1
|
Zhao H. The Science and Art of Structure-Based Virtual Screening. ACS Med Chem Lett 2024; 15:436-440. [PMID: 38628791 PMCID: PMC11017385 DOI: 10.1021/acsmedchemlett.4c00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Structure-based virtual screening has gained momentum again as the high attrition rate at every stage of drug discovery drives the need to explore a greater chemical space. From the Bayesian perspective, its shortcomings as a viable strategy for sustainable hit discovery are discussed, with regard to the prior hit rates of screening libraries and the performance of computational methods. Lessons are shared in selecting virtual hits for experimental validation learned from a series of eight successful campaigns, one of which impacted the discovery of a drug candidate currently in clinical trials.
Collapse
Affiliation(s)
- Hongtao Zhao
- Medicinal Chemistry, Research and Early
Development, Respiratory and Immunology (R&I), BioPharmaceuticals
R&D, AstraZeneca, Gothenburg 43183, Sweden
| |
Collapse
|
2
|
Wang Y, Li S, Yan Z, Guo Y, Zhang L. Computational insights into novel inhibitor indole-heterocycle specific against glycogen phosphorylase isoenzymes interaction mechanism. Future Med Chem 2023; 15:913-922. [PMID: 37395076 DOI: 10.4155/fmc-2023-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Abstract
Background: Glycogen phosphorylase (GP) is a potential drug target. As the three subtypes of GP are highly conserved, it is difficult to research their specificity. However, compound 1 inhibits the GP subtypes differently and was studied to aid in designing specific inhibitors. Results: Molecular docking showed that the ligands in GP subtype complexes had some differences in spatial conformation and binding modes, stabilized by polar and nonpolar interactions. The results were confirmed through kinetic experiments, with affinities of -85.230 (brain GP), -73.809 (liver GP) and -66.061 kJ/mol (muscle GP). Conclusion: The study provides insight into the possible reasons for differences in compound 1's inhibitory activity against the GP subtypes and offers guidance in designing target molecules for regulating selectivity among the subtypes.
Collapse
Affiliation(s)
- Youde Wang
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, 067000, China
| | - Shuai Li
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, 067000, China
| | - Zhiwei Yan
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, 067000, China
| | - Yachun Guo
- Department of Pathogen Biology, Chengde Medical University, Chengde, Hebei, 067000, China
| | - Liying Zhang
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, 067000, China
| |
Collapse
|
3
|
Zhao Y, Wang Q, Xie C, Cai Y, Chen X, Hou Y, He L, Li J, Yao M, Chen S, Wu W, Chen X, Hong A. Peptide ligands targeting FGF receptors promote recovery from dorsal root crush injury via AKT/mTOR signaling. Am J Cancer Res 2021; 11:10125-10147. [PMID: 34815808 PMCID: PMC8581430 DOI: 10.7150/thno.62525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Fibroblast growth factor receptors (FGFRs) are key targets for nerve regeneration and repair. The therapeutic effect of exogenous recombinant FGFs in vivo is limited due to their high molecular weight. Small peptides with low molecular weight, easy diffusion, low immunogenicity, and nontoxic metabolite formation are potential candidates. The present study aimed to develop a novel low-molecular-weight peptide agonist of FGFR to promote nerve injury repair. Methods: Phage display technology was employed to screen peptide ligands targeting FGFR2. The peptide ligand affinity for FGFRs was detected by isothermal titration calorimetry. Structural biology-based computer virtual analysis was used to characterize the interaction between the peptide ligand and FGFR2. The peptide ligand effect on axon growth, regeneration, and behavioral recovery of sensory neurons was determined in the primary culture of sensory neurons and dorsal root ganglia (DRG) explants in vitro and a rat spinal dorsal root injury (DRI) model in vivo. The peptide ligand binding to other membrane receptors was characterized by surface plasmon resonance (SPR) and liquid chromatography-mass spectrometry (LC-MS)/MS. Intracellular signaling pathways primarily affected by the peptide ligand were characterized by phosphoproteomics, and related pathways were verified using specific inhibitors. Results: We identified a novel FGFR-targeting small peptide, CH02, with seven amino acid residues. CH02 activated FGFR signaling through high-affinity binding with the extracellular segment of FGFRs and also had an affinity for several receptor tyrosine kinase (RTK) family members, including VEGFR2. In sensory neurons cultured in vitro, CH02 maintained the survival of neurons and promoted axon growth. Simultaneously, CH02 robustly enhanced nerve regeneration and sensory-motor behavioral recovery after DRI in rats. CH02-induced activation of FGFR signaling promoted nerve regeneration primarily via AKT and ERK signaling downstream of FGFRs. Activation of mTOR downstream of AKT signaling augmented axon growth potential in response to CH02. Conclusion: Our study revealed the significant therapeutic effect of CH02 on strengthening nerve regeneration and suggested a strategy for treating peripheral and central nervous system injuries.
Collapse
|
4
|
Protocatechuic aldehyde protects cardiomycoytes against ischemic injury via regulation of nuclear pyruvate kinase M2. Acta Pharm Sin B 2021; 11:3553-3566. [PMID: 34900536 PMCID: PMC8642444 DOI: 10.1016/j.apsb.2021.03.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/14/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Rescuing cells from stress damage emerges a potential therapeutic strategy to combat myocardial infarction. Protocatechuic aldehyde (PCA) is a major phenolic acid in Chinese herb Danshen (Salvia miltiorrhiza root). This study investigated whether PCA regulated nuclear pyruvate kinase isoform M2 (PKM2) function to protect cardiomyocytes. In rats subjected to isoprenaline, PCA attenuated heart injury and protected cardiomyocytes from apoptosis. Through DARTS and CETSA assays, we identified that PCA bound and promoted PKM2 nuclear translocation in cardiomyocytes exposed to oxygen/glucose deprivation (OGD). In the nucleus, PCA increased the binding of PKM2 to β-catenin via preserving PKM2 acetylation, and the complex, in cooperation with T-cell factor 4 (TCF4), was required for transcriptional induction of genes encoding anti-apoptotic proteins, contributing to rescuing cardiomyocyte survival. In addition, PCA ameliorated mitochondrial dysfunction and prevented mitochondrial apoptosis dependent on PKM2. Consistently, PCA increased the binding of PKM2 to β-catenin, improved heart contractive function, normalized heart structure and attenuated oxidative damage in mice subjected to artery ligation, but the protective effects were lost in Pkm2-deficient heart. Together, we showed that PCA regulated nuclear PKM2 function to rescue cardiomyocyte survival via β-catenin/TCF4 signaling cascade, suggesting the potential of pharmacological intervention of PKM2 shuttle to protect the heart.
Collapse
Key Words
- Apoptosis
- CETSA, cellular thermal shift assay
- CK-MB, creatine kinase isoenzyme-MB
- DARTS, drug affinity responsive target stability
- Heart ischemia
- ISO, isoprenaline
- LDH, lactate dehydrogenase
- Mitochondrial damage
- Myocardial infarction
- NRVMs, neonatal rat ventricular myocytes
- Nuclear translocation
- OGD, oxygen and glucose deprivation
- PCA, protocatechuic aldehyde
- PKM2
- PKM2, pyruvate kinase isoform M2
- Protocatechuic aldehyde
- ROS, reactive oxygen species
- TCF4
- TCF4, T-cell factor 4
- TUNEL, deoxynucleotidyl transferase-mediated dUTP nick end-labeling
- shRNA, short hairpin RNA
- β-Catenin
Collapse
|
5
|
Wang J, Zhu X, Qin X, Jiang H, Gao Y, Gao J. [miR-324-5p inhibits lipopolysaccharide-induced proliferation of rat glomerular mesangial cells by regulating the Syk/Ras/c-fos pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1571-1578. [PMID: 33243745 DOI: 10.12122/j.issn.1673-4254.2020.11.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To investigate the effect of miR-324-5p on the proliferation of rat glomerular mesangial (HBZY-1) cells and the role of Syk/Ras/c-fos signaling pathway in mediating this effect. METHODS HBZY-1 cells cultured in vitro were transiently transfected with miR-324-5p mimics or miR-324-5p-mimics-NC followed by treatment with lipopolysaccharide (LPS). MTT assay was used to detect the proliferation activity of HBZY-1 cells, and RT-qPCR was used to detect the expressions of miR-324-5p and the mRNA expressions of Syk, Ras, MEK1/2, ERK1/2 and c-fos mRNA. The protein expressions of p-Syk, Ras, p-MEK1/2, p-ERK1/2 and c-Fos were detected by Western blotting and immunofluorescence assay. RESULTS MTT assay showed that exposure to LPS significantly enhanced the proliferative activity of HBZY-1 cells. Compared with the cells treated with LPS and LPS + mimics NC, the cells transfected with miR-324-5p mimics prior to LPS exposure exhibited significantly lowered proliferative activity. Transfection with miR-324-5p mimics significantly lowered the mRNA expressions of Syk, Ras, MEK1/2, ERK1/2 and c-fos and the protein expressions of p-Syk, Ras, MEK1/2, ERK1/2 and c-Fos (P < 0.05), and reduced numbers of cells positive for p-Syk, Ras, p-MEK1/2, p-ERK1/2 and c-Fos proteins following LPS exposure. CONCLUSIONS miR-324-5p can inhibit the proliferation of rat chronic glomerulonephritis cells induced by LPS by inhibiting Syk/Ras/c-fos signaling pathway and may potentially serve as a diagnostic indicator and a therapeutic target for chronic glomerulonephritis.
Collapse
Affiliation(s)
- Jing Wang
- First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei 230012, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xiaoli Zhu
- First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei 230012, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xiujuan Qin
- First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Hui Jiang
- First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei 230012, China.,Anhui Provincial Key Laboratory of Chinese Medicine Compounds, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yachen Gao
- First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jiarong Gao
- First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei 230012, China.,Anhui Provincial Key Laboratory of Chinese Medicine Compounds, Anhui University of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
6
|
Wang Q, Zhao Y, Chen X, Hong A. Virtual screening of approved clinic drugs with main protease (3CL pro) reveals potential inhibitory effects on SARS-CoV-2. J Biomol Struct Dyn 2020; 40:685-695. [PMID: 32909528 PMCID: PMC7544985 DOI: 10.1080/07391102.2020.1817786] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
3CLpro is the main protease of the novel coronavirus (SARS-CoV-2) responsible for their intracellular duplication. Based on virtual screening technology and molecular dynamics simulation, we found 23 approved clinical drugs such as Viomycin, Capastat, Carfilzomib and Saquinavir, which showed high affinity with the 3CLpro active sites. These findings showed that there were potential drugs that inhibit SARS-Cov-2's 3CLpro in the current clinical drug library, and these drugs can be further tested or chemically modified for the treatment of COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Qiang Wang
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, China.,Guangdong provincial Engineering Research Center of Biotechnology, Guangzhou, China.,Engineering Research Center of Genetic Medicine of Ministry of Education, Guangzhou, China
| | - Ying Zhao
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, China.,Guangdong provincial Engineering Research Center of Biotechnology, Guangzhou, China.,Engineering Research Center of Genetic Medicine of Ministry of Education, Guangzhou, China
| | - Xiaojia Chen
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, China.,Guangdong provincial Engineering Research Center of Biotechnology, Guangzhou, China.,Engineering Research Center of Genetic Medicine of Ministry of Education, Guangzhou, China
| | - An Hong
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, China.,Guangdong provincial Engineering Research Center of Biotechnology, Guangzhou, China.,Engineering Research Center of Genetic Medicine of Ministry of Education, Guangzhou, China
| |
Collapse
|
7
|
Gong Y, Zhang H, Geng N, Ren X, Giesy JP, Luo Y, Xing L, Wu P, Yu Z, Chen J. Short-chain chlorinated paraffins (SCCPs) disrupt hepatic fatty acid metabolism in liver of male rat via interacting with peroxisome proliferator-activated receptor α (PPARα). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 181:164-171. [PMID: 31185430 DOI: 10.1016/j.ecoenv.2019.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 06/09/2023]
Abstract
Short-chain chlorinated paraffins (SCCPs) are frequently detected in environmental matrices and human tissues. It was hypothesized that SCCPs might interact with the peroxisome proliferator-activated receptor α (PPARα). In the present study, an in vitro, dual-luciferase reporter gene assay and in silico molecular docking analysis were employed together to study the interactions between SCCPs congeners and PPARα. Expressions of genes downstream in pathways activated by PPARα in liver of rats exposed to 1, 10, or 100 mg/kg bm/d of C10-13-CPs (56.5% Cl) for 28 days were examined to confirm activation potencies of SCCPs toward PPARα signaling. Effects of exposure to C10-13-CPs (56.5% Cl) on fatty acid metabolism in rat liver were also explored via a pseudo-targeted metabolomics strategy. Our results showed that C10-13-CPs (56.5% Cl) caused a dose-dependent greater expression of luciferase activity of rat PPARα. Molecular docking modeling revealed that SCCPs had a strong capacity to bind with PPARα only through hydrophobic interactions and the binding affinity was dependent on the degree of chlorination in SCCPs congeners. In livers of male rats, exposure to 100 mg/kg bm/d of C10-13-CPs (56.5% Cl) resulted in up-regulated expressions of 11 genes that are downstream in the PPARα-activated pathway and regulate catabolism of fatty acid. Consistently, accelerated fatty acid oxidation was observed mainly characterized by lesser concentrations of ∑fatty acids in livers of rats. Overall, these results demonstrated, for the first time, that SCCPs could activate rat PPARα signaling and thereby disrupt metabolism of fatty acid in livers of male rats.
Collapse
Affiliation(s)
- Yufeng Gong
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China
| | - Haijun Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China.
| | - Ningbo Geng
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China
| | - Xiaoqian Ren
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - John P Giesy
- Department of Veterinary Biomedical Sciences and Toxicology Centre, University of Saskatchewan, Saskatoon SK, S7N 5B4, Saskatchewan, Canada; Department of Environmental Science, Baylor University, Waco TX, 76706, Texas, United States
| | - Yun Luo
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liguo Xing
- Safety Evaluation Center of Shenyang Research Institute of Chemical Industry Ltd, Shenyang, 110021, Liaoning, China
| | - Ping Wu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China
| | - Zhengkun Yu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China
| | - Jiping Chen
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China
| |
Collapse
|
8
|
Ganjoo A, Prabhakar C. In silico structural anatomization of spleen tyrosine kinase inhibitors: Pharmacophore modeling, 3D QSAR analysis and molecular docking studies. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2019.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Enhancing the atypical esterase promiscuity of the γ-lactamase Sspg from
Sulfolobus solfataricus
by substrate screening. Appl Microbiol Biotechnol 2019; 103:4077-4087. [DOI: 10.1007/s00253-019-09758-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/06/2019] [Indexed: 11/26/2022]
|
10
|
Zheng L, Chen S, Cao Y, Zhao L, Gao Y, Ding X, Wang X, Gu Y, Wang S, Zhu Z, Yuan Y, Chen X, Chai Y. Combination of comprehensive two-dimensional prostate cancer cell membrane chromatographic system and network pharmacology for characterizing membrane binding active components from Radix et Rhizoma Rhei and their targets. J Chromatogr A 2018; 1564:145-154. [DOI: 10.1016/j.chroma.2018.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/02/2018] [Accepted: 06/05/2018] [Indexed: 02/08/2023]
|
11
|
Gong Y, Zhang H, Geng N, Xing L, Fan J, Luo Y, Song X, Ren X, Wang F, Chen J. Short-chain chlorinated paraffins (SCCPs) induced thyroid disruption by enhancement of hepatic thyroid hormone influx and degradation in male Sprague Dawley rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 625:657-666. [PMID: 29304504 DOI: 10.1016/j.scitotenv.2017.12.251] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 05/22/2023]
Abstract
Short-chain chlorinated paraffins (SCCPs) are known to disturb thyroid hormone (TH) homeostasis in rodents. However, the mechanism remains to be fully characterized. In this study, male Sprague Dawley rats received SCCPs (0, 1, 10, or 100mg/kg/day) via gavage once a day for consecutive 28days. Plasma and hepatic TH concentrations, thyrocyte structure, as well as thyroid and hepatic mRNA and protein levels of genes associated with TH homeostasis were examined. Moreover, we performed molecular docking to predict interactions between constitutive androstane receptor (CAR), a key regulator in xenobiotic-induced TH metabolism, with different SCCP molecules. Exposure to SCCPs significantly decreased the circulating free thyroxine (T4) and triiodothyronine (T3) levels, but increased thyroid-stimulating hormone (TSH) levels by a feedback mechanism. Decreased hepatic T4 and increased hepatic T3 levels were also seen after 100mg/kg/day SCCPs exposure. SCCPs didn't show any significant effects on the expression of thyroid TH synthesis genes or thyrocyte structure. However, stimulation effects were observed for mRNA and protein levels of hepatic uridine diphosphoglucuronosyl transferase (UGT) 1A1 and organic anion transporter 2, suggesting an accelerated TH metabolism in rat liver. The increased cytochrome P450 2B1 but not 1A1 mRNA and protein levels indicated that the CAR signaling was activated by SCCPs exposure. According to docking analysis, SCCPs form hydrophobic interactions with CAR and the binding affinity shows dependency on chlorine content. Overall, our data showed that CAR implicated enhancement of hepatic TH influx and degradation could be the main cause for SCCPs induced TH deficiency in male rats.
Collapse
Affiliation(s)
- Yufeng Gong
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Haijun Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Ningbo Geng
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Liguo Xing
- Safety Evaluation Center of Shenyang Research Institute of Chemical Industry Ltd., Shenyang 110021, China
| | - Jingfeng Fan
- National Marine Environmental Monitoring Center, Dalian 116023, China
| | - Yun Luo
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyao Song
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xiaoqian Ren
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feidi Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiping Chen
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
12
|
Discovery of furan carboxylate derivatives as novel inhibitors of ATP-citrate lyase via virtual high-throughput screening. Bioorg Med Chem Lett 2017; 27:929-935. [DOI: 10.1016/j.bmcl.2017.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 11/19/2022]
|
13
|
Muraoka T, Ide M, Irie M, Morikami K, Miura T, Nishihara M, Kashiwagi H. Development of a Method for Converting a TAK1 Type I Inhibitor into a Type II or c-Helix-Out Inhibitor by Structure-Based Drug Design (SBDD). Chem Pharm Bull (Tokyo) 2017; 64:1622-1629. [PMID: 27803473 DOI: 10.1248/cpb.c16-00606] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have developed a method for converting a transforming growth factor-β-activated kinase 1 (TAK1) type I inhibitor into a type II or c-helix-out inhibitor by structure-based drug design (SBDD) to achieve an effective strategy for developing these different types of kinase inhibitor in parallel. TAK1 plays a key role in inflammatory and immune signaling, and is therefore considered to be an attractive molecular target for the treatment of human diseases (inflammatory disease, cancer, etc.). We have already reported novel type I TAK1 inhibitor, so we utilized its X-ray information to design a new chemical class type II and c-helix-out inhibitors. To develop the type II inhibitor, we superimposed the X-ray structure of our reported type I inhibitor onto a type II compound that inhibits multiple kinases, and used SBDD to design a new type II inhibitor. For the TAK1 c-helix-out inhibitor, we utilized the X-ray structure of a b-Raf c-helix-out inhibitor to design compounds, because TAK1 is located close to b-Raf in the Sugen kinase tree, so we considered that TAK1 would, similarly to b-Raf, form a c-helix-out conformation. The X-ray crystal structure of the inhibitors in complex with TAK1 confirmed the binding modes of the compounds we designed. This report is notable for being the first discovery of a c-helix-out inhibitor against TAK1.
Collapse
|
14
|
Zhu F, Liu F, Wu B, He B. Efficient Extracellular Expression of Metalloprotease for Z-Aspartame Synthesis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:9631-9638. [PMID: 27966925 DOI: 10.1021/acs.jafc.6b04164] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Metalloprotease PT121 and its mutant Y114S (Tyr114 was substituted to Ser) are effective catalysts for the synthesis of Z-aspartame (Z-APM). This study presents the selection of a suitable signal peptide for improving expression and extracellular secretion of proteases PT121 and Y114S by Escherichia coli. Co-inducers containing IPTG and arabinose were used to promote protease production and cell growth. Under optimal conditions, the expression levels of PT121 and Y114S reached >500 mg/L, and the extracellular activity of PT121/Y114S accounted for 87/82% of the total activity of proteases. Surprisingly, purer protein was obtained in the supernatant, because arabinose reduced cell membrane permeability, avoiding cell lysis. Comparison of Z-APM synthesis and caseinolysis between proteases PT121 and Y114S showed that mutant Y114S presented remarkably higher activity of Z-APM synthesis and considerably lower activity of caseinolysis. The significant difference in substrate specificity renders these enzymes promising biocatalysts.
Collapse
Affiliation(s)
- Fucheng Zhu
- College of Biotechnology and Pharmaceutical Engineering and ‡School of Pharmaceutical Sciences, Nanjing Tech University , No. 30 Puzhu South Road, Nanjing 211816, China
| | - Feng Liu
- College of Biotechnology and Pharmaceutical Engineering and ‡School of Pharmaceutical Sciences, Nanjing Tech University , No. 30 Puzhu South Road, Nanjing 211816, China
| | - Bin Wu
- College of Biotechnology and Pharmaceutical Engineering and ‡School of Pharmaceutical Sciences, Nanjing Tech University , No. 30 Puzhu South Road, Nanjing 211816, China
| | - Bingfang He
- College of Biotechnology and Pharmaceutical Engineering and ‡School of Pharmaceutical Sciences, Nanjing Tech University , No. 30 Puzhu South Road, Nanjing 211816, China
| |
Collapse
|
15
|
Ding X, Cao Y, Yuan Y, Gong Z, Liu Y, Zhao L, Lv L, Zhang G, Wang D, Jia D, Zhu Z, Hong Z, Chen X, Chai Y. Development of APTES-Decorated HepG2 Cancer Stem Cell Membrane Chromatography for Screening Active Components from Salvia miltiorrhiza. Anal Chem 2016; 88:12081-12089. [PMID: 28193057 DOI: 10.1021/acs.analchem.6b02709] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Xuan Ding
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Yan Cao
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Yongfang Yuan
- Department
of Pharmacy, Shanghai ninth People’s Hospital, No. 280 Mohe
Road, Shanghai 201999, PR China
| | - Zhirong Gong
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Yue Liu
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Liang Zhao
- Department
of Pharmacy, Eastern Hepatobiliary Surgery Hospital, No. 225 Changhai
Road, Shanghai 200438, PR China
| | - Lei Lv
- Department
of Pharmacy, Eastern Hepatobiliary Surgery Hospital, No. 225 Changhai
Road, Shanghai 200438, PR China
| | - Guoqing Zhang
- Department
of Pharmacy, Eastern Hepatobiliary Surgery Hospital, No. 225 Changhai
Road, Shanghai 200438, PR China
| | - Dongyao Wang
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Dan Jia
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Zhenyu Zhu
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Zhanying Hong
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Xiaofei Chen
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| | - Yifeng Chai
- School
of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai 200433, PR China
| |
Collapse
|
16
|
Facilitating the Evolution of Esterase Activity from a Promiscuous Enzyme (Mhg) with Catalytic Functions of Amide Hydrolysis and Carboxylic Acid Perhydrolysis by Engineering the Substrate Entrance Tunnel. Appl Environ Microbiol 2016; 82:6748-6756. [PMID: 27613682 DOI: 10.1128/aem.01817-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/04/2016] [Indexed: 11/20/2022] Open
Abstract
Promiscuous enzymes are generally considered to be starting points in the evolution of offspring enzymes with more specific or even novel catalytic activities, which is the molecular basis of producing new biological functions. Mhg, a typical α/β fold hydrolase, was previously reported to have both γ-lactamase and perhydrolase activities. However, despite having high structural similarity to and sharing an identical catalytic triad with an extensively studied esterase from Pseudomonas fluorescens, this enzyme did not show any esterase activity. Molecular docking and sequence analysis suggested a possible role for the entry of the binding pocket in blocking the entrance tunnel, preventing the ester compounds from entering into the pocket. By engineering the entrance tunnel with only one or two amino acid substitutions, we successfully obtained five esterase variants of Mhg. The variants exhibited a very broad substrate acceptance, hydrolyzing not only the classical p-nitrophenol esters but also various types of chiral esters, which are widely used as drug intermediates. Site 233 at the entrance tunnel of Mhg was found to play a pivotal role in modulating the three catalytic activities by adjusting the size and shape of the tunnel, with different amino acid substitutions at this site facilitating different activities. Remarkably, the variant with the L233G mutation was a very specific esterase without any γ-lactamase and perhydrolase activities. Considering the amino acid conservation and differentiation, this site could be a key target for future protein engineering. In addition, we demonstrate that engineering the entrance tunnel is an efficient strategy to regulate enzyme catalytic capabilities. IMPORTANCE Promiscuous enzymes can act as starting points in the evolution of novel catalytic activities, thus providing a molecular basis for the production of new biological functions. In this study, we identified a critical amino acid residue (Leu233) at the entry of the substrate tunnel of a promiscuous enzyme, Mhg. We found that substitution of this residue with smaller amino acids such as Gly, Ala, Ser, or Pro endowed the enzyme with novel esterase activity. Different amino acids at this site can facilitate different catalytic activities. These findings exhibited universal significance in this subset of α/β fold hydrolases, including Mhg. Furthermore, we demonstrate that engineering the entrance tunnel is an efficient strategy to evolve new enzyme catalytic capabilities. Our study has important implications for the regulation of enzyme catalytic promiscuity and development of protein engineering methodologies.
Collapse
|
17
|
Zhang N, Zhao H. Enriching screening libraries with bioactive fragment space. Bioorg Med Chem Lett 2016; 26:3594-7. [PMID: 27311891 DOI: 10.1016/j.bmcl.2016.06.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/05/2016] [Accepted: 06/06/2016] [Indexed: 02/04/2023]
Abstract
By deconvoluting 238,073 bioactive molecules in the ChEMBL library into extended Murcko ring systems, we identified a set of 2245 ring systems present in at least 10 molecules. These ring systems belong to 2221 clusters by ECFP4 fingerprints with a minimum intracluster similarity of 0.8. Their overlap with ring systems in commercial libraries was further quantified. Our findings suggest that success of a small fragment library is driven by the convergence of effective coverage of bioactive ring systems (e.g., 10% coverage by 1000 fragments vs. 40% by 2million HTS compounds), high enrichment of bioactive ring systems, and low molecular complexity enhancing the probability of a match with the protein targets. Reconciling with the previous studies, bioactive ring systems are underrepresented in screening libraries. As such, we propose a library of virtual fragments with key functionalities via fragmentation of bioactive molecules. Its utility is exemplified by a prospective application on protein kinase CK2, resulting in the discovery of a series of novel inhibitors with the most potent compound having an IC50 of 0.5μM and a ligand efficiency of 0.41kcal/mol per heavy atom.
Collapse
Affiliation(s)
- Na Zhang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Hongtao Zhao
- Lephar Research, Rindögatan 21, 11558 Stockholm, Sweden.
| |
Collapse
|
18
|
Wu X, Chen X, Dan J, Cao Y, Gao S, Guo Z, Zerbe P, Chai Y, Diao Y, Zhang L. Characterization of anti-leukemia components from Indigo naturalis using comprehensive two-dimensional K562/cell membrane chromatography and in silico target identification. Sci Rep 2016; 6:25491. [PMID: 27150638 PMCID: PMC4858665 DOI: 10.1038/srep25491] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/18/2016] [Indexed: 12/30/2022] Open
Abstract
Traditional Chinese Medicine (TCM) has been developed for thousands of years and has formed an integrated theoretical system based on a large amount of clinical practice. However, essential ingredients in TCM herbs have not been fully identified, and their precise mechanisms and targets are not elucidated. In this study, a new strategy combining comprehensive two-dimensional K562/cell membrane chromatographic system and in silico target identification was established to characterize active components from Indigo naturalis, a famous TCM herb that has been widely used for the treatment of leukemia in China, and their targets. Three active components, indirubin, tryptanthrin and isorhamnetin, were successfully characterized and their anti-leukemia effects were validated by cell viability and cell apoptosis assays. Isorhamnetin, with undefined cancer related targets, was selected for in silico target identification. Proto-oncogene tyrosine-protein kinase (Src) was identified as its membrane target and the dissociation constant (Kd) between Src and isorhamnetin was 3.81 μM. Furthermore, anti-leukemia effects of isorhamnetin were mediated by Src through inducing G2/M cell cycle arrest. The results demonstrated that the integrated strategy could efficiently characterize active components in TCM and their targets, which may bring a new light for a better understanding of the complex mechanism of herbal medicines.
Collapse
Affiliation(s)
- Xunxun Wu
- School of Biomedical Science, Institute of Molecular Medicine, Huaqiao University, Quanzhou 362021, PR China.,School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Xiaofei Chen
- School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Jia Dan
- School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Yan Cao
- School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Shouhong Gao
- School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Zhiying Guo
- School of Biomedical Science, Institute of Molecular Medicine, Huaqiao University, Quanzhou 362021, PR China.,School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Philipp Zerbe
- Department of Plant Biology, University of California, Davis, CA 95616, USA
| | - Yifeng Chai
- School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Yong Diao
- School of Biomedical Science, Institute of Molecular Medicine, Huaqiao University, Quanzhou 362021, PR China
| | - Lei Zhang
- School of Pharmacy, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200433, PR China
| |
Collapse
|
19
|
Hudson BM, Nguyen E, Tantillo DJ. The influence of intramolecular sulfur-lone pair interactions on small-molecule drug design and receptor binding. Org Biomol Chem 2016; 14:3975-80. [PMID: 27049933 DOI: 10.1039/c6ob00254d] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sulfur-lone pair interactions are important conformational control elements in sulfur-containing heterocycles that abound in pharmaceuticals, natural products, agrochemicals, polymers and other important classes of organic molecules. Nonetheless, the role of intramolecular sulfur-lone pair interactions in the binding of small molecules to receptors is often overlooked. Here we analyze the magnitudes and origins of these interactions for a variety of biologically relevant small molecules using quantum chemical and automated docking calculations. In most cases examined in this study, the lowest energy conformation of the small molecule displays a sulfur-lone pair close contact. However, docking studies, both published and new, often predict that conformations without sulfur-lone pair contacts have the best binding affinity for their respective receptors. This is a serious problem. Since many of these predicted bound conformations are not actually energetically accessible, pursuing design (e.g., drug design) around these binding modes necessarily will lead, serendipity aside, to dead end designs. Our results constitute a caution that one best not neglect these interactions when predicting the binding affinities of potential ligands (drugs or not) for hosts (enzymes, receptors, DNA, RNA, synthetic hosts). Moreover, a better understanding and awareness of sulfur-lone pair interactions should facilitate the rational modulation of host-guest interactions involving sulfur-containing molecules.
Collapse
Affiliation(s)
- B M Hudson
- Department of Chemistry, University of California, Davis, CA 95618, USA.
| | | | | |
Collapse
|
20
|
Unzue A, Zhao H, Lolli G, Dong J, Zhu J, Zechner M, Dolbois A, Caflisch A, Nevado C. The “Gatekeeper” Residue Influences the Mode of Binding of Acetyl Indoles to Bromodomains. J Med Chem 2016; 59:3087-97. [DOI: 10.1021/acs.jmedchem.5b01757] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Andrea Unzue
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Hongtao Zhao
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Graziano Lolli
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Jing Dong
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Jian Zhu
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Melanie Zechner
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Aymeric Dolbois
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Amedeo Caflisch
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Cristina Nevado
- Department of Chemistry and ‡Department of
Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| |
Collapse
|
21
|
Xu M, Caflisch A, Hamm P. Protein Structural Memory Influences Ligand Binding Mode(s) and Unbinding Rates. J Chem Theory Comput 2016; 12:1393-9. [PMID: 26799675 DOI: 10.1021/acs.jctc.5b01052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The binding of small molecules (e.g., natural ligands, metabolites, and drugs) to proteins governs most biochemical pathways and physiological processes. Here, we use molecular dynamics to investigate the unbinding of dimethyl sulfoxide (DMSO) from two distinct states of a small rotamase enzyme, the FK506-binding protein (FKBP). These states correspond to the FKBP protein relaxed with and without DMSO in the active site. Since the time scale of ligand unbinding (2-20 ns) is faster than protein relaxation (100 ns), a novel methodology is introduced to relax the protein without having to introduce an artificial constraint. The simulation results show that the unbinding time is an order of magnitude longer for dissociation from the DMSO-bound state (holo-relaxed). That is, the actual rate of unbinding depends on the state of the protein, with the protein having a long-lived memory. The rate thus depends on the concentration of the ligand as the apo and holo states reflect low and high concentrations of DMSO, respectively. Moreover, there are multiple binding modes in the apo-relaxed state, while a single binding mode dominates the holo-relaxed state in which DMSO acts as hydrogen bond acceptor from the backbone NH of Ile56, as in the crystal structure of the DMSO/FKBP complex. The solvent relaxes very fast (∼1 ns) close to the NH of Ile56 and with the same time scale of the protein far away from the active site. These results have implications for high-throughput docking, which makes use of a rigid structure of the protein target.
Collapse
Affiliation(s)
- Min Xu
- Department of Biochemistry and ‡Department of Chemistry, University of Zürich , Winterthurerstrasse 190, Zürich CH-8057, Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry and ‡Department of Chemistry, University of Zürich , Winterthurerstrasse 190, Zürich CH-8057, Switzerland
| | - Peter Hamm
- Department of Biochemistry and ‡Department of Chemistry, University of Zürich , Winterthurerstrasse 190, Zürich CH-8057, Switzerland
| |
Collapse
|
22
|
Sun Y, Zhao H, Wang J, Zhu J, Wu S. Identification and regulation of the catalytic promiscuity of (−)-γ-lactamase from Microbacterium hydrocarbonoxydans. Appl Microbiol Biotechnol 2015; 99:7559-68. [DOI: 10.1007/s00253-015-6503-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/17/2015] [Indexed: 02/02/2023]
|
23
|
Zhao H, Caflisch A. Molecular dynamics in drug design. Eur J Med Chem 2015; 91:4-14. [DOI: 10.1016/j.ejmech.2014.08.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/31/2014] [Accepted: 08/03/2014] [Indexed: 11/30/2022]
|
24
|
Li GB, Yang LL, Yuan Y, Zou J, Cao Y, Yang SY, Xiang R, Xiang M. Virtual screening in small molecule discovery for epigenetic targets. Methods 2015; 71:158-66. [DOI: 10.1016/j.ymeth.2014.11.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/27/2014] [Accepted: 11/14/2014] [Indexed: 12/31/2022] Open
|
25
|
Small-molecule inhibitors of spleen tyrosine kinase as therapeutic agents for immune disorders: will promise meet expectations? Future Med Chem 2014; 6:1811-27. [DOI: 10.4155/fmc.14.126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Following on the heels of the US FDA approval of tofacitinib (Xeljanz, Pfizer, USA), an inhibitor of the JAK family members, and ibrutinib (Imbruvica, Janssen, Belgium), an inhibitor of BTK, for the treatment of rheumatoid arthritis and chronic lymphocytic leukemia, respectively, there is now renewed interest in the biopharmaceutical industry in the development of orally active small-molecule agents targeting key protein kinases implicated in immune regulation. One such ‘immunokinase’ target is SYK, a non-receptor tyrosine protein kinase critical for transducing intracellular signaling cascades for various immune recognition receptors, such as the B-cell receptor and the Fc receptor. Here, we review and discuss the progress and challenges in the development of small-molecule inhibitors of SYK and their potential as a new class of disease-modifying immunosuppressive agents for certain inflammatory and autoimmune disorders.
Collapse
|
26
|
Zhao H, Gartenmann L, Dong J, Spiliotopoulos D, Caflisch A. Discovery of BRD4 bromodomain inhibitors by fragment-based high-throughput docking. Bioorg Med Chem Lett 2014; 24:2493-6. [PMID: 24767840 DOI: 10.1016/j.bmcl.2014.04.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/13/2022]
Abstract
Bromodomains (BRDs) recognize acetyl-lysine modified histone tails mediating epigenetic processes. BRD4, a protein containing two bromodomains, has emerged as an attractive therapeutic target for several types of cancer as well as inflammatory diseases. Using a fragment-based in silico screening approach, we identified two small molecules that bind to the first bromodomain of BRD4 with low-micromolar affinity and favorable ligand efficiency (0.37 kcal/mol per non-hydrogen atom), selectively over other families of bromodomains. Notably, the hit rate of the fragment-based in silico approach is about 10% as only 24 putative inhibitors, from an initial library of about 9 million molecules, were tested in vitro.
Collapse
Affiliation(s)
- Hongtao Zhao
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Lisa Gartenmann
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Jing Dong
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Dimitrios Spiliotopoulos
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|