1
|
Zhang D, Han B, Chen XF, Zhao S, Li WX, Zhang H, Zhang ML, Huo MQ, Qiu YS, Ren YJ, Zhang YD, Ren XQ, Wang W, Tang JF. A Biolayer Interferometry-Based SARS-COV-2 Mpro-Targeted Active Ingredients Recognition System: Construction and Application in Ligand Screening From Herbal Medicines. PHYTOCHEMICAL ANALYSIS : PCA 2025; 36:718-731. [PMID: 39568228 DOI: 10.1002/pca.3462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 11/22/2024]
Abstract
INTRODUCTION Drug discovery research targeting SARS-CoV-2 and other emerging pathogens remains critically important. Active compounds derived from plants frequently serve as lead compounds for further drug discovery; however, numerous unrelated chemical constituents in crude extracts may obscure the effective ingredients in LC-MS analysis. OBJECTIVE The aim of this study is to construct a biolayer interferometry (BLI)-based system for recognizing active ingredients that inhibit the main protease (Mpro) of SARS-CoV-2 and to identify the active chemical components binding to Mpro from herbal medicines. METHODOLOGY We developed a novel FRET fluorogenic probe by linking the amino acid sequences of the fluorescent proteins Lssmorange and mKate2 (Ls-mK). The interaction between traditional Chinese medicine and Mpro was analyzed using BLI. Ultrahigh performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) was employed to analyze the composition of herbal medicines. RESULTS Fluorescence detection and spectroscopy confirmed the successful construction of an Mpro inhibitor screening system. Lanqin Oral Liquid (LQL) and Gardeniae fructus exhibited strong inhibitory effects on Mpro. Ten compounds were identified from G. fructus extracts; among them, deacetyl asperulosidic acid methyl ester (DAAME) and Gardoside were found to strongly bind to Mpro, with dissociation constants (KD) of 3.41 μM and 801 nM, respectively. The half-maximal inhibitory concentrations (IC50) of DAAME and Gardoside for Mpro were 27.46 and 13.7 μM, respectively. CONCLUSION This study established a functional Mpro inhibitor screening system. Among the 10 components identified from G. fructus that bind to Mpro, DAAME and Gardoside displayed strong binding and inhibitory activity, indicating their potential as lead compounds for inhibiting SARS-CoV-2 viral replication.
Collapse
Affiliation(s)
- Dai Zhang
- Department of Laboratory Medicine, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Bing Han
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Fei Chen
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Shuai Zhao
- Henan Provincial Key Laboratory of Pediatric Genes and Metabolic Diseases, Zhengzhou University Affiliated Children's Hospital, Zhengzhou, China
| | - Wei-Xia Li
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Department of Pharmacy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Hui Zhang
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ming-Liang Zhang
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Meng-Qi Huo
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yong-Sheng Qiu
- Department of Anesthesiology, Zhengzhou University Affiliated Children's Hospital, Zhengzhou, China
| | - Ying-Jie Ren
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yao-Dong Zhang
- Henan Provincial Key Laboratory of Pediatric Genes and Metabolic Diseases, Zhengzhou University Affiliated Children's Hospital, Zhengzhou, China
| | - Xian-Qing Ren
- Department of Pediatrics, Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Wei Wang
- Department of Infection, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jin-Fa Tang
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Department of Pharmacy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
2
|
Zhang Y, Wang D, Wang X, Ma H, Liu Y, Hong Z, Zhu Z, Chen X, Lv D, Cao Y, Chai Y. A dual-target SPR screening system for simultaneous ligand discovery of SARS-CoV-2 spike protein and its receptor ACE2 from Chinese herbs. J Pharm Biomed Anal 2024; 245:116142. [PMID: 38631070 DOI: 10.1016/j.jpba.2024.116142] [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] [Received: 01/12/2024] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
Traditional Chinese Medicine (TCM) is a supremely valuable resource for the development of drug discovery. Few methods are capable of hunting for potential molecule ligands from TCM towards more than one single protein target. In this study, a novel dual-target surface plasmon resonance (SPR) biosensor was developed to perform targeted compound screening of two key proteins involved in the cellular invasion process of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2): the spike (S) protein receptor binding domain (RBD) and the angiotensin-converting enzyme 2 (ACE2). The screening and identification of active compounds from six Chinese herbs were conducted taking into consideration the multi-component and multi-target nature of Traditional Chinese Medicine (TCM). Puerarin from Radix Puerariae Lobatae was discovered to exhibit specific binding affinity to both S protein RBD and ACE2. The results highlight the efficiency of the dual-target SPR system in drug screening and provide a novel approach for exploring the targeted mechanisms of active components from Chinese herbs for disease treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Dongyao Wang
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Xiying Wang
- Suzhou Innovation Center of Shanghai University, Suzhou 215127, China
| | - Huilin Ma
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Yue Liu
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zhanying Hong
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zhenyu Zhu
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Center for Instrumental Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Xiaofei Chen
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Center for Instrumental Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Diya Lv
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Center for Instrumental Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Yan Cao
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Yifeng Chai
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai 200433, China; Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
3
|
Jin H, Cui D, Fan Y, Li G, Zhong Z, Wang Y. Recent advances in bioaffinity strategies for preclinical and clinical drug discovery: Screening natural products, small molecules and antibodies. Drug Discov Today 2024; 29:103885. [PMID: 38278476 DOI: 10.1016/j.drudis.2024.103885] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/26/2023] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
Bioaffinity drug screening strategies have gained popularity in preclinical and clinical drug discovery for natural products, small molecules and antibodies owing to their superior selectivity, the large number of compounds to be screened and their ability to minimize the time and expenses of the drug discovery process. This paper provides a systematic summary of the principles of commonly used bioaffinity-based screening methods, elaborates on the success of bioaffinity in clinical drug development and summarizes the active compounds, preclinical drugs and marketed drugs obtained through affinity screening methods. Owing to the high demand for new drugs, bioaffinity-guided screening techniques will play a greater part in clinical drug development.
Collapse
Affiliation(s)
- Haochun Jin
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Dianxin Cui
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Yu Fan
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China; Zhuhai UM Science and Technology Research Institute, Zhuhai 519031, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China; Zhuhai UM Science and Technology Research Institute, Zhuhai 519031, China.
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China.
| | - Yitao Wang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China.
| |
Collapse
|
4
|
Damergi B, Essid R, Fares N, Khadraoui N, Ageitos L, Ben Alaya A, Gharbi D, Abid I, Rashed Alothman M, Limam F, Rodríguez J, Jiménez C, Tabbene O. Datura stramonium Flowers as a Potential Natural Resource of Bioactive Molecules: Identification of Anti-Inflammatory Agents and Molecular Docking Analysis. Molecules 2023; 28:5195. [PMID: 37446858 DOI: 10.3390/molecules28135195] [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: 04/25/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
The present study investigated the antioxidant, antibacterial, antiviral and anti-inflammatory activities of different aerial parts (flowers, leaves and seeds) of Datura stramonium. The plant material was extracted with 80% methanol for about 24 h. The sensitivity to microorganisms analysis was performed by the microdilution technique. Antioxidant tests were performed by scavenging the DPPH and ABTS radicals, and by FRAP assay. Anti-inflammatory activity was evaluated through the inhibition of nitric oxide production in activated macrophage RAW 264.7 cells. Cell viability was assessed with an MTT assay. Results show that the flower extract revealed a powerful antimicrobial capacity against Gram-positive bacteria and strong antioxidant and anti-inflammatory activities. No significant cytotoxicity to activated macrophages was recorded. High resolution electrospray ionization mass spectrometry and nuclear magnetic resonance analysis identified two molecules with important anti-inflammatory effects: 12α-hydroxydaturametelin B and daturametelin B. Molecular docking analysis with both pro-inflammatory agents tumor necrosis factor alpha and interleukin-6 revealed that both compounds showed good binding features with the selected target proteins. Our results suggest that D. stramonium flower is a promising source of compounds with potential antioxidant, antibacterial, and anti-inflammatory activities. Isolated withanolide steroidal lactones from D. stramonium flower extract with promising anti-inflammatory activity have therapeutic potential against inflammatory disorders.
Collapse
Affiliation(s)
- Bilel Damergi
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
- Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 2092, Tunisia
| | - Rym Essid
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Nadia Fares
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Nadine Khadraoui
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
- Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 2092, Tunisia
| | - Lucía Ageitos
- Centro Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 Coruña, Spain
| | - Ameni Ben Alaya
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
- Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 2092, Tunisia
| | - Dorra Gharbi
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Islem Abid
- Botany and Microbiology Department, Science College, King Saud University, Riyadh 11451, Saudi Arabia
| | - Monerah Rashed Alothman
- Botany and Microbiology Department, Science College, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ferid Limam
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Jaime Rodríguez
- Centro Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 Coruña, Spain
| | - Carlos Jiménez
- Centro Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 Coruña, Spain
| | - Olfa Tabbene
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| |
Collapse
|
5
|
Li X, Yang W, Chen H, Pan F, Liu W, Qi D, Yu S, Liu H, Chai X, Liu Y, Pan Y, Wang G. Rapid screening and in vivo target occupancy quantitative evaluation of xanthine oxidase inhibitors based on drug-target binding kinetics research strategy: A case study of Chrysanthemum morifolium Ramat. Biomed Pharmacother 2023; 161:114379. [PMID: 36827711 DOI: 10.1016/j.biopha.2023.114379] [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: 11/30/2022] [Revised: 01/26/2023] [Accepted: 02/05/2023] [Indexed: 02/24/2023] Open
Abstract
Chrysanthemum morifolium Ramat. is a kind of food and drug dual-use traditional Chinese medicine possessing multiple pharmacological and biochemical benefits. In our study, a rapid and high-throughput method based on Surface plasmon resonance (SPR) biosensor technology was developed and verified for screening potential xanthine oxidase (XOD) inhibitors exemplarily in the Chrysanthemum morifolium Ramat. Coupled with ultra-high performance liquid chromatography-electrospray ionization-tandem mass spectrometry (UPLC-ESI-MS), 14 XOD-binders were identified. In the SPR-based biosensor and molecular docking analysis, most compounds exhibited a strong affinity and binding kinetic property (association rate constant, Kon and dissociation rate constant, Koff) for XOD and could be regarded as potential inhibitors. More importantly, to further accurately assess target occupancy of candidate compounds in vivo, a mathematical model was established and verified involving three crucial intrinsic kinetic processes (Pharmacokinetics, Binding kinetic and Target kinetic). Overall, the proposed screening and assessment strategy could be proved an effective theoretical basis for further pharmacodynamic evaluation.
Collapse
Affiliation(s)
- Xueyan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenning Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongjiao Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fulu Pan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wei Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongying Qi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shuang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huining Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoyu Chai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yanli Pan
- Institute of Information on Traditional Chinese Medicine China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Guopeng Wang
- Zhongcai Health (Beijing) Biological Technology Development Co., Ltd., Beijing 101500, China.
| |
Collapse
|
6
|
Javaid N, Patra MC, Cho DE, Batool M, Kim Y, Choi GM, Kim MS, Hahm DH, Choi S. An orally active, small-molecule TNF inhibitor that disrupts the homotrimerization interface improves inflammatory arthritis in mice. Sci Signal 2022; 15:eabi8713. [DOI: 10.1126/scisignal.abi8713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Excessive signaling by the proinflammatory cytokine TNF is involved in several autoimmune diseases, including rheumatoid arthritis (RA). However, unlike the approved biologics currently used to treat this and other conditions, commercially available small-molecule inhibitors of TNF trimerization are cytotoxic or exhibit low potency. Here, we report a TNF-inhibitory molecule (TIM) that reduced TNF signaling in vitro and was an effective treatment in a mouse model of RA. The initial lead compound, TIM1, attenuated TNF-induced apoptosis of human and mouse cells by delaying the induction of proinflammatory NF-κB and MAPK signaling and caspase 3– and caspase 8–dependent apoptosis. TIM1 inhibited the secretion of the proinflammatory cytokines IL-6 and IL-8 by disrupting TNF homotrimerization, thereby preventing its association with the TNF receptor. In a mouse model of collagen-induced polyarthritis, the more potent TIM1 analog TIM1c was orally bioavailable and reduced paw swelling, histological indicators of knee joint pathology, inflammatory infiltration of the joint, and the overall arthritis index. Orally delivered TIM1c showed immunological effects similar to those elicited by intraperitoneal injection of the FDA-approved TNF receptor decoy etanercept. Thus, TIM1c is a promising lead compound for the development of small-molecule therapies for the treatment of RA and other TNF-dependent systemic inflammation disorders.
Collapse
Affiliation(s)
- Nasir Javaid
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Mahesh Chandra Patra
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Da-Eun Cho
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Maria Batool
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
| | - Yoongeun Kim
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Gwang Muk Choi
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Dae-Hyun Hahm
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
| |
Collapse
|
7
|
Zhou W, Wang M, Zhang A, Huang D, Guo H, Shen G. Directional screening and identification of potential cytotoxic components from Achnatherum inebrians by a combination of surface palsmon resonance and chromatography. CHINESE HERBAL MEDICINES 2022. [DOI: 10.1016/j.chmed.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
8
|
Li S, Li X, Wang H, Jia X, Mao H, Dong F, Zhao T, Gao Y, Zhang C, Bai R, Liu R, Yan L, Ji Y, Zhang N, Wang W. The Hypoglycemic Effect of JinQi Jiangtang Tablets Is Partially Dependent on the Palmatine-Induced Activation of the Fibroblast Growth Factor Receptor 1 Signaling Pathway. Front Pharmacol 2022; 13:895724. [PMID: 35935824 PMCID: PMC9354937 DOI: 10.3389/fphar.2022.895724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
JinQi Jiangtang tablet (JQJTT) is a Chinese patent medicine that has been shown to be beneficial for patients with diabetes both preclinically and clinically; however, the molecular mechanism underlying the effects of JQJTT remains unclear. In this study, surface plasmon resonance fishing was employed to identify JQJTT constituent molecules that can specifically bind to fibroblast growth factor receptor 1 (FGFR1), leading to the retrieval of palmatine (PAL), a key active ingredient of JQJTT. In vivo and in vitro experiments demonstrated that PAL can significantly stimulate FGFR1 phosphorylation and upregulate glucose transporter type 1 (GLUT-1) expression, thereby facilitating glucose uptake in insulin resistance (IR) HepG2 cells as well as alleviating hyperglycemia in diabetic mice. Our results revealed that PAL functions as an FGFR1 activator and that the hypoglycemic effect of JQJTT is partially dependent on the PAL-induced activation of the FGFR1 pathway. In addition, this study contributed to the understanding the pharmacodynamic basis and mechanism of action of JQJTT and provided a novel concept for future research on PAL.
Collapse
Affiliation(s)
- Siming Li
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Xiaoling Li
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - HeMeng Wang
- College of Life Sciences, Tarim University, Alar, China
- College of Life Sciences, Northeast Agricultural University, Harbin, China
| | - Xinhang Jia
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Haoyang Mao
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Fangxin Dong
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Tingting Zhao
- Aier School of Ophthalmology, Central South University, Changsha, China
| | - Yuan Gao
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Chen Zhang
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Ruisong Bai
- College of Life Sciences, Northeast Agricultural University, Harbin, China
| | - Ruihao Liu
- College of Life Sciences, Northeast Agricultural University, Harbin, China
| | - Lijun Yan
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Yubin Ji
- School of Pharmacy, Harbin University of Commerce, Harbin, China
- *Correspondence: Yubin Ji, ; Wenfei Wang,
| | - Na Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin, China
| | - Wenfei Wang
- College of Life Sciences, Northeast Agricultural University, Harbin, China
- *Correspondence: Yubin Ji, ; Wenfei Wang,
| |
Collapse
|
9
|
Immunosuppressant Therapies in COVID-19: Is the TNF Axis an Alternative? Pharmaceuticals (Basel) 2022; 15:ph15050616. [PMID: 35631442 PMCID: PMC9147078 DOI: 10.3390/ph15050616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/05/2023] Open
Abstract
The study of cytokine storm in COVID-19 has been having different edges in accordance with the knowledge of the disease. Various cytokines have been the focus, especially to define specific treatments; however, there are no conclusive results that fully support any of the options proposed for emergency treatment. One of the cytokines that requires a more exhaustive review is the tumor necrosis factor (TNF) and its receptors (TNFRs) as increased values of soluble formats for both TNFR1 and TNFR2 have been identified. TNF is a versatile cytokine with different impacts at the cellular level depending on the action form (transmembrane or soluble) and the receptor to which it is associated. In that sense, the triggered mechanisms can be diversified. Furthermore, there is the possibility of the joint action provided by synergism between one or more cytokines with TNF, where the detonation of combined cellular processes has been suggested. This review aims to discuss some roles of TNF and its receptors in the pro-inflammatory stage of COVID-19, understand its ways of action, and let to reposition this cytokine or some of its receptors as therapeutic targets.
Collapse
|
10
|
Yao L, Liao M, Wang JK, Wang J, Liu D, Tu PF, Zeng KW. Gold Nanoparticle-Based Photo-Cross-Linking Strategy for Cellular Target Identification of Supercomplex Molecular Systems. Anal Chem 2022; 94:3180-3187. [PMID: 35133791 DOI: 10.1021/acs.analchem.1c04652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cellular target identification plays an essential role in innovative drug development and pharmacological mechanism elucidation. However, very few practical experimental methodologies have been developed for identifying target proteins for supercomplex molecular systems such as biologically active phytochemicals or pharmaceutical compositions. To overcome this limitation, we synthesized gold nanoparticles (AuNPs) as solid scaffolds, which were bound with 4,4'-dihydroxybenzophenone (DHBP) as a photo-cross-linking group on the surface. Then, DHBP-modified AuNPs cross-linked various organic compounds from phytochemicals under ultraviolet radiation via carbene reactions, H-C bond insertion, for catalytic C-C bond formation. We next used the phytochemical-cross-linked AuNPs (phytoAuNPs) to pull down potential binding proteins from brain tissue lysate and identified 13 neuroprotective targets by mass spectrometry analysis. As an exemplary study, we selected Hsp60 as a crucial cellular target to further screen 14 target-binding compounds from phytochemicals through surface plasmon resonance (SPR) analysis, followed by Hsp60 activity detection and neuroprotective effect assay in cells. Collectively, this gold nanoparticle-based photo-cross-linking strategy can serve as a useful platform for discovering novel cellular targets for supercomplex molecular systems and help to explore pharmacological mechanisms and active substances.
Collapse
Affiliation(s)
- Lu Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Min Liao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jing-Kang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Dan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
11
|
Lv D, Xu J, Qi M, Wang D, Xu W, Qiu L, Li Y, Cao Y. A strategy of screening and binding analysis of bioactive components from traditional Chinese medicine based on surface plasmon resonance biosensor. J Pharm Anal 2021; 12:500-508. [PMID: 35811628 PMCID: PMC9257445 DOI: 10.1016/j.jpha.2021.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
Elucidating the active components of traditional Chinese medicine (TCM) is essential for understanding the mechanisms of TCM and promote its rational use as well as TCM-derived drug development. Recent studies have shown that surface plasmon resonance (SPR) technology is promising in this field. In the present study, we propose an SPR-based integrated strategy to screen and analyze the major active components of TCM. We used Radix Paeoniae Alba (RPA) as an example to identify the compounds that can account for its anti-inflammatory mechanism via tumor necrosis factor receptor type 1 (TNF-R1). First, RPA extraction was analyzed using an SPR-based screening system, and the potential active ingredients were collected, enriched, and identified as paeoniflorin and paeonol. Next, the affinity constants of paeoniflorin and paeonol were determined as 4.9 and 11.8 μM, respectively. Then, SPR-based competition assays and molecular docking were performed to show that the two compounds could compete with tumor necrosis factor-α (TNF-α) while binding to the subdomain 1 site of TNF-R1. Finally, in biological assays, the two compounds suppressed cytotoxicity and apoptosis induced by TNF-α in the L929 cell line. These findings prove that SPR technology is a useful tool for determining the active ingredients of TCM at the molecular level and can be used in various aspects of drug development. The SPR-based integrated strategy is reliable and feasible in TCM studies and will shed light on the elucidation of the pharmacological mechanism of TCM and facilitate its modernization. A surface plasmon resonance-based integrated strategy was established to analyze traditional Chinese medicine. Surface plasmon resonance technology can be used for ligand screening, affinity detection, and binding site confirmation. Paeoniflorin and paeonol were identified as TNF-R1-bound ingredients in RPA.
Collapse
Affiliation(s)
- Diya Lv
- Center for Instrumental Analysis, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Jin Xu
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Minyu Qi
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Dongyao Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Weiheng Xu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Lei Qiu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yinghua Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Corresponding author.
| | - Yan Cao
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
- Corresponding author.
| |
Collapse
|
12
|
Tao Y, Chen L, Pan M, Zhu F, Zhu D. Tailored Biosensors for Drug Screening, Efficacy Assessment, and Toxicity Evaluation. ACS Sens 2021; 6:3146-3162. [PMID: 34516080 DOI: 10.1021/acssensors.1c01600] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biosensors have been flourishing in the field of drug discovery with pronounced developments in the past few years. They facilitate the screening and discovery of innovative drugs. However, there is still a lack of critical reviews that compare the merits and shortcomings of these biosensors from a pharmaceutical point of view. This contribution presents a critical and up-to-date overview on the recent progress of tailored biosensors, including surface plasmon resonance, fluorescent, photoelectrochemical, and electrochemical systems with emphasis on their mechanisms and applications in drug screening, efficacy assessment, and toxicity evaluation. Multiple functional nanomaterials have also been incorporated into the biosensors. Representative examples of each type of biosensors are discussed in terms of design strategy, response mechanism, and potential applications. In the end, we also compare the results and summarize the major insights gained from the works, demonstrating the challenges and prospects of biosensors-assisted drug discovery.
Collapse
Affiliation(s)
- Yi Tao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lin Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Meiling Pan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fei Zhu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| |
Collapse
|
13
|
A Novel Competitive Binding Screening Assay Reveals Sennoside B as a Potent Natural Product Inhibitor of TNF-α. Biomedicines 2021; 9:biomedicines9091250. [PMID: 34572435 PMCID: PMC8465676 DOI: 10.3390/biomedicines9091250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 11/17/2022] Open
Abstract
Natural products (NPs) have played a significant role in drug discovery for diverse diseases, and numerous attempts have been made to discover promising NP inhibitors of tumor necrosis factor α (TNF-α), a major therapeutic target in autoimmune diseases. However, NP inhibitors of TNF-α, which have the potential to be developed as new drugs, have not been reported for over a decade. To facilitate the search for new promising inhibitors of TNF-α, we developed an efficient competitive binding screening assay based on analytical size exclusion chromatography coupled with liquid chromatography-tandem mass spectrometry. Application of this screening method to the NP library led to the discovery of a potent inhibitor of TNF-α, sennoside B, with an IC50 value of 0.32 µM in TNF-α induced HeLa cell toxicity assays. Surprisingly, the potency of sennoside B was 5.7-fold higher than that of the synthetic TNF-α inhibitor SPD304. Molecular docking was performed to determine the binding mode of sennoside B to TNF-α. In conclusion, we successfully developed a novel competition binding screening method to discover small molecule TNF-α inhibitors and identified the natural compound sennoside B as having exceptional potency.
Collapse
|
14
|
Dong Q, Hu N, Yue H, Wang H, Ku J. Identification of α-glucosidase inhibitors from the bran of Chenopodium quinoa Willd. by surface plasmon resonance coupled with ultra-performance liquid chromatography and quadrupole-time-of-flight-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1181:122919. [PMID: 34500401 DOI: 10.1016/j.jchromb.2021.122919] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/26/2021] [Accepted: 08/29/2021] [Indexed: 01/17/2023]
Abstract
Extracts from the bran of Chenopodium quinoa Willd. (QBE) were reported to be active in inhibiting α-glycosidase, a promising target for treatment of diabetes mellitus. However, the constituents responsible for the α-glucosidase-inhibiting activity of QBE have not been fully characterized. The present study aimed to set up a method for rapid identification of glycosidase inhibiting compounds from the quinoa bran. With surface plasmon resonance (SPR) coupled with liquid chromatography-mass spectrometry (LC-MS), we identified eight flavonoids and ten triterpenoid saponins that may bind to the α-glycosidase. Analysis of the interaction kinetics by molecular docking supported their α-glucosidase-inhibiting activity and revealed the potential mechanisms for the inhibitory effects. In summary, this study established a SPR and LC-MS-based method for rapid in vitro screening of α-glucosidase inhibitors and suggested the quinoa bran a potential natural source of α-glucosidase inhibitors.
Collapse
Affiliation(s)
- Qi Dong
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research and CAS Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Qinghai 810008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Na Hu
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research and CAS Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Qinghai 810008, China
| | - Huilan Yue
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research and CAS Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Qinghai 810008, China
| | - Honglun Wang
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research and CAS Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Qinghai 810008, China.
| | - Jinliang Ku
- Beijing Tongrentang Health Pharmaceutical (Qinghai) Co. Ltd, Qinghai 817000, China
| |
Collapse
|
15
|
Transmembrane TNF and Its Receptors TNFR1 and TNFR2 in Mycobacterial Infections. Int J Mol Sci 2021; 22:ijms22115461. [PMID: 34067256 PMCID: PMC8196896 DOI: 10.3390/ijms22115461] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor necrosis factor (TNF) is one of the main cytokines regulating a pro-inflammatory environment. It has been related to several cell functions, for instance, phagocytosis, apoptosis, proliferation, mitochondrial dynamic. Moreover, during mycobacterial infections, TNF plays an essential role to maintain granuloma formation. Several effector mechanisms have been implicated according to the interactions of the two active forms, soluble TNF (solTNF) and transmembrane TNF (tmTNF), with their receptors TNFR1 and TNFR2. We review the impact of these interactions in the context of mycobacterial infections. TNF is tightly regulated by binding to receptors, however, during mycobacterial infections, upstream activation signalling pathways may be influenced by key regulatory factors either at the membrane or cytosol level. Detailing the structure and activation pathways used by TNF and its receptors, such as its interaction with solTNF/TNFRs versus tmTNF/TNFRs, may bring a better understanding of the molecular mechanisms involved in activation pathways which can be helpful for the development of new therapies aimed at being more efficient against mycobacterial infections.
Collapse
|
16
|
Cao Y, Cao Y, Shi Y, Cai Y, Chen L, Wang D, Liu Y, Chen X, Zhu Z, Hong Z, Chai Y. Surface plasmon resonance biosensor combined with lentiviral particle stabilization strategy for rapid and specific screening of P-Glycoprotein ligands. Anal Bioanal Chem 2021; 413:2021-2031. [PMID: 33528601 DOI: 10.1007/s00216-021-03170-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/24/2020] [Accepted: 01/07/2021] [Indexed: 11/28/2022]
Abstract
A novel surface plasmon resonance-based P-gp ligand screening system (SPR-PLSS) combined with lentiviral particle (LVP) stabilization strategy was constructed to screen out potential P-gp inhibitors from natural products. Firstly, we constructed LVPs with high and low expression levels of P-gp. The LVPs can ensure the natural conformation of P-gp based on the principle that LVPs germinated from packaging cells will contain cell membrane fragments and P-gp they carry. Then the LVPs with high P-gp expression for active channel and LVPs with low P-gp expression for reference channel were immobilized on CM5 chip respectively. The affinity detection was thus carried out with the signal reduction on the two channels. The P-gp inhibitors, Valspodar (Val) and cyclosporin (CsA), as positive compounds, were detected to characterize the chip's activity, and the KD of Val and CsA were 14.09 μM and 16.41 μM, respectively. Forty compounds from natural product library were screened using the SPR CM5 chip, and magnolol (Mag), honokiol (Hon), and resveratrol (Res) were screened out as potential P-gp ligands, showing a significant response signal. This work presented a novel P-gp ligand screening system based on LVP-immobilized biosensor to rapidly screen out P-gp ligands from natural product library. Compared with traditional cell experiments which the screening time may take up to several days, our method only takes several hours. Furthermore, this study has also provided solid evidences to support that some complicated membrane proteins would apply to the lentivirus-based SPR screening system.
Collapse
Affiliation(s)
- Yuhong Cao
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Yan Cao
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Yiwei Shi
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Ying Cai
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Langdong Chen
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Yue Liu
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Xiaofei Chen
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Zhenyu Zhu
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Zhanying Hong
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China.
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
17
|
Biolayer interferometry provides a robust method for detecting DNA binding small molecules in microbial extracts. Anal Bioanal Chem 2020; 413:1159-1171. [PMID: 33236226 DOI: 10.1007/s00216-020-03079-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/24/2020] [Accepted: 11/17/2020] [Indexed: 10/22/2022]
Abstract
DNA replication is an exceptional point of therapeutic intervention for many cancer types and several small molecules targeting DNA have been developed into clinically used antitumor agents. Many of these molecules are naturally occurring metabolites from plants and microorganisms, such as the widely used chemotherapeutic doxorubicin. While natural product sources contain a vast number of DNA binding small molecules, isolating and identifying these molecules is challenging. Typical screening campaigns utilize time-consuming bioactivity-guided fractionation approaches, which use sequential rounds of cell-based assays to guide the isolation of active compounds. In this study, we explore the use of biolayer interferometry (BLI) as a tool for rapidly screening natural product sources for DNA targeting small molecules. We first verified that BLI robustly detected DNA binding using designed GC- and AT-rich DNA oligonucleotides with known DNA intercalating, groove binding, and covalent binding agents including actinomycin D (1), doxorubicin (2), ethidium bromide (3), propidium iodide (4), Hoechst 33342 (5), and netropsin (6). Although binding varied with the properties of the oligonucleotides, measured binding affinities agreed with previously reported values. We next utilized BLI to screen over 100 bacterial extracts from our microbial library for DNA binding activity and found three highly active extracts. Binding-guided isolation was used to isolate the active principle component from each extract, which were identified as echinomycin (8), actinomycin V (9), and chartreusin (10). This biosensor-based DNA binding screen is a novel, low-cost, easy to use, and sensitive approach for medium-throughput screening of complex chemical libraries. Graphical abstract.
Collapse
|
18
|
Hou X, Sun M, Bao T, Xie X, Wei F, Wang S. Recent advances in screening active components from natural products based on bioaffinity techniques. Acta Pharm Sin B 2020; 10:1800-1813. [PMID: 33163336 PMCID: PMC7606101 DOI: 10.1016/j.apsb.2020.04.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 02/08/2023] Open
Abstract
Natural products have provided numerous lead compounds for drug discovery. However, the traditional analytical methods cannot detect most of these active components, especially at their usual low concentrations, from complex natural products. Herein, we reviewed the recent technological advances (2015–2019) related to the separation and screening bioactive components from natural resources, especially the emerging screening methods based on the bioaffinity techniques, including biological chromatography, affinity electrophoresis, affinity mass spectroscopy, and the latest magnetic and optical methods. These screening methods are uniquely advanced compared to other traditional methods, and they can fish out the active components from complex natural products because of the affinity between target and components, without tedious separation works. Therefore, these new tools can reduce the time and cost of the drug discovery process and accelerate the development of more effective and better-targeted therapeutic agents.
Collapse
Key Words
- AAs, amaryllidaceous alkaloids
- ABCA1, ATP-binding cassette transporter A1
- ACE, affinity capillary electrophoresis
- APTES, 3-aminopropyl-triethoxysilane
- ASMS, affinity selection mass spectrometry
- Active components
- Bioaffinity techniques
- CMC, Cell membrane chromatography
- CMMCNTs, Cell membrane magnetic carbon nanotube
- CMSP, Cell membrane stationary phase
- CNT, carbon nanotubes
- ChE, cholesterol efflux
- EGFR, epidermal growth factor receptor
- FP, fluorescence polarization
- Fe3O4–NH2, aminated magnetic nanoparticles
- HCS, high content screen
- HTS, high throughout screen
- HUVEC, human umbilical vein endothelial cells
- IMER, immobilized enzyme microreactor
- MAO-B, monoamine oxidases B
- MNP, immobilized on nanoparticles
- MPTS, 3-mercaptopropyl-trimethoxysilane
- MS, mass spectrometry
- MSPE, magnetic solid-phase extraction
- Natural products
- PD, Parkinson's disease
- PMG, physcion-8-O-β-d-monoglucoside
- RGD, arginine-glycine-aspartic acid
- SPR, surface plasmon resonance
- STAT3, signal transducer and activator of transcription 3
- Screening
- TCMs, traditional Chinese medicines
- TYR, tyrosinase
- TYR-MNPs, tyrosinase-immobilized magnetic nanoparticles
- Topo I, topoisomerase I
- UF, affinity ultrafiltration
- XOD, xanthine oxidase
- α1A-AR, α1A-adrenergic receptor
Collapse
|
19
|
Zhang N, Wang Z, Zhao Y. Selective inhibition of Tumor necrosis factor receptor-1 (TNFR1) for the treatment of autoimmune diseases. Cytokine Growth Factor Rev 2020; 55:80-85. [PMID: 32327345 DOI: 10.1016/j.cytogfr.2020.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
Anti-TNF biologics have achieved great success in the treatment of autoimmune diseases and have been the most selling biologics on market. However, the anti-TNF biologics have shown some disadvantages such as poor efficacy to some patients and high risk of infection and malignancies during clinical application. Current anti-TNF biologics are antibodies or antibody fragments that bind to TNF-α and subsequently block both TNF-TNFR1 and TNF-TNFR2 signaling. Transgenic animal studies indicate that TNFR1 signaling is responsible for chronic inflammation and cell apoptosis whereas TNFR2 signaling regulates tissue regeneration and inflammation. Recent studies propose to selectively inhibit TNFR1 to enhance efficacy and avoid side effects. In this review, we introduce the biology of TNF-TNFR1 and TNF-TNFR2 signaling, the advantages of selective inhibition of TNF-TNFR1 signaling and research updates on the development of selective inhibitors for TNF-TNFR1 signaling. Antibodies, small molecules and aptamers that selectively inhibit TNFR1 have showed therapeutic potential and less side effects in preclinical studies. Development of selective inhibitors for TNFR1 is a good strategy to enhance the efficacy and reduce the side effects of anti-TNF inhibitors and will be a trend for next-generation of anti-TNF inhibitors.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, HeNan Province, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, HeNan Province, Zhengzhou 450001, Henan, PR China
| | - Ziyi Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Yongxing Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, HeNan Province, Zhengzhou 450001, Henan, PR China; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, HeNan Province, Zhengzhou 450001, Henan, PR China.
| |
Collapse
|
20
|
Therapeutic potential of TNFα inhibitors in chronic inflammatory disorders: Past and future. Genes Dis 2020; 8:38-47. [PMID: 33569512 PMCID: PMC7859422 DOI: 10.1016/j.gendis.2020.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/13/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
In the past 20 years, patients with rheumatoid arthritis (RA), Crohn's disease (CD), and other immune diseases have witnessed the impact of a great treatment advance with the availability of biological TNFα inhibitors. With 5 approved anti-TNFα biologics on the market and soon available biosimilars, patients have more treatment options and have benefited from understanding the biology of TNFα. Nevertheless, many unmet needs remain for people living with TNFα-related diseases, namely some side effects and tolerance of current anti-TNFα biologics and resistance to therapies. Furthermore, common diseases such as osteoarthritis and back/neck pain may respond to anti-TNFα therapies at early onset of symptoms. Development of new TNFα inhibitors focusing on TNFR1 specific inhibitors, preferably small molecules that can be delivered orally, is much needed.
Collapse
|
21
|
Yang L, Hou A, Wang S, Zhang J, Man W, Guo X, Yang B, Wang Q, Jiang H, Kuang H. Screening and quantification of TNF-α ligand from Angelicae Pubescentis Radix by biosensor and UPLC-MS/MS. Anal Biochem 2020; 596:113643. [PMID: 32105738 DOI: 10.1016/j.ab.2020.113643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/29/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022]
Abstract
The aim of this study is to establish a method for rapid screening of active ingredients targeting TNF-α from Chinese herbal medicines. Take Angelicae Pubescentis Radix (APR) as an example, surface plasma resonance technique was used to establish for screening small molecule inhibitors of TNF-α from APR extract. Then UPLC-MS/MS coupled with chemometric was used for quantitative and evaluate the differences of the candidate compounds bound to TNF-α in APR from different sources. In the experiment, TNF-α protein was fixed on the CM5 chip surface of biacore T200 biosensor by amino coupling. A series of small molecular compounds in APR were screened and six phenolic acid compounds had a strong affinity for TNF-α protein and could be used as TNF-α antagonists. In summary, the targeted drug screening method for TNF-α protein based on SPR technology established in this study can be used to screen anti-TNF-α small molecule inhibitors. UPLC-MS/MS can accurately quantify 15 active ingredients, which provides reliable experimental data and new research ideas for targeted drug research on TNF-α protein.
Collapse
Affiliation(s)
- Liu Yang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China
| | - Ajiao Hou
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China
| | - Song Wang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China
| | - Jiaxu Zhang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China
| | - Wenjing Man
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China
| | - Xinyue Guo
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China
| | - Bingyou Yang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China
| | - Qiuhong Wang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 528458, PR China
| | - Hai Jiang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China.
| | - Haixue Kuang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, PR China.
| |
Collapse
|
22
|
Chen L, Lv D, Wang S, Wang D, Chen X, Liu Y, Hong Z, Zhu Z, Cao Y, Chai Y. Surface Plasmon Resonance-Based Membrane Protein-Targeted Active Ingredients Recognition Strategy: Construction and Implementation in Ligand Screening from Herbal Medicines. Anal Chem 2020; 92:3972-3980. [DOI: 10.1021/acs.analchem.9b05479] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Langdong Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Diya Lv
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Shaozhan Wang
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Xiaofei Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Yue Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Zhanying Hong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Zhenyu Zhu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Yan Cao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| |
Collapse
|
23
|
Physcion and physcion 8-O-β-glucopyranoside: A review of their pharmacology, toxicities and pharmacokinetics. Chem Biol Interact 2019; 310:108722. [DOI: 10.1016/j.cbi.2019.06.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/27/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022]
|
24
|
Protective effects of Clematichinenoside AR against inflammation and cytotoxicity induced by human tumor necrosis factor-α. Int Immunopharmacol 2019; 75:105563. [PMID: 31408840 DOI: 10.1016/j.intimp.2019.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 03/31/2019] [Accepted: 04/04/2019] [Indexed: 12/28/2022]
Abstract
Clematichinenoside AR (AR), a major active ingredient extracted from traditional Chinese herb Clematis chinensis Osbeck, has been demonstrated to possess anti-inflammatory and immune-modulatory activities in the treatment of experimental rheumatoid arthritis (RA). The therapeutic potential of AR was supposed to be closely correlated to its ability against tumor necrosis factor-α (TNF-α). Therefore, we aimed to explore the protective effects of Clematichinenoside AR against inflammation and cytotoxicity induced by human TNF-α. AR treatment significantly decreased IL-6 and IL-8 secretion, and attenuated MMP-1 production in human RA-derived fibroblast-like synoviocyte MH7A cells stimulated by recombinant human TNF-α (rhTNF-α). AR might antagonize rhTNF-α-induced responses in MH7A cells through inhibiting p38 and ERK MAPKs signal activation. In TNF-α-sensitive murine fibroblast L929 cells, AR treatment attenuated the proliferation inhibition ratio induced by rhTNF-α/ActD and antagonized rhTNF-α-induced cytotoxicity. The cellular and nuclear morphological alterations in apoptotic characteristics induced by rhTNF-α/ActD in L929 cells were observed to be attenuated by the pretreatment with AR under a phase-contrast and fluorescence microscopy, respectively. The Annexin V-FITC/PI double-staining assay was performed to confirm that AR pretreatment obviously decreased the cell death. The antagonistic effects of AR against rhTNF-α-induced cytotoxicity might be potentially attributed to the degeneration of reactive oxygen species and the increasing of mitochondrial membrane potential, along with the suppression of durative phosphorylation of c-Jun N-terminal kinase (JNK). Collectively, our results indicated that AR antagonizes the inflammatory and cytotoxic activities induced by human TNF-α effectively in vitro, which provided further evidence for a novel mechanism underlying AR for treating RA correlating with excessive TNF-α production.
Collapse
|
25
|
Identification of novel inhibitors for TNFα, TNFR1 and TNFα-TNFR1 complex using pharmacophore-based approaches. J Transl Med 2019; 17:215. [PMID: 31266509 PMCID: PMC6604280 DOI: 10.1186/s12967-019-1965-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tumor necrosis factor α (TNFα) is a multifunctional cytokine with a potent pro-inflammatory effect. It is a validated therapeutic target molecule for several disorders related to autoimmunity and inflammation. TNFα-TNF receptor-1 (TNFR1) signaling contributes to the pathological processes of these disorders. The current study is focused on finding novel small molecules that can directly bind to TNFα and/or TNFR1, preventing the interaction between TNFα or TNFR1, and regulating downstream signaling pathways. METHODS Cheminformatics pipeline (pharmacophore modeling, virtual screening, molecular docking and in silico ADMET analysis) was used to screen for novel TNFα and TNFR1 inhibitors in the Zinc database. The pharmacophore-based models were generated to screen for the best drug like compounds in the Zinc database. RESULTS The 39, 37 and 45 best hit molecules were mapped with the core pharmacophore features of TNFα, TNFR1, and the TNFα-TNFR1 complex respectively. They were further evaluated by molecular docking, protein-ligand interactions and in silico ADMET studies. The molecular docking analysis revealed the binding energies of TNFα, TNFR1 and the TNFα-TNFR1 complex, the basis of which was used to select the top five best binding energy compounds. Furthermore, in silico ADMET studies clearly revealed that all 15 compounds (ZINC09609430, ZINC49467549, ZINC13113075, ZINC39907639, ZINC25251930, ZINC02968981, ZINC09544246, ZINC58047088, ZINC72021182, ZINC08704414, ZINC05462670, ZINC35681945, ZINC23553920, ZINC05328058, and ZINC17206695) satisfied the Lipinski rule of five and had no toxicity. CONCLUSIONS The new selective TNFα, TNFR1 and TNFα-TNFR1 complex inhibitors can serve as anti-inflammatory agents and are promising candidates for further research.
Collapse
|
26
|
Identification of eupatilin and ginkgolide B as p38 ligands from medicinal herbs by surface plasmon resonance biosensor-based active ingredients recognition system. J Pharm Biomed Anal 2019; 171:35-42. [PMID: 30965219 DOI: 10.1016/j.jpba.2019.03.029] [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: 09/07/2018] [Revised: 01/25/2019] [Accepted: 03/14/2019] [Indexed: 12/17/2022]
Abstract
Screening of bioactive ligands for a certain protein target from medicinal herbs is a highly important yet challenging task during drug discovery process. In this study, a surface plasmon resonance biosensor-based active ingredient recognition system (SPR-AIRS) was applied to screen p38 mitogen-activated protein kinase (p38) ligands from herbal extracts. After p38 protein was immobilized on a SPR chip and the suitability of SPR-AIRS was validated, thirty-four p38-related medicinal herbs were selected and pre-screened. Two medicinal herbs having high response signal with p38-immobilized chip, Folium Ginkgo and Herba Artemisiae Scopariae, were injected into SPR system for ligand fishing. Among them, two active compounds, eupatilin (EPT) and ginkgolide B (GKB), were identified as p38 ligands, and then the KD values of EPT and GKB were measured as 21.68 ± 2.21 and 44.71 ± 1.80 μM, respectively. They can inhibit p38 activities significantly and bind to the ATP binding site on p38. Furthermore, EPT and GKB can inhibit cell proliferation (IC50 = 30.31 ± 6.84 and 42.97 ± 0.83 μM), induce apoptosis and G2/M cell cycle arrest against K562 cell line. This is the first time that EPT and GKB are reported as effective p38 binding ligands. These results prove that SPR-AIRS could be an effective method to screen active compounds acting on a specific protein from complex systems.
Collapse
|
27
|
Chen L, Lv D, Chen X, Liu M, Wang D, Liu Y, Hong Z, Zhu Z, Hu X, Cao Y, Yang J, Chai Y. Biosensor-Based Active Ingredients Recognition System for Screening STAT3 Ligands from Medical Herbs. Anal Chem 2018; 90:8936-8945. [PMID: 29953204 DOI: 10.1021/acs.analchem.8b01103] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Langdong Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Diya Lv
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Xiaofei Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Mingdong Liu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Yue Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Zhanying Hong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Zhenyu Zhu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Xiaoxia Hu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Yan Cao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Jianmin Yang
- Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| |
Collapse
|
28
|
Shaikh F, He J, Bhadra P, Chen X, Siu SWI. TNF Receptor Type II as an Emerging Drug Target for the Treatment of Cancer, Autoimmune Diseases, and Graft-Versus-Host Disease: Current Perspectives and In Silico Search for Small Molecule Binders. Front Immunol 2018; 9:1382. [PMID: 29967617 PMCID: PMC6015900 DOI: 10.3389/fimmu.2018.01382] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022] Open
Abstract
There is now compelling evidence that TNF receptor type II (TNFR2) is predominantly expressed on CD4+Foxp3+ regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), and plays a major role in the expansion and function of Tregs and MDSCs. Consequently, targeting of TNFR2 by either antagonists or agonists may represent a novel strategy in the treatment of cancer and autoimmune diseases, by downregulating or upregulating suppressor cell activity. The advance in the understanding of complex structure of TNFR2 and its binding with TNF at molecular levels offers opportunity for structure-guided drug discovery. This article reviews the current evidences regarding the decisive role of TNFR2 in immunosuppressive function of Tregs and MDSCs, and the current effort to develop novel TNFR2-targeting therapeutic agents in the treatment of cancer, autoimmune diseases, and graft-versus-host disease. To shed light on the potential TNFR2-targeting small molecules, we for the first time performed virtual screening of 400,000 natural compounds against the two TNF-binding sites, regions 3 and 4, of TNFR2. Our result showed that the top hits at region 4 had slightly higher docking energies than those at region 3. Nevertheless, free energy calculation from the TNF–TNFR2 molecular dynamics simulation revealed that the binding strength of TNF in region 3 is only one-tenth of that in region 4. This suggests that region 3 is a potentially more viable binding site to be targeted by small molecules than region 4. Therefore, the effectiveness in targeting region 3 of TNFR2 deserves further investigation.
Collapse
Affiliation(s)
- Faraz Shaikh
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Macao, China
| | - Jiang He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Pratiti Bhadra
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Macao, China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Shirley W I Siu
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Macao, China
| |
Collapse
|
29
|
Hinman SS, McKeating KS, Cheng Q. Surface Plasmon Resonance: Material and Interface Design for Universal Accessibility. Anal Chem 2018; 90:19-39. [PMID: 29053253 PMCID: PMC6041476 DOI: 10.1021/acs.analchem.7b04251] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Samuel S. Hinman
- Environmental Toxicology, University of California–Riverside, Riverside, California 92521, United States
| | - Kristy S. McKeating
- Department of Chemistry, University of California–Riverside, Riverside, California 92521, United States
| | - Quan Cheng
- Environmental Toxicology, University of California–Riverside, Riverside, California 92521, United States
- Department of Chemistry, University of California–Riverside, Riverside, California 92521, United States
| |
Collapse
|
30
|
Drescher DG, Selvakumar D, Drescher MJ. Analysis of Protein Interactions by Surface Plasmon Resonance. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 110:1-30. [PMID: 29412994 DOI: 10.1016/bs.apcsb.2017.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Surface plasmon resonance is an optical technique that is utilized for detecting molecular interactions, such as interactions that occur between proteins or other classes of molecules. Binding of a mobile molecule (analyte) to a molecule immobilized on a thin metal film (ligand) changes the refractive index of the film. The angle of extinction of light that is completely reflected after polarized light impinges upon the film, is altered and monitored as a change in detector position for a dip in reflected intensity (the surface plasmon resonance phenomenon). Because the method strictly detects mass, there is no need to label the interacting components, thus eliminating possible changes of their molecular properties. In this chapter, we review essential SPR methodology and present applications to basic science and human disease.
Collapse
Affiliation(s)
- Dennis G Drescher
- Wayne State University School of Medicine, Detroit, MI, United States.
| | | | - Marian J Drescher
- Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
31
|
Chen S, Feng Z, Wang Y, Ma S, Hu Z, Yang P, Chai Y, Xie X. Discovery of Novel Ligands for TNF-α and TNF Receptor-1 through Structure-Based Virtual Screening and Biological Assay. J Chem Inf Model 2017; 57:1101-1111. [PMID: 28422491 PMCID: PMC6732210 DOI: 10.1021/acs.jcim.6b00672] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Tumor necrosis factor α (TNF-α) is overexpressed in various diseases, and it has been a validated therapeutic target for autoimmune diseases. All therapeutics currently used to target TNF-α are biomacromolecules, and limited numbers of TNF-α chemical inhibitors have been reported, which makes the identification of small-molecule alternatives an urgent need. Recent studies have mainly focused on identifying small molecules that directly bind to TNF-α or TNF receptor-1 (TNFR1), inhibit the interaction between TNF-α and TNFR1, and/or regulate related signaling pathways. In this study, we combined in silico methods with biophysical and cell-based assays to identify novel antagonists that bind to TNF-α or TNFR1. Pharmacophore model filtering and molecular docking were applied to identify potential TNF-α antagonists. In regard to TNFR1, we constructed a three-dimensional model of the TNF-α-TNFR1 complex and carried out molecular dynamics simulations to sample the conformations. The residues in TNF-α that have been reported to play important roles in the TNF-α-TNFR1 complex were removed to form a pocket for further virtual screening of TNFR1-binding ligands. We obtained 20 virtual hits and tested them using surface plasmon resonance-based assays, which resulted in one ligand that binds to TNFR1 and four ligands with different scaffolds that bind to TNF-α. T1 and R1, the two most active compounds with Kd values of 11 and 16 μM for TNF-α and TNFR1, respectively, showed activities similar to those of known antagonists. Further cell-based assays also demonstrated that T1 and R1 have similar activities compared to the known TNF-α antagonist C87. Our work has not only produced several TNF-α and TNFR1 antagonists with novel scaffolds for further structural optimization but also showcases the power of our in silico methods for TNF-α- and TNFR1-based drug discovery.
Collapse
Affiliation(s)
- Si Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Yun Wang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Shifan Ma
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Ziheng Hu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Peng Yang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Xiangqun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|