1
|
Abera B, Abdissa N, Endale M, Melaku Y, Shenkute K, Ensermu U, Hunsen M, Rentsch D, Eswaramoorthy R. Evaluation of the Antibacterial and Antioxidant Properties of Chemical Constituents of the Roots of Woodfordia uniflora: An Integrated Approach of Experimental and Computational Study. Biochem Res Int 2024; 2024:1322756. [PMID: 39659700 PMCID: PMC11631344 DOI: 10.1155/bri/1322756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 09/13/2024] [Accepted: 10/04/2024] [Indexed: 12/12/2024] Open
Abstract
Woodfordia uniflora is a medicinal plant used for the treatment of malaria, toothache, and stomach problems. The root parts of the plant are also used for healing liver disorders. Silica gel chromatography separation of CH2Cl2/MeOH (1:1) and MeOH extracts of roots of W. uniflora result in the isolation of three compounds, namely, bergenin (1), β-sitosterol (2), and epigallocatechin 3-gallate (3), reported herein for the first time from the plant. The structure of the isolated compounds was elucidated using NMR (1D and 2D) techniques. Disk diffusion and DPPH assay were used to evaluate the antibacterial and antioxidant activities, respectively. Molecular docking was done by the AutoDock Vina 4.2 program. The pharmacokinetics and toxicity profile of compounds were predicted by Swiss ADME and Pro Tox II online servers. GC-MS analysis roots of W. uniflora result in the identification of five compounds, of which palmitic acid (34.9%) was the major constituent. The antibacterial activity result indicated that the oil extract had promising activity against P. aeruginosa, E. coli, S. pyogenes, and S. aureus with IZ of 14.3 ± 0.81, 15.0 ± 0.0, 15.6 ± 0.47, and 17.6 ± 0.47 mm, respectively, at 5 mg/mL, compared to ciprofloxacin (1Z 27-30.0 ± 0.0 mm) at 30 μg/mL. MeOH and CH2Cl2/MeOH (1:1) extract showed inhibition against E. coli (IZ of 13.6 ± 0.47 mm) and P. aeruginosa (IZ of 10.0 ± 0.0 mm), respectively, at 200 mg/mL. Bergenin (1) and β-sitosterol (2) also displayed maximum inhibition of E. coil (IZ of 11.6 ± 0.47) and S. aureus (11.0 ± 0.0 mm), respectively, at 5 mg/mL. The antioxidant activity results showed that CH2Cl2/MeOH (1:1) and MeOH extracts, bergenin (1), and compound 3 displayed potent scavenging DPPH radical with a percentage of inhibition of 76.8 ± 0.12, 77.8 ± 0.08, 71.4 ± 0.08, and 91.2 ± 0.16, respectively, compared to ascorbic acid (93.2% ± 0.04%) at 100 μg/mL. The molecular docking analysis showed that all compounds (1-3) exhibited minimum binding energy toward PDB ID: 1HD2 (-5.2 to -6.3 kcal/mol), compared to ascorbic acid (-5.6 kcal/mol), and toward PDB ID: 1DNU (-8.0 to -10.7 kcal/mol) receptors, compared to ascorbic acid (-5.7 kcal/mol). Toward the PDB ID: 4FM9 receptor, β-sitosterol (2) and compound 3 exhibited the best binding free energy of -9.1 and -9.8 kcal·mol, respectively, compared to vosaroxin (-7.8 kcal/mol). The drug-likeness analysis result indicated that bergenin (1) and β-sitosterol (2) obeyed four and five criteria of Lipinski's rule, respectively, and are more likely to be administered orally. The in silico toxicity analysis showed none of the compounds would be cytotoxic, mutagenic, or hepatotoxic. The in vitro antioxidant and antibacterial results supported by in silico analysis demonstrated that the roots of W. uniflora have the potential to be therapeutic agents for bacterial infections and free radical-inducing diseases.
Collapse
Affiliation(s)
- Bihon Abera
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Negera Abdissa
- Traditional and Modern Medicine Research and Development Directorate, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Milkyas Endale
- Traditional and Modern Medicine Research and Development Directorate, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Yadessa Melaku
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Kebede Shenkute
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Urgessa Ensermu
- Department of Applied Biology, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Mo Hunsen
- Department of Chemistry, Kenyon College, Gambier, Ohio 43022, USA
| | - Daniel Rentsch
- Laboratory for Functional Polymers, Empa, Swiss Federal Laboratories for Materials Science and Technology, Überlandstrasse 129, Dübendorf 8600, Switzerland
| | - Rajalakshmanan Eswaramoorthy
- Department of Biomaterials, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| |
Collapse
|
2
|
Matos IDA, Dallazen JL, Reis LR, Souza LF, Bevevino RC, de Moura RD, Ronsein GE, Hoch NC, da Costa Júnior NB, Costa SKP, Meotti FC. Targeting Myeloperoxidase Ameliorates Gouty Arthritis: A Virtual Screening Success Story. J Med Chem 2024; 67:12012-12032. [PMID: 38991154 DOI: 10.1021/acs.jmedchem.4c00721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
This study presents a new approach for identifying myeloperoxidase (MPO) inhibitors with strong in vivo efficacy. By combining inhibitor-like rules and structure-based virtual screening, the pipeline achieved a 70% success rate in discovering diverse, nanomolar-potency reversible inhibitors and hypochlorous acid (HOCl) scavengers. Mechanistic analysis identified RL6 as a genuine MPO inhibitor and RL7 as a potent HOCl scavenger. Both compounds effectively suppressed HOCl production in cells and neutrophils, with RL6 showing a superior inhibition of neutrophil extracellular trap release (NETosis). In a gout arthritis mouse model, intraperitoneal RL6 administration reduced edema, peroxidase activity, and IL-1β levels. RL6 also exhibited oral bioavailability, significantly reducing paw edema when administered orally. This study highlights the efficacy of integrating diverse screening methods to enhance virtual screening success, validating the anti-inflammatory potential of potent inhibitors, and advancing the MPO inhibitor research.
Collapse
Affiliation(s)
- Isaac de A Matos
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jorge L Dallazen
- Department of Pharmacology, Institute of Biological Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Lorenna R Reis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Luiz Felipe Souza
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Regina C Bevevino
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Rafael D de Moura
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Graziella E Ronsein
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Nicolas Carlos Hoch
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Soraia Kátia P Costa
- Department of Pharmacology, Institute of Biological Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Flavia C Meotti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
3
|
Nachimuthu L, Desikan R. Development of methyl 5-((cinnamoyloxy)methyl)picolinate, exploring its bio-target potential aiming at CVD mediated by multiple proteins through surface and physiochemical analysis. Sci Rep 2024; 14:13328. [PMID: 38858562 PMCID: PMC11164877 DOI: 10.1038/s41598-024-64165-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
The emphasis on sustainable sources of drug development seems imminent with phytochemicals emerging as promising candidates due to their minimal probability of adverse effects. This study focuses on utilizing simple cinnamic acid and nicotinic acid derivatives as starting materials, employing an efficient synthetic protocol to obtain methyl 5-((cinnamoyloxy)methyl)picolinate targeting CVD mediated by multiple enzymes such as MAPK, PCSK9, MPO, SIRT1 and TNF-α. Comprehensive characterization of synthesized molecule is achieved through 1H, 13C, FT-IR, and HRMS methods. Additionally, the crystal structure was established via SC-XRD. Comparative analysis with the DFT-optimized structure identifies key nucleophilic and electrophilic regions for determining interactions with bio-targets. Notably, Compound 5 adheres to all drug-likeness criteria, further validated through screening similar pharmacophoric drugs from databases. Targeting bio-relevant areas with a specific focus on CVD drug development. The molecular docking studies elucidate ligand-protein interactions for better binding connectivity. This investigation further underscores the importance of sustainable practices, simple chemical synthesis, and computational approaches, contributing to the pursuit of eco-friendly drug development with enhanced safety profiles (MTT assay).
Collapse
Affiliation(s)
- Lenin Nachimuthu
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, 632014, Tamilnadu, India
| | - Rajagopal Desikan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, 632014, Tamilnadu, India.
| |
Collapse
|
4
|
Lin W, Chen H, Chen X, Guo C. The Roles of Neutrophil-Derived Myeloperoxidase (MPO) in Diseases: The New Progress. Antioxidants (Basel) 2024; 13:132. [PMID: 38275657 PMCID: PMC10812636 DOI: 10.3390/antiox13010132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
Myeloperoxidase (MPO) is a heme-containing peroxidase, mainly expressed in neutrophils and, to a lesser extent, in monocytes. MPO is known to have a broad bactericidal ability via catalyzing the reaction of Cl- with H2O2 to produce a strong oxidant, hypochlorous acid (HOCl). However, the overproduction of MPO-derived oxidants has drawn attention to its detrimental role, especially in diseases characterized by acute or chronic inflammation. Broadly speaking, MPO and its derived oxidants are involved in the pathological processes of diseases mainly through the oxidation of biomolecules, which promotes inflammation and oxidative stress. Meanwhile, some researchers found that MPO deficiency or using MPO inhibitors could attenuate inflammation and tissue injuries. Taken together, MPO might be a promising target for both prognostic and therapeutic interventions. Therefore, understanding the role of MPO in the progress of various diseases is of great value. This review provides a comprehensive analysis of the diverse roles of MPO in the progression of several diseases, including cardiovascular diseases (CVDs), neurodegenerative diseases, cancers, renal diseases, and lung diseases (including COVID-19). This information serves as a valuable reference for subsequent mechanistic research and drug development.
Collapse
Affiliation(s)
- Wei Lin
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China;
| | - Huili Chen
- Center of System Pharmacology and Pharmacometrics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA;
| | - Xijing Chen
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China;
| | - Chaorui Guo
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China;
| |
Collapse
|
5
|
Paumann-Page M, Obinger C, Winterbourn CC, Furtmüller PG. Peroxidasin Inhibition by Phloroglucinol and Other Peroxidase Inhibitors. Antioxidants (Basel) 2023; 13:23. [PMID: 38275643 PMCID: PMC10812467 DOI: 10.3390/antiox13010023] [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: 11/14/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Human peroxidasin (PXDN) is a ubiquitous peroxidase enzyme expressed in most tissues in the body. PXDN represents an interesting therapeutic target for inhibition, as it plays a role in numerous pathologies, including cardiovascular disease, cancer and fibrosis. Like other peroxidases, PXDN generates hypohalous acids and free radical species, thereby facilitating oxidative modifications of numerous biomolecules. We have studied the inhibition of PXDN halogenation and peroxidase activity by phloroglucinol and 14 other peroxidase inhibitors. Although a number of compounds on their own potently inhibited PXDN halogenation activity, only five were effective in the presence of a peroxidase substrate with IC50 values in the low μM range. Using sequential stopped-flow spectrophotometry, we examined the mechanisms of inhibition for several compounds. Phloroglucinol was the most potent inhibitor with a nanomolar IC50 for purified PXDN and IC50 values of 0.95 μM and 1.6 μM for the inhibition of hypobromous acid (HOBr)-mediated collagen IV cross-linking in a decellularized extracellular matrix and a cell culture model. Other compounds were less effective in these models. Most interestingly, phloroglucinol was identified to irreversibly inhibit PXDN, either by mechanism-based inhibition or tight binding. Our work has highlighted phloroglucinol as a promising lead compound for the design of highly specific PXDN inhibitors and the assays used in this study provide a suitable approach for high-throughput screening of PXDN inhibitors.
Collapse
Affiliation(s)
- Martina Paumann-Page
- Mātai Hāora Centre for Redox Biology and Medicine, University of Otago Christchurch, Ōtautahi Christchurch 8011, New Zealand;
- Institute of Biochemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria;
| | - Christian Obinger
- Institute of Biochemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria;
| | - Christine C. Winterbourn
- Mātai Hāora Centre for Redox Biology and Medicine, University of Otago Christchurch, Ōtautahi Christchurch 8011, New Zealand;
| | - Paul G. Furtmüller
- Institute of Biochemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria;
| |
Collapse
|
6
|
Soliman AM, Ghorab WM, Lotfy DM, Karam HM, Ghorab MM, Ramadan LA. Novel iodoquinazolinones bearing sulfonamide moiety as potential antioxidants and neuroprotectors. Sci Rep 2023; 13:15546. [PMID: 37730974 PMCID: PMC10511408 DOI: 10.1038/s41598-023-42239-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
In a search for new antioxidants, a set of new iodoquinazolinone derivatives bearing benzenesulfonamide moiety and variable acetamide pharmacophores 5-17 were designed and synthesized. The structures of the synthesized compounds were confirmed based on spectral data. Compounds 5-17 were screened using in vitro assay for their antioxidant potential and acetylcholinesterase (AChE) inhibitory activity. The 2-(6-iodo-4-oxo-3-(4-sulfamoylphenyl)-3,4-dihydroquinazolin-2-ylthio)-N-(pyrazin-2-yl) acetamide 14 was the most active scaffold with potent AChE inhibitory activity. Compound 14 showed relative safety with a median lethal dose of 300 mg/kg (LD50 = 300 mg/kg), in an acute toxicity study. The possible antioxidant and neuroprotective activities of 14 were evaluated in irradiated mice. Compound 14 possessed in vivo AChE inhibitory activity and was able to modify the brain neurotransmitters. It was able to cause mitigation of gamma radiation-induced oxidative stress verified by the decline in Myeloperoxidase (MPO) and increase of glutathione (GSH) levels. Also, 14 restored the alterations in behavioral tests. Molecular docking of 14 was performed inside MPO and AChE active sites and showed the same binding interactions as that of the co-crystallized ligands considering the binding possibilities and energy scores. These findings would support that 14 could be considered a promising antioxidant with a neuromodulatory effect.
Collapse
Affiliation(s)
- Aiten M Soliman
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Walid M Ghorab
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Dina M Lotfy
- Pharmacology and Toxicology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Heba M Karam
- Pharmacology and Toxicology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Mostafa M Ghorab
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt.
| | - Laila A Ramadan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| |
Collapse
|
7
|
Teo A, Chan LLY, Cheung C, Chia PY, Ong SWX, Fong SW, Ng LFP, Renia L, Lye DC, Young BE, Yeo TW. Myeloperoxidase inhibition may protect against endothelial glycocalyx shedding induced by COVID-19 plasma. COMMUNICATIONS MEDICINE 2023; 3:62. [PMID: 37147421 PMCID: PMC10160718 DOI: 10.1038/s43856-023-00293-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/27/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND SARS-CoV-2, the causative agent of COVID-19, is a threat to public health. Evidence suggests increased neutrophil activation and endothelial glycocalyx (EG) damage are independently associated with severe COVID-19. Here, we hypothesised that an increased level of blood neutrophil myeloperoxidase (MPO) is associated with soluble EG breakdown, and inhibiting MPO activity may reduce EG damage. METHODS Analysing a subset of acute and convalescent COVID-19 plasma, 10 from severe and 15 from non-severe COVID-19 cases, and 9 from pre-COVID-19 controls, we determined MPO levels, MPO activity and soluble EG proteins (syndecan-1 and glypican-1) levels by enzyme-linked immunosorbent assay. In vitro primary human aortic endothelial cells were cultured with plasma untreated or treated with specific MPO inhibitors (MPO-IN-28, AZD5904) to determine EG shedding. We then investigated whether inhibiting MPO activity decreased EG degradation. RESULTS In COVID-19 plasma, MPO levels, MPO activity and levels of soluble EG proteins are significantly raised compared to controls, and concentrations increase in proportion to disease severity. Despite clinical recovery, protein concentrations remain significantly elevated. Interestingly, there is a trend of increasing MPO activity in convalescent plasma in both severe and non-severe groups. MPO levels and MPO activity correlate significantly with soluble EG levels and inhibiting MPO activity leads to reduced syndecan-1 shedding, in vitro. CONCLUSIONS Neutrophil MPO may increase EG shedding in COVID-19, and inhibiting MPO activity may protect against EG degradation. Further research is needed to evaluate the utility of MPO inhibitors as potential therapeutics against severe COVID-19.
Collapse
Affiliation(s)
- Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, VIC, Australia.
- National Centre for Infectious Diseases, Singapore, Singapore.
| | - Louisa L Y Chan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science and Technology and Research (A*STAR), Singapore, Singapore
| | - Po Ying Chia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Sean Wei Xiang Ong
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Siew Wai Fong
- A*STAR Infectious Diseases Lab (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Lab (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Singapore
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK
| | - Laurent Renia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- A*STAR Infectious Diseases Lab (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - David Chien Lye
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore, Singapore
| | - Barnaby Edward Young
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tsin Wen Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
8
|
Christie M, Friesen Westley J, Suresh B, Baiazitov Ramil Y, Wu D, Karloff Diane B, Chang-Sun L, Young-Choon M, Hongyu R, Jairo S, Yuki T, Priya V, Welch Ellen M, Xiaojiao X, Jin Z. Guanidino Quinazolines and Pyrimidines Promote Readthrough of Premature Termination Codons in Cells with Native Nonsense Mutations. Bioorg Med Chem Lett 2022; 76:128989. [PMID: 36150638 DOI: 10.1016/j.bmcl.2022.128989] [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: 08/02/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022]
Abstract
Using small molecules to induce readthrough of premature termination codons is a promising therapeutic approach to treating genetic diseases and cancers caused by nonsense mutations, as evidenced by the widespread use of ataluren to treat nonsense mutation Duchene muscular dystrophy. Herein we describe a series of novel guanidino quinazoline and pyrimidine scaffolds that induce readthrough in both HDQ-P1 mammary carcinoma cells and mdx myotubes. Linkage of basic, tertiary amines with aliphatic, hydrophobic substituents to the terminal guanidine nitrogen of these scaffolds led to significant potency increases. Further potency gains were achieved by flanking the pyrimidine ring with hydrophobic substituents, inducing readthrough at concentrations as low as 120 nM and demonstrating the potential of these compounds to be used either in combination with ataluren or as stand-alone therapeutics.
Collapse
Affiliation(s)
- Morrill Christie
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - J Friesen Westley
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Babu Suresh
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Y Baiazitov Ramil
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Du Wu
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - B Karloff Diane
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Lee Chang-Sun
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Moon Young-Choon
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Ren Hongyu
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Sierra Jairo
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Tomizawa Yuki
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Vazirani Priya
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - M Welch Ellen
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Xue Xiaojiao
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| | - Zhuo Jin
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ, 07080, USA
| |
Collapse
|
9
|
Krawczyk L, Semwal S, Soubhye J, Lemri Ouadriri S, Prévost M, Van Antwerpen P, Roos G, Bouckaert J. Native glycosylation and binding of the antidepressant paroxetine in a low-resolution crystal structure of human myeloperoxidase. Acta Crystallogr D Struct Biol 2022; 78:1099-1109. [PMID: 36048150 PMCID: PMC9435594 DOI: 10.1107/s2059798322007082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/10/2022] [Indexed: 11/10/2022] Open
Abstract
Human myeloperoxidase (MPO) utilizes hydrogen peroxide to oxidize organic compounds and as such plays an essential role in cell-component synthesis, in metabolic and elimination pathways, and in the front-line defence against pathogens. Moreover, MPO is increasingly being reported to play a role in inflammation. The enzymatic activity of MPO has also been shown to depend on its glycosylation. Mammalian MPO crystal structures deposited in the Protein Data Bank (PDB) present only a partial identification of their glycosylation. Here, a newly obtained crystal structure of MPO containing four disulfide-linked dimers and showing an elaborate collection of glycans is reported. These are compared with the glycans identified in proteomics studies and from 18 human MPO structures available in the PDB. The crystal structure also contains bound paroxetine, a blocker of serotonin reuptake that has previously been identified as an irreversible inhibitor of MPO, in the presence of thiocyanate, a physiological substrate of MPO.
Collapse
Affiliation(s)
- Lucas Krawczyk
- UGSF, CNRS, 50 Avenue de Halley, 59658 Villeneuve d’Ascq, France
| | - Shubham Semwal
- UGSF, CNRS, 50 Avenue de Halley, 59658 Villeneuve d’Ascq, France
| | - Jalal Soubhye
- Department of Pharmacognosy, Bioanalysis and Drug Discovery, Faculty of Pharmacy, Université Libre De Bruxelles, Brussels, Belgium
| | | | - Martin Prévost
- Structure et Fonction des Membranes Biologiques, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Goedele Roos
- UGSF, CNRS, 50 Avenue de Halley, 59658 Villeneuve d’Ascq, France
| | - Julie Bouckaert
- UGSF, CNRS, 50 Avenue de Halley, 59658 Villeneuve d’Ascq, France
| |
Collapse
|
10
|
Nyssen P, Maho A, Malempre R, Matagne A, Mouithys-Mickalad A, Hoebeke M. Propofol inhibits the myeloperoxidase activity by acting as substrate through a redox process. Biochim Biophys Acta Gen Subj 2022; 1866:130100. [PMID: 35150774 DOI: 10.1016/j.bbagen.2022.130100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Propofol (2,6-diisopropylphenol) is frequently used as intravenous anesthetic agent, especially in its injectable form (Diprivan), to initiate and maintain sedative state during surgery or in intensive care units. Numerous studies have reported the antioxidant and anti-inflammatory effect of propofol. The oxidant enzyme myeloperoxidase (MPO), released from activated neutrophils, plays a key role in host defense. An increase of the circulating MPO concentration has been observed in patients admitted in intensive care unit and presenting a systemic inflammatory response related to septic shock or trauma. METHODS This study investigates the immunomodulatory action of propofol and Diprivan as inhibitor of the oxidant activity of MPO. The understanding of the redox action mechanism of propofol and Diprivan on the myeloperoxidase chlorination and peroxidase activities has been refined using the combination of fluorescence and absorption spectroscopies with docking and cyclic voltammetry. RESULTS Propofol acts as a reversible MPO inhibitor. The molecule interacts as a reducing substrate in the peroxidase cycle and promotes the accumulation of compound II. At acidic pH (5.5), propofol and Diprivan do not inhibit the chlorination activity, but their action increases at physiological pH (7.4). The main inhibitory action of Diprivan could be attributed to its HOCl scavenging property. GENERAL SIGNIFICANCE Propofol can act as a reversible MPO inhibitor at clinical concentrations. This property could, in addition to other previously proven anti-inflammatory actions, induce an immunomodulatory action, beneficial during clinical use, particularly in the treatment of systemic inflammation response syndrome.
Collapse
Affiliation(s)
- P Nyssen
- Biomedical Spectroscopy Laboratory, Department of Physics, CESAM, University of Liège, Building B5a, Quartier Agora, Allée du 6 Août, 19, Sart-Tilman, 4000 Liège, Belgium.
| | - A Maho
- Greenmat, Department of Chemistry, CESAM, University of Liège, Building B6c, Quartier Agora, Allée du 6 Août, 19, Sart-Tilman, 4000 Liège, Belgium
| | - R Malempre
- Laboratory of Enzymology and Protein folding, Centre for Protein Engineering, InBioS, University of Liège, Building B6a, Quartier Agora, Allée du 6 Août, 19, Sart-Tilman, 4000 Liège, Belgium
| | - A Matagne
- Laboratory of Enzymology and Protein folding, Centre for Protein Engineering, InBioS, University of Liège, Building B6a, Quartier Agora, Allée du 6 Août, 19, Sart-Tilman, 4000 Liège, Belgium
| | - A Mouithys-Mickalad
- CORD, Department of Chemistry, CIRM, University of Liège, Building B6a, Quartier Agora, Allée du 6 Août, 13, Sart-Tilman, 4000 Liège, Belgium
| | - M Hoebeke
- Biomedical Spectroscopy Laboratory, Department of Physics, CESAM, University of Liège, Building B5a, Quartier Agora, Allée du 6 Août, 19, Sart-Tilman, 4000 Liège, Belgium
| |
Collapse
|
11
|
Design, synthesis and biochemical evaluation of novel 2-amino-3-(7-methoxybenzo[d][1,3]dioxol-5-yl)propanoic acid using Horseradish peroxidase (HRP) activity, cellular ROS inhibition and molecular docking study. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
12
|
Valadez-Cosmes P, Raftopoulou S, Mihalic ZN, Marsche G, Kargl J. Myeloperoxidase: Growing importance in cancer pathogenesis and potential drug target. Pharmacol Ther 2021; 236:108052. [PMID: 34890688 DOI: 10.1016/j.pharmthera.2021.108052] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
Myeloperoxidase is a heme-peroxidase which makes up approximately 5% of the total dry cell weight of neutrophils where it is predominantly found in the primary (azurophilic) granules. Other cell types, such as monocytes and certain macrophage subpopulations also contain myeloperoxidase, but to a much lesser extent. Initially, the function of myeloperoxidase had been mainly associated with its ability as a catalyzer of reactive oxidants that help to clear pathogens. However, over the past years non-canonical functions of myeloperoxidase have been described both in health and disease. Attention has been specially focused on inflammatory diseases, in which an exacerbate infiltration of leukocytes can favor a poorly-controlled production and release of myeloperoxidase and its oxidants. There is compelling evidence that myeloperoxidase derived oxidants contribute to tissue damage and the development and propagation of acute and chronic vascular inflammation. Recently, neutrophils have attracted much attention within the large diversity of innate immune cells that are part of the tumor microenvironment. In particular, neutrophil-derived myeloperoxidase may play an important role in cancer development and progression. This review article aims to provide a comprehensive overview of the roles of myeloperoxidase in the development and progression of cancer. We propose future research approaches and explore prospects of inhibiting myeloperoxidase as a strategy to fight against cancer.
Collapse
Affiliation(s)
- Paulina Valadez-Cosmes
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Sofia Raftopoulou
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Zala Nikita Mihalic
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
13
|
Siraki AG. The many roles of myeloperoxidase: From inflammation and immunity to biomarkers, drug metabolism and drug discovery. Redox Biol 2021; 46:102109. [PMID: 34455146 PMCID: PMC8403760 DOI: 10.1016/j.redox.2021.102109] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022] Open
Abstract
This review provides a practical guide to myeloperoxidase (MPO) and presents to the reader the diversity of its presence in biology. The review provides a historical background, from peroxidase activity to the discovery of MPO, to its role in disease and drug development. MPO is discussed in terms of its necessity, as specific individuals lack MPO expression. An underlying theme presented throughout brings up the question of the benefit and burden of MPO activity. Enzyme structure is discussed, including accurate masses and glycosylation sites. The catalytic cycle of MPO and its corresponding pathways are presented, with a discussion of the importance of the redox couples of the different states of MPO. Cell lines expressing MPO are discussed and practically summarized for the reader, and locations of MPO (primary and secondary) are provided. Useful methods of MPO detection are discussed, and how these can be used for studying disease processes are implied through the presentation of MPO as a biomarker. The presence of MPO in neutrophil extracellular traps is presented, and the activators of the former are provided. Lastly, the transition from drug metabolism to a target for drug development is where the review concludes.
Collapse
Affiliation(s)
- Arno G Siraki
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
14
|
Etsè KS, Etsè KD, Nyssen P, Mouithys-Mickalad A. Assessment of anti-inflammatory-like, antioxidant activities and molecular docking of three alkynyl-substituted 3-ylidene-dihydrobenzo[d]isothiazole 1,1-dioxide derivatives. Chem Biol Interact 2021; 344:109513. [PMID: 33974901 DOI: 10.1016/j.cbi.2021.109513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/12/2021] [Accepted: 05/05/2021] [Indexed: 11/19/2022]
Abstract
The presence of enyne and benzoisothiazole functions in the molecular architecture of compounds 1, 2 and 3 were expected to provide biochemical activities. In the present work, we first examined the molecular surface contact of three alkynyl-substituted 3-ylidenedihydrobenzo[d] isothiazole 1,1-dioxides. The analysis of the Hirshfeld surfaces reveals that only compound 3 exhibited a well-defined red spots, indicating intermolecular interactions identified as S-O⋯H, C-H⋯O and C-O⋯H contacts. Comparative fingerprint histograms of the three compounds show that close pair interactions are dominated by C-H⋯H-C contact. By UV-visible analysis, compound 1 showed the most intense absorbances at 407 and 441 nm, respectively. The radical scavenging activity explored in the DPPH test, shows that only 1 exhibited low anti-radical activity. Furthermore, cellular antioxidant capacity of benzoisothiazoles 1-3 was investigated with PMA-activated HL-60 cells using chemiluminescence and fluorescence techniques in the presence of L-012 and Amplex Red probe, respectively. Results highlight that compound 1 exhibited moderate anti-ROS capacity while compounds 2 and 3 enhanced ROS production. The cytotoxicity test performed on HL-60 cells, using the MTS assay, confirmed the lack of toxicity of the tested benzoisothiazole 1 compared to 2 and 3 which show low cytotoxicity (≤30%). Anti-catalytic activity was evaluated by following the inhibitory potential of the benzoisothiazoles on MPO activity and depicted benzoisothiazoles-MPO interactions by docking. Both SIEFED and docking studies demonstrated an anti-catalytic activity of the tested benzoisothiazoles towards MPO with the best activity for compound 2.
Collapse
Affiliation(s)
- Koffi Sénam Etsè
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Quartier Ho^pital B36 Av. Hippocrate 15 B-4000 Liège, Belgium
| | - Kodjo Djidjolé Etsè
- Laboratoire de Physiologie et Biotechnologie Végétales (LPBV), Faculté des Sciences (FDS), Université de Lomé (UL), Lomé, Togo
| | - Pauline Nyssen
- Biomedical Spectroscopy Laboratory, Department of Physics, CESAM, ULiège, Sart-Tilman, B-4000 Liège, Belgium
| | - Ange Mouithys-Mickalad
- Center for Oxygen, Research and Development (CORD) and Center for Interdisciplinary Research on Medicine (CIRM) Institute of Chemistry University of Liège, Sart-Tilman (B.6a), 4000 Liège, Belgium.
| |
Collapse
|
15
|
Wang C, Fan X, Chen F, Qian PC, Cheng J. Vinylene carbonate: beyond the ethyne surrogate in rhodium-catalyzed annulation with amidines toward 4-methylquinazolines. Chem Commun (Camb) 2021; 57:3929-3932. [DOI: 10.1039/d1cc00882j] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Vinylene carbonate: acetylation reagent rather than ethyne surrogate in rhodium-catalyzed annulation with amidines toward 4-methylquinazolines.
Collapse
Affiliation(s)
- Chang Wang
- Institute of New Materials & Industry Technology, College of Chemistry & Materials Engineering, Wenzhou University
- Wenzhou
- P. R. China
| | - Xiaodong Fan
- Institute of New Materials & Industry Technology, College of Chemistry & Materials Engineering, Wenzhou University
- Wenzhou
- P. R. China
| | - Fan Chen
- Institute of New Materials & Industry Technology, College of Chemistry & Materials Engineering, Wenzhou University
- Wenzhou
- P. R. China
| | - Peng-Cheng Qian
- Institute of New Materials & Industry Technology, College of Chemistry & Materials Engineering, Wenzhou University
- Wenzhou
- P. R. China
| | - Jiang Cheng
- Institute of New Materials & Industry Technology, College of Chemistry & Materials Engineering, Wenzhou University
- Wenzhou
- P. R. China
| |
Collapse
|
16
|
Abstract
Myeloperoxidase participates in innate immune defense mechanism through formation of microbicidal reactive oxidants and diffusible radical species. A unique activity is its ability to use chloride as a cosubstrate with hydrogen peroxide to generate chlorinating oxidants such as hypochlorous acid, a potent antimicrobial agent. However, chronic MPO activation can lead to indiscriminate protein modification causing tissue damage, and has been associated with chronic inflammatory diseases, atherosclerosis, and acute cardiovascular events. This has attracted considerable interest in the development of therapeutically useful MPO inhibitors. Today, based on the profound knowledge of structure and function of MPO and its biochemical and biophysical differences with the other homologous human peroxidases, various rational and high-throughput screening attempts were performed in developing specific irreversible and reversible inhibitors. The most prominent candidates as well as MPO inhibitors already studied in clinical trials are introduced and discussed.
Collapse
|
17
|
Matos IDA, da Costa Júnior NB, Meotti FC. Integration of an Inhibitor-like Rule and Structure-based Virtual Screening for the Discovery of Novel Myeloperoxidase Inhibitors. J Chem Inf Model 2020; 60:6408-6418. [PMID: 33270445 DOI: 10.1021/acs.jcim.0c00813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Myeloperoxidase (MPO) is an attractive therapeutic target against inflammation. Herein, we developed an inhibitor-like rule, based on known MPO inhibitors, and generated a target database containing 6546 molecules with privileged inhibitory properties. Using a structure-based approach validated by decoys, robust statistical metrics, redocking, and cross-docking, we selected 10 putative MPO inhibitors with high chemical diversity. At 20 μM, six of these 10 compounds (i.e., 60% success rate) inhibited more than 20% of the chlorinating activity of the enzyme. Additionally, we found that compound ZINC9089086 forms hydrogen bonds with Arg233 and with the hemic carboxylate. It makes a π-stacking interaction with the heme group and displays a high affinity for the enzyme active site. When incubated with purified MPO, ZINC9089086 inhibited the chlorinating activity of the enzyme with an IC50 of 2.2 ± 0.1 μM in a reversible manner. Subsequent experiments revealed that ZINC9089086 inhibited hypochlorous acid production in dHL-60 cells and human neutrophils. Furthermore, the theoretical ADME/Tox profile indicated that this compound exhibits low toxicity risks and adequate pharmacokinetic parameters, thus making ZINC9089086 a very promising candidate for preclinical anti-inflammatory studies. Overall, our study shows that integrating an inhibitor-like rule with a validated structure-based methodology is an excellent approach for improving the success rate and molecular diversity of novel MPO inhibitors with good pharmacokinetics and toxicological profiles. By combining these tools, it was possible to increase the assurance rate, which ultimately diminishes the costs and time needed for the acquisition, synthesis, and evaluation of new compounds.
Collapse
Affiliation(s)
- Isaac de Araújo Matos
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil
| | | | - Flavia Carla Meotti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil
| |
Collapse
|
18
|
Myeloperoxidase: A versatile mediator of endothelial dysfunction and therapeutic target during cardiovascular disease. Pharmacol Ther 2020; 221:107711. [PMID: 33137376 DOI: 10.1016/j.pharmthera.2020.107711] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Myeloperoxidase (MPO) is a prominent mammalian heme peroxidase and a fundamental component of the innate immune response against microbial pathogens. In recent times, MPO has received considerable attention as a key oxidative enzyme capable of impairing the bioactivity of nitric oxide (NO) and promoting endothelial dysfunction; a clinically relevant event that manifests throughout the development of inflammatory cardiovascular disease. Increasing evidence indicates that during cardiovascular disease, MPO is released intravascularly by activated leukocytes resulting in its transport and sequestration within the vascular endothelium. At this site, MPO catalyzes various oxidative reactions that are capable of promoting vascular inflammation and impairing NO bioactivity and endothelial function. In particular, MPO catalyzes the production of the potent oxidant hypochlorous acid (HOCl) and the catalytic consumption of NO via the enzyme's NO oxidase activity. An emerging paradigm is the ability of MPO to also influence endothelial function via non-catalytic, cytokine-like activities. In this review article we discuss the implications of our increasing knowledge of the versatility of MPO's actions as a mediator of cardiovascular disease and endothelial dysfunction for the development of new pharmacological agents capable of effectively combating MPO's pathogenic activities. More specifically, we will (i) discuss the various transport mechanisms by which MPO accumulates into the endothelium of inflamed or diseased arteries, (ii) detail the clinical and basic scientific evidence identifying MPO as a significant cause of endothelial dysfunction and cardiovascular disease, (iii) provide an up-to-date coverage on the different oxidative mechanisms by which MPO can impair endothelial function during cardiovascular disease including an evaluation of the contributions of MPO-catalyzed HOCl production and NO oxidation, and (iv) outline the novel non-enzymatic mechanisms of MPO and their potential contribution to endothelial dysfunction. Finally, we deliver a detailed appraisal of the different pharmacological strategies available for targeting the catalytic and non-catalytic modes-of-action of MPO in order to protect against endothelial dysfunction in cardiovascular disease.
Collapse
|
19
|
Casas AI, Nogales C, Mucke HAM, Petraina A, Cuadrado A, Rojo AI, Ghezzi P, Jaquet V, Augsburger F, Dufrasne F, Soubhye J, Deshwal S, Di Sante M, Kaludercic N, Di Lisa F, Schmidt HHHW. On the Clinical Pharmacology of Reactive Oxygen Species. Pharmacol Rev 2020; 72:801-828. [DOI: 10.1124/pr.120.019422] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
20
|
Soubhye J, Van Antwerpen P, Dufrasne F. A patent review of myeloperoxidase inhibitors for treating chronic inflammatory syndromes (focus on cardiovascular diseases, 2013-2019). Expert Opin Ther Pat 2020; 30:595-608. [DOI: 10.1080/13543776.2020.1780210] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Jalal Soubhye
- Department of Pharmacognosy, Bioanalysis and Drug Discovery, Faculty of Pharmacy, Universite Libre De Bruxelles (ULB), Bruxelles, Belgium
| | - Pierre Van Antwerpen
- Department of Pharmacognosy, Bioanalysis and Drug Discovery, Faculty of Pharmacy, Universite Libre De Bruxelles (ULB), Bruxelles, Belgium
| | - François Dufrasne
- Microbiology, Bioorganic and Macromolecular Chemistry, Faculty of Pharmacy, Universite Libre De Bruxelles, Bruxelles, Belgium
| |
Collapse
|
21
|
Patnaik A, Axford L, Deng L, Cohick E, Ren X, Loi S, Kecman S, Hollis-Symynkywicz M, Harrison TJ, Papillon JPN, Dales N, Hamann LG, Lee L, Regard JB, Marcinkeviciene J, Marro ML, Patterson AW. Discovery of a novel indole pharmacophore for the irreversible inhibition of myeloperoxidase (MPO). Bioorg Med Chem 2020; 28:115548. [PMID: 32503688 DOI: 10.1016/j.bmc.2020.115548] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/23/2022]
Abstract
Myeloperoxidase (MPO) activity and subsequent generation of hypochlorous acid has been associated with the killing of host-invading microorganisms (e.g. bacteria, viruses, and fungi). However, during oxidative stress, high MPO activity can damage host tissue and is linked to several chronic inflammatory conditions. Herein, we describe the development of a novel biaryl, indole-pyrazole series of irreversible mechanism-based inhibitors of MPO. Derived from an indole-containing high-throughput screen hit, optimization efforts resulted in potent and selective 6-substituted indoles with good oral bioavailability and in vivo activity.
Collapse
Affiliation(s)
- Anup Patnaik
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States.
| | - Laura Axford
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Lin Deng
- PK Sciences, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Evan Cohick
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Xianglin Ren
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Sally Loi
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Sam Kecman
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Micah Hollis-Symynkywicz
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Tyler J Harrison
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Julien P N Papillon
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Natalie Dales
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Lawrence G Hamann
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Lac Lee
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Jean B Regard
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Jovita Marcinkeviciene
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Martin L Marro
- Cardiovascular and Metabolic Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States
| | - Andrew W Patterson
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, United States.
| |
Collapse
|
22
|
Slater O, Kontoyianni M. The compromise of virtual screening and its impact on drug discovery. Expert Opin Drug Discov 2019; 14:619-637. [PMID: 31025886 DOI: 10.1080/17460441.2019.1604677] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Docking and structure-based virtual screening (VS) have been standard approaches in structure-based design for over two decades. However, our understanding of the limitations, potential, and strength of these techniques has enhanced, raising expectations. Areas covered: Based on a survey of reports in the past five years, we assess whether VS: (1) predicts binding poses in agreement with crystallographic data (when available); (2) is a superior screening tool, as often claimed; (3) is successful in identifying chemical scaffolds that can be starting points for subsequent lead optimization cycles. Data shows that knowledge of the target and its chemotypes in postprocessing lead to viable hits in early drug discovery endeavors. Expert opinion: VS is capable of accurate placements in the pocket for the most part, but does not consistently score screening collections accurately. What matters is capitalization on available resources to get closer to a viable lead or optimizable series. Integration of approaches, subjective hit selection guided by knowledge of the receptor or endogenous ligand, libraries driven by experimental guides, validation studies to identify the best docking/scoring that reproduces experimental findings, constraints regarding receptor-ligand interactions, thoroughly designed methodologies, and predefined cutoff scoring criteria strengthen VS's position in pharmaceutical research.
Collapse
Affiliation(s)
- Olivia Slater
- a Department of Pharmaceutical Sciences , Southern Illinois University Edwardsville , Edwardsville , IL , USA
| | - Maria Kontoyianni
- a Department of Pharmaceutical Sciences , Southern Illinois University Edwardsville , Edwardsville , IL , USA
| |
Collapse
|
23
|
Ti H, Zhou Y, Liang X, Li R, Ding K, Zhao X. Targeted Treatments for Chronic Obstructive Pulmonary Disease (COPD) Using Low-Molecular-Weight Drugs (LMWDs). J Med Chem 2019; 62:5944-5978. [PMID: 30682248 DOI: 10.1021/acs.jmedchem.8b01520] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a very common and frequently fatal airway disease. Current therapies for COPD depend mainly on long-acting bronchodilators, which cannot target the pathogenic mechanisms of chronic inflammation in COPD. New pharmaceutical therapies for the inflammatory processes of COPD are urgently needed. Several anti-inflammatory targets have been identified based on increased understanding of the pathogenesis of COPD, which raises new hopes for targeted treatment of this fatal respiratory disease. In this review, we discuss the recent advances in bioactive low-molecular-weight drugs (LMWDs) for the treatment of COPD and, in addition to the first-line drug bronchodilators, focus particularly on low-molecular-weight anti-inflammatory agents, including modulators of inflammatory mediators, inflammasome inhibitors, protease inhibitors, antioxidants, PDE4 inhibitors, kinase inhibitors, and other agents. We also provide new insights into targeted COPD treatments using LMWDs, particularly small-molecule agents.
Collapse
Affiliation(s)
- Huihui Ti
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China
| | - Yang Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China.,Division of Theoretical Chemistry and Biology, School of Biotechnology , Royal Institute of Technology (KTH) , AlbaNova University Center , Stockholm SE-100 44 , Sweden
| | - Xue Liang
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy , Jinan University , Guangzhou 510632 , P. R. China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China
| | - Xin Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China.,School of Life Sciences , The Chinese University of Hong Kong , Shatin, N.T. , Hong Kong SAR 999077 , P. R. China
| |
Collapse
|
24
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
25
|
Galijasevic S. The development of myeloperoxidase inhibitors. Bioorg Med Chem Lett 2018; 29:1-7. [PMID: 30466896 DOI: 10.1016/j.bmcl.2018.11.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Abstract
Myeloperoxidase (MPO), an abundant hemoprotein present in neutrophils and monocytes, plays a significant role in immune surveillance and host defense mechanisms. However, increased MPO activity has been linked to a number of pathologies with compelling evidence in initiation and progression of inflammatory events. As a result, search for active compounds that can efficiently inhibit MPO activity and subsequently decrease inflammatory events has been focus of the current research. This perspective provides an overview of the development of MPO inhibitors, their mechanism of action and the review of molecules that were in clinical trials as promising MPO inhibitors.
Collapse
Affiliation(s)
- Semira Galijasevic
- University Sarajevo School of Science and Technology, Sarajevo Medical School, Bosnia and Herzegovina.
| |
Collapse
|
26
|
Albrett AM, Ashby LV, Dickerhof N, Kettle AJ, Winterbourn CC. Heterogeneity of hypochlorous acid production in individual neutrophil phagosomes revealed by a rhodamine-based probe. J Biol Chem 2018; 293:15715-15724. [PMID: 30135208 DOI: 10.1074/jbc.ra118.004789] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/12/2018] [Indexed: 01/21/2023] Open
Abstract
The rhodamine-based probe R19-S has been shown to react with hypochlorous acid (HOCl) to yield fluorescent R19, but not with some other oxidants including hydrogen peroxide. Here, we further examined the specificity of R19-S and used it for real-time monitoring of HOCl production in neutrophil phagosomes. We show that it also reacts rapidly with hypobromous acid, bromamines, and hypoiodous acid, indicating that R19-S responds to these reactive halogen species as well as HOCl. Hypothiocyanous acid and taurine chloramine were unreactive, however, and ammonia chloramine and dichloramine reacted only very slowly. MS analyses revealed additional products from the reaction of HOCl with R19-S, including a chlorinated species as a minor product. Of note, phagocytosis of opsonized zymosan or Staphylococcus aureus by neutrophils was accompanied by an increase in R19 fluorescence. This increase depended on NADPH oxidase and myeloperoxidase activities, and detection of chlorinated R19-S confirmed its specificity for HOCl. Using live-cell imaging to track individual phagosomes in single neutrophils, we observed considerable heterogeneity among the phagosomes in the time from ingestion of a zymosan particle to when fluorescence was first detected, ranging from 1 to >30 min. However, once initiated, the subsequent fluorescence increase was uniform, reaching a similar maximum in ∼10 min. Our results confirm the utility of R19-S for detecting HOCl in real-time and provide definitive evidence that isolated neutrophils produce HOCl in phagosomes. The intriguing variability in the onset of HOCl production among phagosomes identified here could influence the way they kill ingested bacteria.
Collapse
Affiliation(s)
- Amelia M Albrett
- From the Department of Pathology and Biomedical Science, Centre for Free Radical Research, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - Louisa V Ashby
- From the Department of Pathology and Biomedical Science, Centre for Free Radical Research, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - Nina Dickerhof
- From the Department of Pathology and Biomedical Science, Centre for Free Radical Research, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - Anthony J Kettle
- From the Department of Pathology and Biomedical Science, Centre for Free Radical Research, University of Otago Christchurch, Christchurch 8140, New Zealand
| | - Christine C Winterbourn
- From the Department of Pathology and Biomedical Science, Centre for Free Radical Research, University of Otago Christchurch, Christchurch 8140, New Zealand
| |
Collapse
|
27
|
Xiao X, Saha P, Yeoh BS, Hipp JA, Singh V, Vijay-Kumar M. Myeloperoxidase deficiency attenuates systemic and dietary iron-induced adverse effects. J Nutr Biochem 2018; 62:28-34. [PMID: 30218980 DOI: 10.1016/j.jnutbio.2018.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/01/2018] [Accepted: 08/11/2018] [Indexed: 02/08/2023]
Abstract
Iron deficiency is routinely treated with oral or systemic iron supplements, which are highly reactive and could induce oxidative stress via augmenting the activity of proinflammatory enzyme myeloperoxidase (MPO). To investigate the extent to which MPO is involved in iron-induced toxicity, acute (24 h) iron toxicity was induced by intraperitoneal administration of FeSO4 (25 mg/kg body weight) to MPO-deficient (MpoKO) mice and their wild-type (WT) littermates. Acute iron toxicity was also assessed in WT mice pretreated with an MPO inhibitor, 4-aminobenzoic acid hydrazide. Systemic iron administration up-regulated circulating MPO and neutrophil elastase and elevated systemic inflammatory and organ damage markers in WT mice. However, genetic deletion of MPO or its inhibition significantly reduced iron-induced organ damage and systemic inflammatory responses. In contrast to the acute model, 8 weeks of 2% carbonyl iron diet feeding to WT mice did not change the levels of circulating MPO and neutrophil elastase but promoted their accumulation in the liver. Even though both MpoKO and WT mice displayed similar levels of diet-induced hyperferremia, MpoKO mice showed significantly reduced inflammatory response and oxidative stress than the WT mice. In addition, WT bone-marrow-derived neutrophils (BMDN) generated more reactive oxygen species than MPO-deficient BMDN upon iron stimulation. Altogether, genetic deficiency or pharmacologic inhibition of MPO substantially attenuated acute and chronic iron-induced toxicity. Our results suggest that targeting MPO during iron supplementation is a promising approach to reduce iron-induced toxicity/side effects in vulnerable population.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Piu Saha
- Department of Physiology & Pharmacology, University of Toledo, OH 43614, USA
| | - Beng San Yeoh
- Graduate Program in Immunology & Infectious Diseases, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jennifer A Hipp
- Department of Pathology, University of Toledo, OH 43614, USA
| | - Vishal Singh
- Department of Physiology & Pharmacology, University of Toledo, OH 43614, USA
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo, OH 43614, USA; Department of Medical Microbiology & Immunology, University of Toledo, OH 43614, USA.
| |
Collapse
|
28
|
Tseng A, Kim K, Li J, Cho J. Myeloperoxidase Negatively Regulates Neutrophil-Endothelial Cell Interactions by Impairing αMβ2 Integrin Function in Sterile Inflammation. Front Med (Lausanne) 2018; 5:134. [PMID: 29780806 PMCID: PMC5946029 DOI: 10.3389/fmed.2018.00134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/20/2018] [Indexed: 01/22/2023] Open
Abstract
Interactions of neutrophils with endothelial cells (ECs) and platelets contribute to tissue damage and vascular occlusion under sterile inflammatory conditions. However, the molecular mechanisms regulating the cell–cell interactions remain poorly understood. Previous studies suggest that reactive oxygen species, such as hydrogen peroxide (H2O2), produced from NADPH oxidase 2 play a critical role in platelet–neutrophil interactions by regulating the function of neutrophil αMβ2 integrin during sterile inflammation. In this study, we further demonstrate a crucial role for myeloperoxidase (MPO) in regulating the adhesive function of neutrophils through αMβ2 integrin. Using real-time fluorescence intravital microscopy and in vitro assays, we showed that loss of MPO promoted neutrophil–EC interactions and neutrophil emigration but did not affect neutrophil–platelet interactions under inflammatory conditions. Using genetic and pharmacologic approaches, we found that following agonist stimulation, MPO knockout (KO) neutrophils exhibited a significant increase in extracellular H2O2 and surface level of αMβ2 integrin and that these effects were dependent on MPO activity. Our in vivo studies using an ischemia/reperfusion-induced hepatic inflammation model revealed that compared to wild-type mice, neutrophils from MPO KO mice—displayed a pro-migratory phenotype while ameliorating tissue damage. These results suggest that MPO plays a negative role in the adhesive and migratory function of neutrophils by impairing αMβ2 integrin function under sterile inflammatory conditions.
Collapse
Affiliation(s)
- Alan Tseng
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| | - Kyungho Kim
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, United States.,Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu, South Korea
| | - Jing Li
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| |
Collapse
|
29
|
New fluorescence-based high-throughput screening assay for small molecule inhibitors of tyrosyl-DNA phosphodiesterase 2 (TDP2). Eur J Pharm Sci 2018; 118:67-79. [PMID: 29574079 DOI: 10.1016/j.ejps.2018.03.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 01/03/2023]
Abstract
Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs topoisomerase II (TOP2) mediated DNA damages and causes resistance to TOP2-targeted cancer therapy. Inhibiting TDP2 could sensitize cancer cells toward TOP2 inhibitors. However, potent TDP2 inhibitors with favorable physicochemical properties are not yet reported. Therefore, there is a need to search for novel molecular scaffolds capable of inhibiting TDP2. We report herein a new simple, robust, homogenous mix-and-read fluorescence biochemical assay based using humanized zebrafish TDP2 (14M_zTDP2), which provides biochemical and molecular structure basis for TDP2 inhibitor discovery. The assay was validated by screening a preselected library of 1600 compounds (Z' ≥ 0.72) in a 384-well format, and by running in parallel gel-based assays with fluorescent DNA substrates. This library was curated via virtual high throughput screening (vHTS) of 460,000 compounds from Chembridge Library, using the crystal structure of the novel surrogate protein 14M_zTDP2. From this primary screening, we selected the best 32 compounds (2% of the library) to further assess their TDP2 inhibition potential, leading to the IC50 determination of 10 compounds. Based on the dose-response curve profile, pan-assay interference compounds (PAINS) structure identification, physicochemical properties and efficiency parameters, two hit compounds, 11a and 19a, were tested using a novel secondary fluorescence gel-based assay. Preliminary structure-activity relationship (SAR) studies identified guanidine derivative 12a as an improved hit with a 6.4-fold increase in potency over the original HTS hit 11a. This study highlights the importance of the development of combination approaches (biochemistry, crystallography and high throughput screening) for the discovery of TDP2 inhibitors.
Collapse
|
30
|
Forbes LV, Kettle AJ. A multi-substrate assay for finding physiologically effective inhibitors of myeloperoxidase. Anal Biochem 2018; 544:13-21. [DOI: 10.1016/j.ab.2017.12.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/23/2022]
|