1
|
Rudrapal M, Kirboga KK, Abdalla M, Maji S. Explainable artificial intelligence-assisted virtual screening and bioinformatics approaches for effective bioactivity prediction of phenolic cyclooxygenase-2 (COX-2) inhibitors using PubChem molecular fingerprints. Mol Divers 2024; 28:2099-2118. [PMID: 38200203 DOI: 10.1007/s11030-023-10782-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/22/2023] [Indexed: 01/12/2024]
Abstract
Cyclooxygenase-2 (COX-2) inhibitors are nonsteroidal anti-inflammatory drugs that treat inflammation, pain and fever. This study determined the interaction mechanisms of COX-2 inhibitors and the molecular properties needed to design new drug candidates. Using machine learning and explainable AI methods, the inhibition activity of 1488 molecules was modelled, and essential properties were identified. These properties included aromatic rings, nitrogen-containing functional groups and aliphatic hydrocarbons. They affected the water solubility, hydrophobicity and binding affinity of COX-2 inhibitors. The binding mode, stability and ADME properties of 16 ligands bound to the Cyclooxygenase active site of COX-2 were investigated by molecular docking, molecular dynamics simulation and MM-GBSA analysis. The results showed that ligand 339,222 was the most stable and effective COX-2 inhibitor. It inhibited prostaglandin synthesis by disrupting the protein conformation of COX-2. It had good ADME properties and high clinical potential. This study demonstrated the potential of machine learning and bioinformatics methods in discovering COX-2 inhibitors.
Collapse
Affiliation(s)
- Mithun Rudrapal
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan's Foundation for Science, Technology & Research (Deemed to Be University), Guntur, 522213, India.
| | - Kevser Kübra Kirboga
- Informatics Institute, Istanbul Technical University, 34469, Maslak, Istanbul, Turkey.
- Bioengineering Department, BilecikSeyhEdebali University, 11230, Bilecik, Turkey.
| | - Mohnad Abdalla
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, 250022, Shandong, People's Republic of China
| | - Siddhartha Maji
- Department of Chemistry, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
2
|
Agafonova I, Chingizova E, Chaikina E, Menchinskaya E, Kozlovskiy S, Likhatskaya G, Sabutski Y, Polonik S, Aminin D, Pislyagin E. Protection Activity of 1,4-Naphthoquinones in Rotenone-Induced Models of Neurotoxicity. Mar Drugs 2024; 22:62. [PMID: 38393033 PMCID: PMC10890484 DOI: 10.3390/md22020062] [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: 12/06/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
The MTS cell viability test was used to screen a mini library of natural and synthetic 1,4-naphthoquinone derivatives (1,4-NQs) from marine sources. This screening identified two highly effective compounds, U-443 and U-573, which showed potential in protecting Neuro-2a neuroblastoma cells from the toxic effects of rotenone in an in vitro model of neurotoxicity. The selected 1,4-NQs demonstrated the capability to reduce oxidative stress by decreasing the levels of reactive oxygen species (ROS) and nitric oxide (NO) in Neuro-2a neuroblastoma cells and RAW 264.7 macrophage cells and displayed significant antioxidant properties in mouse brain homogenate. Normal mitochondrial function was restored and the mitochondrial membrane potential was also regained by 1,4-NQs after exposure to neurotoxins. Furthermore, at low concentrations, these compounds were found to significantly reduce levels of proinflammatory cytokines TNF and IL-1β and notably inhibit the activity of cyclooxygenase-2 (COX-2) in RAW 264.7 macrophages. The results of docking studies showed that the 1,4-NQs were bound to the active site of COX-2, analogically to a known inhibitor of this enzyme, SC-558. Both substances significantly improved the behavioral changes in female CD1 mice with rotenone-induced early stage of Parkinson's disease (PD) in vivo. It is proposed that the 1,4-NQs, U-443 and U-573, can protect neurons and microglia through their potent anti-ROS and anti-inflammatory activities.
Collapse
Affiliation(s)
- Irina Agafonova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Ekaterina Chingizova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Elena Chaikina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Ekaterina Menchinskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Sergey Kozlovskiy
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Galina Likhatskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Yuri Sabutski
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Sergey Polonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| | - Dmitry Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shin-Chuan 1st Road, Sanmin District, Kaohsiung City 80708, Taiwan
| | - Evgeny Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia; (I.A.); (E.C.); (E.C.); (E.M.); (S.K.); (G.L.); (Y.S.); (S.P.); (D.A.)
| |
Collapse
|
3
|
Sharma A, Sharma R, Sharma M, Panadan JJ, Ansari MA, Dalai S. Comparative evaluation of effect of different premedication agents on efficacy of Articaine: A randomized contol trial. J Oral Biol Craniofac Res 2023; 13:218-223. [PMID: 36741857 PMCID: PMC9894783 DOI: 10.1016/j.jobcr.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/18/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023] Open
Abstract
Type of study Original Research. Aims To comparatively evaluate the effect of different premedication agents on the efficacy of 4% Articaine in teeth with symptomatic irreversible pulpitis. Materials and methods The primary objective of our study is to evaluate the effect of premedication agents on efficacy of Articaine as an oral anesthetic. Our secondary objective is to comparatively evaluate the efficacy of Diclofenac patch, Ibuprofen tablet, Paracetamol tablet and Placebo as a premedication agent. Patients with 25-40 years age, no systemic disease, no history of medication for that complaint, with pain on Heft Parker Visual Analog Scale between 55 mm and 170 mm (VAS), no tenderness on percussion, cold test and EPT negative- Positive, giving proper consent, coming to the Department of Conservative Dentistry and Endodontics were allowed to participate. The exclusion criteria include the following- Non-vital teeth, pregnant and lactating women, allergic to Articaine and NSAIDs, active systemic disease, immune-compromised patients, taken analgesics in last 24 h, root fractures, restoration extending to pulp10 and periapical pathologies (except periodontal ligament widening).Preoperatively pain was recorded using Heft Parker VAS (Visual Analog Scale). Cold testing, palpation, percussion and EPT were carried out. 40 patients having symptomatic irreversible pulpitis were randomly divided into 4 groups: group 1 Placebo (n = 10), group 2-Diclofenac patch (n = 10), group-3 Ibuprofen tablets (n = 10), group 4-Paracetamol tablets (n = 10). After 1 h of premedication, all patients were administered IANB injection using 4% Articaine (Septanest with adrenaline 1/100000, Septodont, France) containing epinephrine 1:100000. 15 mins after administration of IANB, patients were asked about symptomatic numbness and was tested with Endo frost and EPT and Outcome was recorded. If lip numbness was present, Electric Pulp Testing and Cold Test give negative result then endodontic access opening was performed and pain was recorded using visual analog scale. The study was conducted for a period of 1.5 years. Results During the access cavity preparation only 1 subject in the Group III reported pain while in other groups none of the subjects reported pain of any type. When the intergroup comparison was made of intensity of pain 15 min after LA and during access cavity preparation, the difference between the groups was statistically non-significant when analyzed using One Way ANOVA. The intragroup comparison between three time intervals revealed significant reduction in the pain scores from the pre-treatment levels in all the four groups. Conclusions The results of the study showed that there is no significant effect of different premedication agents on the efficacy of 4% Articaine in teeth with symptomatic irreversible pulpitis.
Collapse
Affiliation(s)
- Anjali Sharma
- Department of Conservative Dentistry and Endodontics, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, India
| | - Rohit Sharma
- Department of Conservative Dentistry and Endodontics, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, India
| | - Madhurima Sharma
- Department of Prosthodontics and crown and bridge, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, India
| | - John Johnson Panadan
- Department of Conservative Dentistry and Endodontics, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, India
| | - M Abbas Ansari
- Department of Conservative Dentistry and Endodontics, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, India
| | - Sasmita Dalai
- Department of Conservative Dentistry and Endodontics, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, India
- Department of Public Health Detistry, India
| |
Collapse
|
4
|
El-Dershaby NH, El-Hawash SA, Kassab SE, Daabees HG, Abdel Moneim AE, El-Miligy MMM. Rational Design and Synthesis of New Selective COX-2 Inhibitors with In Vivo PGE2-Lowering Activity by Tethering Benzenesulfonamide and 1,2,3-Triazole Pharmacophores to Some NSAIDs. Pharmaceuticals (Basel) 2022; 15:ph15101165. [PMID: 36297278 PMCID: PMC9609428 DOI: 10.3390/ph15101165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/01/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
New selective COX-2 inhibitors were designed and synthesized by tethering 1,2,3-triazole and benzenesulfonamide pharmacophores to some NSAIDs. Compounds 6b and 6j showed higher in vitro COX-2 selectivity and inhibitory activity (IC50 = 0.04 µM and S.I. = 329 and 312, respectively) than celecoxib (IC50 = 0.05 µM and S.I. = 294). Compound 6e revealed equipotent in vitro COX-2 inhibitory activity to celecoxib. Furthermore, 6b and 6j expressed more potent relief of carrageenan-induced paw edema thickness in mice than celecoxib, with ED50 values of 11.74 µmol/kg and 13.38 µmol/kg vs. 16.24 µmol/kg, respectively. Compounds 6b and 6j inhibited the production of PGE2 with a % inhibition of PGE2 production of 90.70% and 86.34%, respectively, exceeding celecoxib’s percentage (78.62%). Moreover, 6b and 6j demonstrated a gastric safety profile comparable to celecoxib. In conclusion, compounds 6b and 6j better achieved the target goal as more potent and selective COX-2 inhibitors than celecoxib in vitro and in vivo.
Collapse
Affiliation(s)
- Nada H. El-Dershaby
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour 22516, Egypt
| | - Soad A. El-Hawash
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Shaymaa E. Kassab
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour 22516, Egypt
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt
| | - Hoda G. Daabees
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour 22516, Egypt
| | - Ahmed E. Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Mostafa M. M. El-Miligy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Correspondence:
| |
Collapse
|
5
|
Mori T, Abe I. Structural basis for endoperoxide-forming oxygenases. Beilstein J Org Chem 2022; 18:707-721. [PMID: 35821691 PMCID: PMC9235837 DOI: 10.3762/bjoc.18.71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
Endoperoxide natural products are widely distributed in nature and exhibit various biological activities. Due to their chemical features, endoperoxide and endoperoxide-derived secondary metabolites have attracted keen attention in the field of natural products and organic synthesis. In this review, we summarize the structural analyses, mechanistic investigations, and proposed reaction mechanisms of endoperoxide-forming oxygenases, including cyclooxygenase, fumitremorgin B endoperoxidase (FtmOx1), and the asnovolin A endoperoxygenase NvfI.
Collapse
Affiliation(s)
- Takahiro Mori
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
- PRESTO, Japan Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Ikuro Abe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
6
|
Papa A, Pasquini S, Contri C, Gemma S, Campiani G, Butini S, Varani K, Vincenzi F. Polypharmacological Approaches for CNS Diseases: Focus on Endocannabinoid Degradation Inhibition. Cells 2022; 11:cells11030471. [PMID: 35159280 PMCID: PMC8834510 DOI: 10.3390/cells11030471] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Polypharmacology breaks up the classical paradigm of “one-drug, one target, one disease” electing multitarget compounds as potential therapeutic tools suitable for the treatment of complex diseases, such as metabolic syndrome, psychiatric or degenerative central nervous system (CNS) disorders, and cancer. These diseases often require a combination therapy which may result in positive but also negative synergistic effects. The endocannabinoid system (ECS) is emerging as a particularly attractive therapeutic target in CNS disorders and neurodegenerative diseases including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), stroke, traumatic brain injury (TBI), pain, and epilepsy. ECS is an organized neuromodulatory network, composed by endogenous cannabinoids, cannabinoid receptors type 1 and type 2 (CB1 and CB2), and the main catabolic enzymes involved in the endocannabinoid inactivation such as fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL). The multiple connections of the ECS with other signaling pathways in the CNS allows the consideration of the ECS as an optimal source of inspiration in the development of innovative polypharmacological compounds. In this review, we focused our attention on the reported polypharmacological examples in which FAAH and MAGL inhibitors are involved.
Collapse
Affiliation(s)
- Alessandro Papa
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Silvia Pasquini
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.P.); (S.G.); (G.C.)
- Correspondence: ; Tel.: +39-0577-234161
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.); (F.V.)
| |
Collapse
|
7
|
In silico characterisation of olive phenolic compounds as potential cyclooxygenase modulators. Part 1. J Mol Graph Model 2020; 101:107719. [PMID: 32898836 DOI: 10.1016/j.jmgm.2020.107719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/09/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used to reduce pain. These target cyclooxygenase (COX) enzymes which produce inflammatory mediators. Adverse effects associated with the use of traditional NSAIDs have led to a rise in the development of alternative therapies. Derived from Olea Europaea, olive oil is a main component of the Mediterranean diet, containing phenolic compounds that contribute to its antioxidant and anti-inflammatory properties. It has previously been found that oleocanthal, a phenolic compound derived from the olive, had similar effects to ibuprofen, a commonly used NSAID. There is an abundance of olive phenolic compounds that have yet to be investigated for their anti-inflammatory properties. In this study, it was sought to identify potential olive-derived compounds with the ability to inhibit COX enzymes, and study the mechanisms using in silico approaches. Molecular docking was employed to determine the COX inhibitory potential of an olive phenolic compound library. From docking, it was determined that 1-oleyltyrosol (1OL) and ligstroside derivative 2 (LG2) demonstrated the greatest binding affinity to both COX-1 and COX-2. Interactions with these compounds were further examined using molecular dynamics simulations. The residue contributions to binding free energy were computed using Molecular Mechanics-Poisson Boltzmann Surface Area (MM-PBSA) methods, revealing that residues Leu93, Val116, Leu352, and Ala527 in COX-1 and COX-2 were key determinants of potential inhibition. Along with part 2 of this study, this work aims to identify and characterise novel phenolic compounds which may possess COX inhibitory properties.
Collapse
|
8
|
Rouzer CA, Marnett LJ. Structural and Chemical Biology of the Interaction of Cyclooxygenase with Substrates and Non-Steroidal Anti-Inflammatory Drugs. Chem Rev 2020; 120:7592-7641. [PMID: 32609495 PMCID: PMC8253488 DOI: 10.1021/acs.chemrev.0c00215] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cyclooxgenases are key enzymes of lipid signaling. They carry out the first step in the production of prostaglandins, important mediators of inflammation, pain, cardiovascular disease, and cancer, and they are the molecular targets for nonsteroidal anti-inflammatory drugs, which are among the oldest and most chemically diverse set of drugs known. Homodimeric proteins that behave as allosterically modulated, functional heterodimers, the cyclooxygenases exhibit complex kinetic behavior, requiring peroxide-dependent activation and undergoing suicide inactivation. Due to their important physiological and pathophysiological roles and keen interest on the part of the pharmaceutical industry, the cyclooxygenases have been the focus of a vast array of structural studies, leading to the publication of over 80 crystal structures of the enzymes in complex with substrates or inhibitors supported by a wealth of functional data generated by site-directed mutation experiments. In this review, we explore the chemical biology of the cyclooxygenases through the lens of this wealth of structural and functional information. We identify key structural features of the cyclooxygenases, break down their active site into regional binding pockets to facilitate comparisons between structures, and explore similarities and differences in the binding modes of the wide variety of ligands (both substrates and inhibitors) that have been characterized in complex with the enzymes. Throughout, we correlate structure with function whenever possible. Finally, we summarize what can and cannot be learned from the currently available structural data and discuss the critical intriguing questions that remain despite the wealth of information that has been amassed in this field.
Collapse
Affiliation(s)
- Carol A Rouzer
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J Marnett
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
9
|
Yadav AK, Reinhardt CJ, Arango AS, Huff HC, Dong L, Malkowski MG, Das A, Tajkhorshid E, Chan J. An Activity-Based Sensing Approach for the Detection of Cyclooxygenase-2 in Live Cells. Angew Chem Int Ed Engl 2020; 59:3307-3314. [PMID: 31854058 PMCID: PMC7416425 DOI: 10.1002/anie.201914845] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Indexed: 01/05/2023]
Abstract
Cyclooxygenase-2 (COX-2) overexpression is prominent in inflammatory diseases, neurodegenerative disorders, and cancer. Directly monitoring COX-2 activity within its native environment poses an exciting approach to account for and illuminate the effect of the local environments on protein activity. Herein, we report the development of CoxFluor, the first activity-based sensing approach for monitoring COX-2 within live cells with confocal microscopy and flow cytometry. CoxFluor strategically links a natural substrate with a dye precursor to engage both the cyclooxygenase and peroxidase activities of COX-2. This catalyzes the release of resorufin and the natural product, as supported by molecular dynamics and ensemble docking. CoxFluor enabled the detection of oxygen-dependent changes in COX-2 activity that are independent of protein expression within live macrophage cells.
Collapse
Affiliation(s)
- Anuj K Yadav
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Christopher J Reinhardt
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Andres S Arango
- Center for Biophysics and Quantitative Biology, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hannah C Huff
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Liang Dong
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Michael G Malkowski
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Aditi Das
- Center for Biophysics and Quantitative Biology, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Emad Tajkhorshid
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Center for Biophysics and Quantitative Biology, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jefferson Chan
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
10
|
Yadav AK, Reinhardt CJ, Arango AS, Huff HC, Dong L, Malkowski MG, Das A, Tajkhorshid E, Chan J. An Activity‐Based Sensing Approach for the Detection of Cyclooxygenase‐2 in Live Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914845] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Anuj K. Yadav
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Christopher J. Reinhardt
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Andres S. Arango
- Center for Biophysics and Quantitative Biology Department of Biochemistry University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Hannah C. Huff
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Liang Dong
- Department of Structural Biology Jacobs School of Medicine and Biomedical Sciences University at Buffalo Buffalo NY 14203 USA
| | - Michael G. Malkowski
- Department of Structural Biology Jacobs School of Medicine and Biomedical Sciences University at Buffalo Buffalo NY 14203 USA
| | - Aditi Das
- Center for Biophysics and Quantitative Biology Department of Biochemistry University of Illinois at Urbana-Champaign Urbana IL 61801 USA
- Department of Comparative Biosciences University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Emad Tajkhorshid
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
- Center for Biophysics and Quantitative Biology Department of Biochemistry University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Jefferson Chan
- Department of Chemistry Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| |
Collapse
|
11
|
Venugopala KN, Al-Attraqchi OHA, Tratrat C, Nayak SK, Morsy MA, Aldhubiab BE, Attimarad M, Nair AB, Sreeharsha N, Venugopala R, Haroun M, Girish MB, Chandrashekharappa S, Alwassil OI, Odhav B. Novel Series of Methyl 3-(Substituted Benzoyl)-7-Substituted-2-Phenylindolizine-1-Carboxylates as Promising Anti-Inflammatory Agents: Molecular Modeling Studies. Biomolecules 2019; 9:E661. [PMID: 31661893 PMCID: PMC6920857 DOI: 10.3390/biom9110661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 01/24/2023] Open
Abstract
The cyclooxygenase-2 (COX-2) enzyme is considered to be an important target for developing novel anti-inflammatory agents. Selective COX-2 inhibitors offer the advantage of lower adverse effects that are commonly associated with non-selective COX inhibitors. In this work, a novel series of methyl 3-(substituted benzoyl)-7-substituted-2-phenylindolizine-1-carboxylates was synthesized and evaluated for COX-2 inhibitory activity. Compound 4e was identified as the most active compound of the series with an IC50 of 6.71 M, which is comparable to the IC50 of indomethacin, a marketed non-steroidal anti-inflammatory drug (NSAID). Molecular modeling and crystallographic studies were conducted to further characterize the compounds and gain better understanding of the binding interactions between the compounds and the residues at the active site of the COX-2 enzyme. The pharmacokinetic properties and potential toxic effects were predicted for all the synthesized compounds, which indicated good drug-like properties. Thus, these synthesized compounds can be considered as potential lead compounds for developing effective anti-inflammatory therapeutic agents.
Collapse
Affiliation(s)
- Katharigatta N Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
- Department of Biotechnology and Food Technology, Durban University of Technology, Durban 4001, South Africa.
| | | | - Christophe Tratrat
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Susanta K Nayak
- Department of Chemistry, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra 440010, India.
| | - Mohamed A Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt.
| | - Bandar E Aldhubiab
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Mahesh Attimarad
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Anroop B Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Rashmi Venugopala
- Department of Public Health Medicine, University of KwaZulu-Natal, Howard College Campus, Durban 4001, South Africa.
| | - Michelyne Haroun
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Meravanige B Girish
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Sandeep Chandrashekharappa
- Institute for Stem Cell Biology and Regenerative Medicine, NCBS, TIFR, GKVK, Bellary Road, Bangalore 560065, India.
| | - Osama I Alwassil
- Department of Pharmaceutical Sciences, College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia.
| | - Bharti Odhav
- Department of Biotechnology and Food Technology, Durban University of Technology, Durban 4001, South Africa.
| |
Collapse
|
12
|
Kaur B, Kaur M, Kaur N, Garg S, Bhatti R, Singh P. Engineered Substrate for Cyclooxygenase-2: A Pentapeptide Isoconformational to Arachidonic Acid for Managing Inflammation. J Med Chem 2019; 62:6363-6376. [PMID: 31244108 DOI: 10.1021/acs.jmedchem.9b00823] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Beyond the conventional mode of working of anti-inflammatory agents through enzyme inhibition, herein, COX-2 was provided with an alternate substrate. A proline-centered pentapeptide isoconformational to arachidonic acid, which exhibited appreciable selectivity for COX-2, overcoming acetic acid- and formalin-induced pain in rats to almost 80%, was treated as a substrate by the enzyme. Remarkably, COX-2 metabolized the pentapeptide into small fragments consisting mainly of di- and tripeptides that ensured the safe breakdown of the peptide under in vivo conditions. The kinetic parameter Kcat/Km for COX-2-mediated metabolism of the peptide (6.3 × 105 M-1 s-1) was quite similar to 9.5 × 105 M-1 s-1 for arachidonic acid. Evidenced by the molecular dynamic studies and the use of Y385F COX-2, it was observed that the breakage of the pentapeptide has probably been taken place through H-bond activation of the peptide bond by the side chains of Y385 and S530.
Collapse
|
13
|
Smith WL, Malkowski MG. Interactions of fatty acids, nonsteroidal anti-inflammatory drugs, and coxibs with the catalytic and allosteric subunits of cyclooxygenases-1 and -2. J Biol Chem 2019; 294:1697-1705. [PMID: 30710016 DOI: 10.1074/jbc.tm118.006295] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Prostaglandin endoperoxide H synthases-1 and -2, commonly called cyclooxygenases-1 and -2 (COX-1 and -2), catalyze the committed step in prostaglandin biosynthesis-the conversion of arachidonic acid to prostaglandin endoperoxide H2 Both COX isoforms are sequence homodimers that function as conformational heterodimers having allosteric (Eallo) and catalytic (Ecat) subunits. At least in the case of COX-2, the enzyme becomes folded into a stable Eallo/Ecat pair. Some COX inhibitors (i.e. nonsteroidal anti-inflammatory drugs and coxibs) and common fatty acids (FAs) modulate Ecat activity by binding Eallo. However, the interactions and outcomes often differ between isoforms. For example, naproxen directly and completely inhibits COX-1 by binding Ecat but indirectly and incompletely inhibits COX-2 by binding Eallo. Additionally, COX-1 is allosterically inhibited up to 50% by common FAs like palmitic acid, whereas COX-2 is allosterically activated 2-fold by palmitic acid. FA binding to Eallo also affects responses to COX inhibitors. Thus, COXs are physiologically and pharmacologically regulated by the FA tone of the milieu in which each operates-COX-1 in the endoplasmic reticulum and COX-2 in the Golgi apparatus. Cross-talk between Eallo and Ecat involves a loop in Eallo immediately downstream of Arg-120. Mutational studies suggest that allosteric modulation requires a direct interaction between the carboxyl group of allosteric effectors and Arg-120 of Eallo; however, structural studies show some allosterically active FAs positioned in COX-2 in a conformation lacking an interaction with Arg-120. Thus, many details about the biological consequences of COX allosterism and how ligand binding to Eallo modulates Ecat remain to be resolved.
Collapse
Affiliation(s)
- William L Smith
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109.
| | - Michael G Malkowski
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, New York 14203.
| |
Collapse
|
14
|
Biological evaluation and in silico molecular docking study of a new series of thiazol-2-yl-hydrazone conglomerates. RESEARCH ON CHEMICAL INTERMEDIATES 2018. [DOI: 10.1007/s11164-018-3261-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Goodman MC, Xu S, Rouzer CA, Banerjee S, Ghebreselasie K, Migliore M, Piomelli D, Marnett LJ. Dual cyclooxygenase-fatty acid amide hydrolase inhibitor exploits novel binding interactions in the cyclooxygenase active site. J Biol Chem 2018; 293:3028-3038. [PMID: 29326169 DOI: 10.1074/jbc.m117.802058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/13/2017] [Indexed: 12/29/2022] Open
Abstract
The cyclooxygenases COX-1 and COX-2 oxygenate arachidonic acid (AA) to prostaglandin H2 (PGH2). COX-2 also oxygenates the endocannabinoids 2-arachidonoylglycerol (2-AG) and arachidonoylethanolamide (AEA) to the corresponding PGH2 analogs. Both enzymes are targets of nonsteroidal anti-inflammatory drugs (NSAIDs), but NSAID-mediated COX inhibition is associated with gastrointestinal toxicity. One potential strategy to counter this toxicity is to also inhibit fatty acid amide hydrolase (FAAH), which hydrolyzes bioactive fatty acid ethanolamides (FAEs) into fatty acids and ethanolamine. Here, we investigated the mechanism of COX inhibition by ARN2508, an NSAID that inhibits both COXs and FAAH with high potency, target selectivity, and decreased gastrointestinal toxicity in mouse models, presumably due to its ability to increase levels of FAEs. A 2.27-Å-resolution X-ray crystal structure of the COX-2·(S)-ARN2508 complex reveals that ARN2508 adopts a binding pose similar to that of its parent NSAID flurbiprofen. However, ARN2508's alkyl tail is inserted deep into the top channel, an active site region not exploited by any previously reported NSAID. As for flurbiprofen, ARN2508's potency is highly dependent on the configuration of the α-methyl group. Thus, (S)-ARN2508 is more potent than (R)-ARN2508 for inhibition of AA oxygenation by both COXs and 2-AG oxygenation by COX-2. Also, similarly to (R)-flurbiprofen, (R)-ARN2508 exhibits substrate selectivity for inhibition of 2-AG oxygenation. Site-directed mutagenesis confirms the importance of insertion of the alkyl tail into the top channel for (S)-ARN2508's potency and suggests a role for Ser-530 as a determinant of the inhibitor's slow rate of inhibition compared with that of (S)-flurbiprofen.
Collapse
Affiliation(s)
- Michael C Goodman
- From the A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology and Center in Molecular Toxicology, Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Shu Xu
- From the A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology and Center in Molecular Toxicology, Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Carol A Rouzer
- From the A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology and Center in Molecular Toxicology, Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Surajit Banerjee
- the Northeastern Collaborative Access Team, Argonne National Laboratory, Argonne, Illinois 60439
| | - Kebreab Ghebreselasie
- From the A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology and Center in Molecular Toxicology, Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Marco Migliore
- the Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Daniele Piomelli
- the Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, 16163 Genoa, Italy.,the Departments of Anatomy, Neurobiology, Pharmacology, and Biological Chemistry, University of California, Irvine, California 92697, and
| | - Lawrence J Marnett
- From the A. B. Hancock, Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology and Center in Molecular Toxicology, Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232,
| |
Collapse
|
16
|
Chandrashekharappa S, Venugopala KN, Tratrat C, Mahomoodally FM, Aldhubiab BE, Haroun M, Venugopala R, Mohan MK, Kulkarni RS, Attimarad MV, Harsha S, Odhav B. Efficient synthesis and characterization of novel indolizines: exploration of in vitro COX-2 inhibitory activity and molecular modelling studies. NEW J CHEM 2018. [DOI: 10.1039/c7nj05010k] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Novel indolizine scaffolds as COX-2 inhibiting agents.
Collapse
|
17
|
Kassab SE, Khedr MA, Ali HI, Abdalla MM. Discovery of new indomethacin-based analogs with potentially selective cyclooxygenase-2 inhibition and observed diminishing to PGE2 activities. Eur J Med Chem 2017; 141:306-321. [PMID: 29031075 DOI: 10.1016/j.ejmech.2017.09.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 11/29/2022]
Abstract
New ring-extended analogs of indomethacin were designed based on the structure of active binding site of both COX-1 and COX-2 isoenzymes and the interaction pattern required for selective inhibition of COX-2 to improve its selectivity against COX-2. The strategy adopted for designing the new inhibitors involved i) ring extension of indomethacin to reduce the possibility of analogs to be accommodated into the narrow hydrophobic tunnel of COX-1, ii) deletion of carboxylic acid to reduce the possibility of inhibitor to form salt bridge with Arg120 and eventually prevent COX-1 inhibition, and iii) introduction of methylsulfonyl group to increase the opportunity of the analogs to interact with the polar side pocket that's is crucial for inhibition process of COX-2. The three series of tetrahydrocarbazoles involving 4, 5, 9, 10 and 12 were synthesized in quantitative yields adopting limited number of reaction steps, and applying laboratory friendly reaction conditions. In vitro and in vivo assays for data profiling the new candidates revealed the significant improvement in the potency and selectivity against COX-2 of 6-methoxytetrahydrocarbazole 4 (IC50 = 0.97 μmol) to verify the effect of ring extension in comparison to indomethacin (IC50 = 2.63 μmol), and 6-methylsulfonyltetrahydrocarbazole 10a (IC50 = 0.28 μmol) to verify the effect of ring extension and introduction of methylsulfonyl group. 9-(4-chlorobenzoyl)-6-(methylsulfonyl)-1,2,3,9-tetrahydro-4H-carbazol-4-one 12a showed the most potential and selective activity against COX-2 (IC50 = 0.23 μmol) to be with superior potency to Celecoxib (IC50 = 0.30 μmol). Consistently, 12a was the most active with all the other anti-inflammatory test descriptors and its activity in diminishing the PGE2 with the other analogs confirmed the elaboration of new class of selective COX-2 inhibitors beyond the diarylsulfonamides as a previously common class of selective COX-2 inhibitors. Molecular docking study revealed the high binding score of compound 12a (-30.78 kcal/mol), with less clash contribution (7.2) that is close to indomethacin. Also, 12a showed low conformation entropy score (1.40). Molecular dynamic (MD) simulation identified the equilibrium of both potential and kinetic energies.
Collapse
Affiliation(s)
- Shaymaa E Kassab
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira 22516, Egypt.
| | - Mohammed A Khedr
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt
| | - Hamed I Ali
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt; Department of Pharmaceutical Sciences, Texas A&M University Irma Lerma Rangel College of Pharmacy, Kingsville 78363, Texas, USA
| | - Mohamed M Abdalla
- Research Unit, Saco Pharm. Co., 6th of October City, Giza 68330, Egypt
| |
Collapse
|
18
|
Narsinghani T, Sharma R. Synthesis, anti-inflammatory activities and docking studies of amide derivatives of meclofenamic acid. CHEMICAL PAPERS 2016. [DOI: 10.1007/s11696-016-0102-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
19
|
Dong L, Yuan C, Orlando BJ, Malkowski MG, Smith WL. Fatty Acid Binding to the Allosteric Subunit of Cyclooxygenase-2 Relieves a Tonic Inhibition of the Catalytic Subunit. J Biol Chem 2016; 291:25641-25655. [PMID: 27756840 DOI: 10.1074/jbc.m116.757310] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/13/2016] [Indexed: 01/24/2023] Open
Abstract
Prostaglandin endoperoxide H synthase-2 (PGHS-2), also called cyclooxygenase-2 (COX-2), converts arachidonic acid to PGH2 PGHS-2 is a conformational heterodimer composed of allosteric (Eallo) and catalytic (Ecat) subunits. Fatty acids (FAs) bind to Arg-120 of Eallo increasing to different degrees, depending on the FA, the Vmax of its Ecat partner. We report here that movement of helical residues 120-122 and loop residues 123-129 of Eallo underlies the allosteric effects of FAs and allosteric COX-2 inhibitors, including naproxen and flurbiprofen. An S121P substitution in both PGHS-2 monomers yields a variant (S121P/S121P PGHS-2) that has 1.7-1.8 times the Vmax of native PGHS-2 and is relatively insensitive to activation by FAs or inhibition by allosteric inhibitors. The S121P substitution in Eallo is primarily responsible for these effects. In X-ray crystal structures, the Cα atoms of helical residues 119-122 of S121P/S121P PGHS-2 are displaced from their normal positions. Additionally, the S121P/S121P PGHS-2 variants in which Pro-127 and Ser-541 are replaced by cysteines spontaneously forms Cys-127 to Cys-541 cross-links between monomers. This is unlike the corresponding native PGHS-2 variant and suggests that S121P substitutions also unhinge the loop involving residues 123-129. We conclude the following: (a) the region involving residues 120-129 of unoccupied Eallo tonically inhibits Ecat; (b) binding of an activating FA (e.g. arachidonic, palmitic, or oleic acid) to Eallo or an S121P substitution in Eallo repositions this region to increase Ecat activity; and (c) allosteric COX inhibitors act by preventing FA binding to Eallo and additionally by relocating Eallo residues to inhibit Ecat.
Collapse
Affiliation(s)
- Liang Dong
- From the Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Chong Yuan
- From the Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Benjamin J Orlando
- the Department of Structural Biology, University at Buffalo, The State University of New York, and the Hauptman-Woodward Medical Research Institute, Buffalo, New York 14203
| | - Michael G Malkowski
- the Department of Structural Biology, University at Buffalo, The State University of New York, and the Hauptman-Woodward Medical Research Institute, Buffalo, New York 14203
| | - William L Smith
- From the Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| |
Collapse
|
20
|
TRPV1-FAAH-COX: TheCouples Gamein Pain Treatment. ChemMedChem 2016; 11:1686-94. [DOI: 10.1002/cmdc.201600111] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/31/2016] [Indexed: 12/11/2022]
|
21
|
Singh P, Kaur S, Kaur J, Singh G, Bhatti R. Rational Design of Small Peptides for Optimal Inhibition of Cyclooxygenase-2: Development of a Highly Effective Anti-Inflammatory Agent. J Med Chem 2016; 59:3920-34. [DOI: 10.1021/acs.jmedchem.6b00134] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Palwinder Singh
- UGC
Sponsored Centre for Advanced Studies - Department of Chemistry and ‡Department of Pharmaceutical
Sciences, Guru Nanak Dev University, Amritsar 143005, India
| | - Sukhmeet Kaur
- UGC
Sponsored Centre for Advanced Studies - Department of Chemistry and ‡Department of Pharmaceutical
Sciences, Guru Nanak Dev University, Amritsar 143005, India
| | - Jagroop Kaur
- UGC
Sponsored Centre for Advanced Studies - Department of Chemistry and ‡Department of Pharmaceutical
Sciences, Guru Nanak Dev University, Amritsar 143005, India
| | - Gurjit Singh
- UGC
Sponsored Centre for Advanced Studies - Department of Chemistry and ‡Department of Pharmaceutical
Sciences, Guru Nanak Dev University, Amritsar 143005, India
| | - Rajbir Bhatti
- UGC
Sponsored Centre for Advanced Studies - Department of Chemistry and ‡Department of Pharmaceutical
Sciences, Guru Nanak Dev University, Amritsar 143005, India
| |
Collapse
|
22
|
Chaudhary N, Aparoy P. Deciphering the mechanism behind the varied binding activities of COXIBs through Molecular Dynamic Simulations, MM-PBSA binding energy calculations and per-residue energy decomposition studies. J Biomol Struct Dyn 2016; 35:868-882. [PMID: 26982261 DOI: 10.1080/07391102.2016.1165736] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
COX-2 is a well-known drug target in inflammatory disorders. COX-1/COX-2 selectivity of NSAIDs is crucial in assessing the gastrointestinal side effects associated with COX-1 inhibition. Celecoxib, rofecoxib, and valdecoxib are well-known specific COX-2 inhibiting drugs. Recently, polmacoxib, a COX-2/CA-II dual inhibitor has been approved by the Korean FDA. These COXIBs have similar structure with diverse activity range. Present study focuses on unraveling the mechanism behind the 10-fold difference in the activities of these sulfonamide-containing COXIBs. In order to obtain insights into their binding with COX-2 at molecular level, molecular dynamics simulations studies, and MM-PBSA approaches were employed. Further, per-residue decomposition of these energies led to the identification of crucial amino acids and interactions contributing to the differential binding of COXIBs. The results clearly indicated that Leu338, Ser339, Arg499, Ile503, Phe504, Val509, and Ser516 (Leu352, Ser353, Arg513, Ile517, Phe518, Val523, and Ser530 in PGHS-1 numbering) were imperative in determining the activity of these COXIBs. The binding energies and energy contribution of various residues were similar in all the three simulations. The results suggest that hydrogen bond interaction between the hydroxyl group of Ser516 and five-membered ring of diarylheterocycles augments the affinity in COXIBs. The SAR of the inhibitors studied and the per-residue energy decomposition values suggested the importance of Ser516. Additionally, the positive binding energy obtained with Arg106 explains the binding of COXIBs in hydrophobic channel deep in the COX-2 active site. The findings of the present work would aid in the development of potent COX-2 inhibitors.
Collapse
Affiliation(s)
- Neha Chaudhary
- a Centre for Computational Biology and Bioinformatics, School of Life Sciences , Central University of Himachal Pradesh , Dharamshala , Himachal Pradesh 176215 , India
| | - Polamarasetty Aparoy
- a Centre for Computational Biology and Bioinformatics, School of Life Sciences , Central University of Himachal Pradesh , Dharamshala , Himachal Pradesh 176215 , India
| |
Collapse
|
23
|
Saito MS, Lourenço AL, Dias LRS, Freitas ACC, Vitorino MI, Albuquerque MG, Rodrigues CR, Cabral LM, Dias EP, Castro HC, Satlher PC. Antiplatelet pyrazolopyridines derivatives: pharmacological, biochemical and toxicological characterization. J Enzyme Inhib Med Chem 2016; 31:1591-601. [DOI: 10.3109/14756366.2016.1158712] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Max Seidy Saito
- Laboratório de Antibióticos, Bioquímica, Ensino e Modelagem Molecular (LABiEMol) – Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil,
- Programa de Pós-Graduação em Patologia (PPG-UFF) – Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil,
| | - André Luiz Lourenço
- Laboratório de Antibióticos, Bioquímica, Ensino e Modelagem Molecular (LABiEMol) – Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil,
- Programa de Pós-Graduação em Patologia (PPG-UFF) – Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil,
| | - Luiza Rosaria Sousa Dias
- Laboratório de Química Medicinal (LQMed) – Faculdade de Farmácia, Universidade Federal Fluminense, Niterói, Brazil,
| | | | - Maíra Ingrid Vitorino
- Laboratório de Antibióticos, Bioquímica, Ensino e Modelagem Molecular (LABiEMol) – Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil,
| | | | | | - Lúcio Mendes Cabral
- Laboratório de Tecnologia Industrial Farmacêutica (LabTIF) – Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliane Pedra Dias
- Programa de Pós-Graduação em Patologia (PPG-UFF) – Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil,
| | - Helena Carla Castro
- Laboratório de Antibióticos, Bioquímica, Ensino e Modelagem Molecular (LABiEMol) – Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil,
- Programa de Pós-Graduação em Patologia (PPG-UFF) – Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil,
| | - Plínio Cunha Satlher
- Laboratório de Antibióticos, Bioquímica, Ensino e Modelagem Molecular (LABiEMol) – Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil,
- Laboratório de Tecnologia Industrial Farmacêutica (LabTIF) – Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Synthesis and in silico investigation of thiazoles bearing pyrazoles derivatives as anti-inflammatory agents. Comput Biol Chem 2016; 61:86-96. [PMID: 26844536 DOI: 10.1016/j.compbiolchem.2016.01.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 12/15/2015] [Accepted: 01/21/2016] [Indexed: 11/23/2022]
Abstract
Searching novel, safe and effective anti-inflammatory agents has remained an evolving research enquiry in the mainstream of inflammatory disorders. In the present investigation series of thiazoles bearing pyrazole as a possible pharmacophore were synthesized and assessed for their anti inflammatory activity using in vitro and in vivo methods. In order to decipher the possible anti-inflammatory mechanism of action of the synthesized compounds, cyclooxygenase I and II (COX-I and COX-II) inhibition assays were also carried out. The results obtained clearly focus the significance of compounds 5d, 5h and 5i as selective COX-II inhibitors. Moreover, compound 5h was also identified as a lead molecule for inhibition of the carrageenin induced rat paw edema in animal model studies. Molecular docking results revealed significant interactions of the test compounds with the active site of COX-II, which perhaps can be explored for design and development of novel COX-II selective anti-inflammatory agents.
Collapse
|
25
|
Characterisation of (R)-2-(2-Fluorobiphenyl-4-yl)-N-(3-Methylpyridin-2-yl)Propanamide as a Dual Fatty Acid Amide Hydrolase: Cyclooxygenase Inhibitor. PLoS One 2015; 10:e0139212. [PMID: 26406890 PMCID: PMC4583449 DOI: 10.1371/journal.pone.0139212] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/10/2015] [Indexed: 12/20/2022] Open
Abstract
Background Increased endocannabinoid tonus by dual-action fatty acid amide hydrolase (FAAH) and substrate selective cyclooxygenase (COX-2) inhibitors is a promising approach for pain-relief. One such compound with this profile is 2-(2-fluorobiphenyl-4-yl)-N-(3-methylpyridin-2-yl)propanamide (Flu-AM1). These activities are shown by Flu-AM1 racemate, but it is not known whether its two single enantiomers behave differently, as is the case towards COX-2 for the parent flurbiprofen enantiomers. Further, the effects of the compound upon COX-2-derived lipids in intact cells are not known. Methodology/Principal Findings COX inhibition was determined using an oxygraphic method with arachidonic acid and 2-arachidonoylglycerol (2-AG) as substrates. FAAH was assayed in mouse brain homogenates using anandamide (AEA) as substrate. Lipidomic analysis was conducted in unstimulated and lipopolysaccharide + interferon γ- stimulated RAW 264.7 macrophage cells. Both enantiomers inhibited COX-2 in a substrate-selective and time-dependent manner, with IC50 values in the absence of a preincubation phase of: (R)-Flu-AM1, COX-1 (arachidonic acid) 6 μM; COX-2 (arachidonic acid) 20 μM; COX-2 (2-AG) 1 μM; (S)-Flu-AM1, COX-1 (arachidonic acid) 3 μM; COX-2 (arachidonic acid) 10 μM; COX-2 (2-AG) 0.7 μM. The compounds showed no enantiomeric selectivity in their FAAH inhibitory properties. (R)-Flu-AM1 (10 μM) greatly inhibited the production of prostaglandin D2 and E2 in both unstimulated and lipopolysaccharide + interferon γ- stimulated RAW 264.7 macrophage cells. Levels of 2-AG were not affected either by (R)-Flu-AM1 or by 10 μM flurbiprofen, either alone or in combination with the FAAH inhibitor URB597 (1 μM). Conclusions/Significance Both enantiomers of Flu-AM1 are more potent inhibitors of 2-AG compared to arachidonic acid oxygenation by COX-2. Inhibition of COX in lipopolysaccharide + interferon γ- stimulated RAW 264.7 cells is insufficient to affect 2-AG levels despite the large induction of COX-2 produced by this treatment.
Collapse
|
26
|
Lei J, Zhou Y, Xie D, Zhang Y. Mechanistic insights into a classic wonder drug--aspirin. J Am Chem Soc 2015; 137:70-3. [PMID: 25514511 PMCID: PMC4309031 DOI: 10.1021/ja5112964] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Indexed: 01/24/2023]
Abstract
Aspirin, one of the oldest and most common anti-inflammatory agents, has recently been shown to reduce cancer risks. The principal pharmacological effects of aspirin are known to arise from its covalent modification of cyclooxygenase-2 (COX-2) through acetylation of Ser530, but the detailed mechanism of its biochemical action and specificity remains to be elucidated. In this work, we have filled this gap by employing a state-of-the-art computational approach, Born-Oppenheimer molecular dynamics simulations with ab initio quantum mechanical/molecular mechanical potential and umbrella sampling. Our studies have characterized a substrate-assisted inhibition mechanism for aspirin acetylating COX: it proceeds in two successive stages with a metastable tetrahedral intermediate, in which the carboxyl group of aspirin serves as the general base. The computational results confirmed that aspirin would be 10-100 times more potent against COX-1 than against COX-2, and revealed that this inhibition specificity between the two COX isoforms can be attributed mainly to the difference in kinetics rate of the covalent inhibition reaction, not the aspirin-binding step. The structural origin of this differential inhibition of the COX enzymes by aspirin has also been elucidated.
Collapse
Affiliation(s)
- Jinping Lei
- Institute
of Theoretical and Computational Chemistry, Laboratory of Mesoscopic
Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Yanzi Zhou
- Institute
of Theoretical and Computational Chemistry, Laboratory of Mesoscopic
Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Daiqian Xie
- Institute
of Theoretical and Computational Chemistry, Laboratory of Mesoscopic
Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
- Synergetic
Innovation Center of Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026,China
| | - Yingkai Zhang
- Department
of Chemistry, New York University, New York, New York 10003 United States
- NYU-ECNU
Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
| |
Collapse
|
27
|
Orlando BJ, Lucido MJ, Malkowski MG. The structure of ibuprofen bound to cyclooxygenase-2. J Struct Biol 2014; 189:62-6. [PMID: 25463020 DOI: 10.1016/j.jsb.2014.11.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/14/2014] [Accepted: 11/18/2014] [Indexed: 02/04/2023]
Abstract
The cyclooxygenases (COX-1 and COX-2) catalyze the rate-limiting step in the biosynthesis of prostaglandins, and are the pharmacological targets of non-steroidal anti-inflammatory drugs (NSAIDs) and COX-2 selective inhibitors (coxibs). Ibuprofen (IBP) is one of the most commonly available over-the-counter pharmaceuticals in the world. The anti-inflammatory and analgesic properties of IBP are thought to arise from inhibition of COX-2 rather than COX-1. While an X-ray crystal structure of IBP bound to COX-1 has been solved, no such structure exists for the cognate isoform COX-2. We have determined the crystal structure of muCOX-2 with a racemic mixture of (R/S)-IBP. Our structure reveals that only the S-isomer of IBP was bound, indicating that the S-isomer possesses higher affinity for COX-2 than the R-isomer. Mutational analysis of Arg-120 and Tyr-355 at the entrance of the cyclooxygenase channel confirmed their role in binding and inhibition of COX-2 by IBP. Our results provide the first atomic level detail of the interaction between IBP and COX-2.
Collapse
Affiliation(s)
- Benjamin J Orlando
- Department of Structural Biology, The State University of New York at Buffalo and the Hauptman-Woodward Medical Research Institute, 700 Ellicott Street, Buffalo, NY 14203, USA
| | - Michael J Lucido
- Department of Structural Biology, The State University of New York at Buffalo and the Hauptman-Woodward Medical Research Institute, 700 Ellicott Street, Buffalo, NY 14203, USA
| | - Michael G Malkowski
- Department of Structural Biology, The State University of New York at Buffalo and the Hauptman-Woodward Medical Research Institute, 700 Ellicott Street, Buffalo, NY 14203, USA.
| |
Collapse
|
28
|
Bacterial and algal orthologs of prostaglandin H₂synthase: novel insights into the evolution of an integral membrane protein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:83-94. [PMID: 25281773 DOI: 10.1016/j.bbamem.2014.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/09/2014] [Accepted: 09/23/2014] [Indexed: 01/01/2023]
Abstract
Prostaglandin H₂synthase (PGHS; EC 1.14.99.1), a bi-functional heme enzyme that contains cyclooxygenase and peroxidase activities, plays a central role in the inflammatory response, pain, and blood clotting in higher eukaryotes. In this review, we discuss the progenitors of the mammalian enzyme by using modern bioinformatics and homology modeling to draw comparisons between this well-studied system and its orthologs from algae and bacterial sources. A clade of bacterial and algal orthologs is described that have salient structural features distinct from eukaryotic counterparts, including the lack of a dimerization and EGF-like domains, the absence of gene duplicates, and minimal membrane-binding domains. The functional implications of shared and variant features are discussed.
Collapse
|
29
|
Zhang H, Fan J, Wang K, Li J, Wang C, Nie Y, Jiang T, Mu H, Peng X, Jiang K. Highly Sensitive Naphthalene-Based Two-Photon Fluorescent Probe for in Situ Real-Time Bioimaging of Ultratrace Cyclooxygenase-2 in Living Biosystems. Anal Chem 2014; 86:9131-8. [DOI: 10.1021/ac501944y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Hua Zhang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Jiangli Fan
- State
Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Hi-Tech Zone, Dalian 116024, P.R. China
| | - Kui Wang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Jing Li
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Caixia Wang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Yamin Nie
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Tao Jiang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| | - Huiying Mu
- State
Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Hi-Tech Zone, Dalian 116024, P.R. China
| | - Xiaojun Peng
- State
Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Hi-Tech Zone, Dalian 116024, P.R. China
| | - Kai Jiang
- School
of Chemistry and Chemical Engineering, Henan Normal University, 46
Jianshe Road, Muye Zone, Xinxiang 453007, P.R. China
| |
Collapse
|
30
|
Vitale P, Perrone MG, Malerba P, Lavecchia A, Scilimati A. Selective COX-1 inhibition as a target of theranostic novel diarylisoxazoles. Eur J Med Chem 2014; 74:606-18. [PMID: 24531199 DOI: 10.1016/j.ejmech.2013.12.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 12/05/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022]
Abstract
Cyclooxygenase(COX)-1 role in some diseases is increasingly studied. 3-(5-Chlorofuran-2-yl)-5-methyl-4-phenylisoxazole (P6), a highly selective cyclooxygenase-1 inhibitor, was used as a "lead" to design new isoxazoles (2a-m), differently selective towards COX-1. Those isoxazoles might be useful as novel theranostic agents and also to better clarify COX-1 role in the human physiology and diseases. 2a-m were prepared in fair to good yields developing suitable synthetic strategies. They were evaluated in vitro for their COX-inhibitory activity and selectivity. Structure-activity relationship studies of the novel set of diarylisoxazoles allowed to identify new key determinants for COX-1 selectivity, and to uncover compounds appropriate for a deep pharmacokinetic and pharmacodynamic investigation. 3-(5-Chlorofuran-2yl)-4-phenylisoxazol-5-amine (2f) was the most active compound of the series, its inhibitory activity was assessed in purified enzyme (COX-1 IC₅₀ = 1.1 μM; COX-2 IC₅₀ > 50 μM) and in the ovarian cancer cell line (OVCAR-3) expressing only COX-1 (IC₅₀ = 0.58 μM). Furthermore, the high inhibitory potency of 2f was rationalized through docking simulations in terms of interactions with a crystallographic model of the COX-1 binding site. We found critical interactions between the inhibitor and constriction residues R120 and Y355 at the base of the active site, as well as with S530 at the top of the side pocket.
Collapse
Affiliation(s)
- Paola Vitale
- Department of Pharmacy & Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| | - Maria Grazia Perrone
- Department of Pharmacy & Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| | - Paola Malerba
- Department of Pharmacy & Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy
| | - Antonio Lavecchia
- Department of Pharmacy, "Drug Discovery" Laboratory, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy.
| | - Antonio Scilimati
- Department of Pharmacy & Pharmaceutical Sciences, University of Bari "A. Moro", Via Orabona 4, 70125 Bari, Italy.
| |
Collapse
|
31
|
Zhang H, Fan J, Wang J, Dou B, Zhou F, Cao J, Qu J, Cao Z, Zhao W, Peng X. Fluorescence Discrimination of Cancer from Inflammation by Molecular Response to COX-2 Enzymes. J Am Chem Soc 2013; 135:17469-75. [DOI: 10.1021/ja4085308] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
| | | | | | | | - Fan Zhou
- Key Laboratories of Optoelectronic
Devices and Systems of Ministry of Education, Shenzhen University, Shenzhen 518060, People’s Republic of China
| | | | - Junle Qu
- Key Laboratories of Optoelectronic
Devices and Systems of Ministry of Education, Shenzhen University, Shenzhen 518060, People’s Republic of China
| | | | | | | |
Collapse
|
32
|
Windsor MA, Valk PL, Xu S, Banerjee S, Marnett LJ. Exploring the molecular determinants of substrate-selective inhibition of cyclooxygenase-2 by lumiracoxib. Bioorg Med Chem Lett 2013; 23:5860-4. [PMID: 24060487 DOI: 10.1016/j.bmcl.2013.08.097] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/22/2013] [Accepted: 08/26/2013] [Indexed: 01/22/2023]
Abstract
Lumiracoxib is a substrate-selective inhibitor of endocannabinoid oxygenation by cyclooxygenase-2 (COX-2). We assayed a series of lumiracoxib derivatives to identify the structural determinants of substrate-selective inhibition. The hydrogen-bonding potential of the substituents at the ortho positions of the aniline ring dictated the potency and substrate selectivity of the inhibitors. The presence of a 5'-methyl group on the phenylacetic acid ring increased the potency of molecules with a single ortho substituent. Des-fluorolumiracoxib (2) was the most potent and selective inhibitor of endocannabinoid oxygenation. The positioning of critical substituents in the binding site was identified from a 2.35Å crystal structure of lumiracoxib bound to COX-2.
Collapse
Affiliation(s)
- Matthew A Windsor
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Department of Biochemistry, Department of Chemistry, Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | | | | | | | | |
Collapse
|
33
|
Dong L, Sharma NP, Jurban BJ, Smith WL. Pre-existent asymmetry in the human cyclooxygenase-2 sequence homodimer. J Biol Chem 2013; 288:28641-55. [PMID: 23955344 DOI: 10.1074/jbc.m113.505503] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Prostaglandin endoperoxide H synthase-2 (PGHS-2), also known as cyclooxygenase-2 (COX-2), is a sequence homodimer. However, the enzyme exhibits half-site heme and inhibitor binding and functions as a conformational heterodimer having a catalytic subunit (Ecat) with heme bound and an allosteric subunit (Eallo) lacking heme. Some recombinant heterodimers composed of a COX-deficient mutant subunit and a native subunit (i.e. Mutant/Native PGHS-2) have COX activities similar to native PGHS-2. This suggests that the presence of heme plus substrate leads to the subunits becoming lodged in a semi-stable Eallo-mutant/Ecat-Native∼heme form during catalysis. We examined this concept using human PGHS-2 dimers composed of combinations of Y385F, R120Q, R120A, and S530A mutant or native subunits. With some heterodimers (e.g. Y385F/Native PGHS-2), heme binds with significantly higher affinity to the native subunit. This correlates with near native COX activity for the heterodimer. With other heterodimers (e.g. S530A/Native PGHS-2), heme binds with similar affinities to both subunits, and the COX activity approximates that expected for an enzyme in which each monomer contributes equally to the net COX activity. With or without heme, aspirin acetylates one-half of the subunits of the native PGHS-2 dimer, the Ecat subunits. Subunits having an S530A mutation are refractory to acetylation. Curiously, aspirin acetylates only one-quarter of the monomers of S530A/Native PGHS-2 with or without heme. This implies that there are comparable amounts of two noninterchangeable species of apoenzymes, Eallo-S530A/Ecat-Native and Eallo-Native/Ecat-S530A. These results suggest that native PGHS-2 assumes a reasonably stable, asymmetric Eallo/Ecat form during its folding and processing.
Collapse
Affiliation(s)
- Liang Dong
- From the Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | | | | | | |
Collapse
|
34
|
Vitale P, Tacconelli S, Perrone MG, Malerba P, Simone L, Scilimati A, Lavecchia A, Dovizio M, Marcantoni E, Bruno A, Patrignani P. Synthesis, Pharmacological Characterization, and Docking Analysis of a Novel Family of Diarylisoxazoles as Highly Selective Cyclooxygenase-1 (COX-1) Inhibitors. J Med Chem 2013; 56:4277-99. [DOI: 10.1021/jm301905a] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Paola Vitale
- Dipartimento di Farmacia-Scienze
del Farmaco, Università degli Studi di Bari “A. Moro”, Via Orabona 4, 70125 Bari, Italy
| | | | - Maria Grazia Perrone
- Dipartimento di Farmacia-Scienze
del Farmaco, Università degli Studi di Bari “A. Moro”, Via Orabona 4, 70125 Bari, Italy
| | - Paola Malerba
- Dipartimento di Farmacia-Scienze
del Farmaco, Università degli Studi di Bari “A. Moro”, Via Orabona 4, 70125 Bari, Italy
| | - Laura Simone
- Dipartimento di Farmacia-Scienze
del Farmaco, Università degli Studi di Bari “A. Moro”, Via Orabona 4, 70125 Bari, Italy
| | - Antonio Scilimati
- Dipartimento di Farmacia-Scienze
del Farmaco, Università degli Studi di Bari “A. Moro”, Via Orabona 4, 70125 Bari, Italy
| | - Antonio Lavecchia
- Dipartimento di Farmacia, “Drug
Discovery” Laboratory, Università di Napoli “Federico II”, Via D. Montesano 49, 80131
Napoli, Italy
| | | | | | | | | |
Collapse
|
35
|
Blobaum AL, Uddin MJ, Felts AS, Crews BC, Rouzer CA, Marnett LJ. The 2'-Trifluoromethyl Analogue of Indomethacin Is a Potent and Selective COX-2 Inhibitor. ACS Med Chem Lett 2013; 4:486-490. [PMID: 23687559 PMCID: PMC3654564 DOI: 10.1021/ml400066a] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 03/24/2013] [Indexed: 11/29/2022] Open
Abstract
![]()
Indomethacin is a potent, time-dependent,
nonselective inhibitor
of the cyclooxygenase enzymes (COX-1 and COX-2). Deletion of the 2′-methyl
group of indomethacin produces a weak, reversible COX inhibitor, leading
us to explore functionality at that position. Here, we report that
substitution of the 2′-methyl group of indomethacin with trifluoromethyl
produces CF3–indomethacin, a tight-binding inhibitor
with kinetic properties similar to those of indomethacin and unexpected
COX-2 selectivity (IC50 mCOX-2 = 267 nM; IC50 oCOX-1 > 100 μM). Studies with site-directed mutants reveal
that COX-2 selectivity results from insertion of the CF3 group into a small hydrophobic pocket formed by Ala-527, Val-349,
Ser-530, and Leu-531 and projection of the methoxy group toward a
side pocket bordered by Val-523. CF3–indomethacin
inhibited COX-2 activity in human head and neck squamous cell carcinoma
cells and exhibited in vivo anti-inflammatory activity in the carrageenan-induced
rat paw edema model with similar potency to that of indomethacin.
Collapse
Affiliation(s)
- Anna L. Blobaum
- The A. B.
Hancock Jr. Memorial Laboratory for Cancer
Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United
States
| | - Md. Jashim Uddin
- The A. B.
Hancock Jr. Memorial Laboratory for Cancer
Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United
States
| | - Andrew S. Felts
- The A. B.
Hancock Jr. Memorial Laboratory for Cancer
Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United
States
| | - Brenda C. Crews
- The A. B.
Hancock Jr. Memorial Laboratory for Cancer
Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United
States
| | - Carol A. Rouzer
- The A. B.
Hancock Jr. Memorial Laboratory for Cancer
Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United
States
| | - Lawrence J. Marnett
- The A. B.
Hancock Jr. Memorial Laboratory for Cancer
Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United
States
| |
Collapse
|
36
|
Varvas K, Kasvandik S, Hansen K, Järving I, Morell I, Samel N. Structural and catalytic insights into the algal prostaglandin H synthase reveal atypical features of the first non-animal cyclooxygenase. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:863-71. [DOI: 10.1016/j.bbalip.2012.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 11/06/2012] [Accepted: 11/27/2012] [Indexed: 10/27/2022]
|
37
|
Hirz T, Khalaf A, El-Hachem N, Mrad MF, Abdallah H, Créminon C, Grée R, Merhi RA, Habib A, Hachem A, Hamade E. New analogues of 13-hydroxyocatdecadienoic acid and 12-hydroxyeicosatetraenoic acid block human blood platelet aggregation and cyclooxygenase-1 activity. Chem Cent J 2012; 6:152. [PMID: 23228056 PMCID: PMC3582601 DOI: 10.1186/1752-153x-6-152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 12/03/2012] [Indexed: 12/05/2022] Open
Abstract
Background Thromboxane A2 is derived from arachidonic acid through the action of cyclooxygenases and thromboxane synthase. It is mainly formed in blood platelets upon activation and plays an important role in aggregation. Aspirin is effective in reducing the incidence of complications following acute coronary syndrome and stroke. The anti-thrombotic effect of aspirin is obtained through the irreversible inhibition of cyclooxygenases. Analogues of 12-hydroxyeicosatetraenoic acid and 13-hydroxyocatdecadienoic acid were shown previously to modulate platelet activation and to block thromboxane receptors. Results and discussion We synthesized 10 compounds based on the structures of analogues of 12-hydroxyeicosatetraenoic acid and 13-hydroxyocatdecadienoic acid and evaluated their effect on platelet aggregation triggered by arachidonic acid. The structure activity relationship was evaluated. Five compounds showed a significant inhibition of platelet aggregation and highlighted the importance of the lipidic hydrophobic hydrocarbon chain and the phenol group. Their IC50 ranged from 7.5 ± 0.8 to 14.2 ± 5.7 μM (Mean ± S.E.M.). All five compounds decreased platelet aggregation and thromboxane synthesis in response to collagen whereas no modification of platelet aggregation in response to thromboxane receptor agonist, U46619, was observed. Using COS-7 cells overexpressing human cyclooxygenase-1, we showed that these compounds are specific inhibitors of cyclooxygenase-1 with IC50 ranging from 1.3 to 12 μM. Docking observation of human recombinant cyclooxygenase-1 supported a role of the phenol group in the fitting of cyclooxygenase-1, most likely related to hydrogen bonding with the Tyr 355 of cyclooxygenase-1. Conclusions In conclusion, the compounds we synthesized at first based on the structures of analogues of 12 lipoxygenase metabolites showed a role of the phenol group in the anti-platelet and anti-cyclooxygenase-1 activities. These compounds mediate their effects via blockade of cyclooxygenase-1.
Collapse
Affiliation(s)
- Taghreed Hirz
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, AUB, Beirut, POBox 11-236, Lebanon.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Windsor MA, Hermanson DJ, Kingsley PJ, Xu S, Crews BC, Ho W, Keenan CM, Banerjee S, Sharkey KA, Marnett LJ. Substrate-Selective Inhibition of Cyclooxygenase-2: Development and Evaluation of Achiral Profen Probes. ACS Med Chem Lett 2012; 3:759-763. [PMID: 22984634 PMCID: PMC3441040 DOI: 10.1021/ml3001616] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/13/2012] [Indexed: 11/30/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) oxygenates arachidonic acid and the endocannabinoids 2-arachidonoylglycerol (2-AG) and arachidonoylethanolamide (AEA). We recently reported that (R)-profens selectively inhibit endocannabinoid oxygenation but not arachidonic acid oxygenation. In this work, we synthesized achiral derivatives of five profen scaffolds and evaluated them for substrate-selective inhibition using in vitro and cellular assays. The size of the substituents dictated the inhibitory strength of the analogs, with smaller substituents enabling greater potency but less selectivity. Inhibitors based on the flurbiprofen scaffold possessed the greatest potency and selectivity, with desmethylflurbiprofen (3a) exhibiting an IC(50) of 0.11 μM for inhibition of 2-AG oxygenation. The crystal structure of desmethylflurbiprofen complexed to mCOX-2 demonstrated a similar binding mode to other profens. Desmethylflurbiprofen exhibited a half-life in mice comparable to that of ibuprofen. The data presented suggest that achiral profens can act as lead molecules toward in vivo probes of substrate-selective COX-2 inhibition.
Collapse
Affiliation(s)
- Matthew A. Windsor
- A.B. Hancock Jr. Memorial Laboratory
for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt−Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee,
United States
| | - Daniel J. Hermanson
- A.B. Hancock Jr. Memorial Laboratory
for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt−Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee,
United States
| | - Philip J. Kingsley
- A.B. Hancock Jr. Memorial Laboratory
for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt−Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee,
United States
| | - Shu Xu
- A.B. Hancock Jr. Memorial Laboratory
for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt−Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee,
United States
| | - Brenda C. Crews
- A.B. Hancock Jr. Memorial Laboratory
for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt−Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee,
United States
| | - Winnie Ho
- Hotchkiss
Brain Institute and
Snyder Institute for Chronic Diseases, Department of Physiology and
Pharmacology, University of Calgary, Calgary,
Alberta, Canada
| | - Catherine M. Keenan
- Hotchkiss
Brain Institute and
Snyder Institute for Chronic Diseases, Department of Physiology and
Pharmacology, University of Calgary, Calgary,
Alberta, Canada
| | - Surajit Banerjee
- Northeastern
Collaborative Access
Team and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States
- Argonne National Laboratory, Argonne, Illinois, United States
| | - Keith A. Sharkey
- Hotchkiss
Brain Institute and
Snyder Institute for Chronic Diseases, Department of Physiology and
Pharmacology, University of Calgary, Calgary,
Alberta, Canada
| | - Lawrence J. Marnett
- A.B. Hancock Jr. Memorial Laboratory
for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology,
Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology,
and Vanderbilt−Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee,
United States
| |
Collapse
|
39
|
|
40
|
Cui PH, Rawling T, Bourget K, Kim T, Duke CC, Doddareddy MR, Hibbs DE, Zhou F, Tattam BN, Petrovic N, Murray M. Antiproliferative and Antimigratory Actions of Synthetic Long Chain n-3 Monounsaturated Fatty Acids in Breast Cancer Cells That Overexpress Cyclooxygenase-2. J Med Chem 2012; 55:7163-72. [DOI: 10.1021/jm300673z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nenad Petrovic
- Pharmacy and Medical Sciences, University of South Australia, GPO Box 2471, Adelaide
SA 5001, Australia
| | | |
Collapse
|
41
|
Estevão MS, Carvalho LCR, Freitas M, Gomes A, Viegas A, Manso J, Erhardt S, Fernandes E, Cabrita EJ, Marques MMB. Indole based cyclooxygenase inhibitors: synthesis, biological evaluation, docking and NMR screening. Eur J Med Chem 2012; 54:823-33. [PMID: 22796043 DOI: 10.1016/j.ejmech.2012.06.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 06/19/2012] [Accepted: 06/21/2012] [Indexed: 02/02/2023]
Abstract
The close structural similarity between the two cyclooxygenase (COXs) isoforms and the absence of selective inhibitors without side effects continues to stimulate the development of novel approaches towards selective anti-inflammatory drugs. In the present study a small library of new indolic compounds involving two different substitutions patterns at the indole scaffold was synthesized. In order to establish a relation between the spatial distribution of known functional groups related with inhibitory activity, two substitution patterns were explored: one with substituents at N-1, C-3, C-5 positions and another at C-2, C-3 and C5 positions. Accordingly, indole positions C-5, C-3 and N-1 were substituted with: sulfonamide or methylsulfone at C-5, p-halo-benzyl group at C-3, and an alkyl chain with a trifluoromethyl group at N-1. Alternatively, a p-halo-benzyl group was introduced at C-2, leaving the indolic nitrogen free. Inhibitory studies were performed and the activity results obtained against both COXs isoforms were rationalized based on docking and NMR studies. Docking studies show that dialkyation at C-2 and C-3 favors a binding with an orientation similar to that of the known selective inhibitor SC-558. From the tested compounds, this substitution pattern is correlated with the highest inhibitory activity and selectivity: 70% COX-2 inhibition at 50 μM, and low COX-1 inhibition (18 ± 9%). Additionally, Saturation Transfer Difference NMR experiments reveal different interaction patterns with both COXs isoforms that may be related with different orientations of the sulfonamide group in the binding pocket. Despite the moderated inhibitory activities found, this study represents an innovative approach towards COXs inhibitory activity rationalization and to the design of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Mónica S Estevão
- REQUIMTE-CQFB, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Monte de Caparica, 2829-516 Caparica, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Vecchio AJ, Orlando BJ, Nandagiri R, Malkowski MG. Investigating substrate promiscuity in cyclooxygenase-2: the role of Arg-120 and residues lining the hydrophobic groove. J Biol Chem 2012; 287:24619-30. [PMID: 22637474 DOI: 10.1074/jbc.m112.372243] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cyclooxygenases (COX-1 and COX-2) generate prostaglandin H(2) from arachidonic acid (AA). In its catalytically productive conformation, AA binds within the cyclooxygenase channel with its carboxylate near Arg-120 and Tyr-355 and ω-end located within a hydrophobic groove above Ser-530. Although AA is the preferred substrate for both isoforms, COX-2 can oxygenate a broad spectrum of substrates. Mutational analyses have established that an interaction of the carboxylate of AA with Arg-120 is required for high affinity binding by COX-1 but not COX-2, suggesting that hydrophobic interactions between the ω-end of substrates and cyclooxygenase channel residues play a significant role in COX-2-mediated oxygenation. We used structure-function analyses to investigate the role that Arg-120 and residues lining the hydrophobic groove play in the binding and oxygenation of substrates by murine (mu) COX-2. Mutations to individual amino acids within the hydrophobic groove exhibited decreased rates of oxygenation toward AA with little effect on binding. R120A muCOX-2 oxygenated 18-carbon ω-6 and ω-3 substrates albeit at reduced rates, indicating that an interaction with Arg-120 is not required for catalysis. Structural determinations of Co(3+)-protoporphyrin IX-reconstituted muCOX-2 with α-linolenic acid and G533V muCOX-2 with AA indicate that proper bisallylic carbon alignment is the major determinant for efficient substrate oxygenation by COX-2. Overall, these findings implicate Arg-120 and hydrophobic groove residues as determinants that govern proper alignment of the bisallylic carbon below Tyr-385 for catalysis in COX-2 and confirm nuances between COX isoforms that explain substrate promiscuity.
Collapse
Affiliation(s)
- Alex J Vecchio
- Hauptman-Woodward Medical Research Institute and Department of Structural Biology, The State University of New York, Buffalo, New York 14203, USA
| | | | | | | |
Collapse
|
43
|
Duggan KC, Hermanson DJ, Musee J, Prusakiewicz JJ, Scheib JL, Carter BD, Banerjee S, Oates JA, Marnett LJ. (R)-Profens are substrate-selective inhibitors of endocannabinoid oxygenation by COX-2. Nat Chem Biol 2011; 7:803-9. [PMID: 22053353 DOI: 10.1038/nchembio.663] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cyclooxygenase-2 (COX-2) catalyzes the oxygenation of arachidonic acid and the endocannabinoids 2-arachidonoylglycerol and arachidonoylethanolamide. Evaluation of a series of COX-2 inhibitors revealed that many weak competitive inhibitors of arachidonic acid oxygenation are potent inhibitors of endocannabinoid oxygenation. (R) enantiomers of ibuprofen, naproxen and flurbiprofen, which are considered to be inactive as COX-2 inhibitors, are potent 'substrate-selective inhibitors' of endocannabinoid oxygenation. Crystal structures of the COX-2–(R)-naproxen and COX-2–(R)-flurbiprofen complexes verified this unexpected binding and defined the orientation of the (R) enantiomers relative to (S) enantiomers. (R)-Profens selectively inhibited endocannabinoid oxygenation by lipopolysaccharide-stimulated dorsal root ganglion (DRG) cells. Substrate-selective inhibition provides new tools for investigating the role of COX-2 in endocannabinoid oxygenation and a possible explanation for the ability of (R)-profens to maintain endocannabinoid tone in models of neuropathic pain.
Collapse
Affiliation(s)
- Kelsey C Duggan
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mukherjee A, Angeles-Boza AM, Huff GS, Roth JP. Catalytic mechanism of a heme and tyrosyl radical-containing fatty acid α-(di)oxygenase. J Am Chem Soc 2010; 133:227-38. [PMID: 21166399 DOI: 10.1021/ja104180v] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The steady-state catalytic mechanism of a fatty acid α-(di)oxygenase is examined, revealing that a persistent tyrosyl radical (Tyr379(•)) effects O(2) insertion into C(α)-H bonds of fatty acids. The initiating C(α)-H homolysis step is characterized by apparent rate constants and deuterium kinetic isotope effects (KIEs) that increase hyperbolically upon raising the concentration of O(2). These results are consistent with H(•) tunneling, transitioning from a reversible to an irreversible regime. The limiting deuterium KIEs increase from ∼30 to 120 as the fatty acid chain is shortened from that of the native substrate. In addition, activation barriers increase in a manner that reflects decreased fatty acid binding affinities. Anaerobic isotope exchange experiments provide compelling evidence that Tyr379(•) initiates catalysis by H(•) abstraction. C(α)-H homolysis is kinetically driven by O(2) trapping of the α-carbon radical and reduction of a putative peroxyl radical intermediate to a 2(R)-hydroperoxide product. These findings add to a body of work which establishes large-scale hydrogen tunneling in proteins. This particular example is novel because it involves a protein-derived amino acid radical.
Collapse
Affiliation(s)
- Arnab Mukherjee
- Department of Chemistry, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | | | | | | |
Collapse
|
45
|
Duggan KC, Walters MJ, Musee J, Harp JM, Kiefer JR, Oates JA, Marnett LJ. Molecular basis for cyclooxygenase inhibition by the non-steroidal anti-inflammatory drug naproxen. J Biol Chem 2010; 285:34950-9. [PMID: 20810665 DOI: 10.1074/jbc.m110.162982] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Naproxen ((S)-6-methoxy-α-methyl-2-naphthaleneacetic acid) is a powerful non-selective non-steroidal anti-inflammatory drug that is extensively used as a prescription and over-the-counter medication. Naproxen exhibits gastrointestinal toxicity, but its cardiovascular toxicity may be reduced compared with other drugs in its class. Despite the fact that naproxen has been marketed for many years, the molecular basis of its interaction with cyclooxygenase (COX) enzymes is unknown. We performed a detailed study of naproxen-COX-2 interactions using site-directed mutagenesis, structure-activity analysis, and x-ray crystallography. The results indicate that each of the pendant groups of the naphthyl scaffold are essential for COX inhibition, and only minimal substitutions are tolerated. Mutation of Trp-387 to Phe significantly reduced inhibition by naproxen, a result that appears unique to this inhibitor. Substitution of S or CH(2) for the O atom of the p-methoxy group yielded analogs that were not affected by the W387F substitution and that exhibited increased COX-2 selectivity relative to naproxen. Crystallization and x-ray analysis yielded structures of COX-2 complexed to naproxen and its methylthio analog at 1.7 and 2.3 Å resolution, respectively. The combination of mutagenesis, structure analysis, and x-ray crystallography provided comprehensive information on the unique interactions responsible for naproxen binding to COX-2.
Collapse
Affiliation(s)
- Kelsey C Duggan
- AB Hancock Jr Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt Institute for Chemical Biology, the Center in Molecular Toxicology, Nashville, Tennessee 37232-0146
| | | | | | | | | | | | | |
Collapse
|
46
|
Uddin MJ, Crews BC, Blobaum AL, Kingsley PJ, Gorden DL, McIntyre JO, Matrisian LM, Subbaramaiah K, Dannenberg AJ, Piston DW, Marnett LJ. Selective visualization of cyclooxygenase-2 in inflammation and cancer by targeted fluorescent imaging agents. Cancer Res 2010; 70:3618-27. [PMID: 20430759 DOI: 10.1158/0008-5472.can-09-2664] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Effective diagnosis of inflammation and cancer by molecular imaging is challenging because of interference from nonselective accumulation of the contrast agents in normal tissues. Here, we report a series of novel fluorescence imaging agents that efficiently target cyclooxygenase-2 (COX-2), which is normally absent from cells, but is found at high levels in inflammatory lesions and in many premalignant and malignant tumors. After either i.p. or i.v. injection, these reagents become highly enriched in inflamed or tumor tissue compared with normal tissue and this accumulation provides sufficient signal for in vivo fluorescence imaging. Further, we show that only the intact parent compound is found in the region of interest. COX-2-specific delivery was unambiguously confirmed using animals bearing targeted deletions of COX-2 and by blocking the COX-2 active site with high-affinity inhibitors in both in vitro and in vivo models. Because of their high specificity, contrast, and detectability, these fluorocoxibs are ideal candidates for detection of inflammatory lesions or early-stage COX-2-expressing human cancers, such as those in the esophagus, oropharynx, and colon.
Collapse
Affiliation(s)
- Md Jashim Uddin
- AB Hancock, Jr Memorial Laboratory for Cancer Research, Department of Biochemistry, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Vecchio AJ, Simmons DM, Malkowski MG. Structural basis of fatty acid substrate binding to cyclooxygenase-2. J Biol Chem 2010; 285:22152-63. [PMID: 20463020 DOI: 10.1074/jbc.m110.119867] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The cyclooxygenases (COX-1 and COX-2) are membrane-associated heme-containing homodimers that generate prostaglandin H(2) from arachidonic acid (AA). Although AA is the preferred substrate, other fatty acids are oxygenated by these enzymes with varying efficiencies. We determined the crystal structures of AA, eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) bound to Co(3+)-protoporphyrin IX-reconstituted murine COX-2 to 2.1, 2.4, and 2.65 A, respectively. AA, EPA, and docosahexaenoic acid bind in different conformations in each monomer constituting the homodimer in their respective structures such that one monomer exhibits nonproductive binding and the other productive binding of the substrate in the cyclooxygenase channel. The interactions identified between protein and substrate when bound to COX-1 are conserved in our COX-2 structures, with the only notable difference being the lack of interaction of the carboxylate of AA and EPA with the side chain of Arg-120. Leu-531 exhibits a different side chain conformation when the nonproductive and productive binding modes of AA are compared. Unlike COX-1, mutating this residue to Ala, Phe, Pro, or Thr did not result in a significant loss of activity or substrate binding affinity. Determination of the L531F:AA crystal structure resulted in AA binding in the same global conformation in each monomer. We speculate that the mobility of the Leu-531 side chain increases the volume available at the opening of the cyclooxygenase channel and contributes to the observed ability of COX-2 to oxygenate a broad spectrum of fatty acid and fatty ester substrates.
Collapse
Affiliation(s)
- Alex J Vecchio
- Hauptman-Woodward Medical Research Institute, Buffalo, New York 14203, USA
| | | | | |
Collapse
|
48
|
Kleiman NS. Resistance or resistors? Has Ohm's law come to antiplatelet therapy? Am Heart J 2010; 159:713-715. [PMID: 20435177 DOI: 10.1016/j.ahj.2010.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 02/02/2010] [Indexed: 05/29/2023]
|
49
|
Sharma NP, Dong L, Yuan C, Noon KR, Smith WL. Asymmetric acetylation of the cyclooxygenase-2 homodimer by aspirin and its effects on the oxygenation of arachidonic, eicosapentaenoic, and docosahexaenoic acids. Mol Pharmacol 2010; 77:979-86. [PMID: 20194532 DOI: 10.1124/mol.109.063115] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prostaglandin endoperoxide H synthases (PGHS)-1 and -2, also called cyclooxygenases, convert arachidonic acid (AA) to prostaglandin H(2) (PGH(2)) in the committed step of prostaglandin biosynthesis. Both enzymes are homodimers, but the monomers often behave asymmetrically as conformational heterodimers during catalysis and inhibition. Here we report that aspirin maximally acetylates one monomer of human (hu) PGHS-2. The acetylated monomer of aspirin-treated huPGHS-2 forms 15-hydroperoxyeicosatetraenoic acid from AA, whereas the nonacetylated partner monomer forms mainly PGH(2) but only at 15 to 20% of the rate of native huPGHS-2. These latter conclusions are based on the findings that the nonsteroidal anti-inflammatory drug diclofenac binds a single monomer of native huPGHS-2, having an unmodified Ser530 to inhibit the enzyme, and that diclofenac inhibits PGH(2) but not 15-hydroperoxyeicosatraenoic acid formation by acetylated huPGHS-2. The 18R- and 17R-resolvins putatively involved in resolution of inflammation are reportedly formed via aspirin-acetylated PGHS-2 from eicosapentaenoic acid and docosahexaenoic acid, respectively, so we also characterized the oxygenation of these omega-3 fatty acids by aspirin-treated huPGHS-2. Our in vitro studies suggest that 18R- and 17R-resolvins could be formed only at low rates corresponding to less than 1 and 5%, respectively, of the rates of formation of PGH(2) by native PGHS-2.
Collapse
Affiliation(s)
- Narayan P Sharma
- Department of Biological Chemistry, University of Michigan Medical School, 1150 West Medical Center Drive, 5301 MSRB III, Ann Arbor, MI 48109-0606, USA
| | | | | | | | | |
Collapse
|
50
|
Genome wide analysis and comparative docking studies of new diaryl furan derivatives against human cyclooxygenase-2, lipoxygenase, thromboxane synthase and prostacyclin synthase enzymes involved in inflammatory pathway. J Mol Graph Model 2009; 28:313-29. [DOI: 10.1016/j.jmgm.2009.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 08/19/2009] [Accepted: 08/20/2009] [Indexed: 11/21/2022]
|