimo Valoti. Key Targets for Multi-Target Ligands Designed to Combat Neurodegeneration. Front. Neurosci. 2016;10
    https://doi.org/10.3389/fnins.2016.00375
  28. Sudhir Raghavan, David S. Baskin, Martyn A. Sharpe. Encyclopedia of Biological Chemistry III. 2016.
    https://doi.org/10.1016/B978-0-12-819460-7.00343-1
  29. María Eugenia Prados, Carmen Navarrete, Adela García-Martín, Isabel Lastres-Cubillo, Francisco Ponce-Díaz, José Martínez-Orgado, Eduardo Muñoz. VCE-005.1, an hypoxia mimetic betulinic acid derivative, induces angiogenesis and shows efficacy in a murine model of traumatic brain injury. Biomedicine & Pharmacotherapy 2023;162:114715
    https://doi.org/10.1016/j.biopha.2023.114715
  30. Awwad A. Radwan, Fars K. Alanazi, Mohammad Raish, Joazaizulfazli Jamalis. Design and synthesis of multi-functional small-molecule based inhibitors of amyloid-β aggregation: Molecular modeling and in vitro evaluation. PLoS ONE 2023;18:e0286195
    https://doi.org/10.1371/journal.pone.0286195
  31. Yash Pal Singh, Amruta Pandey, Swati Vishwakarma, Gyan Modi. A review on iron chelators as potential therapeutic agents for the treatment of Alzheimer’s and Parkinson’s diseases. Mol Divers 2019;23:509
    https://doi.org/10.1007/s11030-018-9878-4
  32. Paolo Guglielmi, Simone Carradori, Ilaria D’Agostino, Cristina Campestre, Jacobus P. Petzer. An updated patent review on monoamine oxidase (MAO) inhibitors. Expert Opinion on Therapeutic Patents 2022;32:849
    https://doi.org/10.1080/13543776.2022.2083501
  33. Ganesh N. Pandian, Rhys D. Taylor, Syed Junetha, Abhijit Saha, Chandran Anandhakumar, Thangavel Vaijayanthi, Hiroshi Sugiyama. Alteration of epigenetic program to recover memory and alleviate neurodegeneration: prospects of multi-target molecules. Biomater. Sci. 2014;2:1043
    https://doi.org/10.1039/C4BM00068D
  34. Paolo Guglielmi, Simone Carradori, Alessandra Ammazzalorso, Daniela Secci. Novel approaches to the discovery of selective human monoamine oxidase-B inhibitors: is there room for improvement?. Expert Opinion on Drug Discovery 2019;14:995
    https://doi.org/10.1080/17460441.2019.1637415
  35. Bilqees Sameem, Mina Saeedi, Mohammad Mahdavi, Abbas Shafiee. A review on tacrine-based scaffolds as multi-target drugs (MTDLs) for Alzheimer's disease. European Journal of Medicinal Chemistry 2017;128:332
    https://doi.org/10.1016/j.ejmech.2016.10.060
  36. David S. Goldstein, Irwin J. Kopin, Yehonatan Sharabi. Catecholamine autotoxicity. Implications for pharmacology and therapeutics of Parkinson disease and related disorders. Pharmacology & Therapeutics 2014;144:268
    https://doi.org/10.1016/j.pharmthera.2014.06.006
  37. Cristian Follmer. Monoamine oxidase and α-synuclein as targets in Parkinson’s disease therapy. Expert Review of Neurotherapeutics 2014;14:703
    https://doi.org/10.1586/14737175.2014.920235
  38. Salvatore Bongarzone, Matteo Staderini, Maria Laura Bolognesi. Multitarget ligands and theranostics: sharpening the medicinal chemistry sword against prion diseases. Future Medicinal Chemistry 2014;6:1017
    https://doi.org/10.4155/fmc.14.56
  39. Hugo Juárez Olguín, David Calderón Guzmán, Ernestina Hernández García, Gerardo Barragán Mejía. The Role of Dopamine and Its Dysfunction as a Consequence of Oxidative Stress. Oxidative Medicine and Cellular Longevity 2016;2016:1
    https://doi.org/10.1155/2016/9730467
  40. Veda Prachayasittikul, Ratchanok Pingaew, Supaluk Prachayasittikul, Virapong Prachayasittikul. 8-Hydroxyquinolines: A Promising Pharmacophore Potentially Developed as Disease-Modifying Agents for Neurodegenerative Diseases: A Review. HETEROCYCLES 2022;105:202
    https://doi.org/10.3987/REV-22-SR(R)6
  41. Maria Laura Bolognesi, Andrea Cavalli. Multitarget Drug Discovery and Polypharmacology. ChemMedChem 2016;11:1190
    https://doi.org/10.1002/cmdc.201600161
  42. S. O. Bachurin. A review of drugs for treatment of Alzheimer’s disease in clinical trials: main trends. Z. nevrol. psikhiatr. im. S.S. Korsakova 2016;116:77
    https://doi.org/10.17116/jnevro20161168177-87
  43. Jing Xu, Jing Kang, Xiaocong Sun, Xiangrong Cao, Kasimu Rena, Dongho Lee, Quanhui Ren, Shen Li, Yasushi Ohizumi, Yuanqiang Guo. Di- and Triterpenoids from the Leaves of Casearia balansae and Neurite Outgrowth Promoting Effects of PC12 Cells. J. Nat. Prod. 2016;79:170
    https://doi.org/10.1021/acs.jnatprod.5b00815
  44. Banibrata Das, Ashoka Kandegedara, Liping Xu, Tamara Antonio, Timothy Stemmler, Maarten E. A. Reith, Aloke K. Dutta. A Novel Iron(II) Preferring Dopamine Agonist Chelator as Potential Symptomatic and Neuroprotective Therapeutic Agent for Parkinson’s Disease. ACS Chem. Neurosci. 2017;8:723
    https://doi.org/10.1021/acschemneuro.6b00356
  45. Anastasios Fotinos, Yongjin Zhu, Lilly L. J. Mao, Nazem Atassi, Edward W. Zhou, Sarfraz Ahmad, Yingjun Guan, James D. Berry, Merit E. Cudkowicz, Xin Wang. Melatonin, Neuroprotective Agents and Antidepressant Therapy. 2017.
    https://doi.org/10.1007/978-81-322-2803-5_51
  46. Payel Bhattacharjee, Jyotirmoy Mitra, Debasish Bhattacharyya. Toxins and Drug Discovery. 2017.
    https://doi.org/10.1007/978-94-007-6452-1_11
  47. Pablo Duarte, Antonio Cuadrado, Rafael León. Reactive Oxygen Species. 2017.
    https://doi.org/10.1007/164_2020_384
  48. JessicaD Panes, Aline Wendt, Oscar Ramirez-Molina, PatricioA Castro, Jorge Fuentealba. Deciphering the role of PGC-1α in neurological disorders: from mitochondrial dysfunction to synaptic failure. Neural Regen Res 2022;17:237
    https://doi.org/10.4103/1673-5374.317957
  49. Niraj Kumar Jha, Saurabh Kumar Jha, Renu Sharma, Dhiraj Kumar, Rashmi K. Ambasta, Pravir Kumar. Hypoxia-Induced Signaling Activation in Neurodegenerative Diseases: Targets for New Therapeutic Strategies. JAD 2018;62:15
    https://doi.org/10.3233/JAD-170589
  50. Mrudang Shah, Subramanian Rajagopalan, Liping Xu, Chandrashekhar Voshavar, Yevgeniya Shurubor, Flint Beal, Julie K. Andersen, Aloke K. Dutta. The high‐affinity D2/D3 agonist D512 protects PC12 cells from 6‐OHDA‐induced apoptotic cell death and rescues dopaminergic neurons in the MPTP mouse model of Parkinson's disease. Journal of Neurochemistry 2014;131:74
    https://doi.org/10.1111/jnc.12767
  51. Andrew Holt, Dmitriy Matveychuk, Darrell D. Mousseau, Glen B. Baker. Encyclopedia of Psychopharmacology. 2014.
    https://doi.org/10.1007/978-3-642-27772-6_371-2
  52. Amirhossein Maghsoudi, Saideh Fakharzadeh, Maryam Hafizi, Maryam Abbasi, Fatemeh Kohram, Shima Sardab, Abbas Tahzibi, Somayeh Kalanaky, Mohammad Hassan Nazaran. Neuroprotective effects of three different sizes nanochelating based nano complexes in MPP(+) induced neurotoxicity. Apoptosis 2015;20:298
    https://doi.org/10.1007/s10495-014-1069-x
  53. Rebecca E. Hughes, Katarina Nikolic, Rona R. Ramsay. One for All? Hitting Multiple Alzheimer's Disease Targets with One Drug. Front. Neurosci. 2016;10
    https://doi.org/10.3389/fnins.2016.00177
  54. Satoka Kasai, Toru Yoshihara, Olga Lopatina, Katsuhiko Ishihara, Haruhiro Higashida. Selegiline Ameliorates Depression-Like Behavior in Mice Lacking the CD157/BST1 Gene, a Risk Factor for Parkinson’s Disease. Front. Behav. Neurosci. 2017;11
    https://doi.org/10.3389/fnbeh.2017.00075
  55. Nerea Alonso, Olga Caamaño, Francisco J. Romero-Duran, Feng Luan, M. Natália D. S. Cordeiro, Matilde Yañez, Humberto González-Díaz, Xerardo García-Mera. Model for High-Throughput Screening of Multitarget Drugs in Chemical Neurosciences: Synthesis, Assay, and Theoretic Study of Rasagiline Carbamates. ACS Chem. Neurosci. 2013;4:1393
    https://doi.org/10.1021/cn400111n
  56. Payel Bhattacharjee, Jyotirmoy Mitra, Debasish Bhattacharyya. Toxins and Drug Discovery. 2013.
    https://doi.org/10.1007/978-94-007-6726-3_11-1
  57. Hiroko Munakata, Risa Ishikawa, Toshiaki Saitoh, Toshie Kambe, Terumasa Chiba, Kyoji Taguchi, Kenji Abe. Preventative effects of 1-methyl-1,2,3,4-tetrahydroisoquinoline derivatives (N-functional group loading) on MPTP-induced parkinsonism in mice. Can. J. Physiol. Pharmacol. 2022;100:594
    https://doi.org/10.1139/cjpp-2021-0659
  58. Akhil Kumar, Ashok Sharma. Computational Modeling of Drugs Against Alzheimer’s Disease. 2022.
    https://doi.org/10.1007/978-1-4939-7404-7_19
  59. . Parkinson's Disease. 2022.
    https://doi.org/10.1017/CBO9781107284210.001
  60. Jan Korábečný, Eugenie Nepovimová, Tereza Cikánková, Katarína Špilovská, Lucie Vašková, Eva Mezeiová, Kamil Kuča, Jana Hroudová. Newly Developed Drugs for Alzheimer’s Disease in Relation to Energy Metabolism, Cholinergic and Monoaminergic Neurotransmission. Neuroscience 2018;370:191
    https://doi.org/10.1016/j.neuroscience.2017.06.034
  61. Jiqing Cao, Jianwei Hou, Jing Ping, Dongming Cai. Advances in developing novel therapeutic strategies for Alzheimer’s disease. Mol Neurodegeneration 2018;13
    https://doi.org/10.1186/s13024-018-0299-8
  62. Shinghung Mak, Wenming Li, Hongjun Fu, Jialie Luo, Wei Cui, Shengquan Hu, Yuanping Pang, Paul R. Carlier, Karl Wahkeung Tsim, Rongbiao Pi, Yifan Han. Promising tacrine/huperzine A‐based dimeric acetylcholinesterase inhibitors for neurodegenerative disorders: From relieving symptoms to modifying diseases through multitarget. Journal of Neurochemistry 2021;158:1381
    https://doi.org/10.1111/jnc.15379
  63. Lovedeep Singh, Harpreet Kaur, Girish Chandra Arya, Rajbir Bhatti. Neuroprotective potential of formononetin, a naturally occurring isoflavone phytoestrogen. Chem Biol Drug Des 2024;103
    https://doi.org/10.1111/cbdd.14353
  64. Anne Robert, Yan Liu, Michel Nguyen, Bernard Meunier. Regulation of Copper and Iron Homeostasis by Metal Chelators: A Possible Chemotherapy for Alzheimer’s Disease. Acc. Chem. Res. 2015;48:1332
    https://doi.org/10.1021/acs.accounts.5b00119
  65. Kristin Part, Kai Künnis-Beres, Helen Poska, Tiit Land, Ruth Shimmo, Sandra Zetterström Fernaeus. Amyloid β25–35 induced ROS-burst through NADPH oxidase is sensitive to iron chelation in microglial Bv2 cells. Brain Research 2015;1629:282
    https://doi.org/10.1016/j.brainres.2015.09.034
  66. Damijan Knez, Matej Sova, Urban Košak, Stanislav Gobec. Dual inhibitors of cholinesterases and monoamine oxidases for Alzheimer’s disease. Future Medicinal Chemistry 2017;9:811
    https://doi.org/10.4155/fmc-2017-0036
  67. Cheng-Xin Gong, Fei Liu, Khalid Iqbal, G. Perry, J. Avila, P.I. Moreira, A.A. Sorensen, M. Tabaton. Multifactorial Hypothesis and Multi-Targets for Alzheimer’s Disease. JAD 2018;64:S107
    https://doi.org/10.3233/JAD-179921
  68. Sven-Eric Pålhagen. Parkinson's Disease. 2018.
    https://doi.org/10.1017/CBO9781107284210.004
  69. Chun-Sing Lam, Jing-Jie Li, George Lim Tipoe, Moussa B. H. Youdim, Man-Lung Fung, Muzamil Ahmad. Monoamine oxidase A upregulated by chronic intermittent hypoxia activates indoleamine 2,3-dioxygenase and neurodegeneration. PLoS ONE 2017;12:e0177940
    https://doi.org/10.1371/journal.pone.0177940
  70. K. Lutsenko, S. Hagenow, A. Affini, D. Reiner, H. Stark. Rasagiline derivatives combined with histamine H3 receptor properties. Bioorganic & Medicinal Chemistry Letters 2019;29:126612
    https://doi.org/10.1016/j.bmcl.2019.08.016
  71. Dawid Panek, Tomasz Wichur, Justyna Godyń , Anna Pasieka, Barbara Malawska. Advances toward multifunctional cholinesterase and β-amyloid aggregation inhibitors. Future Medicinal Chemistry 2017;9:1835
    https://doi.org/10.4155/fmc-2017-0094
  72. Carlos A. D. Sousa, Ivo E. Sampaio-Dias, Fabio Rizzo-Aguiar, Xerardo Garcia-Mera, José E. Rodríguez-Borges. Enantiopure synthesis of 7-(1-pyrindanyl)propargyl ethers as rasagiline analogues via chemical or enzymatic resolution of 1-pyrindan-7-ol. RSC Adv. 2015;5:104509
    https://doi.org/10.1039/C5RA24868J
  73. Ashi Mannan, Thakur Gurjeet Singh, Varinder Singh, Nikhil Garg, Amarjot Kaur, Manjinder Singh. Insights into the Mechanism of the Therapeutic Potential of Herbal Monoamine Oxidase Inhibitors in Neurological Diseases. CDT 2022;23:286
    https://doi.org/10.2174/1389450122666210707120256
  74. Rona R. Ramsay, Giuseppe Di Giovanni. Interdisciplinary Chemical Approaches for Neuropathology. CNS Neurosci Ther 2014;20:571
    https://doi.org/10.1111/cns.12297
  75. Robert Otto, Robert Penzis, Friedemann Gaube, Thomas Winckler, Dorothea Appenroth, Christian Fleck, Christian Tränkle, Jochen Lehmann, Christoph Enzensperger. Beta and gamma carboline derivatives as potential anti-Alzheimer agents: A comparison. European Journal of Medicinal Chemistry 2014;87:63
    https://doi.org/10.1016/j.ejmech.2014.09.048
  76. Rona Ramsay, Keith Tipton. Assessment of Enzyme Inhibition: A Review with Examples from the Development of Monoamine Oxidase and Cholinesterase Inhibitory Drugs. Molecules 2017;22:1192
    https://doi.org/10.3390/molecules22071192
  77. Zhi-Bin Tong, Ruili Huang, Yuhong Wang, Carleen A. Klumpp-Thomas, John C. Braisted, Zina Itkin, Paul Shinn, Menghang Xia, Anton Simeonov, David L. Gerhold. The Toxmatrix: Chemo-Genomic Profiling Identifies Interactions That Reveal Mechanisms of Toxicity. Chem. Res. Toxicol. 2018;31:127
    https://doi.org/10.1021/acs.chemrestox.7b00290
  78. Viktoriya Ivasiv, Claudia Albertini, Ana E. Gonçalves, Michele Rossi, Maria L. Bolognesi. Molecular Hybridization as a Tool for Designing Multitarget Drug Candidates for Complex Diseases. CTMC 2019;19:1694
    https://doi.org/10.2174/1568026619666190619115735
  79. J. Wallach, T. Colestock, A. Adejare. Drug Discovery Approaches for the Treatment of Neurodegenerative Disorders. 2019.
    https://doi.org/10.1016/B978-0-12-802810-0.00006-4
  80. M. S. Kukharsky, R. K. Ovchinnikov, S. O. Bachurin. Molecular aspects of the pathogenesis and current approaches to pharmacological correction of Alzheimer’s disease. Z. nevrol. psikhiatr. im. S.S. Korsakova 2015;115:103
    https://doi.org/10.17116/jnevro20151156103-114
  81. Chun-Sing Lam, George Lim Tipoe, Johnny Kong-Ching Wong, Moussa B. H. Youdim, Man-Lung Fung, Kenji Hashimoto. M30 Antagonizes Indoleamine 2,3-Dioxygenase Activation and Neurodegeneration Induced by Corticosterone in the Hippocampus. PLoS ONE 2016;11:e0166966
    https://doi.org/10.1371/journal.pone.0166966
  82. Qi Mao, Wang-zhi Qin, Ao Zhang, Na Ye. Recent advances in dopaminergic strategies for the treatment of Parkinson’s disease. Acta Pharmacol Sin 2020;41:471
    https://doi.org/10.1038/s41401-020-0365-y