ACS Publications. Most Trusted. Most Cited. Most Read
Structural Basis for the Synthesis of Indirubins as Potent and Selective Inhibitors of Glycogen Synthase Kinase-3 and Cyclin-Dependent Kinases
My Activity
    Article

    Structural Basis for the Synthesis of Indirubins as Potent and Selective Inhibitors of Glycogen Synthase Kinase-3 and Cyclin-Dependent Kinases
    Click to copy article linkArticle link copied!

    View Author Information
    Laboratory of Pharmacognosy and Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, University of Athens, Panepistimiopolis Zografou, GR-15771 Athens, Greece, Structural Biology Unit, Department of Experimental Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milano, Italy, 3, Centre for Structural Biology, Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, U.K., C.N.R.S., Cell Cycle Group, Station Biologique, B.P. 74, 29682 ROSCOFF Cedex, Bretagne, France, and Laboratory of Molecular & Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, New York 10021
    Other Access Options

    Journal of Medicinal Chemistry

    Cite this: J. Med. Chem. 2004, 47, 4, 935–946
    Click to copy citationCitation copied!
    https://doi.org/10.1021/jm031016d
    Published January 8, 2004
    Copyright © 2004 American Chemical Society

    Abstract

    Click to copy section linkSection link copied!
    Abstract Image

    Pharmacological inhibitors of glycogen synthase kinase-3 (GSK-3) and cyclin-dependent kinases have a promising potential for applications against several neurodegenerative diseases such as Alzheimer's disease. Indirubins, a family of bis-indoles isolated from various natural sources, are potent inhibitors of several kinases, including GSK-3. Using the cocrystal structures of various indirubins with GSK-3β, CDK2 and CDK5/p25, we have modeled the binding of indirubins within the ATP-binding pocket of these kinases. This modeling approach provided some insight into the molecular basis of indirubins' action and selectivity and allowed us to forecast some improvements of this family of bis-indoles as kinase inhibitors. Predicted molecules, including 6-substituted and 5,6-disubstituted indirubins, were synthesized and evaluated as CDK and GSK-3 inhibitors. Control, kinase-inactive indirubins were obtained by introduction of a methyl substitution on N1.

    Copyright © 2004 American Chemical Society

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. Add or change your institution or let them know you’d like them to include access.

     University of Athens.

    *

     Corresponding authors:  A. L. Skaltsounis (tel:  +3210 7274598, fax:  +3210 7274594, e-mail:  [email protected]) or L. Meijer (e-mail:  [email protected]).

     European Institute of Oncology.

    §

     Chester Beatty Laboratories.

     C.N.R.S.

     The Rockefeller University.

    Cited By

    Click to copy section linkSection link copied!

    This article is cited by 309 publications.

    1. Nicolai A. Aksenov, Alexander V. Aksenov, Lidiya A. Prityko, Dmitrii A. Aksenov, Daria S. Aksenova, Mezvah A. Nobi, Michael Rubin. Oxidative Cyclization of 4-(2-Aminophenyl)-4-oxo-2-phenylbutanenitriles into 2-(3-Oxoindolin-2-ylidene)acetonitriles. ACS Omega 2022, 7 (16) , 14345-14356. https://doi.org/10.1021/acsomega.2c01238
    2. Zhuoxian Cao, Fenfen Yang, Jie Wang, Zhicheng Gu, Shuxian Lin, Pan Wang, Jianxiong An, Ting Liu, Yan Li, Yongjun Li, Hening Lin, Yonglong Zhao, Bin He. Indirubin Derivatives as Dual Inhibitors Targeting Cyclin-Dependent Kinase and Histone Deacetylase for Treating Cancer. Journal of Medicinal Chemistry 2021, 64 (20) , 15280-15296. https://doi.org/10.1021/acs.jmedchem.1c01311
    3. Huifang Yin, Hongping Chen, Meng Yan, Zhikun Li, Rongdi Yang, Yanjiao Li, Yanfang Wang, Jianyi Guan, Huili Mao, Yan Wang, Yuyang Zhang. Efficient Bioproduction of Indigo and Indirubin by Optimizing a Novel Terpenoid Cyclase XiaI in Escherichia coli. ACS Omega 2021, 6 (31) , 20569-20576. https://doi.org/10.1021/acsomega.1c02679
    4. Weigang Huang, Adam J. Carr, Nicole Hajicek, Miri Sokolovski, Edhriz Siraliev-Perez, P. Brian Hardy, Kenneth H. Pearce, John Sondek, Qisheng Zhang. A High-Throughput Assay to Identify Allosteric Inhibitors of the PLC-γ Isozymes Operating at Membranes. Biochemistry 2020, 59 (41) , 4029-4038. https://doi.org/10.1021/acs.biochem.0c00511
    5. Luke N. Latimer, Zachary N. Russ, James Lucas, John E. Dueber. Exploration of Acetylation as a Base-Labile Protecting Group in Escherichia coli for an Indigo Precursor. ACS Synthetic Biology 2020, 9 (10) , 2775-2783. https://doi.org/10.1021/acssynbio.0c00297
    6. Nikitia Mexia, Stamatis Koutrakis, Guochun He, Alexios-Leandros Skaltsounis, Michael S. Denison, Prokopios Magiatis. A Biomimetic, One-Step Transformation of Simple Indolic Compounds to Malassezia-Related Alkaloids with High AhR Potency and Efficacy. Chemical Research in Toxicology 2019, 32 (11) , 2238-2249. https://doi.org/10.1021/acs.chemrestox.9b00270
    7. Joonwon Kim, Jeongchan Lee, Pyung-Gang Lee, Eun-Jung Kim, Wolfgang Kroutil, Byung-gee Kim. Elucidating Cysteine-Assisted Synthesis of Indirubin by a Flavin-Containing Monooxygenase. ACS Catalysis 2019, 9 (10) , 9539-9544. https://doi.org/10.1021/acscatal.9b02613
    8. Xue-Bing Chen, Sheng-Li Xiong, Zhi-Xu Xie, Yong-Chao Wang, Wei Liu. Three-Component One-Pot Synthesis of Highly Functionalized Bis-Indole Derivatives. ACS Omega 2019, 4 (7) , 11832-11837. https://doi.org/10.1021/acsomega.9b01159
    9. Solomon Tadesse, Elizabeth C. Caldon, Wayne Tilley, Shudong Wang. Cyclin-Dependent Kinase 2 Inhibitors in Cancer Therapy: An Update. Journal of Medicinal Chemistry 2019, 62 (9) , 4233-4251. https://doi.org/10.1021/acs.jmedchem.8b01469
    10. Najat Aourz, Ann-Sophie K. Serruys, Joëlle N. Chabwine, Pascal Byenda Balegamire, Tatiana Afrikanova, RuAngelie Edrada-Ebel, Alexander I. Grey, Appolinary R. Kamuhabwa, Laura Walrave, Camila V. Esguerra, Fred van Leuven, Peter A. M. de Witte, Ilse Smolders, Alexander D. Crawford. Identification of GSK-3 as a Potential Therapeutic Entry Point for Epilepsy. ACS Chemical Neuroscience 2019, 10 (4) , 1992-2003. https://doi.org/10.1021/acschemneuro.8b00281
    11. Radek Jorda, Denisa Hendrychová, Jiří Voller, Eva Řezníčková, Tomáš Gucký, Vladimír Kryštof. How Selective Are Pharmacological Inhibitors of Cell-Cycle-Regulating Cyclin-Dependent Kinases?. Journal of Medicinal Chemistry 2018, 61 (20) , 9105-9120. https://doi.org/10.1021/acs.jmedchem.8b00049
    12. Jisook Park, Alexandre Jean, David Y.-K. Chen. Organocatalytic and Late-Stage CH-Functionalization Enabled Asymmetric Synthesis of Communesin F and Putative Communesins. The Journal of Organic Chemistry 2018, 83 (13) , 6936-6957. https://doi.org/10.1021/acs.joc.7b02426
    13. Nicolas Gaboriaud-Kolar, Vasillios Myrianthopoulos, Konstantina Vougogiannopoulou, Panagiotis Gerolymatos, David A. Horne, Richard Jove, Emmanuel Mikros, Sangkil Nam, and Alexios-Leandros Skaltsounis . Natural-Based Indirubins Display Potent Cytotoxicity toward Wild-Type and T315I-Resistant Leukemia Cell Lines. Journal of Natural Products 2016, 79 (10) , 2464-2471. https://doi.org/10.1021/acs.jnatprod.6b00285
    14. Stewart A. Low, Chris V. Galliford, Jiyuan Yang, Philip S. Low, and Jindřich Kopeček . Biodistribution of Fracture-Targeted GSK3β Inhibitor-Loaded Micelles for Improved Fracture Healing. Biomacromolecules 2015, 16 (10) , 3145-3153. https://doi.org/10.1021/acs.biomac.5b00777
    15. Christopher Merritt, Lisseth E. Silva, Angela L. Tanner, Kenneth Stuart, and Michael P. Pollastri . Kinases as Druggable Targets in Trypanosomatid Protozoan Parasites. Chemical Reviews 2014, 114 (22) , 11280-11304. https://doi.org/10.1021/cr500197d
    16. Lukas K. Filak, Danuta S. Kalinowski, Theresa J. Bauer, Des R. Richardson, and Vladimir B. Arion . Effect of the Piperazine Unit and Metal-Binding Site Position on the Solubility and Anti-Proliferative Activity of Ruthenium(II)- and Osmium(II)- Arene Complexes of Isomeric Indolo[3,2-c]quinoline—Piperazine Hybrids. Inorganic Chemistry 2014, 53 (13) , 6934-6943. https://doi.org/10.1021/ic500825j
    17. Carlo Pergola, Nicolas Gaboriaud-Kolar, Nadine Jestädt, Stefanie König, Marina Kritsanida, Anja M. Schaible, Haokun Li, Ulrike Garscha, Christina Weinigel, Dagmar Barz, Kai F. Albring, Otmar Huber, Alexios L. Skaltsounis, and Oliver Werz . Indirubin Core Structure of Glycogen Synthase Kinase-3 Inhibitors as Novel Chemotype for Intervention with 5-Lipoxygenase. Journal of Medicinal Chemistry 2014, 57 (9) , 3715-3723. https://doi.org/10.1021/jm401740w
    18. Valeria La Pietra, Giuseppe La Regina, Antonio Coluccia, Valeria Famiglini, Sveva Pelliccia, Batya Plotkin, Hagit Eldar-Finkelman, Andrea Brancale, Carlo Ballatore, Alex Crowe, Kurt R. Brunden, Luciana Marinelli, Ettore Novellino, and Romano Silvestri . Design, Synthesis, and Biological Evaluation of 1-Phenylpyrazolo[3,4-e]pyrrolo[3,4-g]indolizine-4,6(1H,5H)-diones as New Glycogen Synthase Kinase-3β Inhibitors. Journal of Medicinal Chemistry 2013, 56 (24) , 10066-10078. https://doi.org/10.1021/jm401466v
    19. Vassilios Myrianthopoulos, Marina Kritsanida, Nicolas Gaboriaud-Kolar, Prokopios Magiatis, Yoan Ferandin, Emilie Durieu, Olivier Lozach, Daniel Cappel, Meera Soundararajan, Panagis Filippakopoulos, Woody Sherman, Stefan Knapp, Laurent Meijer, Emmanuel Mikros, and Alexios-Leandros Skaltsounis . Novel Inverse Binding Mode of Indirubin Derivatives Yields Improved Selectivity for DYRK Kinases. ACS Medicinal Chemistry Letters 2013, 4 (1) , 22-26. https://doi.org/10.1021/ml300207a
    20. Tania Tahtouh, Jonathan M. Elkins, Panagis Filippakopoulos, Meera Soundararajan, Guillaume Burgy, Emilie Durieu, Claude Cochet, Ralf S. Schmid, Donald C. Lo, Florent Delhommel, Anselm E. Oberholzer, Laurence H. Pearl, François Carreaux, Jean-Pierre Bazureau, Stefan Knapp, and Laurent Meijer . Selectivity, Cocrystal Structures, and Neuroprotective Properties of Leucettines, a Family of Protein Kinase Inhibitors Derived from the Marine Sponge Alkaloid Leucettamine B. Journal of Medicinal Chemistry 2012, 55 (21) , 9312-9330. https://doi.org/10.1021/jm301034u
    21. Haixia Zou, Liyan Zhou, Yuanzhen Li, Yi Cui, Hanbing Zhong, Zhengying Pan, Zhen Yang and Junmin Quan . Benzo[e]isoindole-1,3-diones as Potential Inhibitors of Glycogen Synthase Kinase-3 (GSK-3). Synthesis, Kinase Inhibitory Activity, Zebrafish Phenotype, and Modeling of Binding Mode. Journal of Medicinal Chemistry 2010, 53 (3) , 994-1003. https://doi.org/10.1021/jm9013373
    22. Marina Kritsanida, Prokopios Magiatis, Alexios-Leandros Skaltsounis, Youyi Peng, Peng Li and Lawrence P. Wennogle. Synthesis and Antiproliferative Activity of 7-Azaindirubin-3′-oxime, a 7-Aza Isostere of the Natural Indirubin Pharmacophore. Journal of Natural Products 2009, 72 (12) , 2199-2202. https://doi.org/10.1021/np9003905
    23. Morihisa Saitoh, Jun Kunitomo, Eiji Kimura, Hiroki Iwashita, Yumiko Uno, Tomohiro Onishi, Noriko Uchiyama, Tomohiro Kawamoto, Toshimasa Tanaka, Clifford D. Mol, Douglas R. Dougan, Garret P. Textor, Gyorgy P. Snell, Masayuki Takizawa, Fumio Itoh and Masakuni Kori . 2-{3-[4-(Alkylsulfinyl)phenyl]-1-benzofuran-5-yl}-5-methyl-1,3,4-oxadiazole Derivatives as Novel Inhibitors of Glycogen Synthase Kinase-3β with Good Brain Permeability. Journal of Medicinal Chemistry 2009, 52 (20) , 6270-6286. https://doi.org/10.1021/jm900647e
    24. Gordon M. Cragg, Paul G. Grothaus and David J. Newman. Impact of Natural Products on Developing New Anti-Cancer Agents. Chemical Reviews 2009, 109 (7) , 3012-3043. https://doi.org/10.1021/cr900019j
    25. Konstantina Vougogiannopoulou, Yoan Ferandin, Karima Bettayeb, Vassilios Myrianthopoulos, Olivier Lozach, Yunzhen Fan, Carl Hirschie Johnson, Prokopios Magiatis, Alexios-Leandros Skaltsounis, Emmanuel Mikros and Laurent Meijer . Soluble 3′,6-Substituted Indirubins with Enhanced Selectivity toward Glycogen Synthase Kinase -3 Alter Circadian Period. Journal of Medicinal Chemistry 2008, 51 (20) , 6421-6431. https://doi.org/10.1021/jm800648y
    26. Hendrik Stukenbrock, Rainer Mussmann, Marcus Geese, Yoan Ferandin, Olivier Lozach, Thomas Lemcke, Simone Kegel, Alexander Lomow, Ulrike Burk, Cord Dohrmann, Laurent Meijer, Matthias Austen and Conrad Kunick. 9-Cyano-1-azapaullone (Cazpaullone), a Glycogen Synthase Kinase-3 (GSK-3) Inhibitor Activating Pancreatic β Cell Protection and Replication. Journal of Medicinal Chemistry 2008, 51 (7) , 2196-2207. https://doi.org/10.1021/jm701582f
    27. Chao Che, Jing Xiang, Guo-Xin Wang, Reza Fathi, Jun-Min Quan and Zhen Yang . One-Pot Synthesis of Quinoline-Based Tetracycles by a Tandem Three-Component Reaction. Journal of Combinatorial Chemistry 2007, 9 (6) , 982-989. https://doi.org/10.1021/cc070058a
    28. Vassilios Myrianthopoulos,, Prokopios Magiatis,, Yoan Ferandin,, Alexios-Leandros Skaltsounis,, Laurent Meijer, and, Emmanuel Mikros. An Integrated Computational Approach to the Phenomenon of Potent and Selective Inhibition of Aurora Kinases B and C by a Series of 7-Substituted Indirubins. Journal of Medicinal Chemistry 2007, 50 (17) , 4027-4037. https://doi.org/10.1021/jm070077z
    29. Filippo Marchioni,, Jian Yang,, Wesley Walker, and, Fred Wudl. A Low Band Gap Conjugated Polymer for Supercapacitor Devices. The Journal of Physical Chemistry B 2006, 110 (44) , 22202-22206. https://doi.org/10.1021/jp063849w
    30. Yoan Ferandin,, Karima Bettayeb,, Marina Kritsanida,, Olivier Lozach,, Panagiotis Polychronopoulos,, Prokopios Magiatis,, Alexios-Leandros Skaltsounis, and, Laurent Meijer. 3‘-Substituted 7-Halogenoindirubins, a New Class of Cell Death Inducing Agents. Journal of Medicinal Chemistry 2006, 49 (15) , 4638-4649. https://doi.org/10.1021/jm060314i
    31. Tímea Polgár,, Andrea Baki,, Györgyi I. Szendrei, and, György M. Keserűu. Comparative Virtual and Experimental High-Throughput Screening for Glycogen Synthase Kinase-3β Inhibitors. Journal of Medicinal Chemistry 2005, 48 (25) , 7946-7959. https://doi.org/10.1021/jm050504d
    32. Ana Martinez,, Mercedes Alonso,, Ana Castro,, Isabel Dorronsoro,, J. Luis Gelpí,, F. Javier Luque,, Concepción Pérez, and, Francisco J. Moreno. SAR and 3D-QSAR Studies on Thiadiazolidinone Derivatives:  Exploration of Structural Requirements for Glycogen Synthase Kinase 3 Inhibitors. Journal of Medicinal Chemistry 2005, 48 (23) , 7103-7112. https://doi.org/10.1021/jm040895g
    33. Anna Vulpetti,, Patrizia Crivori,, Alexander Cameron,, Jay Bertrand,, Maria Gabriella Brasca,, Roberto D'Alessio, and, Paolo Pevarello. Structure-Based Approaches to Improve Selectivity:  CDK2−GSK3β Binding Site Analysis. Journal of Chemical Information and Modeling 2005, 45 (5) , 1282-1290. https://doi.org/10.1021/ci0500280
    34. William M. Rockey and, Adrian H. Elcock. Rapid Computational Identification of the Targets of Protein Kinase Inhibitors. Journal of Medicinal Chemistry 2005, 48 (12) , 4138-4152. https://doi.org/10.1021/jm049461b
    35. Marina Mapelli,, Lucia Massimiliano,, Claudia Crovace,, Markus A. Seeliger,, Li-Huei Tsai,, Laurent Meijer, and, Andrea Musacchio. Mechanism of CDK5/p25 Binding by CDK Inhibitors. Journal of Medicinal Chemistry 2005, 48 (3) , 671-679. https://doi.org/10.1021/jm049323m
    36. Francis X. Tavares,, Joyce A. Boucheron,, Scott H. Dickerson,, Robert J. Griffin,, Frank Preugschat,, Stephen A. Thomson,, Tony Y. Wang, and, Hui-Qiang Zhou. N-Phenyl-4-pyrazolo[1,5-b]pyridazin-3-ylpyrimidin-2-amines as Potent and Selective Inhibitors of Glycogen Synthase Kinase 3 with Good Cellular Efficacy. Journal of Medicinal Chemistry 2004, 47 (19) , 4716-4730. https://doi.org/10.1021/jm040063i
    37. Tanja Stiller, Martin Hein, Peter Langer. Synthesis of electron-withdrawing group substituted isatin-N-glycosides by cyclization of formylated aniline-N-glycosides with oxalyl chloride. Tetrahedron Letters 2025, 156 , 155442. https://doi.org/10.1016/j.tetlet.2024.155442
    38. Heba T. Abdel-Mohsen. Oxindole–benzothiazole hybrids as CDK2 inhibitors and anticancer agents: design, synthesis and biological evaluation. BMC Chemistry 2024, 18 (1) https://doi.org/10.1186/s13065-024-01277-1
    39. Öznur Eyilcim, Fulya Günay, Yuk Yin Ng, Özlem Ulucan Açan, Zuhal Turgut, Ömer Tahir Günkara. Design, Synthesis, Biological Evaluation and Molecular Docking Studies of a New Series of Maleimide Derivatives. ChemistryOpen 2024, 13 (12) https://doi.org/10.1002/open.202400058
    40. Heba T. Abdel‐Mohsen, Yasmin M. Syam, Mahmoud S. Abd El‐Ghany, Somaia S. Abd El‐Karim. Benzimidazole‐oxindole hybrids: A novel class of selective dual CDK2 and GSK‐3β inhibitors of potent anticancer activity. Archiv der Pharmazie 2024, 357 (10) https://doi.org/10.1002/ardp.202300721
    41. Mohamed T.M. Nemr, Ahmed Elshewy, Mohammed L. Ibrahim, Ahmed M. El Kerdawy, Peter A. Halim. Design, synthesis, antineoplastic activity of new pyrazolo[3,4-d]pyrimidine derivatives as dual CDK2/GSK3β kinase inhibitors; molecular docking study, and ADME prediction. Bioorganic Chemistry 2024, 150 , 107566. https://doi.org/10.1016/j.bioorg.2024.107566
    42. Md Mustafiz Ansari, Sanjeev Kumar Sahu, Thakur Gurjeet Singh, Sovia R.J. Singh, Paranjeet Kaur. Evolving significance of kinase inhibitors in the management of Alzheimer's disease. European Journal of Pharmacology 2024, 979 , 176816. https://doi.org/10.1016/j.ejphar.2024.176816
    43. Chenchen Ma, Siyuan Cui, Ruirong Xu. Developments of Fms-like Tyrosine Kinase 3 Inhibitors as Anticancer Agents for AML Treatment. Current Medicinal Chemistry 2024, 31 (29) , 4657-4686. https://doi.org/10.2174/0109298673277543231205072556
    44. Katrin M. Dyballa, Stefanie Libnow, Martin Hein, Peter Langer. Synthesis of oxo- and carboindirubin-N-glycosides. Tetrahedron Letters 2024, 144 , 155151. https://doi.org/10.1016/j.tetlet.2024.155151
    45. Quan Tang, Sheng-Chao Huang, Hua Wang, Tianyu Long, Han-Han Kong, Qing-Qing Yang, Nianyu Huang, Long Wang. Metal-Free Synthesis of 2-Benzylideneindolin-3-ones via a Nucleophilic/Rearrangement/Azide–Alkene Cascade Reaction. Synlett 2024, 9 https://doi.org/10.1055/a-2341-9185
    46. Esraa Abdo Moustafa, Heba Abdelrasheed Allam, Marwa A. Fouad, Ahmed M. El Kerdawy, Nahed Nasser Eid El-Sayed, Christoph Wagner, Hatem A. Abdel-Aziz, Manal Abdel Fattah Ezzat. Discovery of novel quinolin-2-one derivatives as potential GSK-3β inhibitors for treatment of Alzheimer’s disease: Pharmacophore-based design, preliminary SAR, in vitro and in vivo biological evaluation. Bioorganic Chemistry 2024, 146 , 107324. https://doi.org/10.1016/j.bioorg.2024.107324
    47. Daniela N. S. dos Santos, Nilanjon Naskar, Kirsten Reess, Estefanía Delgado-Pinar, João Sérgio Seixas de Melo, Angelika Rueck, , , . Metabolic imaging with FLIM/PLIM indirubin derivatives: an ultrafast window into cellular metabolism and oxygen consumption. 2024, 40. https://doi.org/10.1117/12.3002527
    48. Danîela C. Nobre, Estefanía Delgado-Pinar, Carla Cunha, J. Sérgio Seixas de Melo. The role of the oxime group in the excited state deactivation processes of indirubin. Physical Chemistry Chemical Physics 2024, 26 (9) , 7416-7423. https://doi.org/10.1039/D3CP05260E
    49. Hari Sharan Adhikari, Aditya Garai, Chetana Khanal, Paras Nath Yadav. Synthesis and Comprehensive Characterization with Anticancer Activity Assessment of Salicylaldehyde and 2-Acetylphenol Based Chitosan Thiosemicarbazones and Their Copper(II) Complexes. Carbohydrate Polymer Technologies and Applications 2024, 34 , 100469. https://doi.org/10.1016/j.carpta.2024.100469
    50. D.N.S. dos Santos, N. Naskar, E. Delgado-Pinar, K. Reess, J.S. Seixas de Melo, A. Rueck. Bromine indirubin FLIM/PLIM sensors to measure oxygen in normoxic and hypoxic PDT conditions. Photodiagnosis and Photodynamic Therapy 2024, 45 , 103964. https://doi.org/10.1016/j.pdpdt.2024.103964
    51. Erin McMoneagle, Jin Zhou, Shiyao Zhang, Weixue Huang, Sunday Solomon Josiah, Ke Ding, Yun Wang, Jinwei Zhang. Neuronal K+-Cl- cotransporter KCC2 as a promising drug target for epilepsy treatment. Acta Pharmacologica Sinica 2024, 45 (1) , 1-22. https://doi.org/10.1038/s41401-023-01149-9
    52. Jiahao Sun, Shanshan Liang, Xinguo Liu, Shaolong Zhang, Meng Li, Qinggang Zhang, Jianzhong Chen. Insights into the selectivity of a brain-penetrant CDK4/6 vs CDK1/2 inhibitor for glioblastoma used in multiple replica molecular dynamics simulations. Journal of Biomolecular Structure and Dynamics 2023, , 1-20. https://doi.org/10.1080/07391102.2023.2294175
    53. Khadidja Adjir, Amina C. Berrekhchi-Berrahma, Majda Sekkal-Rahal. Theoretical characterization and biological activity investigation of indirubins, cyclin dependent kinases inhibitors. Journal of Biomolecular Structure and Dynamics 2023, 53 , 1-10. https://doi.org/10.1080/07391102.2023.2294182
    54. Nguyen Dinh Thanh, Nguyen Thi Kim Giang, Vu Ngoc Toan, Hoang Thi Kim Van, Do Son Hai, Nguyen Minh Tri, Duong Ngoc Toan. Substituted isatin-thiosemicarbazones containing d-galactose moiety: Synthesis, antimicrobial inhibition assay and molecular simulation study. Chemical Papers 2023, 77 (12) , 7813-7834. https://doi.org/10.1007/s11696-023-03083-w
    55. Siyuan Wan, Xinye Chen, Fucheng Yin, Shang Li, Yonglei Zhang, Heng Luo, Zhongwen Luo, Ningjie Cui, Yifan Chen, Xinxin Li, Lingyi Kong, Xiaobing Wang. Indirubin derivatives as bifunctional molecules inducing DNA damage and targeting PARP for the treatment of cancer. European Journal of Medicinal Chemistry 2023, 261 , 115843. https://doi.org/10.1016/j.ejmech.2023.115843
    56. Nguyen Dinh Thanh, Nguyen Thi Kim Giang, Vu Ngoc Toan, Hoang Thi Kim Van, Nguyen Minh Tri, Hoang Huu Anh, Do Son Hai. Synthesis and Potential Dual Inhibitory Activity Against Acetylcholinesterase and Butyrylcholinesterase of Galactose‐Conjugated Isatin β‐Thiosemicarbazones. ChemistrySelect 2023, 8 (22) https://doi.org/10.1002/slct.202301632
    57. Dake Qi, Haiyang Lin, Bingying Hu, Yang Wei. A review on in vitro model of the blood-brain barrier (BBB) based on hCMEC/D3 cells. Journal of Controlled Release 2023, 358 , 78-97. https://doi.org/10.1016/j.jconrel.2023.04.020
    58. Hari Sharan Adhikari, Aditya Garai, Paras Nath Yadav. Synthesis, characterization, and anticancer activity of chitosan functionalized isatin based thiosemicarbazones, and their copper(II) complexes. Carbohydrate Research 2023, 526 , 108796. https://doi.org/10.1016/j.carres.2023.108796
    59. Haoting Li, Anne E. Mattingly, Richard D. Smith, Roberta J. Melander, Robert K. Ernst, Christian Melander. 6-Bromoindirubin-3′-oxime derivatives are highly active colistin adjuvants against Klebsiella pneumoniae. RSC Medicinal Chemistry 2023, 14 (2) , 247-252. https://doi.org/10.1039/D2MD00370H
    60. Hongorzul Davaapil, Madeline McNamara, Alessandra Granata, Robyn G.C. Macrae, Mei Hirano, Martina Fitzek, J.A. Aragon-Martin, Anne Child, David M. Smith, Sanjay Sinha. A phenotypic screen of Marfan syndrome iPSC-derived vascular smooth muscle cells uncovers GSK3β as a new target. Stem Cell Reports 2023, 18 (2) , 555-569. https://doi.org/10.1016/j.stemcr.2022.12.014
    61. Jeongchan Lee, Joonwon Kim, Hyun Kim, HyunA Park, Jin Young Kim, Eun‐Jung Kim, Yung‐Hun Yang, Kwon‐Young Choi, Byung‐Gee Kim. Constructing multi‐enzymatic cascade reactions for selective production of 6‐bromoindirubin from tryptophan in Escherichia coli. Biotechnology and Bioengineering 2022, 119 (10) , 2938-2949. https://doi.org/10.1002/bit.28188
    62. Arvind Kumar Jain, Arindam Gupta, C. Karthikeyan, Piyush Trivedi, Anita Dutt Konar. Substituent Orchestration in Dimethylquinoxaline Derivatives: A Tool for Fishing Out Appropriate CDK5 Inhibitors as Potential Therapeutics for Alzheimer's. Chemistry & Biodiversity 2022, 19 (9) https://doi.org/10.1002/cbdv.202200116
    63. Sukanya Sukanya, Bhanwar Singh Choudhary, Pakhuri Mehta, Slawomir Filipek, Ruchi Malik. Identification of CNS compatible small molecules as glycogen synthase kinase-3β (GSK-3β) inhibitors through structure-based virtual screening. Medicinal Chemistry Research 2022, 31 (9) , 1545-1557. https://doi.org/10.1007/s00044-022-02912-z
    64. Fen-Fen Yang, Ming-Shan Shuai, Xiang Guan, Mao Zhang, Qing-Qing Zhang, Xiao-Zhong Fu, Zong-Qin Li, Da-Peng Wang, Meng Zhou, Yuan-Yong Yang, Ting Liu, Bin He, Yong-Long Zhao. Synthesis and antibacterial activity studies in vitro of indirubin-3′-monoximes. RSC Advances 2022, 12 (38) , 25068-25080. https://doi.org/10.1039/D2RA01035F
    65. Lin Yang, Xinyun Li, Wei Huang, Xiangshu Rao, Yu Lai. Pharmacological properties of indirubin and its derivatives. Biomedicine & Pharmacotherapy 2022, 151 , 113112. https://doi.org/10.1016/j.biopha.2022.113112
    66. Nicolai A. Aksenov, Alexander V. Aksenov, Igor A. Kurenkov, Nikita K. Kirillov, Dmitrii A. Aksenov, Nikolai A. Arutiunov, Daria S. Aksenova, Michael Rubin. One-Pot Synthesis of (E)-2-(3-Oxoindolin-2-ylidene)-2-arylacetonitriles. Molecules 2022, 27 (9) , 2808. https://doi.org/10.3390/molecules27092808
    67. Serena Montanari, Claudia Seidl, Lara Davani, Eleonora Gianquinto, Eliska Emrichova, Cristina Terenzi, Vincenza Andrisano, Angela De Simone. Natural products as novel scaffolds for the design of glycogen synthase kinase 3β inhibitors. Expert Opinion on Drug Discovery 2022, 17 (4) , 377-396. https://doi.org/10.1080/17460441.2022.2043845
    68. Vijaya Verma, M. J. Vijay Kumar, Kavita Sharma, Sridhar Rajaram, Ravi Muddashetty, Ravi Manjithaya, Thomas Behnisch, James P. Clement. Pharmacological intervention in young adolescents rescues synaptic physiology and behavioural deficits in Syngap1+/− mice. Experimental Brain Research 2022, 240 (1) , 289-309. https://doi.org/10.1007/s00221-021-06254-x
    69. Jung Sun Park, In Ae Jung, Hong Sang Choi, Dong-Hyun Kim, Hoon In Choi, Eun Hui Bae, Seong Kwon Ma, Soo Wan Kim. Anti-fibrotic effect of 6-bromo-indirubin-3′-oxime (6-BIO) via regulation of activator protein-1 (AP-1) and specificity protein-1 (SP-1) transcription factors in kidney cells. Biomedicine & Pharmacotherapy 2022, 145 , 112402. https://doi.org/10.1016/j.biopha.2021.112402
    70. Hezhen Wang, Zhiyuan Wang, Chunyong Wei, Jing Wang, Yingshu Xu, Guohui Bai, Qizheng Yao, Lei Zhang, Yongzheng Chen. Anticancer potential of indirubins in medicinal chemistry: Biological activity, structural modification, and structure-activity relationship. European Journal of Medicinal Chemistry 2021, 223 , 113652. https://doi.org/10.1016/j.ejmech.2021.113652
    71. Sandipty Kayastha, Laxmi Sagwan-Barkdoll, Aldwin Anterola, Lahiru N. Jayakody. Developing synthetic microbes to produce indirubin-derivatives. Biocatalysis and Agricultural Biotechnology 2021, 37 , 102162. https://doi.org/10.1016/j.bcab.2021.102162
    72. Dandan Qiao, Qian He, Xiaowei Cheng, Yongxiu Yao, Venugopal Nair, Hongxia Shao, Aijian Qin, Kun Qian. Regulation of Avian Leukosis Virus Subgroup J Replication by Wnt/β-Catenin Signaling Pathway. Viruses 2021, 13 (10) , 1968. https://doi.org/10.3390/v13101968
    73. Igor A. Schepetkin, Mark B. Plotnikov, Andrei I. Khlebnikov, Tatiana M. Plotnikova, Mark T. Quinn. Oximes: Novel Therapeutics with Anticancer and Anti-Inflammatory Potential. Biomolecules 2021, 11 (6) , 777. https://doi.org/10.3390/biom11060777
    74. Mo-Ran Sun, Hong-Liang Li, Meng-Yu Ba, Weyland Cheng, Hai-Liang Zhu, Yong-Tao Duan. Cyclopropyl Scaffold: A Generalist for Marketed Drugs. Mini-Reviews in Medicinal Chemistry 2021, 21 (2) , 150-170. https://doi.org/10.2174/1389557520666200729161150
    75. Wagdy M. Eldehna, Sara T. Al-Rashood, Tarfah Al-Warhi, Razan O. Eskandrani, Amal Alharbi, Ahmed M. El Kerdawy. Novel oxindole/benzofuran hybrids as potential dual CDK2/GSK-3β inhibitors targeting breast cancer: design, synthesis, biological evaluation, and in silico studies. Journal of Enzyme Inhibition and Medicinal Chemistry 2021, 36 (1) , 271-286. https://doi.org/10.1080/14756366.2020.1862101
    76. Yosif Almoshari, Rongguo Ren, Haipeng Zhang, Zhenshan Jia, Xin Wei, Ningrong Chen, Guojuan Li, Sangjin Ryu, Subodh M. Lele, Richard A. Reinhardt, Dong Wang. GSK3 inhibitor-loaded osteotropic Pluronic hydrogel effectively mitigates periodontal tissue damage associated with experimental periodontitis. Biomaterials 2020, 261 , 120293. https://doi.org/10.1016/j.biomaterials.2020.120293
    77. Suprim Tha, Sapana Shakya, Rajani Malla, Pramod Aryal. Prospects of Indole derivatives as methyl transfer inhibitors: antimicrobial resistance managers. BMC Pharmacology and Toxicology 2020, 21 (1) https://doi.org/10.1186/s40360-020-00402-9
    78. Bing Fu, Guorui Yin, Ke Song, Xiurui Mu, Bo Xu, Xubin Zhang. Indirubin-3′-Oxime (IDR3O) Inhibits Proliferation of Osteosarcoma Cells in vitro and Tumor Growth in vivo Through AMPK-Activation and PGC-1α/TFAM Up-Regulation. Doklady Biochemistry and Biophysics 2020, 495 (1) , 354-360. https://doi.org/10.1134/S1607672920060022
    79. Elena Catanzaro, Nibal Betari, Jose M. Arencibia, Serena Montanari, Claudia Sissi, Angela De Simone, Ivano Vassura, Alan Santini, Vincenza Andrisano, Vincenzo Tumiatti, Marco De Vivo, Dmitri V. Krysko, Marco B.L. Rocchi, Carmela Fimognari, Andrea Milelli. Targeting topoisomerase II with trypthantrin derivatives: Discovery of 7-((2-(dimethylamino)ethyl)amino)indolo[2,1-b]quinazoline-6,12-dione as an antiproliferative agent and to treat cancer. European Journal of Medicinal Chemistry 2020, 202 , 112504. https://doi.org/10.1016/j.ejmech.2020.112504
    80. Luminita Crisan, Sorin Avram, Ludovic Kurunczi, Liliana Pacureanu. Partial Least Squares Discriminant Analysis and 3D Similarity Perspective Applied to Analyze Comprehensively the Selectivity of Glycogen Synthase Kinase 3 Inhibitors. Molecular Informatics 2020, 39 (6) https://doi.org/10.1002/minf.201900142
    81. Pyeonghwa Jeong, Yeongyu Moon, Je-Heon Lee, So-Deok Lee, Jiyeon Park, Jungeun Lee, Jiheon Kim, Hyo Jeong Lee, Na Yoon Kim, Jungil Choi, Jeong Doo Heo, Ji Eun Shin, Hyun Woo Park, Yoon-Gyoon Kim, Sun-Young Han, Yong-Chul Kim. Discovery of orally active indirubin-3′-oxime derivatives as potent type 1 FLT3 inhibitors for acute myeloid leukemia. European Journal of Medicinal Chemistry 2020, 195 , 112205. https://doi.org/10.1016/j.ejmech.2020.112205
    82. Nicolai A. Aksenov, Dmitrii A. Aksenov, Nikolai A. Arutiunov, Daria S. Aksenova, Alexander V. Aksenov, Michael Rubin. Unexpected cyclization of ortho -nitrochalcones into 2-alkylideneindolin-3-ones. RSC Advances 2020, 10 (31) , 18440-18450. https://doi.org/10.1039/D0RA03520C
    83. Kenneth Chen, Thienlong Phan, Angel Lin, Luca Sardo, Anthony R. Mele, Michael R. Nonnemacher, Zachary Klase, . Morphine exposure exacerbates HIV-1 Tat driven changes to neuroinflammatory factors in cultured astrocytes. PLOS ONE 2020, 15 (3) , e0230563. https://doi.org/10.1371/journal.pone.0230563
    84. Kandasamy Saravanan, Govindasamy Hunday, Poomani Kumaradhas. Binding and stability of indirubin-3-monoxime in the GSK3β enzyme: a molecular dynamics simulation and binding free energy study. Journal of Biomolecular Structure and Dynamics 2020, 38 (4) , 957-974. https://doi.org/10.1080/07391102.2019.1591301
    85. Jasmine Speranza, Natalizia Miceli, Maria Fernanda Taviano, Salvatore Ragusa, Inga Kwiecień, Agnieszka Szopa, Halina Ekiert. Isatis tinctoria L. (Woad): A Review of Its Botany, Ethnobotanical Uses, Phytochemistry, Biological Activities, and Biotechnological Studies. Plants 2020, 9 (3) , 298. https://doi.org/10.3390/plants9030298
    86. Nilesh Kahar, Pankaj Jadhav, R. V. Ramana Reddy, Sudam Dawande. A rhodium( ii ) catalysed domino synthesis of azepino fused diindoles from isatin tethered N -sulfonyl-1,2,3-triazoles and indoles. Chemical Communications 2020, 56 (8) , 1207-1210. https://doi.org/10.1039/C9CC08377D
    87. Carmen Laura Sayas. Tau-based therapies for Alzheimer’s disease: Promising novel neuroprotective approaches. 2020, 245-272. https://doi.org/10.1016/B978-0-12-814037-6.00005-7
    88. ANTONIA EFSTATHIOU, Despina Smirlis. A Radioactive-free Kinase Inhibitor Discovery Assay Against the Trypanosoma brucei Glycogen Synthase Kinase-3 short (TbGSK-3s). BIO-PROTOCOL 2020, 10 (2) https://doi.org/10.21769/BioProtoc.3493
    89. Takeshi Sugai, Kengo Hanaya, Shuhei Higashibayashi. Syntheses of Indirubins by Aldol Condensation of Isatins with Indoxyl Anion Generated in situ by Lipase-Catalyzed Deacetylation of Indoxyl Acetate. HETEROCYCLES 2020, 100 (1) , 129. https://doi.org/10.3987/COM-19-14118
    90. Yoshimi Ichimaru, Makoto Sano, Ichie Kajiwara, Takao Tobe, Hiroki Yoshioka, Kazuhiko Hayashi, Hideaki Ijichi, Shinichi Miyairi. Indirubin 3′-Oxime Inhibits Migration, Invasion, and Metastasis in Mice Bearing Spontaneously Occurring Pancreatic Cancer via Blocking the RAF/ERK, AKT, and SAPK/JNK Pathways. Translational Oncology 2019, 12 (12) , 1574-1582. https://doi.org/10.1016/j.tranon.2019.08.010
    91. Rino Ragno. www.3d-qsar.com: a web portal that brings 3-D QSAR to all electronic devices—the Py-CoMFA web application as tool to build models from pre-aligned datasets. Journal of Computer-Aided Molecular Design 2019, 33 (9) , 855-864. https://doi.org/10.1007/s10822-019-00231-x
    92. Xin Tang, Jesse Drotar, Keji Li, Cullen D. Clairmont, Anna Sophie Brumm, Austin J. Sullins, Hao Wu, Xiaoxiao Shawn Liu, Jinhua Wang, Nathanael S. Gray, Mriganka Sur, Rudolf Jaenisch. Pharmacological enhancement of KCC2 gene expression exerts therapeutic effects on human Rett syndrome neurons and Mecp2 mutant mice. Science Translational Medicine 2019, 11 (503) https://doi.org/10.1126/scitranslmed.aau0164
    93. Liliana Pacureanu, Sorin Avram, Alina Bora, Ludovic Kurunczi, Luminita Crisan. Portraying the selectivity of GSK-3 inhibitors towards CDK-2 by 3D similarity and molecular docking. Structural Chemistry 2019, 30 (3) , 911-923. https://doi.org/10.1007/s11224-018-1224-z
    94. Panagiota-Efstathia Nikolaou, Kerstin Boengler, Panagiotis Efentakis, Konstantina Vouvogiannopoulou, Anastasia Zoga, Nicholas Gaboriaud-Kolar, Vassilios Myrianthopoulos, Pavlos Alexakos, Nikolaos Kostomitsopoulos, Ioannis Rerras, Anna Tsantili-Kakoulidou, Alexios Leandros Skaltsounis, Andreas Papapetropoulos, Efstathios K Iliodromitis, Rainer Schulz, Ioanna Andreadou. Investigating and re-evaluating the role of glycogen synthase kinase 3 beta kinase as a molecular target for cardioprotection by using novel pharmacological inhibitors. Cardiovascular Research 2019, 115 (7) , 1228-1243. https://doi.org/10.1093/cvr/cvz061
    95. Antonia Efstathiou, Nicolas Gaboriaud-Kolar, Vassilios Myrianthopoulos, Konstantina Vougogiannopoulou, Ines Subota, Stephanie Aicher, Emmanuel Mikros, Philippe Bastin, Alexios-Leandros Skaltsounis, Ketty Soteriadou, Despina Smirlis. Indirubin Analogues Inhibit Trypanosoma brucei Glycogen Synthase Kinase 3 Short and T. brucei Growth. Antimicrobial Agents and Chemotherapy 2019, 63 (6) https://doi.org/10.1128/AAC.02065-18
    96. Natalia A. Lozinskaya, Denis A. Babkov, Ekaterina V. Zaryanova, Elena N. Bezsonova, Alexander M. Efremov, Michael D. Tsymlyakov, Lada V. Anikina, Olga Yu. Zakharyascheva, Alexander V. Borisov, Valentina N. Perfilova, Ivan N. Tyurenkov, Marina V. Proskurnina, Alexander A. Spasov. Synthesis and biological evaluation of 3-substituted 2-oxindole derivatives as new glycogen synthase kinase 3β inhibitors. Bioorganic & Medicinal Chemistry 2019, 27 (9) , 1804-1817. https://doi.org/10.1016/j.bmc.2019.03.028
    97. Job Tchoumtchoua, Maria Halabalaki, Evangelos Gikas, Anthony Tsarbopoulos, Nikoletta Fotaki, Lucy Liu, Sangkil Nam, Richard Jove, Leandros A. Skaltsounis. Preliminary pharmacokinetic study of the anticancer 6BIO in mice using an UHPLC-MS/MS approach. Journal of Pharmaceutical and Biomedical Analysis 2019, 164 , 317-325. https://doi.org/10.1016/j.jpba.2018.10.039
    98. Ahmed Karam Farag, Eun Joo Roh. Death‐associated protein kinase (DAPK) family modulators: Current and future therapeutic outcomes. Medicinal Research Reviews 2019, 39 (1) , 349-385. https://doi.org/10.1002/med.21518
    99. Long Zhao, Raz Ben-Yair, Caroline E. Burns, C. Geoffrey Burns. Endocardial Notch Signaling Promotes Cardiomyocyte Proliferation in the Regenerating Zebrafish Heart through Wnt Pathway Antagonism. Cell Reports 2019, 26 (3) , 546-554.e5. https://doi.org/10.1016/j.celrep.2018.12.048
    100. Antonia Efstathiou, Cássio Santana Meira, Nicolas Gaboriaud-Kolar, Tanira Matutino Bastos, Vinícius Pinto Costa Rocha, Konstantina Vougogiannopoulou, Alexios-Leandros Skaltsounis, Despina Smirlis, Milena Botelho Pereira Soares. Indirubin derivatives are potent and selective anti- Trypanosoma cruzi agents. Virulence 2018, 9 (1) , 1658-1668. https://doi.org/10.1080/21505594.2018.1532242
    Load more citations

    Journal of Medicinal Chemistry

    Cite this: J. Med. Chem. 2004, 47, 4, 935–946
    Click to copy citationCitation copied!
    https://doi.org/10.1021/jm031016d
    Published January 8, 2004
    Copyright © 2004 American Chemical Society

    Article Views

    6327

    Altmetric

    -

    Citations

    Learn about these metrics

    Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

    Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

    The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.