1
|
Fotie J, Matherne CM, Mather JB, Wroblewski JE, Johnson K, Boudreaux LG, Perez AA. The Fundamental Role of Oxime and Oxime Ether Moieties in Improving the Physicochemical and Anticancer Properties of Structurally Diverse Scaffolds. Int J Mol Sci 2023; 24:16854. [PMID: 38069175 PMCID: PMC10705934 DOI: 10.3390/ijms242316854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The present review explores the critical role of oxime and oxime ether moieties in enhancing the physicochemical and anticancer properties of structurally diverse molecular frameworks. Specific examples are carefully selected to illustrate the distinct contributions of these functional groups to general strategies for molecular design, modulation of biological activities, computational modeling, and structure-activity relationship studies. An extensive literature search was conducted across three databases, including PubMed, Google Scholar, and Scifinder, enabling us to create one of the most comprehensive overviews of how oximes and oxime ethers impact antitumor activities within a wide range of structural frameworks. This search focused on various combinations of keywords or their synonyms, related to the anticancer activity of oximes and oxime ethers, structure-activity relationships, mechanism of action, as well as molecular dynamics and docking studies. Each article was evaluated based on its scientific merit and the depth of the study, resulting in 268 cited references and more than 336 illustrative chemical structures carefully selected to support this analysis. As many previous reviews focus on one subclass of this extensive family of compounds, this report represents one of the rare and fully comprehensive assessments of the anticancer potential of this group of molecules across diverse molecular scaffolds.
Collapse
Affiliation(s)
- Jean Fotie
- Department of Chemistry and Physics, Southeastern Louisiana University, SLU 10878, Hammond, LA 70402-0878, USA; (C.M.M.); (J.B.M.); (J.E.W.); (K.J.); (L.G.B.); (A.A.P.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Zhang L, Luo B, Lu Y, Chen Y. Targeting Death-Associated Protein Kinases for Treatment of Human Diseases: Recent Advances and Future Directions. J Med Chem 2023; 66:1112-1136. [PMID: 36645394 DOI: 10.1021/acs.jmedchem.2c01606] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The death-associated protein kinase (DAPK) family is a member of the calcium/calmodulin-regulated serine/threonine protein kinase family, and studies have shown that its role, as its name suggests, is mainly to regulate cell death. The DAPK family comprises five members, including DAPK1, DAPK2, DAPK3, DRAK1 and DRAK2, which show high homology in the common N-terminal kinase domain but differ in the extra-catalytic domain. Notably, previous research has suggested that the DAPK family plays an essential role in both the development and regulation of human diseases. However, only a few small-molecule inhibitors have been reported. In this Perspective, we mainly discuss the structure, biological function, and role of DAPKs in diseases and the currently discovered small-molecule inhibitors, providing valuable information for the development of the DAPK field.
Collapse
Affiliation(s)
- Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Boqin Luo
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Yi Chen
- State Key Laboratory of Biotherapy and Cancer Center and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
3
|
Park S, Kye S, Jung ME, Chae CH, Yang K, Kim S, Choi G, Lee K. Discovery of TRD‐93 as a novel
DRAK2
inhibitor. B KOREAN CHEM SOC 2023. [DOI: 10.1002/bkcs.12680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Sangjun Park
- Bio & Drug Discovery Division Korea Research Institute of Chemical Technology Daejeon South Korea
- Medicinal Chemistry & Pharmacology University of Science & Technology Daejeon South Korea
| | - Seungmin Kye
- Bio & Drug Discovery Division Korea Research Institute of Chemical Technology Daejeon South Korea
- Medicinal Chemistry & Pharmacology University of Science & Technology Daejeon South Korea
| | - Myoung Eun Jung
- Bio & Drug Discovery Division Korea Research Institute of Chemical Technology Daejeon South Korea
| | - Chong Hak Chae
- Bio & Drug Discovery Division Korea Research Institute of Chemical Technology Daejeon South Korea
| | | | | | - Gildon Choi
- Bio & Drug Discovery Division Korea Research Institute of Chemical Technology Daejeon South Korea
- Medicinal Chemistry & Pharmacology University of Science & Technology Daejeon South Korea
| | - Kwangho Lee
- Bio & Drug Discovery Division Korea Research Institute of Chemical Technology Daejeon South Korea
- Medicinal Chemistry & Pharmacology University of Science & Technology Daejeon South Korea
| |
Collapse
|
4
|
Zheng Y, Li X, Kuang L, Wang Y. New insights into the characteristics of DRAK2 and its role in apoptosis: From molecular mechanisms to clinically applied potential. Front Pharmacol 2022; 13:1014508. [PMID: 36386181 PMCID: PMC9649744 DOI: 10.3389/fphar.2022.1014508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/12/2022] [Indexed: 11/27/2022] Open
Abstract
As a member of the death-associated protein kinase (DAPK) family, DAP kinase-associated apoptosis-inducing kinase 2 (DRAK2) performs apoptosis-related functions. Compelling evidence suggests that DRAK2 is involved in regulating the activation of T lymphocytes as well as pancreatic β-cell apoptosis in type I diabetes. In addition, DRAK2 has been shown to be involved in the development of related tumor and non-tumor diseases through a variety of mechanisms, including exacerbation of alcoholic fatty liver disease (NAFLD) through SRSF6-associated RNA selective splicing mechanism, regulation of chronic lymphocytic leukemia and acute myeloid leukemia, and progression of colorectal cancer. This review focuses on the structure, function, and upstream pathways of DRAK2 and discusses the potential and challenges associated with the clinical application of DRAK2-based small-molecule inhibitors, with the aim of advancing DRAK2 research.
Collapse
Affiliation(s)
| | | | | | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Ferjancic Z, Kukuruzar A, Bihelovic F. Total Synthesis of (+)‐Alstonlarsine A. Angew Chem Int Ed Engl 2022; 61:e202210297. [DOI: 10.1002/anie.202210297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Indexed: 12/31/2022]
Affiliation(s)
- Zorana Ferjancic
- University of Belgrade— Faculty of Chemistry Studentski trg 16, POB 51 11158 Belgrade 118 Serbia
| | - Andrej Kukuruzar
- University of Belgrade— Faculty of Chemistry Studentski trg 16, POB 51 11158 Belgrade 118 Serbia
| | - Filip Bihelovic
- University of Belgrade— Faculty of Chemistry Studentski trg 16, POB 51 11158 Belgrade 118 Serbia
| |
Collapse
|
6
|
Wu QW, Kapfhammer JP. The Emerging Key Role of the mGluR1-PKCγ Signaling Pathway in the Pathogenesis of Spinocerebellar Ataxias: A Neurodevelopmental Viewpoint. Int J Mol Sci 2022; 23:ijms23169169. [PMID: 36012439 PMCID: PMC9409119 DOI: 10.3390/ijms23169169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/19/2022] Open
Abstract
Spinocerebellar ataxias (SCAs) are a heterogeneous group of autosomal dominantly inherited progressive disorders with degeneration and dysfunction of the cerebellum. Although different subtypes of SCAs are classified according to the disease-associated causative genes, the clinical syndrome of the ataxia is shared, pointing towards a possible convergent pathogenic pathway among SCAs. In this review, we summarize the role of SCA-associated gene function during cerebellar Purkinje cell development and discuss the relationship between SCA pathogenesis and neurodevelopment. We will summarize recent studies on molecules involved in SCA pathogenesis and will focus on the mGluR1-PKCγ signaling pathway evaluating the possibility that this might be a common pathway which contributes to these diseases.
Collapse
|
7
|
Ferjancic Z, Kukuruzar A, Bihelovic F. Total Synthesis of (+)‐Alstonlarsine A. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202210297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Zorana Ferjancic
- Univerzitet u Beogradu Hemijski fakultet Faculty of Chemistry 11158 Belgrade SERBIA
| | - Andrej Kukuruzar
- Univerzitet u Beogradu Hemijski fakultet Faculty of Chemistry 11158 Belgrade SERBIA
| | - Filip Bihelovic
- University of Belgrade Faculty of Chemistry Studentski trg 12-16 11158 Belgrade SERBIA
| |
Collapse
|
8
|
Pharmacological properties of indirubin and its derivatives. Biomed Pharmacother 2022; 151:113112. [PMID: 35598366 DOI: 10.1016/j.biopha.2022.113112] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/23/2022] [Accepted: 05/10/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Indirubin is the main bioactive component of the traditional Chinese medicine Indigo naturalis and is a bisindole alkaloid. Multiple studies have shown that indirubin exhibits good anticancer, anti-inflammatory and neuroprotective properties. METHODS The purpose of this review is to provide a summary of the pharmacological mechanisms of indirubin and its derivatives. RESULTS Indirubin and its derivatives exert anticancer effects by regulating the expression of cyclin-dependent kinases (CDKs), GSK-3β, Bax, Bcl-2, C-MYC, matrix metalloproteinases (MMPs), and focal adhesion kinase (FAK) through the PI3K/AKT/mTOR, nuclear factor (NF)-κB, mitogen-activated protein kinase (MAPK), JAK/signal transducer and activator of transcription 3 (STAT3) pathways and other signaling pathways. We also reviewed the anti-inflammatory and neuroprotective properties of indirubin and its derivatives. CONCLUSION The findings of recent studies assessing indirubin and its derivatives suggest that these compounds can be used as potential drugs to treat tumors, inflammation, neuropathy and bacterial infection.
Collapse
|
9
|
Wu QW, Kapfhammer JP. Serine/threonine kinase 17b (STK17B) signalling regulates Purkinje cell dendritic development and is altered in multiple spinocerebellar ataxias. Eur J Neurosci 2021; 54:6673-6684. [PMID: 34536317 PMCID: PMC9292345 DOI: 10.1111/ejn.15465] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 11/28/2022]
Abstract
Serine/threonine kinase 17b (STK17B, also known as DRAK2) is known to be a downstream effector of protein kinase C (PKC) in the immune system, in particular T lymphocytes. PKC activity also plays a critical role for dendritic development and synaptic maturation and plasticity in cerebellar Purkinje cells. We present evidence that STK17B is strongly expressed in mouse cerebellar Purkinje cells starting in the early postnatal period and remaining highly expressed throughout adult stages and that STK17B is a target of PKC phosphorylation in the cerebellum. STK17B overexpression potentiates the morphological changes of Purkinje cells seen after PKC activation, suggesting that it is a downstream effector of PKC. A phosphorylation mimetic STK17B variant induced a marked reduction of Purkinje cell dendritic tree size, whereas the inhibition of STK17B with the novel compound 16 (Cpd16) could partially rescue the morphological changes of the Purkinje cell dendritic tree after PKC activation. These findings show that STK17B signalling is an important downstream effector of PKC activation in Purkinje cells. Furthermore, STK17B was identified as a molecule being transcriptionally downregulated in mouse models of SCA1, SCA7, SCA14 and SCA41. The reduced expression of STK17B in these mouse models might protect Purkinje cell dendrites from the negative effects of overactivated PKC signalling. Our findings provide new insights in the role of STK17B for Purkinje cell dendritic development and the pathology of SCAs.
Collapse
Affiliation(s)
- Qin-Wei Wu
- Institute of Anatomy, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Josef P Kapfhammer
- Institute of Anatomy, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Wang H, Wang Z, Wei C, Wang J, Xu Y, Bai G, Yao Q, Zhang L, Chen Y. Anticancer potential of indirubins in medicinal chemistry: Biological activity, structural modification, and structure-activity relationship. Eur J Med Chem 2021; 223:113652. [PMID: 34161865 DOI: 10.1016/j.ejmech.2021.113652] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/13/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Indirubin is the crucial ingredient of Danggui Longhui Wan and Qing-Dai, traditional Chinese medicine herbal formulas used for the therapy of chronic myelocytic leukemia in China for hundreds of years. Although the monomeric indirubin has been used in China for the treatment human chronic myelocytic leukemia. However, due to low water solubility, poor pharmacokinetic properties and low therapeutic effects are the major obstacle, and had significantly limited its clinical application. Consequently, the attractive anticancer profile of indirubin has enthused numerous researchers to discover novel indirubin derivatives with improved pharmacodynamic activity as well as good pharmacokinetic property. In this paper, we comprehensively review the recent progress of anticancer potential of indirubins, structural modification and structure-activity relationship, which may provide useful direction for the further development of novel indirubins with improved pharmacological profiles for the treatment of various types of cancer.
Collapse
Affiliation(s)
- Hezhen Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Zhiyuan Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Chunyong Wei
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Jing Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Yingshu Xu
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Guohui Bai
- Key Laboratory of Oral Disease of Higher Schools in Guizhou Province, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| | - Qizheng Yao
- School of Pharmacy, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing, 210009, PR China.
| | - Lei Zhang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| | - Yongzheng Chen
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| |
Collapse
|
11
|
Schepetkin IA, Plotnikov MB, Khlebnikov AI, Plotnikova TM, Quinn MT. Oximes: Novel Therapeutics with Anticancer and Anti-Inflammatory Potential. Biomolecules 2021; 11:biom11060777. [PMID: 34067242 PMCID: PMC8224626 DOI: 10.3390/biom11060777] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Oximes have been studied for decades because of their significant roles as acetylcholinesterase reactivators. Over the last twenty years, a large number of oximes have been reported with useful pharmaceutical properties, including compounds with antibacterial, anticancer, anti-arthritis, and anti-stroke activities. Many oximes are kinase inhibitors and have been shown to inhibit over 40 different kinases, including AMP-activated protein kinase (AMPK), phosphatidylinositol 3-kinase (PI3K), cyclin-dependent kinase (CDK), serine/threonine kinases glycogen synthase kinase 3 α/β (GSK-3α/β), Aurora A, B-Raf, Chk1, death-associated protein-kinase-related 2 (DRAK2), phosphorylase kinase (PhK), serum and glucocorticoid-regulated kinase (SGK), Janus tyrosine kinase (JAK), and multiple receptor and non-receptor tyrosine kinases. Some oximes are inhibitors of lipoxygenase 5, human neutrophil elastase, and proteinase 3. The oxime group contains two H-bond acceptors (nitrogen and oxygen atoms) and one H-bond donor (OH group), versus only one H-bond acceptor present in carbonyl groups. This feature, together with the high polarity of oxime groups, may lead to a significantly different mode of interaction with receptor binding sites compared to corresponding carbonyl compounds, despite small changes in the total size and shape of the compound. In addition, oximes can generate nitric oxide. This review is focused on oximes as kinase inhibitors with anticancer and anti-inflammatory activities. Oximes with non-kinase targets or mechanisms of anti-inflammatory activity are also discussed.
Collapse
Affiliation(s)
- Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA;
| | - Mark B. Plotnikov
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, 634028 Tomsk, Russia;
| | - Andrei I. Khlebnikov
- Kizhner Research Center, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Scientific Research Institute of Biological Medicine, Altai State University, 656049 Barnaul, Russia
| | - Tatiana M. Plotnikova
- Department of Pharmacology, Siberian State Medical University, 634050 Tomsk, Russia;
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA;
- Correspondence: ; Tel.: +1-406-994-4707; Fax: +1-406-994-4303
| |
Collapse
|
12
|
Small Molecules in the Treatment of Squamous Cell Carcinomas: Focus on Indirubins. Cancers (Basel) 2021; 13:cancers13081770. [PMID: 33917267 PMCID: PMC8068014 DOI: 10.3390/cancers13081770] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary In this review, the genetic landscape of squamous cell carcinoma is related to the potential targets of indirubin-based small molecules in cancer therapy. Being a component of traditional Chinese medicine, indirubins are used to treat chronic or inflammatory diseases, and have received increasing attention in cancer treatment due to their proapoptotic and antiproliferative activity. Frequent genetic alterations of squamous cell carcinomas are summarized, and it is discussed how these may render tumors susceptible to indirubin-based small molecule inhibitors. Abstract Skin cancers are the most common malignancies in the world. Among the most frequent skin cancer entities, squamous cell carcinoma (SCC) ranks second (~20%) after basal cell carcinoma (~77%). In early stages, a complete surgical removal of the affected tissue is carried out as standard therapy. To treat advanced and metastatic cancers, targeted therapies with small molecule inhibitors are gaining increasing attention. Small molecules are a heterogeneous group of protein regulators, which are produced by chemical synthesis or fermentation. The majority of them belong to the group of receptor tyrosine kinase inhibitors (RTKIs), which specifically bind to certain RTKs and directly influence the respective signaling pathway. Knowledge of characteristic molecular alterations in certain cancer entities, such as SCC, can help identify tumor-specific substances for targeted therapies. Most frequently, altered genes in SCC include TP53, NOTCH, EGFR, and CCND1. For example, the gene CCND1, which codes for cyclin D1 protein, is upregulated in nearly half of SCC cases and promotes proliferation of affected cells. A treatment with the small molecule 5′-nitroindirubin-monoxime (INO) leads to inhibition of cyclin D1 and thus inhibition of proliferation. As a component of Danggui Longhui Wan, a traditional Chinese medicine, indirubins are used to treat chronic diseases and have been shown to inhibit inflammatory reactions. Indirubins are pharmacologically relevant small molecules with proapoptotic and antiproliferative activity. In this review, we discuss the current literature on indirubin-based small molecules in cancer treatment. A special focus is on the molecular biology of squamous cell carcinomas, their alterations, and how these are rendered susceptible to indirubin-based small molecule inhibitors. The potential molecular mechanisms of the efficacy of indirubins in killing SCC cells will be discussed as well.
Collapse
|
13
|
Hou X, Li JY, Zhao M, Dai C, Li Y, Liu Y. Synthesis, Characterization, and DRAK2 Inhibitory Activities of Hydroxyaurone Derivatives. HETEROCYCLES 2021. [DOI: 10.3987/com-21-14481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
14
|
Zhang Z, Dai C, Wu H, Li J, Nan F. Design and Synthesis of Alkyl Phenols Inhibitors of Death Associated Apoptotic Protein Kinase 2 (DRAK2). CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202103056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
15
|
Picado A, Chaikuad A, Wells CI, Shrestha S, Zuercher WJ, Pickett JE, Kwarcinski FE, Sinha P, de Silva CS, Zutshi R, Liu S, Kannan N, Knapp S, Drewry DH, Willson TM. A Chemical Probe for Dark Kinase STK17B Derives Its Potency and High Selectivity through a Unique P-Loop Conformation. J Med Chem 2020; 63:14626-14646. [PMID: 33215924 PMCID: PMC7816213 DOI: 10.1021/acs.jmedchem.0c01174] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
STK17B is a member of the death-associated protein kinase family and has been genetically linked to the development of diverse diseases. However, the role of STK17B in normal and disease pathology is poorly defined. Here, we present the discovery of thieno[3,2-d] pyrimidine SGC-STK17B-1 (11s), a high-quality chemical probe for this understudied "dark" kinase. 11s is an ATP-competitive inhibitor that showed remarkable selectivity over other kinases including the closely related STK17A. X-ray crystallography of 11s and related thieno[3,2-d]pyrimidines bound to STK17B revealed a unique P-loop conformation characterized by a salt bridge between R41 and the carboxylic acid of the inhibitor. Molecular dynamic simulations of STK17B revealed the flexibility of the P-loop and a wide range of R41 conformations available to the apo-protein. The isomeric thieno[2,3-d]pyrimidine SGC-STK17B-1N (19g) was identified as a negative control compound. The >100-fold lower activity of 19g on STK17B was attributed to the reduced basicity of its pyrimidine N1.
Collapse
Affiliation(s)
- Alfredo Picado
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599-7264
| | - Apirat Chaikuad
- Institute of Pharmaceutical Chemistry, Max-von-Laue-Straße 9, Goethe University Frankfurt, 60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Straße 15, 60438 Frankfurt, Germany
| | - Carrow I. Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599-7264
| | - Safal Shrestha
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - William J. Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599-7264
| | - Julie E. Pickett
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599-7264
| | | | - Parvathi Sinha
- Luceome Biotechnologies, 1665 E. 18th Street, Suite 106, Tucson, AZ 85719
| | - Chandi S. de Silva
- Luceome Biotechnologies, 1665 E. 18th Street, Suite 106, Tucson, AZ 85719
| | - Reena Zutshi
- Luceome Biotechnologies, 1665 E. 18th Street, Suite 106, Tucson, AZ 85719
| | - Shubin Liu
- Research Computing Center, University of North Carolina, Chapel Hill, NC 27599-3420
| | - Natarajan Kannan
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Max-von-Laue-Straße 9, Goethe University Frankfurt, 60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Straße 15, 60438 Frankfurt, Germany
- German Translational Cancer Network (DKTK) site Frankfurt/Mainz
- Frankfurt Cancer Institute (FCI), Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599-7264
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599-7264
| |
Collapse
|
16
|
|
17
|
Synthesis of new indirubin derivatives and their in vitro anticancer activity. CHEMICAL PAPERS 2018. [DOI: 10.1007/s11696-018-0659-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
18
|
Farag AK, Roh EJ. Death-associated protein kinase (DAPK) family modulators: Current and future therapeutic outcomes. Med Res Rev 2018; 39:349-385. [DOI: 10.1002/med.21518] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 05/06/2018] [Accepted: 06/03/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Ahmed Karam Farag
- Chemical Kinomics Research Center; Korea Institute of Science and Technology (KIST); Seoul Republic of Korea
- Division of Bio-Medical Science &Technology, Korea Institute of Science and Technology (KIST) School; University of Science and Technology; Seoul Republic of Korea
| | - Eun Joo Roh
- Chemical Kinomics Research Center; Korea Institute of Science and Technology (KIST); Seoul Republic of Korea
- Division of Bio-Medical Science &Technology, Korea Institute of Science and Technology (KIST) School; University of Science and Technology; Seoul Republic of Korea
| |
Collapse
|
19
|
Investigation of indirubin derivatives: a combination of 3D-QSAR, molecular docking, and ADMET towards the design of new DRAK2 inhibitors. Struct Chem 2018. [DOI: 10.1007/s11224-018-1134-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
20
|
Aouidate A, Ghaleb A, Ghamali M, Ousaa A, Choukrad M, Sbai A, Bouachrine M, Lakhlifi T. 3D QSAR studies, molecular docking and ADMET evaluation, using thiazolidine derivatives as template to obtain new inhibitors of PIM1 kinase. Comput Biol Chem 2018; 74:201-211. [PMID: 29635214 DOI: 10.1016/j.compbiolchem.2018.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/03/2018] [Accepted: 03/10/2018] [Indexed: 02/04/2023]
Abstract
Proviral Integration site for Moloney murine leukemia virus-1 (PIM1) belongs to the serine/threonine kinase family of Ca2+-calmodulin-dependent protein kinase (CAMK) group, which is involved in cell survival and proliferation as well as a number of other signal transduction pathways. Thus, PIM1 is regarded as a promising target for treatment of cancers. In the present paper, a three-dimensional quantitative structure activity relationship (3D-QSAR) and molecular docking were performed to investigate the binding between PIM1 and thiazolidine inhibitors in order to design potent inhibitors. The comparative molecular similarity indices analysis (CoMSIA) was developed using twenty-six molecules having pIC50 ranging from 8.854 to 6.011 (IC50 in nM). The best CoMSIA model gave significant statistical quality. The determination coefficient (R2) and Leave-One-Out cross-validation coefficient (Q2) are 0.85 and 0.58, respectively. Furthermore, the predictive ability of this model was evaluated by external validation((n = 11, R2test = 0.72, and MAE = 0.170 log units). The graphical contour maps could provide structural features to improve inhibitory activity. Furthermore, a good consistency between contour maps and molecular docking strongly demonstrates that the molecular modeling is reliable. Based on these satisfactory results, we designed several new potent PIM1 inhibitors and their inhibitory activities were predicted by the molecular models. Additionally, those newly designed inhibitors, showed promising results in the preliminary in silico ADMET evaluations, compared to the best inhibitor from the studied dataset. The results expand our understanding of thiazolidines as inhibitors of PIM1 and could be of great help in lead optimization for early drug discovery of highly potent inhibitors.
Collapse
Affiliation(s)
- Adnane Aouidate
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Adib Ghaleb
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Mounir Ghamali
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Abdellah Ousaa
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - M'barek Choukrad
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Abdelouahid Sbai
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | | | - Tahar Lakhlifi
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| |
Collapse
|
21
|
Lan Y, Han J, Wang Y, Wang J, Yang G, Li K, Song R, Zheng T, Liang Y, Pan S, Liu X, Zhu M, Liu Y, Meng F, Mohsin M, Cui Y, Zhang B, Subash S, Liu L. STK17B promotes carcinogenesis and metastasis via AKT/GSK-3β/Snail signaling in hepatocellular carcinoma. Cell Death Dis 2018; 9:236. [PMID: 29445189 PMCID: PMC5833726 DOI: 10.1038/s41419-018-0262-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/10/2017] [Accepted: 11/24/2017] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is a lethal malignancy worldwide with frequent intrahepatic and distant metastasis. Elucidating the underlying molecular mechanism that modulates HCC progression is critical for exploring novel therapeutic strategies. Serine/Threonine Kinase 17B (STK17B) is upregulated in HCC tissues, but its role in HCC progression remains elusive. In the present studies, we reported that STK17B had a critical role in HCC progression. STK17B was significantly upregulated in HCC cell lines and specimens, and patients with ectopic STK17B expression characterized with poor clinicopathological features. In vitro and in vivo assay demonstrated that inhibition of STK17B markedly inhibits HCC tumorigenesis and metastasis, while STK17B overexpression promoted these processes. Furthermore, we found that STK17B promoted EMT process via activating AKT/GSK-3β/Snail signal pathway, and miR-455-3p was identified as the upstream regulator of STK17B. Combination of high level of STK17B and low level of miR-455-3p predicted poor prognosis with higher accuracy for HCC patients. In conclusion, our research demonstrated that STK17B promotes HCC progression, induces EMT process via activating AKT/GSK-3β/Snail signal and predicts poor prognosis in HCC.
Collapse
Affiliation(s)
- Yaliang Lan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Jihua Han
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yan Wang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Jiabei Wang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Guangchao Yang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Keyu Li
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Ruipeng Song
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, The Affiliated Tumor Hospital of Harbin Medical University, Harbin, China
| | - Yingjian Liang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Shangha Pan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Xirui Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Mingxi Zhu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yao Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Fanzheng Meng
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Manzoor Mohsin
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yifeng Cui
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Bo Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Sharma Subash
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Lianxin Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China.
| |
Collapse
|
22
|
Khan MM, Khan S, Saigal, Sahoo SC. Efficient and Eco-Friendly One-Pot Synthesis of Functionalized Furan-2-one, Pyrrol-2-one, and Tetrahydropyridine Using Lemon Juice as a Biodegradable Catalyst. ChemistrySelect 2018. [DOI: 10.1002/slct.201702933] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- M. Musawwer Khan
- Department of Chemistry; Aligarh Muslim University; Aligarh- 202002 India
| | - Sarfaraz Khan
- Department of Chemistry; Aligarh Muslim University; Aligarh- 202002 India
| | - Saigal
- Department of Chemistry; Aligarh Muslim University; Aligarh- 202002 India
| | - Subash C. Sahoo
- Department of Chemistry; Punjab University; Chandigarh- 160014 India
| |
Collapse
|
23
|
Fogaça MV, Cândido-Bacani PDM, Benicio LM, Zapata LM, Cardoso PDF, de Oliveira MT, Calvo TR, Varanda EA, Vilegas W, de Syllos Cólus IM. Effects of indirubin and isatin on cell viability, mutagenicity, genotoxicity and BAX/ERCC1 gene expression. PHARMACEUTICAL BIOLOGY 2017; 55:2005-2014. [PMID: 28738722 PMCID: PMC7011876 DOI: 10.1080/13880209.2017.1354387] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT Indigofera suffruticosa Miller (Fabaceae) and I. truxillensis Kunth produce compounds, such as isatin (ISA) and indirubin (IRN), which possess antitumour properties. Their effects in mammalian cells are still not very well understood. OBJECTIVE We evaluated the activities of ISA and/or IRN on cell viability and apoptosis in vitro, their genotoxic potentials in vitro and in vivo, and the IRN- and ISA-induced expression of ERCC1 or BAX genes. MATERIALS AND METHODS HeLa and/or CHO-K1 cell lines were tested (3 or 24 h) in the MTT, Trypan blue exclusion, acridine orange/ethidium bromide, cytokinesis-blocked micronucleus (CBMN) and comet (36, 24 and 72 h) tests after treatment with IRN (0.1 to 200 μM) or ISA (0.5 to 50 μM). Gene expression was measured by RT-qPCR in HeLa cells. Swiss albino mice received IRN (3, 4 or 24 h) by gavage (50, 100 and 150 mg/kg determined from the LD50 - 1 g/kg b.w.) and submitted to comet assay in vivo. RESULTS IRN reduced the viability of CHO-K1 (24 h; 5 to 200 μM) and HeLa cells (10 to 200 μM), and was antiproliferative in the CBMN test (CHO-K1: 0.5 to 10 μM; HeLa: 5 and 10 μM). The drug did not induce apoptosis, micronucleus neither altered gene expression. IRN and ISA were genotoxic for HeLa cells (3 and 24 h) at all doses tested. IRN (100 and 150 mg/kg) also induced genotoxicity in vivo (4 h). CONCLUSION IRN and ISA have properties that make them candidates as chemotherapeutics for further pharmacological investigations.
Collapse
Affiliation(s)
- Manoela Viar Fogaça
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | | | - Lucas Milanez Benicio
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Lara Martinelli Zapata
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | | | | | - Tamara Regina Calvo
- Araraquara Institute of Chemistry, São Paulo State University, Araraquara, Brazil
| | - Eliana Aparecida Varanda
- Araraquara Faculty of Pharmaceutical Sciences, Department of Biological Sciences, São Paulo State University, Araraquara, Brazil
| | - Wagner Vilegas
- Araraquara Institute of Chemistry, São Paulo State University, Araraquara, Brazil
- Experimental Campus of the Paulista Coast, São Paulo State University, São Vicente, Brazil
| | - Ilce Mara de Syllos Cólus
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
24
|
Wang S, Xu L, Lu YT, Liu YF, Han B, Liu T, Tang J, Li J, Wu J, Li JY, Yu LF, Yang F. Discovery of benzofuran-3(2H)-one derivatives as novel DRAK2 inhibitors that protect islet β-cells from apoptosis. Eur J Med Chem 2017; 130:195-208. [PMID: 28249207 DOI: 10.1016/j.ejmech.2017.02.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 12/11/2022]
Abstract
Death-associated protein kinase-related apoptosis-inducing kinase-2 (DRAK2) is a serine/threonine kinase that plays a key role in a wide variety of cell death signaling pathways. Inhibition of DRAK2 was found to efficiently protect islet β-cells from apoptosis and hence DRAK2 inhibitors represent a promising therapeutic strategy for the treatment of diabetes. Only very few chemical entities targeting DRAK2 are currently known. We carried out a high throughput screening and identified compound 4 as a moderate DRAK2 inhibitor with an IC50 value of 3.15 μM. Subsequent SAR studies of hit compound 4 led to the development of novel benzofuran-3(2H)-one series of DRAK2 inhibitors with improved potency and favorable selectivity profiles against 26 selected kinases. Importantly, most potent compounds 40 (IC50 = 0.33 μM) and 41 (IC50 = 0.25 μM) were found to protect islet β-cells from apoptosis in dose-dependent manners. These data support the notion that small molecule inhibitors of DRAK2 represents a promising strategy for the treatment of diabetes.
Collapse
Affiliation(s)
- Sheng Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Lei Xu
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Yu-Ting Lu
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Yu-Fei Liu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Bing Han
- Laboratory of Immunology and Cardiovascular Research, Centre Hospitalier de l'Université de Montréal, 900 rue St-Denis, Montréal, Québec, Canada
| | - Ting Liu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Jie Tang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China; Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Jia Li
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Jiangping Wu
- Laboratory of Immunology and Cardiovascular Research, Centre Hospitalier de l'Université de Montréal, 900 rue St-Denis, Montréal, Québec, Canada.
| | - Jing-Ya Li
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China.
| | - Li-Fang Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China.
| | - Fan Yang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China.
| |
Collapse
|