1
|
Muhammad I, Rahman N, Gul-E-Nayab, Niaz S, Basharat Z, Rastrelli L, Jayanthi S, Efferth T, Khan H. Screening of potent phytochemical inhibitors against SARS-CoV-2 protease and its two Asian mutants. Comput Biol Med 2021; 133:104362. [PMID: 33894500 PMCID: PMC8051016 DOI: 10.1016/j.compbiomed.2021.104362] [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: 11/10/2020] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022]
Abstract
Background COVID-19, declared a pandemic in March 2020 by the World Health Organization is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). The virus has already killed more than 2.3 million people worldwide. Object The principal intent of this work was to investigate lead compounds by screening natural product library (NPASS) for possible treatment of COVID-19. Methods Pharmacophore features were used to screen a large database to get a small dataset for structure-based virtual screening of natural product compounds. In the structure-based screening, molecular docking was performed to find a potent inhibitor molecule against the main protease (Mpro) of SARS-CoV-2 (PDB ID: 6Y7M). The predicted lead compound was further subjected to Molecular Dynamics (MD) simulation to check the stability of the leads compound with the evolution of time. Results In pharmacophore-based virtual screening, 2,361 compounds were retained out of 30,927. In the structure-based screening, the lead compounds were filtered based on their docking scores. Among the 2,360 compounds, 12 lead compounds were selected based on their docking score. Kazinol T with NPASS ID: NPC474104 showed the highest docking score of −14.355 and passed criteria of Lipinski's drug-like parameters. Monitoring ADMET properties, Kazinol T showed its safety for consumption. Docking of Kazinol T with two Asian mutants (R60C and I152V) showed variations in binding and energy parameters. Normal mode analysis for ligand-bound and unbound form of protease along with its mutants, revealed displacement and correlation parameters for C-alpha atoms. MD simulation results showed that all ligand-protein complexes remained intact and stable in a dynamic environment with negative Gibbs free energy. Conclusions The natural product Kazinol T was a predicted lead compound against the main protease of SARS-CoV-2 and will be the possible treatment for COVID-19.
Collapse
Affiliation(s)
- Ijaz Muhammad
- Department of Zoology, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Noor Rahman
- Department of Biochemistry, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Gul-E-Nayab
- Department of Zoology, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Sadaf Niaz
- Department of Zoology, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Zarrin Basharat
- Jamil-ur-Rahman Center for Genome Research, PCMD, ICCBS, University of Karachi, Karachi, 75270, Pakistan
| | - Luca Rastrelli
- Dipartimento di Farmacia, University of Salerno, Via Giovanni Paolo II, 84084, Fisciano, SA, Italy
| | - Sivaraman Jayanthi
- Computational Drug Design Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
2
|
Santos RVC, de Sena WLB, Dos Santos FA, da Silva Filho AF, da Rocha Pitta MG, da Rocha Pitta MG, de Melo Rego MB, Pereira MC. Potential Therapeutic Agents Against Par-4 Target for Cancer Treatment: Where Are We Going? Curr Drug Targets 2020; 20:635-654. [PMID: 30474528 DOI: 10.2174/1389450120666181126122440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 02/06/2023]
Abstract
One of the greatest challenges of cancer therapeutics nowadays is to find selective targets successfully. Prostate apoptosis response-4 (Par-4) is a selective tumor suppressor protein with an interesting therapeutic potential due to its specificity on inducing apoptosis in cancer cells. Par-4 activity and levels can be downregulated in several tumors and cancer cell types, indicating poor prognosis and treatment resistance. Efforts to increase Par-4 expression levels have been studied, including its use as a therapeutic protein by transfection with adenoviral vectors or plasmids. However, gene therapy is very complex and still presents many hurdles to be overcome. We decided to review molecules and drugs with the capacity to upregulate Par-4 and, thereby, be an alternative to reach this druggable target. In addition, Par-4 localization and function are reviewed in some cancers, clarifying how it can be used as a therapeutic target.
Collapse
Affiliation(s)
- Renata Virgínia Cavalcanti Santos
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Wanessa Layssa Batista de Sena
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Flaviana Alves Dos Santos
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Antônio Felix da Silva Filho
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | | | - Maira Galdino da Rocha Pitta
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Moacyr Barreto de Melo Rego
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Michelly Cristiny Pereira
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
3
|
Fang JD, Yan XB, Lin WJ, Zhao YC, Liu XY. Diphenyl-Diselenide-Mediated Domino Claisen-Type Rearrangement/Cyclization of Propargylic Aryl Ethers: Synthesis of Naphthofuran-2-carboxaldehyde Derivatives. Org Lett 2019; 21:7635-7638. [PMID: 31507178 DOI: 10.1021/acs.orglett.9b02942] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A diphenyl-diselenide-mediated Claisen-type rearrangement/cyclization of propargylic aryl ethers under metal-free conditions is developed, affording various naphthofuran-2-carboxaldehydes in moderate to excellent yield. The broad substrate scope and excellent functional group compatibility suggest that it can be a straightforward and powerful method to access naphthofuran-2-carboxaldehydes in a highly regioselective manner. Moreover, this reaction can be scaled up to the gram scale.
Collapse
Affiliation(s)
- Jun-Dan Fang
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering , Lanzhou University , 222 South Tianshui Road , Lanzhou 730000 , China
| | - Xiao-Biao Yan
- School of Pharmacy , Anhui Medical University , 81 Meishan Road , Hefei 230032 , China
| | - Wu-Jie Lin
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering , Lanzhou University , 222 South Tianshui Road , Lanzhou 730000 , China
| | - Yi-Chuan Zhao
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering , Lanzhou University , 222 South Tianshui Road , Lanzhou 730000 , China
| | - Xue-Yuan Liu
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering , Lanzhou University , 222 South Tianshui Road , Lanzhou 730000 , China
| |
Collapse
|
4
|
Manousopoulou A, Saito S, Yamamoto Y, Al-Daghri NM, Ihara M, Carare RO, Garbis SD. Hemisphere Asymmetry of Response to Pharmacologic Treatment in an Alzheimer's Disease Mouse Model. J Alzheimers Dis 2016; 51:333-8. [PMID: 26836196 PMCID: PMC4927832 DOI: 10.3233/jad-151078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aim of this study was to examine hemisphere asymmetry of response to pharmacologic treatment in an Alzheimer's disease mouse model using cilostazol as a chemical stimulus. Eight-month-old mice were assigned to vehicle or cilostazol treatment for three months and hemispheres were analyzed using quantitative proteomics. Bioinformatics interpretation showed that following treatment, aggregation of blood platelets significantly decreased in the right hemisphere whereas neurodegeneration significantly decreased and synaptic transmission increased in the left hemisphere only. Our study provides novel evidence on cerebral laterality of pharmacologic activity, with important implications in deciphering regional pharmacodynamic effects of existing drugs thus uncovering novel hemisphere-specific therapeutic targets.
Collapse
Affiliation(s)
- Antigoni Manousopoulou
- Institute for Life Sciences, University of Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Satoshi Saito
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Japan
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Japan
| | - Nasser M Al-Daghri
- Biomarkers Research Program, Department of Biochemistry College of Science, King Saud University, KSA.,Prince Mutaib Chair for Biomarkers of Osteoporosis, Department of Biochemistry King Saud University, KSA
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Japan
| | - Roxana O Carare
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Spiros D Garbis
- Institute for Life Sciences, University of Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK.,Cancer Sciences, Faculty of Medicine, University of Southampton, UK
| |
Collapse
|
5
|
Yu S, Yan C, Yang X, He S, Liu J, Qin C, Huang C, Lu Y, Tian Z, Jia L. Pharmacoproteomic analysis reveals that metapristone (RU486 metabolite) intervenes E-cadherin and vimentin to realize cancer metastasis chemoprevention. Sci Rep 2016; 6:22388. [PMID: 26932781 PMCID: PMC4773818 DOI: 10.1038/srep22388] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/10/2016] [Indexed: 12/28/2022] Open
Abstract
Metapristone is the most predominant biological active metabolite of mifepristone, and being developed as a novel cancer metastasis chemopreventive agent by us. Despite its prominent metastasis chemopreventive effect, the underlying mechanism remains elusive. Our study, for the first time, demonstrated that metapristone had the ability to prevent breast cancer cells from migration, invasion, and interfere with their adhesion to endothelial cells. To explore the underlying mechanism of metapristone, we employed the iTRAQ technique to assess the effect of metapristone on MDA-MB-231 cells. In total, 5,145 proteins were identified, of which, 311 proteins showed significant differences in metapristone-treated cells compared to the control group (P-value < 0.05). Bioinformatic analysis showed many differentially expressed proteins (DEPs) functionally associated with post-translational modification, chaperones, translation, transcription, replication, signal transduction, etc. Importantly, many of the DEPs, such as E-cadherin, vimentin, TGF-β receptor I/II, smad2/3, β-catenin, caveolin, and dystroglycan were associated with TGF-β and Wnt signaling pathways, which were also linked to epithelial-to-mesenchymal transition (EMT) process. Further validation of the epithelial marker "E-caderin" and mesenchymal marker "vimetin" were carried out using immunoblot and immunofluorescence. These results have revealed a novel mechanism that metapristone-mediated metastasis chemoprevention is through intervening the EMT-related signaling pathways.
Collapse
Affiliation(s)
- Suhong Yu
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| | - Cuicui Yan
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| | - Xingtian Yang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| | - Sudang He
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| | - Jian Liu
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| | - Chongtao Qin
- School of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou Fujian, 350108, China
| | - Chuanzhong Huang
- Internal Oncology Laboratory, Fujian Provincial Key Laboratory of Translational Medicine Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian, 350002, China
| | - Yusheng Lu
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| | - Zhongping Tian
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou, 350002, China
| |
Collapse
|
6
|
Xu T, Zhang E, Wang D, Wang Y, Zou Y. Cu-Catalyzed Consecutive Hydroxylation and Aerobic Oxidative Cycloetherification under Microwave Conditions: Entry to 2-Arylbenzofuran-3-carboxylic Acids. J Org Chem 2015; 80:4313-24. [DOI: 10.1021/jo502802k] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Tianlong Xu
- Guangzhou
Institute of Chemistry, Chinese Academy of Science, Guangzhou 510650, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Ensheng Zhang
- Guangzhou
Institute of Chemistry, Chinese Academy of Science, Guangzhou 510650, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Dejian Wang
- Guangzhou
Institute of Chemistry, Chinese Academy of Science, Guangzhou 510650, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yan Wang
- New
Drug Research and Development Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yong Zou
- School
of Pharmaceutical Science, Sun Yat-sun University, Guangzhou 510006, China
| |
Collapse
|
7
|
Al-Daghri NM, Al-Attas OS, Johnston HE, Singhania A, Alokail MS, Alkharfy KM, Abd-Alrahman SH, Sabico SL, Roumeliotis TI, Manousopoulou-Garbis A, Townsend PA, Woelk CH, Chrousos GP, Garbis SD. Whole serum 3D LC-nESI-FTMS quantitative proteomics reveals sexual dimorphism in the milieu intérieur of overweight and obese adults. J Proteome Res 2014; 13:5094-105. [PMID: 25072778 DOI: 10.1021/pr5003406] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Linking gender-specific differences to the molecular etiology of obesity has been largely based on genomic and transcriptomic evidence lacking endophenotypic insight and is not applicable to the extracellular fluid compartments, or the milieu intérieur, of the human body. To address this need, this study profiled the whole serum proteomes of age-matched nondiabetic overweight and obese females (n = 28) and males (n = 31) using a multiplex design with pooled biological and technical replicates. To bypass basic limitations of immunodepletion-based strategies, subproteome enrichment by size-exclusion chromatography (SuPrE-SEC) followed by iTRAQ 2D-LC-nESI-FTMS analysis was used. The study resulted in the reproducible analysis of 2472 proteins (peptide FDR < 5%, q < 0.05). A total of 248 proteins exhibited significant modulation between men and women (p < 0.05) that mapped to pathways associated with β-estradiol, lipid and prostanoid metabolism, vitamin D function, immunity/inflammation, and the complement and coagulation cascades. This novel endophenotypic signature of gender-specific differences in whole serum confirmed and expanded the results of previous physiologic and pharmacologic studies exploring sexual dimorphism at the genomic and transcriptomic level in tissues and cells. Conclusively, the multifactorial and pleiotropic nature of human obesity exhibits sexual dimorphism in the circulating proteome of importance to clinical study design.
Collapse
Affiliation(s)
- Nasser M Al-Daghri
- Biomarkers Research Program, Biochemistry Department, College of Science, ‡Prince Mutaib Chair for Biomarkers of Osteoporosis, Biochemistry Department, College of Science, §Center of Excellence in Biotechnology Research, and #Department of Clinical Pharmacy, College of Pharmacy, King Saud University , Riyadh 12372, Kingdom of Saudi Arabia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Huang Q, Zhang J, Martin FL, Peng S, Tian M, Mu X, Shen H. Perfluorooctanoic acid induces apoptosis through the p53-dependent mitochondrial pathway in human hepatic cells: a proteomic study. Toxicol Lett 2013; 223:211-20. [PMID: 24035753 DOI: 10.1016/j.toxlet.2013.09.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 08/28/2013] [Accepted: 09/04/2013] [Indexed: 12/23/2022]
Abstract
Perfluorooctanoic acid (PFOA) is one of the most commonly used perfluorinated compounds, and exposure to it has been associated with a number of adverse health effects. However, the molecular mechanisms involved in PFOA toxicity are still not well characterized. In the present study, flow cytometry analysis revealed that PFOA induced oxidative stress, cell cycle arrest and apoptosis in human non-tumor hepatic cells (L-02). Furthermore, we investigated the alterations in protein profile within L-02 cells exposed to PFOA, aiming to explore the mechanisms underlying PFOA hepatotoxicity on the proteome level. Of the 28 proteins showing significant differential expression in response to PFOA, 24 were down-regulated and 4 were up-regulated. This proteomic study proposed that the inhibition of some proteins, including GRP78, HSP27, CTSD and hnRNPC may be involved in the activation of p53, which consequently triggered the apoptotic process in L-02 cells. Induction of apoptosis via the p53-dependent mitochondrial pathway is further suggested as one of the key toxicological events occurring in L-02 cells under PFOA stress. We hope these data will shed new light on the molecular mechanisms responsible for PFOA-mediated toxicity in human liver cells, and from such studies useful biomarkers indicative of PFOA exposure could be developed.
Collapse
Affiliation(s)
- Qingyu Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | | | | | | | | | | | | |
Collapse
|