1
|
El-Miligy MMM, Al-Kubeisi AK, Nassra RA, El-Zemity SR, Hazzaa AA. Discovery of new thymol-3,4-disubstituted thiazole hybrids as dual COX-2/5-LOX inhibitors with in vivo proof. J Enzyme Inhib Med Chem 2024; 39:2309171. [PMID: 38291670 PMCID: PMC10833116 DOI: 10.1080/14756366.2024.2309171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/14/2024] [Indexed: 02/01/2024] Open
Abstract
New thymol-3,4-disubstitutedthiazole hybrids were synthesised as dual COX-2/5-LOX inhibitors. Compounds 6b, 6d, 6e, and 6f displayed in vitro inhibitory activity against COX-2 (IC50= 0.037, 0.042, 0.046, and 0.039 µM) nearly equal to celecoxib (IC50= 0.045 µM). 6b, 6d, and 6f showed SI (379, 341, and 374, respectively) higher than that of celecoxib (327). 6a-l elicited in vitro 5-LOX inhibitory activity higher than quercetin. 6a-f, 6i-l, 7a, and 7c possessed in vivo inhibition of formalin induced paw edoema higher than celecoxib. 6a, 6b, 6f, 6h-l, and 7b showed gastrointestinal safety profile as celecoxib and diclofenac sodium in the population of fasted rats. Induced fit docking and molecular dynamics simulation predicted good fitting of 6b and 6f without changing the packing and globularity of the apo protein. In conclusion, 6b and 6f achieved the target goal as multitarget inhibitors of inflammation.
Collapse
Affiliation(s)
- Mostafa M. M. El-Miligy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | - Rasha A. Nassra
- Medical Biochemistry Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Saad R. El-Zemity
- Department of Chemistry and Technology of Pesticides, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Aly A. Hazzaa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
El-Miligy MMM, Al-Kubeisi AK, Bekhit MG, El-Zemity SR, Nassra RA, Hazzaa AA. Towards safer anti-inflammatory therapy: synthesis of new thymol–pyrazole hybrids as dual COX-2/5-LOX inhibitors. J Enzyme Inhib Med Chem 2023; 38:294-308. [DOI: 10.1080/14756366.2022.2147164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Mostafa M. M. El-Miligy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | - Mohamed G. Bekhit
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Saad R. El-Zemity
- Department of Chemistry and Technology of Pesticides, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Rasha A. Nassra
- Medical Biochemistry Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Aly A. Hazzaa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
3
|
Di Micco S, Terracciano S, Pierri M, Cantone V, Liening S, König S, Garscha U, Hofstetter RK, Koeberle A, Werz O, Bruno I, Bifulco G. Identification of 2,4-Dinitro-Biphenyl-Based Compounds as MAPEG Inhibitors. ChemMedChem 2022; 17:e202200327. [PMID: 36111583 PMCID: PMC9827972 DOI: 10.1002/cmdc.202200327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/15/2022] [Indexed: 01/14/2023]
Abstract
We identified 2,4-dinitro-biphenyl-based compounds as new inhibitors of leukotriene C4 synthase (LTC4 S) and 5-lipoxygenase-activating protein (FLAP), both members of the "Membrane Associated Proteins in Eicosanoid and Glutathione metabolism" (MAPEG) family involved in the biosynthesis of pro-inflammatory eicosanoids. By molecular docking we evaluated the putative binding against the targets of interest, and by applying cell-free and cell-based assays we assessed the inhibition of LTC4 S and FLAP by the small molecules at low micromolar concentrations. The present results integrate the previously observed inhibitory profile of the tested compounds against another MAPEG member, i. e., microsomal prostaglandin E2 synthase (mPGES)-1, suggesting that the 2,4-dinitro-biphenyl scaffold is a suitable molecular platform for a multitargeting approach to modulate pro-inflammatory mediators in inflammation and cancer treatment.
Collapse
Affiliation(s)
- Simone Di Micco
- European Biomedical Research Institute of Salerno (EBRIS)Via Salvatore De Renzi 5084125SalernoItaly
| | - Stefania Terracciano
- Department of PharmacyUniversity of SalernoVia Giovanni Paolo II 13284084FiscianoSAItaly
| | - Martina Pierri
- Department of PharmacyUniversity of SalernoVia Giovanni Paolo II 13284084FiscianoSAItaly
| | - Vincenza Cantone
- Department of PharmacyUniversity of SalernoVia Giovanni Paolo II 13284084FiscianoSAItaly,Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich-Schiller-University JenaPhilosophenweg 147743JenaGermany
| | - Stefanie Liening
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich-Schiller-University JenaPhilosophenweg 147743JenaGermany
| | - Stefanie König
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich-Schiller-University JenaPhilosophenweg 147743JenaGermany
| | - Ulrike Garscha
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich-Schiller-University JenaPhilosophenweg 147743JenaGermany
| | - Robert Klaus Hofstetter
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich-Schiller-University JenaPhilosophenweg 147743JenaGermany
| | - Andreas Koeberle
- Michael Popp Research InstituteUniversity of InnsbruckMitterweg 246020InnsbruckAustria
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich-Schiller-University JenaPhilosophenweg 147743JenaGermany
| | - Ines Bruno
- Department of PharmacyUniversity of SalernoVia Giovanni Paolo II 13284084FiscianoSAItaly
| | - Giuseppe Bifulco
- Department of PharmacyUniversity of SalernoVia Giovanni Paolo II 13284084FiscianoSAItaly
| |
Collapse
|
4
|
Looking at NSAIDs from a historical perspective and their current status in drug repurposing for cancer treatment and prevention. J Cancer Res Clin Oncol 2022; 149:2095-2113. [PMID: 35876951 PMCID: PMC9310000 DOI: 10.1007/s00432-022-04187-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most frequently prescribed drug classes with wide therapeutic applications over the centuries. Starting from the use of salicylate-containing willow leaves to the recent rise and fall of highly selective cyclooxygenase-2 (COX-2) inhibitors and the latest dual-acting anti-inflammatory molecules, they have displayed a rapid and ongoing evolution. Despite the enormous advances in the last twenty years, investigators are still in search of the design and development of more potent and safer therapy against inflammatory conditions. This challenge has been increasingly attractive as the emergence of inflammation as a common seed and unifying mechanism for most chronic diseases. Indeed, this fact put the NSAIDs in the spotlight for repurposing against inflammation-related disorders. This review attempts to present a historical perspective on the evolution of NSAIDs, regarding their COX-dependent/independent mode of actions, structural and mechanism-based classifications, and adverse effects. Additionally, a systematic review of previous studies was carried out to show the current situation in drug repurposing, particularly in cancers associated with the GI tract such as gastric and colorectal carcinoma. In the case of non-GI-related cancers, preclinical studies elucidating the effects and modes of action were collected and summarized.
Collapse
|
5
|
Effects of nitro-butoxyl- and butyl-esters of non-steroidal anti-inflammatory drugs compared with parent compounds on the contractility of digital arterial smooth muscle from the fallow deer (Dama dama). Inflammopharmacology 2021; 29:1459-1473. [PMID: 34532846 PMCID: PMC8514390 DOI: 10.1007/s10787-021-00858-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/26/2021] [Indexed: 10/28/2022]
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs (NSAIDs) are a major cause of upper gastro-intestinal (GI) ulceration and bleeding as well as cardiovascular (CV) diseases (e.g., myocardial infarction and stroke). A feature common to both these adverse events is a variety of vascular reactions. One approach to overcome these side effects has been the development of nitric-oxide (NO)-donating NSAIDs. The NO is considered to overcome some of these vascular reactions caused by NSAIDs. Unfortunately, the NO-NSAIDs developed so far have not had the expected benefits compared with NSAIDs alone. OBJECTIVES Using in vitro preparations it is hoped to gain insight into the vascular and smooth muscle reactions induced by NO-NSAIDs compared with NSAIDs as a basis for improving the protective responses attributed to the NO-donating properties of these drugs. METHODS A range of NO-NSAIDs was synthesized based on the esterification of NSAIDs with the nitro-butoxylate as a prototype of an NO-donor. These compounds, as well as NO-donor agents and NSAIDS, were examined for their possible effects on isolated segments of digital arteries of fallow deer, which provide a robust model for determining the effects of vasodilator and vasoconstrictor activities, in comparison with those of standard pharmacological agents. RESULTS The NO-NSAIDs were found to antagonise the smooth muscle contractions produced by 5-hydroxytryptamine (serotonin, 5-HT). However, while almost all their parent NSAIDs had little or no effect, with the exception of the R-(-)-isomers of both ibuprofen and flurbiprofen, which caused vasodilatation, all the NO-NSAIDs tested antagonised the increase in tension produced by 5-HT. CONCLUSIONS R-(-)-ibuprofen and R-(-)-flurbiprofen, along with the nitro-butoxyl esters of the NSAIDs examined, produce relaxation of segments of deer digital artery smooth muscle in vitro. The evidence presented suggests that their mechanism involves the release of NO or its products.
Collapse
|
6
|
Mashayekhi-sardoo H, Razavi BM, Ekhtiari M, Kheradmand N, Imenshahidi M. Gastroprotective effects of both aqueous and ethanolic extracts of Lemon verbena leaves against indomethacin-induced gastric ulcer in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:1639-1646. [PMID: 33489039 PMCID: PMC7811806 DOI: 10.22038/ijbms.2020.44341.10377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/05/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Regarding Lemon verbena gastroprotective effects, we investigated the protective effects of Lemon verbena extracts on reducing gastric ulcer induced by indomethacin. MATERIALS AND METHODS Rats received aqueous and ethanolic extracts of Lemon verbena (50, 100, and 200 mg/kg), zileuton (100 mg/kg), montelukast (10 mg/kg), or 1% Tween 80 in presence or absence of indomethacin (100 mg/kg). RESULTS Indomethacin produced stomach ulcer and increased neutrophils percentage and MDA level compared with the control group (P<0.001). Co-administration of indomethacin and zileuton, montelukast and ethanolic (200 mg/kg) (P<0.001), aqueous extract (200 mg/kg) (P<0.05) reduced ulcer compared with the indomethacin group (P<0.001). Ethanolic extracts (100 and 200 mg/kg) and aqueous extract (200 mg/kg) reduced the MDA level (P<0.001). Ethanolic (50, 100, and 200 mg/kg) and aqueous extracts (200 mg/kg) significantly decreased neutrophils percentage compared with the indomethacin group (P<0.001). CONCLUSION Aqueous and particularly ethanolic extracts of Lemon verbena have protective effects on indomethacin-induced gastric ulcers.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-sardoo
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Targeted Drug Delivery Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Ekhtiari
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Kheradmand
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Imenshahidi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Gerstmeier J, Seegers J, Witt F, Waltenberger B, Temml V, Rollinger JM, Stuppner H, Koeberle A, Schuster D, Werz O. Ginkgolic Acid is a Multi-Target Inhibitor of Key Enzymes in Pro-Inflammatory Lipid Mediator Biosynthesis. Front Pharmacol 2019; 10:797. [PMID: 31379572 PMCID: PMC6650749 DOI: 10.3389/fphar.2019.00797] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction: Lipid mediators (LMs) comprise bioactive metabolites of polyunsaturated fatty acids, including pro-inflammatory prostaglandins (PGs), thromboxanes (TXs), and leukotrienes (LTs), as well as specialized pro-resolving mediators (SPMs). They are essentially biosynthesized via cyclooxygenase (COX) and lipoxygenase (LO) pathways in complex networks and regulate the progression as well as the resolution of inflammatory disorders including inflammation-triggered cancer. Ginkgolic acid (GA) is a phenolic acid contained in Ginkgo biloba L. with neuroprotective, antimicrobial, and antitumoral properties. Although LMs regulate microbial infections and tumor progression, whether GA affects LM biosynthesis is unknown and was investigated here in detail. Methods: Pharmacophore-based virtual screening was performed along with docking simulations. Activity assays were conducted for isolated human recombinant 5-LO, cytosolic phospholipase (PLA)2α, COX-2, and ovine COX-1. The activity of human mPGES-1 and thromboxane A2 synthase (TXAS) was determined in crude cellular fractions. Cellular LM formation was studied using human monocytes, neutrophils, platelets, and M1- and M2-like macrophages. LMs were identified after (ultra)high-performance liquid chromatography by UV detection or ESI-tandem mass spectrometry. Results: GA was identified as virtual hit in an mPGES-1 pharmacophore-based virtual screening. Cell-free assays revealed potent suppression of mPGES-1 activity (IC50 = 0.7 µM) that is fully reversible and essentially independent of the substrate concentration. Moreover, cell-free assays revealed COX-1 and TXAS as additional targets of GA with lower affinity (IC50 = 8.1 and 5.2 µM). Notably, 5-LO, the key enzyme in LT biosynthesis, was potently inhibited by GA (IC50 = 0.2 µM) in a reversible and substrate-independent manner. Docking simulations support the molecular interaction of GA with mPGES-1 and 5-LO and suggest concrete binding sites. Interestingly, interference of GA with mPGES-1, COX-1, TXAS, and 5-LO was evident also in intact cells with IC50 values of 2.1-3.8 µM; no radical scavenging or cytotoxic properties were obvious. Analysis of LM profiles from bacteria-stimulated human M1- and M2-like macrophages confirmed the multi-target features of GA and revealed LM redirection towards the formation of 12-/15-LO products including SPM. Conclusions: We reveal GA as potent multi-target inhibitor of key enzymes in the biosynthesis of pro-inflammatory LMs that contribute to the complex pharmacological and toxicological properties of GA.
Collapse
Affiliation(s)
- Jana Gerstmeier
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Julia Seegers
- Department of Pharmaceutical Analytics, Pharmaceutical Institute, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - Finja Witt
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Veronika Temml
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Judith M. Rollinger
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Daniela Schuster
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
8
|
Youssif BGM, Mohamed MFA, Al-Sanea MM, Moustafa AH, Abdelhamid AA, Gomaa HAM. Novel aryl carboximidamide and 3-aryl-1,2,4-oxadiazole analogues of naproxen as dual selective COX-2/15-LOX inhibitors: Design, synthesis and docking studies. Bioorg Chem 2019; 85:577-584. [PMID: 30878890 DOI: 10.1016/j.bioorg.2019.02.043] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/09/2019] [Accepted: 02/19/2019] [Indexed: 12/25/2022]
Abstract
A series of novel naproxen analogues containing 3-aryl-1,2,4-oxadiazoles moiety (4b-g) and their reaction intermediates aryl carboximidamides moiety (3b-g) was synthesized and evaluated in vitro as dual COXs/15-LOX inhibitors. Compounds 3b-g exhibited superior inhibitory activity than celecoxib as COX-2 inhibitors. Compounds 3b-d and 3g were the most potent COX-2 inhibitors with IC50 range of 6.4 - 8.13 nM and higher selectivity indexes (3b, SI = 26.19; 3c, SI = 13.73; 3d, SI = 29.27; 3g, SI = 18.00) comparing to celecoxib (IC50 = 42.60 nM, SI = 8.05). Regarding 15-LOX inhibitory activity, compounds belonging to aryl carboximidamide backbone 3b-e and 3g were the most potent with IC50 range of 1.77-4.91 nM comparing to meclofenamate sodium (IC50 = 5.64 µM). Data revealed that The levels of NO released by aryl carboximidamides 3b-g were more higher than 3-aryl-1,2,4-oxadiazole derivatives 4b-g, which correlated well with their COX-2 inhibitory activities.
Collapse
Affiliation(s)
- Bahaa G M Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Assiut, Egypt; Pharmaceutical Chemistry Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 2014, Saudi Arabia.
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, 82524 Sohag, Egypt.
| | - Mohammad M Al-Sanea
- Pharmaceutical Chemistry Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 2014, Saudi Arabia
| | - Amr H Moustafa
- Department of Chemistry, Faculty of Science, Sohag University, Sohag 82524, Egypt
| | - Antar A Abdelhamid
- Department of Chemistry, Faculty of Science, Sohag University, Sohag 82524, Egypt
| | - Hesham A M Gomaa
- Pharmacology Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 2014, Saudi Arabia; Biochemistry Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| |
Collapse
|
9
|
Gaba M, Singh S, Mohan C, Dhingra R, Chauhan M, Rana P, Dhingra N. Design, Synthesis and Pharmacological Evaluation of Gastro- Protective Anti-inflammatory Analgesic Agents based on Dual Oxidative Stress / Cyclooxygenase Inhibition. Antiinflamm Antiallergy Agents Med Chem 2019; 19:268-290. [PMID: 30914035 DOI: 10.2174/1871523018666190325155244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/12/2019] [Accepted: 03/20/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs (NSAIDs) derived local generation of reactive oxygen species (ROS) plays a crucial role in the formation of gastric ulceration. OBJECTIVE Therefore, anti-inflammatory analgesics with potent antioxidant activity could be a potential therapeutic strategy for the treatment of pain and inflammatory disorders without gastrointestinal (GI) side effects. METHODS In an effort to develop gastroprotective analgesic and anti-inflammatory agents, a series of 2-methylamino-substituted-1H-benzo[d] imidazol-1-yl) (phenyl) methanone derivatives were synthesized and evaluated in vitro for cyclooxygenase (COX) inhibition as well as anti-oxidant potential by the FRAP assay. The compounds with significant in vitro COX-1/COX-2 inhibitory activity and antioxidant activity were further screened in vivo for their anti-inflammatory and analgesic activities. Moreover, the ulcerogenic potential of test compounds was also studied. To gain insight into the plausible mode of interaction of compounds within the active sites of COX-1 and COX-2, molecular docking simulations were performed. RESULTS Among the various synthesized molecules, most of the compounds showed good cyclooxygenase inhibitory activity and efficient antioxidant activity in FRAP assay. After preliminary and indicative in vitro assays, three compounds exhibited most significant antiinflammatory and analgesic activity with better gastric tolerability during their in vivo evaluation. Ligand interaction studies indicated highest dock score -43.05 of 1,2- disubstituted benzimidazole derivatives in comparison to the reference ligand -30.70. Overall studies provided us (2-((4-methoxyphenylamino) methyl) -1h-benzo [d] imidazol- 1-yl) (phenyl) methanone as a lead with potent gastro-protective anti-inflammatory and analgesic activities that can be used for future research. CONCLUSION From the above results, it can be concluded that designing of multifunctional molecules with COX-1/COX-2 inhibitory and anti-oxidant activities could hold a great promise for further development of GI-safer NSAIDs.
Collapse
Affiliation(s)
- Monika Gaba
- Department of Pharmaceutical Sciences, ASBASJSM College of Pharmacy, Bela, Ropar, Punjab, India
| | - Sarbjot Singh
- Drug Discovery Research, Panacea Biotec Pvt. Ltd., Mohali, Punjab, India
| | - Chander Mohan
- Rayat-Bahra Institute of Pharmacy, Hoshiarpur, Punjab, India
| | - Richa Dhingra
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh-160014, India
| | - Monika Chauhan
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh-160014, India
| | - Priyanka Rana
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh-160014, India
| | - Neelima Dhingra
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh-160014, India
| |
Collapse
|
10
|
Novel benzoxanthene lignans that favorably modulate lipid mediator biosynthesis: A promising pharmacological strategy for anti-inflammatory therapy. Biochem Pharmacol 2019; 165:263-274. [PMID: 30836057 DOI: 10.1016/j.bcp.2019.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/01/2019] [Indexed: 02/08/2023]
Abstract
Lipid mediators (LM) encompass pro-inflammatory prostaglandins (PG) and leukotrienes (LT) but also specialized pro-resolving mediators (SPM) which display pivotal bioactivities in health and disease. Pharmacological intervention with inflammatory disorders such as osteoarthritis and rheumatoid arthritis commonly employs anti-inflammatory drugs that can suppress PG and LT formation, which however, possess limited effectiveness and side effects. Here, we report on the discovery and characterization of the two novel benzoxanthene lignans 1 and 2 that modulate select LM biosynthetic enzymes enabling the switch from pro-inflammatory LT to SPM biosynthesis as potential pharmacological strategy to intervene with inflammation. In cell-free assays, compound 1 and 2 inhibit microsomal prostaglandin E2 synthase-1 and leukotriene C4 synthase (IC50 ∼ 0.6-3.4 µM) and potently interfere with 5-lipoxygenase (5-LOX), the key enzyme in LT biosynthesis (IC50 = 0.04 and 0.09 µM). In human neutrophils, monocytes and M1 and M2 macrophages, compound 1 and 2 efficiently suppress LT biosynthesis (IC50 < 1 µM), accompanied by elevation of 15-LOX-derived LM including SPM. In zymosan-induced murine peritonitis, compound 1 and 2 ameliorated self-limited inflammation along with suppression of early LT formation and elevation of subsequent SPM biosynthesis in vivo. Together, these novel benzoxanthene lignans promote the LM class switch from pro-inflammatory towards pro-resolving LM to terminate inflammation, suggesting their suitability as novel leads for pharmacotherapy of arthritis and related inflammatory disorders.
Collapse
|
11
|
Werner M, Jordan PM, Romp E, Czapka A, Rao Z, Kretzer C, Koeberle A, Garscha U, Pace S, Claesson HE, Serhan CN, Werz O, Gerstmeier J. Targeting biosynthetic networks of the proinflammatory and proresolving lipid metabolome. FASEB J 2019; 33:6140-6153. [PMID: 30735438 DOI: 10.1096/fj.201802509r] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nonsteroidal anti-inflammatory drugs interfere with the metabolism of arachidonic acid to proinflammatory prostaglandins and leukotrienes by targeting cyclooxygenases (COXs), 5-lipoxygenase (LOX), or the 5-LOX-activating protein (FLAP). These and related enzymes act in conjunction with marked crosstalk within a complex lipid mediator (LM) network where also specialized proresolving LMs (SPMs) are formed. Here, we present how prominent LM pathways can be differentially modulated in human proinflammatory M1 and proresolving M2 macrophage phenotypes that, upon exposure to Escherichia coli, produce either abundant prostaglandins and leukotrienes (M1) or SPMs (M2). Targeted liquid chromatography-tandem mass spectrometry-based metabololipidomics was applied to analyze and quantify the specific LM profiles. Besides expected on-target actions, we found that: 1) COX or 15-LOX-1 inhibitors elevate inflammatory leukotriene levels, 2) FLAP and 5-LOX inhibitors reduce leukotrienes in M1 but less so in M2 macrophages, 3) zileuton blocks resolution-initiating SPM biosynthesis, whereas FLAP inhibition increases SPM levels, and 4) that the 15-LOX-1 inhibitor 3887 suppresses SPM formation in M2 macrophages. Conclusively, interference with discrete LM biosynthetic enzymes in different macrophage phenotypes considerably affects the LM metabolomes with potential consequences for inflammation-resolution pharmacotherapy. Our data may allow better appraisal of the therapeutic potential of these drugs to intervene with inflammatory disorders.-Werner, M., Jordan, P. M., Romp, E., Czapka, A., Rao, Z., Kretzer, C., Koeberle, A., Garscha, U., Pace, S., Claesson, H.-E., Serhan, C. N., Werz, O., Gerstmeier, J. Targeting biosynthetic networks of the proinflammatory and proresolving lipid metabolome.
Collapse
Affiliation(s)
- Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Erik Romp
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Anna Czapka
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Zhigang Rao
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Christian Kretzer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Ulrike Garscha
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Simona Pace
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Hans-Erik Claesson
- Division of Hematology, Department of Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Charles N Serhan
- Department of Anesthesia, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital-Harvard Medical School, Boston, Massachusetts, USA
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Jana Gerstmeier
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
12
|
Roos J, Peters M, Maucher IV, Kühn B, Fettel J, Hellmuth N, Brat C, Sommer B, Urbschat A, Piesche M, Vogel A, Proschak E, Blöcher R, Buscató E, Häfner AK, Matrone C, Werz O, Heidler J, Wittig I, Angioni C, Geisslinger G, Parnham MJ, Zacharowski K, Steinhilber D, Maier TJ. Drug-Mediated Intracellular Donation of Nitric Oxide Potently Inhibits 5-Lipoxygenase: A Possible Key to Future Antileukotriene Therapy. Antioxid Redox Signal 2018; 28:1265-1285. [PMID: 28699354 DOI: 10.1089/ars.2017.7155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
AIMS 5-Lipoxygenase (5-LO) is the key enzyme of leukotriene (LT) biosynthesis and is critically involved in a number of inflammatory diseases such as arthritis, gout, bronchial asthma, atherosclerosis, and cancer. Because 5-LO contains critical nucleophilic amino acids, which are sensitive to electrophilic modifications, we determined the consequences of a drug-mediated intracellular release of nitric oxide (NO) on 5-LO product formation by human granulocytes and on 5-LO-dependent pulmonary inflammation in vivo. RESULTS Clinically relevant concentrations of NO-releasing nonsteroidal anti-inflammatory drugs and other agents releasing NO intracellularly suppress 5-LO product synthesis in isolated human granulocytes via direct S-nitrosylation of 5-LO at the catalytically important cysteines 416 and 418. Furthermore, suppression of 5-LO product formation was observed in ionophore-stimulated human whole blood and in an animal model of pulmonary inflammation. INNOVATION Here, we report for the first time that drugs releasing NO intracellularly are efficient 5-LO inhibitors in vitro and in vivo at least equivalent to approved 5-LO inhibitors. CONCLUSION Our findings provide a novel mechanistic strategy for the development of a new class of drugs suppressing LT biosynthesis by site-directed nitrosylation. The results may also help to better understand the well-recognized anti-inflammatory clinically relevant actions of NO-releasing drugs. Furthermore, our study describes in detail a novel molecular mode of action of NO. Rebound Track: This work was rejected during standard peer review and rescued by Rebound Peer Review (Antioxid Redox Signal 16: 293-296, 2012) with the following serving as open reviewers: Angel Lanas, Hartmut Kühn, Joan Clària, Orina Belton. Antioxid. Redox Signal. 28, 1265-1285.
Collapse
Affiliation(s)
- Jessica Roos
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany .,2 Department for Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt , Frankfurt, Germany
| | - Marcus Peters
- 3 Department of Experimental Pneumology, Ruhr University Bochum , Bochum, Germany
| | - Isabelle V Maucher
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Benjamin Kühn
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Jasmin Fettel
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Nadine Hellmuth
- 2 Department for Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt , Frankfurt, Germany
| | - Camilla Brat
- 2 Department for Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt , Frankfurt, Germany
| | - Benita Sommer
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Anja Urbschat
- 4 Department of Urology and Pediatric Urology, University Hospital Marburg, Philipps-University Marburg , Marburg, Germany .,5 Department of Biomedicine, Aarhus University , Aarhus C, Denmark
| | - Matthias Piesche
- 5 Department of Biomedicine, Aarhus University , Aarhus C, Denmark .,6 Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule , Talca, Chile
| | - Anja Vogel
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany .,7 Project Group for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME , Frankfurt, Germany
| | - Ewgenij Proschak
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - René Blöcher
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Estella Buscató
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Ann-Kathrin Häfner
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Carmela Matrone
- 5 Department of Biomedicine, Aarhus University , Aarhus C, Denmark
| | - Oliver Werz
- 8 Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Friedrich Schiller University Jena , Jena, Germany
| | - Juliana Heidler
- 9 Department of Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe-University , Frankfurt, Germany
| | - Ilka Wittig
- 9 Department of Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe-University , Frankfurt, Germany
| | - Carlo Angioni
- 10 Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University , Frankfurt, Germany
| | - Gerd Geisslinger
- 7 Project Group for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME , Frankfurt, Germany .,10 Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University , Frankfurt, Germany
| | - Michael J Parnham
- 7 Project Group for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME , Frankfurt, Germany
| | - Kai Zacharowski
- 2 Department for Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt , Frankfurt, Germany
| | - Dieter Steinhilber
- 1 Institute of Pharmaceutical Chemistry, Goethe-University , Frankfurt, Germany
| | - Thorsten J Maier
- 2 Department for Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt , Frankfurt, Germany .,5 Department of Biomedicine, Aarhus University , Aarhus C, Denmark
| |
Collapse
|
13
|
Mohamed MS, Mansour YE, Amin HK, El-Araby ME. Molecular modelling insights into a physiologically favourable approach to eicosanoid biosynthesis inhibition through novel thieno[2,3-b]pyridine derivatives. J Enzyme Inhib Med Chem 2018; 33:755-767. [PMID: 29651867 PMCID: PMC6009894 DOI: 10.1080/14756366.2018.1457657] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In this research, we exploited derivatives of thieno[2,3-b]pyridine as dual inhibitors of the key enzymes in eicosanoid biosynthesis, cyclooxygenase (COX, subtypes 1 and 2) and 5-lipoxygensase (5-LOX). Testing these compounds in a rat paw oedema model revealed potency higher than ibuprofen. The most active compounds 7a, 7b, 8b, and 8c were screened against COX-1/2 and 5-LOX enzymes. Compound 7a was the most powerful inhibitor of 5-LOX with IC50 = 0.15 µM, while its p-chloro analogue 7b was more active against COX-2 (IC50 = 7.5 µM). The less desirable target COX-1 was inhibited more potently by 8c with IC50 = 7.7 µM. Surflex docking programme predicted that the more stable anti- conformer of compound (7a) formed a favourable complex with the active site of 5-LOX but not COX-1. This is in contrast to the binding mode of 8c, which resembles the syn-conformer of series 7 and binds favourably to COX-1.
Collapse
Affiliation(s)
- Mosaad S Mohamed
- a Department of Pharmaceutical Organic Chemistry , Faculty of Pharmacy, Helwan University , Cairo , Egypt
| | - Yara E Mansour
- a Department of Pharmaceutical Organic Chemistry , Faculty of Pharmacy, Helwan University , Cairo , Egypt
| | - Hatem K Amin
- b Department of Biochemistry and Molecular Biology , Faculty of Pharmacy, Helwan University , Cairo , Egypt
| | - Moustafa E El-Araby
- a Department of Pharmaceutical Organic Chemistry , Faculty of Pharmacy, Helwan University , Cairo , Egypt
| |
Collapse
|
14
|
Ghosh R, Alajbegovic A, Gomes AV. NSAIDs and Cardiovascular Diseases: Role of Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:536962. [PMID: 26457127 PMCID: PMC4592725 DOI: 10.1155/2015/536962] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/24/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most commonly used drugs worldwide. NSAIDs are used for a variety of conditions including pain, rheumatoid arthritis, and musculoskeletal disorders. The beneficial effects of NSAIDs in reducing or relieving pain are well established, and other benefits such as reducing inflammation and anticancer effects are also documented. The undesirable side effects of NSAIDs include ulcers, internal bleeding, kidney failure, and increased risk of heart attack and stroke. Some of these side effects may be due to the oxidative stress induced by NSAIDs in different tissues. NSAIDs have been shown to induce reactive oxygen species (ROS) in different cell types including cardiac and cardiovascular related cells. Increases in ROS result in increased levels of oxidized proteins which alters key intracellular signaling pathways. One of these key pathways is apoptosis which causes cell death when significantly activated. This review discusses the relationship between NSAIDs and cardiovascular diseases (CVD) and the role of NSAID-induced ROS in CVD.
Collapse
Affiliation(s)
- Rajeshwary Ghosh
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Azra Alajbegovic
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Aldrin V. Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA
| |
Collapse
|
15
|
Meng H, McClendon CL, Dai Z, Li K, Zhang X, He S, Shang E, Liu Y, Lai L. Discovery of Novel 15-Lipoxygenase Activators To Shift the Human Arachidonic Acid Metabolic Network toward Inflammation Resolution. J Med Chem 2015; 59:4202-9. [DOI: 10.1021/acs.jmedchem.5b01011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | - Christopher L. McClendon
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Shabbir A, Shahzad M, Ali A, Zia-ur-Rehman M. Anti-arthritic activity of N'-[(2,4-dihydroxyphenyl)methylidene]-2-(3,4-dimethyl-5,5-dioxidopyrazolo[4,3-c][1,2]benzothiazin-1(4H)-yl)acetohydrazide. Eur J Pharmacol 2014; 738:263-272. [PMID: 24943733 DOI: 10.1016/j.ejphar.2014.05.045] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 11/21/2022]
Abstract
Benzothiazine and pyrazole derivatives possess anti-inflammatory properties. Previously, synergism of both heterocyclic moieties into a single nucleus has shown to produce biologically active N'-arylmethylidene-2-(3,4-dimethyl-5,5-dioxidopyrazolo(4,3-c)(1,2)benzothiazin-2(4H)yl)acetohydrazides) compound. Present study investigates the anti-arthritic effect and possible mechanism of 2,4-dihydroxyphenyl derivative (DHP) in Freund's complete adjuvant-induced arthritic rat model. Ankle joint histopathology was performed with Hematoxylin & Eosin staining, while serum C-reactive protein (CRP) levels were measured by agglutination method. mRNA expression levels and protein levels of proinflammatory markers were measured by real time reverse transcription polymerase chain reaction and Enzyme linked immunosorbent assay (ELISA), respectively. in vitro Concanavalin A (ConA)-stimulated splenocyte proliferation was also measured by ELISA reader. DHP treatment reduced the macroscopic arthritic score, CRP levels, synovial inflammation, bone erosion and pannus formation. Levels of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), Prostaglandin-E2 (PGE2), and 5-lipoxygenase (5-LOX), were significantly attenuated by DHP. It also significantly decreased the levels of toll-like receptor 2, nuclear factor-kappaB (NF-ĸB), and tissue necrosis factor-α (TNF-α) and non-significantly elevated interleukin-4 (IL-4) levels. Piroxicam, used as reference drug, significantly reduced the levels of COX-1, COX-2, PGE2, NF-ĸB, and TNF-α, but did not show reduction in 5-LOX and toll-like receptor 2 levels. However piroxicam significantly enhanced the levels of IL-4. Both DHP and piroxicam suppressed ConA-stimulated splenocyte proliferation. DHP normalized all altered hematological markers and did not show any sign of hepatotoxicity or nephrotoxicity as determined by alanine transaminase, aspartate aminotransferase, urea, and creatinine levels. Results showed that DHP possesses significant anti-arthritic activity which may be attributed to its immunomodulatory and anti-inflammatory effects.
Collapse
Affiliation(s)
- Arham Shabbir
- Department of Pharmacology, University of Health Sciences, Lahore, Pakistan; Department of Pharmacy, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Muhammad Shahzad
- Department of Pharmacology, University of Health Sciences, Lahore, Pakistan.
| | - Akbar Ali
- Department of Pharmacology, University of Health Sciences, Lahore, Pakistan
| | - Muhammad Zia-ur-Rehman
- Applied Chemistry Research Centre, PCSIR Laboratories Complex, Ferozpur Road, Lahore 54600, Pakistan
| |
Collapse
|
17
|
Flavocoxid, a nutraceutical approach to blunt inflammatory conditions. Mediators Inflamm 2014; 2014:790851. [PMID: 25242871 PMCID: PMC4158568 DOI: 10.1155/2014/790851] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/06/2014] [Accepted: 08/06/2014] [Indexed: 12/15/2022] Open
Abstract
Flavonoids, from Scutellaria baicalensis (Chinese skullcap) and Acacia catechu (black catechu), have been shown to exert a variety of therapeutic effects, including anti-inflammatory, antiviral, antibacterial, and anticancer activities. Flavocoxid is a mixed extract containing baicalin and catechin and it acts as a dual balanced inhibitor of cyclooxygenase-1 (COX-1) and COX-2 peroxidase enzyme activities with a significant inhibition of 5-lipoxygenase (5-LOX) enzyme activity in vitro. Flavocoxid downregulates gene or protein expression of several inflammatory markers and exerts also strong antioxidant activity in several experimental models. Controlled clinical trials and a postmarketing study have clearly shown that flavocoxid is as effective as naproxen in managing the signs and symptoms of osteoarthritis of the knee and it has better upper gastrointestinal, renal, and respiratory safety profile than naproxen. Flavocoxid may therefore provide a potential therapeutic approach to the treatment of chronic inflammatory conditions.
Collapse
|
18
|
Dual and selective lipid inhibitors of cyclooxygenases and lipoxygenase: a molecular docking study. Med Chem Res 2014. [DOI: 10.1007/s00044-014-0919-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
19
|
Meng H, Liu Y, Zhai Y, Lai L. Optimization of 5-hydroxytryptamines as dual function inhibitors targeting phospholipase A2 and leukotriene A4 hydrolase. Eur J Med Chem 2013; 59:160-7. [DOI: 10.1016/j.ejmech.2012.10.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 10/26/2012] [Accepted: 10/27/2012] [Indexed: 12/13/2022]
|
20
|
KuKanich B, Bidgood T, Knesl O. Clinical pharmacology of nonsteroidal anti-inflammatory drugs in dogs. Vet Anaesth Analg 2012; 39:69-90. [PMID: 22151877 DOI: 10.1111/j.1467-2995.2011.00675.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVES To discuss the clinical pharmacology of currently licensed veterinary NSAIDs and to review gastrointestinal and renal adverse effects as well as drug-drug interactions that have been reported with these drugs. To review the use of NSAIDs in the peri-operative setting and their use in patients with osteoarthritis. To further review the reported effects of NSAIDs on canine articular cartilage and liver as well as the clinical relevance of a washout period. DATABASES USED PubMed, CAB abstracts and Google Scholar using dog, dogs, nonsteroidal anti-inflammatory drugs and NSAID(s) as keywords. CONCLUSIONS A good understanding of the mechanisms by which NSAIDs elicit their analgesic effect is essential in order to minimize adverse effects and drug-drug interactions. Cyclooxygenase (COX) is present in at least two active isoforms in the body and is the primary pharmacologic target of NSAIDs. Inhibition of COX is associated with the analgesic effects of NSAIDs. COX is present in the gastrointestinal tract and kidneys, along with other areas of the body, and is also the likely reason for many adverse effects including gastrointestinal and renal adverse effects. The newer veterinary approved NSAIDs have a lower frequency of gastrointestinal adverse effects in dogs compared to drugs such as aspirin, ketoprofen and flunixin, which may be due to differential effects on the COX isoforms. There are currently no published reports demonstrating that the newer NSAIDs are associated with fewer renal or hepatic adverse effects in dogs. NSAIDs remain the cornerstone of oral therapy for osteoarthritis unless contraindicated by intolerance, concurrent therapies or underlying medical conditions. NSAIDs are also effective and frequently used for the management of post-operative pain.
Collapse
Affiliation(s)
- Butch KuKanich
- Kansas State University College of Veterinary Medicine, Manhattan, KS, USA
| | | | | |
Collapse
|
21
|
Sampalis JS, Brownell LA. A randomized, double blind, placebo and active comparator controlled pilot study of UP446, a novel dual pathway inhibitor anti-inflammatory agent of botanical origin. Nutr J 2012; 11:21. [PMID: 22480204 PMCID: PMC3352039 DOI: 10.1186/1475-2891-11-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 04/05/2012] [Indexed: 11/11/2022] Open
Abstract
Background Current use of prescribed or over the counter non-steroidal anti-inflammatory drugs (NSAIDs) for pain and osteoarthritis (OA) have untoward gastrointestinal and cardiovascular related side effects, as a result the need for a safe and effective alternative has become unequivocally crucial. Method A randomized, double blind, placebo and active controlled pilot study of a novel dual pathway, COX1/2 and LOX, inhibitor anti-inflammatory agent of botanical origin, UP446 was conducted. Sixty subjects (age 40-75) with symptomatic OA of the hip or knee were assigned to 4 treatment groups (n = 15); Group A0 (Placebo, CMC capsule), Group A1 (UP446 250 mg/day), Group A2 (UP446 500 mg/day) and Group A3 (Celecoxib, 200 mg/day). MOS-SF-36 and Western Ontario and McMaster University Osteoarthritis Index (WOMAC) data were collected at baseline and after 30, 60 and 90 days of treatment as a measure of efficacy. Erythrocyte sedimentation rate, C-reactive protein, plasma thrombin time (PTT), fructosamine, Hematology, clinical chemistry and fecal occult blood were monitored for safety. Results Statistically significant decrease in WOMAC pain score were observed for Group A1 at day 90, Group A2 at 30 and 90 days and Group A3 at 60 and 90 days. Statistically significant decrease in WOMAC stiffness score were observed for Group A1 and Group A2 at 30, 60 and 90 days; but not for Group A0 and Group A3. The mean change in WOMAC functional impairment scores were statistically significant for Group A1 and Group A2 respectively at 30 days (p = 0.006 and p = 0.006), at 60 days (p = 0.016 and p = 0.002) and at 90 days (p = 0.018 and p = 0.002), these changes were not significant for Group A0 and Group A3. Based on MOS -SF-36 questionnaires, statistically significant improvements in physical function, endurance and mental health scores were observed for all active treatment groups compared to placebo. No significant changes suggestive of toxicity in routine hematologies, serum chemistries, liver enzymes or PTT were noted in any of the treatment groups. Conclusion Based on current findings UP446 is safe and efficacious alternative to established anti-inflammatory medications for alleviating OA symptoms as measured by the WOMAC Index.
Collapse
|
22
|
Cuniberti B, Odore R, Barbero R, Cagnardi P, Badino P, Girardi C, Re G. In vitro and ex vivo pharmacodynamics of selected non-steroidal anti-inflammatory drugs in equine whole blood. Vet J 2012; 191:327-33. [DOI: 10.1016/j.tvjl.2011.03.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 03/15/2011] [Accepted: 03/20/2011] [Indexed: 01/27/2023]
|
23
|
Burnett BP, Levy RM. 5-Lipoxygenase metabolic contributions to NSAID-induced organ toxicity. Adv Ther 2012; 29:79-98. [PMID: 22351432 DOI: 10.1007/s12325-011-0100-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Indexed: 01/01/2023]
Abstract
Cyclooxygenase (COX)-1, COX-2, and 5-lipoxygenase (5-LOX) enzymes produce effectors of pain and inflammation in osteoarthritis (OA) and many other diseases. All three enzymes play a key role in the metabolism of arachidonic acid (AA) to inflammatory fatty acids, which contribute to the deterioration of cartilage. AA is derived from both phospholipase A(2) (PLA(2)) conversion of cell membrane phospholipids and dietary consumption of omega-6 fatty acids. Nonsteroidal antiinflammatory drugs (NSAIDs) inhibit the COX enzymes, but show no anti-5-LOX activity to prevent the formation of leukotrienes (LTs). Cysteinyl LTs, such as LTC(4), LTD(4), LTE(4), and leukoattractive LTB(4) accumulate in several organs of mammals in response to NSAID consumption. Elevated 5-LOX-mediated AA metabolism may contribute to the side-effect profile observed for NSAIDs in OA. Current therapeutics under development, so-called "dual inhibitors" of COX and 5-LOX, show improved side-effect profiles and may represent a new option in the management of OA.
Collapse
Affiliation(s)
- Bruce P Burnett
- Department of Medical Education and Scientific Affairs, Primus Pharmaceuticals, Inc., Scottsdale, Arizona, USA.
| | | |
Collapse
|
24
|
Giorgi M, Cuniberti B, Ye G, Barbero R, Sgorbini M, Vercelli C, Corazza M, Re G. Oral administration of tepoxalin in the horse: A PK/PD study. Vet J 2011; 190:143-9. [DOI: 10.1016/j.tvjl.2010.09.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 09/03/2010] [Accepted: 09/19/2010] [Indexed: 10/18/2022]
|
25
|
Flavocoxid inhibits phospholipase A2, peroxidase moieties of the cyclooxygenases (COX), and 5-lipoxygenase, modifies COX-2 gene expression, and acts as an antioxidant. Mediators Inflamm 2011; 2011:385780. [PMID: 21765617 PMCID: PMC3134205 DOI: 10.1155/2011/385780] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 03/11/2011] [Indexed: 12/26/2022] Open
Abstract
The multiple mechanisms of action for flavocoxid relating to arachidonic acid (AA) formation and metabolism were studied in vitro. Flavocoxid titrated into rat peritoneal macrophage cultures inhibited cellular phospholipase A2 (PLA(2)) (IC(50) = 60 μg/mL). In in vitro enzyme assays, flavocoxid showed little anti-cyclooxygenase (CO) activity on COX-1/-2 enzymes, but inhibited the COX-1 (IC(50) = 12.3) and COX-2 (IC(50) = 11.3 μg/mL) peroxidase (PO) moieties as well as 5-lipoxygenase (5-LOX) (IC(50) = 110 μg/mL). No detectable 5-LOX inhibition was found for multiple traditional and COX-2 selective NSAIDs. Flavocoxid also exhibited strong and varied antioxidant capacities in vitro and decreased nitrite levels (IC(50) = 38 μg/mL) in rat peritoneal macrophages. Finally, in contrast to celecoxib and ibuprofen, which upregulated the cox-2 gene, flavocoxid strongly decreased expression. This work suggests that clinically favourable effects of flavocoxid for management of osteoarthritis (OA) are achieved by simultaneous modification of multiple molecular pathways relating to AA metabolism, oxidative induction of inflammation, and neutralization of reactive oxygen species (ROS).
Collapse
|
26
|
Polito F, Bitto A, Irrera N, Squadrito F, Fazzari C, Minutoli L, Altavilla D. Flavocoxid, a dual inhibitor of cyclooxygenase-2 and 5-lipoxygenase, reduces pancreatic damage in an experimental model of acute pancreatitis. Br J Pharmacol 2011; 161:1002-11. [PMID: 20977452 DOI: 10.1111/j.1476-5381.2010.00933.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Acute pancreatitis is an autodigestive process resulting in acute inflammation of the pancreas. Accumulating evidence indicates the essential contribution of cyclooxygenase (COX)-2 and 5-lipoxygenase (5-LOX) to acute pancreatitis. We studied the effects of flavocoxid, a plant-derived dual inhibitor of COX-2 and 5-LOX, in a model of caerulein (CER)-induced acute pancreatitis. EXPERIMENTAL APPROACH Rats were given CER (80 µg·kg⁻¹ for each of four injections at hourly intervals) or vehicle (Sham-CER). Animals were then randomized to receive flavocoxid (20 mg·kg⁻¹ i.p.) or vehicle, 30 min after the first CER injection. Two hours after the last CER injection, we evaluated damage to the pancreas by histological methods; serum levels of amylase, lipase, leukotriene (LT)B₄ and prostaglandin (PG)E₂ ; pancreatic expression of COX-2 and 5-LOX and tumour necrosis factor-α (TNF-α) gene expression by real-time polymerase chain reaction. KEY RESULTS Caerulein induced inflammatory changes in the pancreas and raised values of the other variables measured. In CER-treated animals, but not in those given saline, flavocoxid inhibited COX-2 and 5-LOX expression, reduced serum levels of lipase and amylase and the degree of pancreatic oedema. Treatment with flavocoxid blunted the increased pancreatic TNF-α mRNA expression, serum leukotriene B₄ and prostaglandin E₂ levels, and protected against histological damage in terms of vacuolization and leukocyte infiltration. CONCLUSIONS AND IMPLICATIONS Our results confirm the key role of both COX-2 and 5-LOX in the inflammatory response to acute pancreatitis. Flavocoxid may provide a potential therapeutic approach to the treatment of patients at high risk of developing this life-threatening condition.
Collapse
Affiliation(s)
- F Polito
- Department of Clinical and Experimental Medicine and Pharmacology, Section of Pharmacology, University of Messina, Messina, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Steinbrink SD, Pergola C, Bühring U, George S, Metzner J, Fischer AS, Häfner AK, Wisniewska JM, Geisslinger G, Werz O, Steinhilber D, Maier TJ. Sulindac sulfide suppresses 5-lipoxygenase at clinically relevant concentrations. Cell Mol Life Sci 2010; 67:797-806. [PMID: 20091083 PMCID: PMC11115735 DOI: 10.1007/s00018-009-0206-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2009] [Revised: 10/25/2009] [Accepted: 11/06/2009] [Indexed: 11/30/2022]
Abstract
Sulindac is a non-selective inhibitor of cyclooxygenases (COX) used to treat inflammation and pain. Additionally, non-COX targets may account for the drug's chemo-preventive efficacy against colorectal cancer and reduced gastrointestinal toxicity. Here, we demonstrate that the pharmacologically active metabolite of sulindac, sulindac sulfide (SSi), targets 5-lipoxygenase (5-LO), the key enzyme in the biosynthesis of proinflammatory leukotrienes (LTs). SSi inhibited 5-LO in ionophore A23187- and LPS/fMLP-stimulated human polymorphonuclear leukocytes (IC(50) approximately 8-10 microM). Importantly, SSi efficiently suppressed 5-LO in human whole blood at clinically relevant plasma levels (IC(50) = 18.7 microM). SSi was 5-LO-selective as no inhibition of related lipoxygenases (12-LO, 15-LO) was observed. The sulindac prodrug and the other metabolite, sulindac sulfone (SSo), failed to inhibit 5-LO. Mechanistic analysis demonstrated that SSi directly suppresses 5-LO with an IC(50) of 20 muM. Together, these findings may provide a novel molecular basis to explain the COX-independent pharmacological effects of sulindac under therapy.
Collapse
Affiliation(s)
- Svenja D. Steinbrink
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| | - Carlo Pergola
- Department of Pharmaceutical Analytics, Pharmaceutical Institute, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Ulrike Bühring
- Department of Pharmaceutical Analytics, Pharmaceutical Institute, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Sven George
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| | - Julia Metzner
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| | - Astrid S. Fischer
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| | - Ann-Kathrin Häfner
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| | - Joanna M. Wisniewska
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| | - Gerd Geisslinger
- Pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt/Main, Germany
| | - Oliver Werz
- Department of Pharmaceutical Analytics, Pharmaceutical Institute, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| | - Thorsten J. Maier
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt/Main, Germany
| |
Collapse
|
28
|
Williams O, Houseman BT, Kunkel EJ, Aizenstein B, Hoffman R, Knight ZA, Shokat KM. Discovery of dual inhibitors of the immune cell PI3Ks p110delta and p110gamma: a prototype for new anti-inflammatory drugs. CHEMISTRY & BIOLOGY 2010; 17:123-34. [PMID: 20189103 PMCID: PMC2858875 DOI: 10.1016/j.chembiol.2010.01.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/19/2009] [Accepted: 01/04/2010] [Indexed: 01/10/2023]
Abstract
PI3Kdelta and PI3Kgamma regulate immune cell signaling, while the related PI3Kalpha and PI3Kbeta regulate cell survival and metabolism. Selective inhibitors of PI3Kdelta/gamma represent a potential class of anti-inflammatory agents lacking the antiproliferative effects associated with PI3Kalpha/beta inhibition. Here we report the discovery of PI3Kdelta/gamma inhibitors that display up to 1000-fold selectivity over PI3Kalpha/beta and evaluate these compounds in a high-content inflammation assay using mixtures of primary human cells. We find selective inhibition of only PI3Kdelta is weakly anti-inflammatory, but PI3Kdelta/gamma inhibitors show superior inflammatory marker suppression through suppression of lipopolysaccharide-induced TNFalpha production and T cell activation. Moreover, PI3Kdelta/gamma inhibition yields an anti-inflammatory signature distinct from pan-PI3K inhibition and known anti-inflammatory drugs, yet bears striking similarities to glucocorticoid receptor agonists. These results highlight the potential of selectively designing drugs that target kinases with shared biological function.
Collapse
Affiliation(s)
- Olusegun Williams
- Graduate Program in Chemistry and Chemical Biology, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, USA
| | - Benjamin T. Houseman
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 600 16 St., San Francisco, CA 94158
| | - Eric J. Kunkel
- BioSeek, Inc., 310 Utah Ave., Suite 100, South San Francisco, CA 94080
| | - Brian Aizenstein
- Invitrogen Corporation, 501 Charmany Drive, Madison, Wisconsin 53719, USA
| | - Randy Hoffman
- Invitrogen Corporation, 501 Charmany Drive, Madison, Wisconsin 53719, USA
| | - Zachary A. Knight
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 600 16 St., San Francisco, CA 94158
| | - Kevan M. Shokat
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 600 16 St., San Francisco, CA 94158
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
29
|
Discovery of 3-(4-bromophenyl)-6-nitrobenzo[1.3.2]dithiazolium ylide 1,1-dioxide as a novel dual cyclooxygenase/5-lipoxygenase inhibitor that also inhibits tumor necrosis factor-α production. Bioorg Med Chem 2010; 18:597-604. [DOI: 10.1016/j.bmc.2009.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2009] [Revised: 11/26/2009] [Accepted: 12/02/2009] [Indexed: 11/22/2022]
|
30
|
Kwak HJ, Park KM, Choi HE, Lim HJ, Park JH, Park HY. The cardioprotective effects of zileuton, a 5-lipoxygenase inhibitor, are mediated by COX-2 via activation of PKCδ. Cell Signal 2010; 22:80-7. [DOI: 10.1016/j.cellsig.2009.09.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 09/12/2009] [Accepted: 09/14/2009] [Indexed: 10/20/2022]
|
31
|
Horvath-Ungerboeck C, Thoday KL, Shaw DJ, van den Broek AHM. Tepoxalin reduces pruritus and modified CADESI-01 scores in dogs with atopic dermatitis: a prospective, randomized, double-blinded, placebo-controlled, cross-over study. Vet Dermatol 2009; 20:233-42. [DOI: 10.1111/j.1365-3164.2009.00739.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Goodman L, Coles TB, Budsberg S. Leukotriene inhibition in small animal medicine. J Vet Pharmacol Ther 2009; 31:387-98. [PMID: 19000257 DOI: 10.1111/j.1365-2885.2008.00963.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Leukotrienes are important mediators of inflammatory and allergic conditions in people and are suspected to play an important role in tumorigenesis and tumor growth of several different tumor types. Based on this, researchers are making great progress in identifying novel pharmacologic targets for several human diseases. Leukotriene inhibition has resulted in therapeutic benefit in clinical trials involving people with osteoarthritis, allergic asthma, and atopic dermatitis. Despite this progress and the possibility that leukotriene inhibition may also play an important therapeutic role in veterinary patients, parallel advances have not yet been made in veterinary medicine. This article summarizes leukotriene function and synthesis. It also reviews the published literature regarding potential therapeutic applications of leukotriene inhibition in both human and veterinary medicine, focusing primarily on osteoarthritis, NSAID induced gastrointestinal mucosal damage, allergic asthma, atopic dermatitis, and cancer.
Collapse
Affiliation(s)
- L Goodman
- Small Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, Athens, GA 30602, USA.
| | | | | |
Collapse
|
33
|
Yang K, Bai H, Ouyang Q, Lai L, Tang C. Finding multiple target optimal intervention in disease-related molecular network. Mol Syst Biol 2008; 4:228. [PMID: 18985027 PMCID: PMC2673713 DOI: 10.1038/msb.2008.60] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 09/14/2008] [Indexed: 11/09/2022] Open
Abstract
Drugs against multiple targets may overcome the many limitations of single targets and achieve a more effective and safer control of the disease. Numerous high-throughput experiments have been performed in this emerging field. However, systematic identification of multiple drug targets and their best intervention requires knowledge of the underlying disease network and calls for innovative computational methods that exploit the network structure and dynamics. Here, we develop a robust computational algorithm for finding multiple target optimal intervention (MTOI) solutions in a disease network. MTOI identifies potential drug targets and suggests optimal combinations of the target intervention that best restore the network to a normal state, which can be customer designed. We applied MTOI to an inflammation-related network. The well-known side effects of the traditional non-steriodal anti-inflammatory drugs and the recently recalled Vioxx were correctly accounted for in our network model. A number of promising MTOI solutions were found to be both effective and safer.
Collapse
Affiliation(s)
- Kun Yang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
34
|
Maxis K, Delalandre A, Martel-Pelletier J, Pelletier JP, Duval N, Lajeunesse D. The shunt from the cyclooxygenase to lipoxygenase pathway in human osteoarthritic subchondral osteoblasts is linked with a variable expression of the 5-lipoxygenase-activating protein. Arthritis Res Ther 2007; 8:R181. [PMID: 17156456 PMCID: PMC1794527 DOI: 10.1186/ar2092] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 11/24/2006] [Accepted: 12/08/2006] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is characterized by articular cartilage degradation and hypertrophic bone changes with osteophyte formation and abnormal bone remodeling. Two groups of OA patients were identified via the production of variable and opposite levels of prostaglandin E2 (PGE2) or leukotriene B4 (LTB4) by subchondral osteoblasts, PGE2 levels discriminating between low and high subgroups. We studied whether the expression of 5-lipoxygenase (5-LO) or 5-LO-activating protein (FLAP) is responsible for the shunt from prostaglandins to leukotrienes. FLAP mRNA levels varied in low and high OA groups compared with normal, whereas mRNA levels of 5-LO were similar in all osteoblasts. Selective inhibition of cyclooxygenase-2 (COX-2) with NS-398-stimulated FLAP expression in the high OA osteoblasts subgroup, whereas it was without effect in the low OA osteoblasts subgroup. The addition of PGE2 to the low OA osteoblasts subgroup decreased FLAP expression but failed to affect it in the high OA osteoblasts subgroup. LTB4 levels in OA osteoblasts were stimulated about twofold by 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) plus transforming growth factor-β (TGF-β), a situation corresponding to their effect on FLAP mRNA levels. Treatments with 1,25(OH)2D3 and TGF-β also modulated PGE2 production. TGF-β stimulated PGE2 production in both OA osteoblast groups, whereas 1,25(OH)2D3 alone had a limited effect but decreased the effect of TGF-β in the low OA osteoblasts subgroup. This modulation of PGE2 production was mirrored by the synthesis of COX-2. IL-18 levels were only slightly increased in a subgroup of OA osteoblasts compared with normal; however, no relationship was observed overall between IL-18 and PGE2 levels in normal and OA osteoblasts. These results suggest that the shunt from the production of PGE2 to LTB4 is through regulation of the expression of FLAP, not 5-LO, in OA osteoblasts. The expression of FLAP in OA osteoblasts is also modulated differently by 1,25(OH)2D3 and TGF-β depending on their endogenous low and high PGE2 levels.
Collapse
Affiliation(s)
- Kelitha Maxis
- Unité de recherche en Arthrose, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 rue Sherbrooke Est, Montréal, Québec, Canada, H2L 4M1
| | - Aline Delalandre
- Unité de recherche en Arthrose, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 rue Sherbrooke Est, Montréal, Québec, Canada, H2L 4M1
| | - Johanne Martel-Pelletier
- Unité de recherche en Arthrose, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 rue Sherbrooke Est, Montréal, Québec, Canada, H2L 4M1
| | - Jean-Pierre Pelletier
- Unité de recherche en Arthrose, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 rue Sherbrooke Est, Montréal, Québec, Canada, H2L 4M1
| | - Nicolas Duval
- Unité de recherche en Arthrose, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 rue Sherbrooke Est, Montréal, Québec, Canada, H2L 4M1
| | - Daniel Lajeunesse
- Unité de recherche en Arthrose, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 rue Sherbrooke Est, Montréal, Québec, Canada, H2L 4M1
| |
Collapse
|
35
|
Hall AJ, Tripp M, Howell T, Darland G, Bland JS, Babish JG. Gastric mucosal cell model for estimating relative gastrointestinal toxicity of non-steroidal anti-inflammatory drugs. Prostaglandins Leukot Essent Fatty Acids 2006; 75:9-17. [PMID: 16806870 DOI: 10.1016/j.plefa.2006.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Accepted: 04/27/2006] [Indexed: 11/15/2022]
Abstract
The study objective was to characterize the AGS human gastric mucosal cell line as a model for estimating gastrointestinal toxicity of COX-inhibiting compounds. Rofecoxib, celecoxib, nimesulide, ibuprofen, indomethacin, aspirin, salicylic acid, naproxen and acetaminophen were tested for inhibition of COX-2-mediated prostaglandin E2 synthesis in A549 and AGS cells. The IC50 ratio AGS/A549 was calculated as an estimate of the therapeutic index (TI) for gastrointestinal toxicity. Calculated IC50 values of non-steroidal anti-inflammatory drugs (NSAIDs) in A549 cells were in excellent agreement with published values (r = 0.996; P < 0.005). Calcium ionophore induction of arachidonic acid release in AGS cells provided TI similar to those using platelets and A549 cells (r = 0.918; P < 0.01). The AGS/A549 model exhibited lower TI than the platelet/A549 model. Spearman ranking correlated clinical NSAID gastropathy with lower AGS TI values. The AGS cell line has excellent potential to serve as a model for assessing the gastrointestinal effects of COX-inhibiting compounds.
Collapse
Affiliation(s)
- A J Hall
- Metagenics, 9770 44th Avenue, Gig Harbor, WA 98332, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Lee ES, Park BC, Paek SH, Lee YS, Basnet A, Jin DQ, Choi HG, Yong CS, Kim JA. Potent Analgesic and Anti-inflammatory Activities of 1-Furan-2-yl-3-pyridin-2-yl-propenone with Gastric Ulcer Sparing Effect. Biol Pharm Bull 2006; 29:361-4. [PMID: 16462046 DOI: 10.1248/bpb.29.361] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most frequently prescribed drug for the treatment of inflammation and pain. However, conventional NSAIDs and selective COX-2 inhibitors have shown many side effects such as gastric mucosal damage and cardiovascular problems. Recently, the use of dual acting inhibitors of cyclooxygenases (COX) and lipoxygenase (LOX) has been highlighted for their minimized side effects compared to NSAIDs. The objective of the present study was to examine the efficacy and the gastric side effects of 1-furan-2-yl-3-pyridin-2-yl-propenone (FPP-3), a synthetic dual inhibitor of COX/5-LOX. Indomethacin (1-50 mg/kg, p.o.), a non-selective COX inhibitor, and FPP-3 (0.5-50 mg/kg, p.o.), a dual inhibitor, significantly suppressed the carrageen-induced paw edema with different pharmacological profiles. The concentrations of FPP-3 and indomethacin showing 50% inhibition of the maximum paw edema in rats were 10 mg/kg and 20 mg/kg, respectively. More importantly, there were no gastric ulcers formed in FPP-3-treated rats and mice, whereas indomethacin caused gastric mucosal bleeding in a concentration-dependent manner. In addition, FPP-3 showed an analgesic effect in acetic acid-induced writhing response in mice in a dose-dependent manner. The results suggest that FPP-3 may have a benefit in combatting inflammation and pain by dual inhibition of COX and LOX.
Collapse
Affiliation(s)
- Eung-Seok Lee
- College of Pharmacy, Yeungnam University, 214-1 Dae-dong, Gyeongsan 712-749, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Agnello KA, Reynolds LR, Budsberg SC. In vivo effects of tepoxalin, an inhibitor of cyclooxygenase and lipoxygenase, on prostanoid and leukotriene production in dogs with chronic osteoarthritis. Am J Vet Res 2005; 66:966-72. [PMID: 16008217 DOI: 10.2460/ajvr.2005.66.966] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate in vivo effects of tepoxalin, an inhibitor of cyclooxygenase (COX) and lipoxygenase (LOX), on prostaglandin (PG) and leukotriene production in osteoarthritic dogs. ANIMALS 7 mixed-breed adult dogs with chronic unilateral arthritis of a stifle joint. PROCEDURE Dogs were treated in accordance with a randomized 3-way crossover design. Each dog received an inert substance, meloxicam, or tepoxalin for 10 days. On day 0 (baseline), 3, and 10, dogs were anesthetized and samples of blood, stifle joint synovial fluid, and gastric mucosa were collected. Concentrations of PGE2 were measured in synovial fluid and after lipopolysaccharide stimulation of whole blood; PGE1 and PGE2 synthesis was measured in gastric mucosa. Thromboxane B2 (TxB2) concentration was measured in whole blood. Leukotriene B4 (LTB4) concentration was determined in gastric mucosa and in whole blood after ex vivo stimulation with a calcium ionophore. RESULTS Tepoxalin significantly decreased LTB4 concentrations in the blood and gastric mucosa at day 10 and TxB2 concentrations in the blood and PGE2 in the gastric mucosa and synovial fluid at days 3 and 10, compared with baseline values. Meloxicam significantly decreased PGE2 concentrations in the blood at days 3 and 10 and synovial fluid at day 3. Meloxicam also decreased PGE1 and PGE2 synthesis in the gastric mucosa at day 3. Meloxicam did not affect LTB4 synthesis in the blood or LTB4 concentrations in the gastric mucosa. CONCLUSIONS AND CLINICAL RELEVANCE Tepoxalin has in vivo inhibitory activity against COX-1, COX-2, and 5-LOX in dogs at the current approved recommended dosage.
Collapse
Affiliation(s)
- Kimberly A Agnello
- Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | | | | |
Collapse
|
38
|
Moreau M, Daminet S, Martel-Pelletier J, Fernandes J, Pelletier JP. Superiority of the gastroduodenal safety profile of licofelone over rofecoxib, a COX-2 selective inhibitor, in dogs. J Vet Pharmacol Ther 2005; 28:81-6. [PMID: 15720519 DOI: 10.1111/j.1365-2885.2004.00640.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study assessed the gastroduodenal safety profile of licofelone, a new nonsteroidal anti-inflammatory drug with dual inhibitory activity against 5-lipoxygenase and cyclo-oxygenase (COX), by using endoscopic evaluations and by comparing licofelone to rofecoxib, a selective COX-2 inhibitor. Twenty-one dogs underwent blinded gastroduodenoscopies, during which the mucosa of the gastroduodenal tract was assessed and scored. Blood analyses were monitored on days 0 (baseline), 14, 28, 42, and 56. Examinations to detect fecal occult blood were performed daily. Dogs were randomly assigned to three groups that received either a placebo, licofelone at a dose of 2.5 mg/kg twice daily, or rofecoxib at a dose of 0.5 mg/kg daily, respectively. Significant differences between the groups in gastric (P = 0.003), duodenal (P = 0.009), and gastroduodenal (P = 0.002) endoscopic lesion scores were observed at day 56. Rofecoxib-treated dogs had more lesions in all areas when compared with placebo-treated dogs, more duodenal lesions when compared with licofelone-treated dogs and more lesions than they had at baseline. In contrast to licofelone, rofecoxib was found to induce significant gastric and gastroduodenal lesions in dogs that lacked pre-existing lesions at baseline. Blood analyses and fecal examinations did not reveal abnormalities in any of the experimental groups. Treatment with licofelone was well tolerated and was shown to be safer than rofecoxib in terms of upper gastrointestinal damage. In this way, this study demonstrates the gastroduodenal safety profile of licofelone for chronic treatment.
Collapse
Affiliation(s)
- M Moreau
- The Companion Animal Research Group, Faculty of Veterinary Medicine, University of Montreal, St Hyacinthe, QC, Canada
| | | | | | | | | |
Collapse
|
39
|
Bauer H, Märker-Hermann E. [Therapy with nonsteroidal anti-inflammatory drugs]. DER ORTHOPADE 2004; 32:1088-94. [PMID: 14655005 DOI: 10.1007/s00132-003-0559-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most frequently prescribed drugs worldwide. Their analgetic and antiphlogistic effect results from an efficient inhibition of prostaglandin synthesis. The risk of side effects is remarkable. This concerns namely side effects in the gastrointestinal tract and the kidneys. The common NSAIDs inhibit both isoforms of the cyclooxygenase (COX). The analgetic and antiphlogistic effect results from the inhibition of COX-2, whereas the inhibition of COX-1 is responsible for the majority of side effects. If NSAID therapy is necessary, patients at risk should receive an effective prophylaxis, or be treated with COX-2-selective substances. The analgetic effects of COX-2 inhibitors are comparable to common NSAIDs, with a significant reduction of the gastrointestinal risks, except in cases of additional risk factors, e.g., low-dose ASS therapy, etc. In cases of renal impairment both substances should be used with caution, and subtle monitoring is necessary.
Collapse
Affiliation(s)
- H Bauer
- Schwerpunkt Rheumatologie, Klinische Immunologie, Klinik Nephrologie, Innere Medizin IV, Dr.-Horst-Schmidt-Kliniken GmbH, Wiesbaden.
| | | |
Collapse
|
40
|
Thiéfin G, Bannwarth B. [Management of the digestive risk in patients treated with NSAIDs. Synthesis and perspectives]. GASTROENTEROLOGIE CLINIQUE ET BIOLOGIQUE 2004; 28 Spec No 3:C96-102. [PMID: 15366681 DOI: 10.1016/s0399-8320(04)95285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Gérard Thiéfin
- Service d'Hépato-Gastroentérologie, CHU Robert-Debré, rue Gńéral-Koenig, 51092 Reims Cedex.
| | | |
Collapse
|
41
|
Martel-Pelletier J, Lajeunesse D, Reboul P, Pelletier JP. Therapeutic role of dual inhibitors of 5-LOX and COX, selective and non-selective non-steroidal anti-inflammatory drugs. Ann Rheum Dis 2003; 62:501-9. [PMID: 12759283 PMCID: PMC1754580 DOI: 10.1136/ard.62.6.501] [Citation(s) in RCA: 304] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Dual 5-LOX/COX inhibitors are potential new drugs to treat inflammation. They act by blocking the formation of both prostaglandins and leucotrienes but do not affect lipoxin formation. Such combined inhibition avoids some of the disadvantages of selective COX-2 inhibitors and spares the gatrointestinal mucosa.
Collapse
Affiliation(s)
- J Martel-Pelletier
- Osteoarthritis Research Unit, Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, Montréal, Québec H2L 4M1 Canada.
| | | | | | | |
Collapse
|
42
|
Paredes Y, Massicotte F, Pelletier JP, Martel-Pelletier J, Laufer S, Lajeunesse D. Study of the role of leukotriene B()4 in abnormal function of human subchondral osteoarthritis osteoblasts: effects of cyclooxygenase and/or 5-lipoxygenase inhibition. ARTHRITIS AND RHEUMATISM 2002; 46:1804-12. [PMID: 12124864 DOI: 10.1002/art.10357] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To compare the effect of licofelone, NS-398 (an inhibitor of cyclooxygenase 2 [COX-2]), and BayX-1005 (an inhibitor of 5-lipoxygenase activating protein) on the production of leukotriene B(4) (LTB(4)) and prostaglandin E(2) (PGE(2)), and on cell biomarkers by human osteoarthritis (OA) subchondral osteoblasts. METHODS Primary in vitro osteoblasts were prepared from subchondral bone specimens obtained from OA patients and autopsy subjects. LTB(4) and PGE(2) levels were measured by enzyme-linked immunosorbent assay in conditioned media of osteoblasts incubated in the presence or absence of licofelone, NS-398, or BayX-1005. The effect of these drugs or of the addition of LTB(4) on alkaline phosphatase (AP) activity and osteocalcin release by OA and normal osteoblasts was determined. The presence of LTB(4) receptors in normal and OA osteoblasts was evaluated by Western blot analysis. RESULTS OA osteoblasts produced variable levels of PGE(2) and LTB(4) compared with normal osteoblasts. Licofelone, at the maximal dose used, inhibited production of PGE(2) and LTB(4) by OA osteoblasts by a mean +/- SEM of 61.2 +/- 6.4% and 67.0 +/- 7.6%, respectively. NS-398 reduced PGE(2) production by 75.8 +/- 5.3%. BayX-1005 inhibited LTB(4) production in OA osteoblasts by 38.7 +/- 14.5% and marginally affected PGE(2) levels (reduction of 14.8 +/- 5.3%). Licofelone dose-dependently stimulated 1,25-dihydroxyvitamin D-induced AP activity while inhibiting osteocalcin release. BayX-1005 partly reproduced these effects, but NS-398 failed to affect them. LTB(4) dose-dependently inhibited AP activity in OA osteoblasts, while its effect on osteocalcin depended on endogenous LTB(4) levels in these cells. In normal osteoblasts, LTB(4) dose-dependently stimulated osteocalcin, whereas it failed to influence AP. LTB(4) receptors BLT1 and BLT2 were present in normal and OA osteoblasts. CONCLUSION Licofelone inhibits the production of PGE(2) and LTB(4). Selective effects of licofelone on AP and osteocalcin occur via its role on LTB(4) production. Because LTB(4) can modify cell biomarkers in OA and normal osteoblasts, our results suggest licofelone could modify abnormal bone remodeling in OA.
Collapse
Affiliation(s)
- Yosabeth Paredes
- Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Rheumatic diseases are the most prevalent causes of disability in western countries, and non-steroidal anti-inflammatory drugs (NSAIDs) are still the most commonly used remedies. However, NSAIDs cause several serious adverse effects, the most important being from gastric injury to gastric ulceration and renal damage. Attempts to develop non-steroidal anti-inflammatory remedies devoid of these shortcomings-especially gastrointestinal toxicity-have followed several strategies. Non-steroidal anti-inflammatory drugs have, therefore, been associated with gastroprotective agents that counteract the damaging effects of prostaglandin synthesis suppression; however, a combination therapy introduces other problems of pharmacokinetics, toxicity, and patient's compliance. More recently, incorporation of a nitric oxide (NO)-generating moiety into the molecule of several NSAIDs was shown to greatly attenuate their ulcerogenic activity; however, several findings suggest a possible involvement of NO in the pathogenesis of arthritis and subsequent tissue destruction. A most promising approach seemed to be the preparation of novel NSAIDs, targeted at the inducible isoform of prostaglandin synthase (COX-2); they appear to be devoid of gastrointestinal toxicity, in that they spare mucosal prostaglandin synthesis. However, a number of recent studies have raised serious questions about the two central tenets that support this approach, namely that the prostaglandins that mediate inflammation and pain are produced solely via COX-2 and that the prostaglandins that are important in gastrointestinal and renal function are produced solely via COX-1. So, a growing body of evidence shows that COX-2 (not only COX-1) also plays a physiological role in several body functions and that, conversely, COX-1 (not only COX-2) may also be induced at sites of inflammation. More recent and puzzling data shows that COX-2 is induced during the resolution of an inflammatory response, and at this point it produces anti-inflammatory (PGD2 and PGF2alpha), but not proinflammatory (PGE2) prostaglandins; inhibition of COX-2 at this point thus results in persistence of the inflammation. Moreover, COX-2 selective NSAIDs have lost the cardiovascular protective effects of non-selective NSAIDs, effects which are mediated through COX-1 inhibition (in addition, COX-2 has a role in sustaining vascular prostacyclin production). The generation of other very important products of the arachidonic acid cascade (besides cyclooxygenase-produced metabolites) is inhibited neither by non-selective nor by COX-2 selective NSAIDs. The products generated by the 5-lipoxygenase pathway (leukotrienes) are particularly important in inflammation; indeed, leukotrienes increase microvascular permeability and are potent chemotactic agents. Moreover, inhibition of 5-lipoxygenase indirectly reduces the expression of TNF-alpha (a cytokine that plays a key role in inflammation). These data and considerations explain the efforts to obtain drugs able to inhibit both 5-lipoxygenase and cyclooxygenases, the so-called dual acting anti-inflammatory drugs. Such compounds retain the activity of classical NSAIDs, while avoiding their main drawbacks, in that curtailed production of gastroprotective prostaglandins is associated with a concurrent curtailed production of the gastro-damaging and bronchoconstrictive leukotrienes. Moreover, thanks to their mechanism of action, dual acting anti-inflammatory drugs could not merely alleviate symptoms of rheumatic diseases, but might also satisfy, at least in part, the criteria of a more definitive treatment. Indeed, leukotrienes are pro-inflammatory, increase microvascular permeability, are potent chemotactic agents and attract eosinophils, neutrophils and monocytes into the synovium.
Collapse
Affiliation(s)
- A Bertolini
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia, Via G. Campi 287, 41100 Modena, Italy.
| | | | | |
Collapse
|
44
|
Tries S, Laufer S. The pharmacological profile of ML3000: A new pyrrolizine derivative inhibiting the enzymes cyclo-oxygenase and 5-lipoxygenase. Inflammopharmacology 2001. [DOI: 10.1163/156856001300248380] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
45
|
Abdel-Salam OM, Czimmer J, Debreceni A, Szolcsányi J, Mózsik G. Gastric mucosal integrity: gastric mucosal blood flow and microcirculation. An overview. JOURNAL OF PHYSIOLOGY, PARIS 2001; 95:105-127. [PMID: 11595425 DOI: 10.1016/s0928-4257(01)00015-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The stomach is in a state of continuous exposure to potentially hazardous agents. Hydrochloric acid together with pepsin constitutes a major and serious threat to the gastric mucosa. Reflux of alkaline duodenal contents containing bile and pancreatic enzymes are additional important injurious factors of endogenous origin. Alcohol, cigarette smoking, drugs and particularly aspirin and aspirin-like drugs, and steroids are among exogenous mucosal irritants that can inflict mucosal injury. The ability of the stomach to defend itself against these noxious agents has been ascribed to a number of factors constituting the gastric mucosal defense. These include mucus and bicarbonate secreted by surface epithelial cells, prostaglandins, sulfhydryl compounds and gastric mucosal blood flow. The latter is considered by several researchers to be of paramount importance in maintaining gastric mucosal integrity. The aim of this paper is to review the experimental and clinical data dealing with the role of mucosal blood flow and in particular the microcirculation in both damage and protection of the gastric mucosa.
Collapse
Affiliation(s)
- O M Abdel-Salam
- Department of Pharmacology, National Research Centre, PO Box 12311, El-Tahrir St., Dokki, Cairo, Egypt.
| | | | | | | | | |
Collapse
|
46
|
Koga H, Aoyagi K, Matsumoto T, Iida M, Fujishima M. Experimental enteropathy in athymic and euthymic rats: synergistic role of lipopolysaccharide and indomethacin. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:G576-82. [PMID: 10070032 DOI: 10.1152/ajpgi.1999.276.3.g576] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The aim of this study was to investigate the immunologic and microbiological bases of indomethacin enteropathy. Athymic nude and euthymic specific pathogen-free (SPF) rats were reared under conventional or SPF conditions. In each group, indomethacin was given intrarectally for 2 days. Indomethacin enteropathy was evaluated using a previously described ulcer index and tissue myeloperoxidase activity. Both euthymic and athymic nude rats developed intestinal ulcers to the same degree under conventional conditions but no or minimal ulcer under SPF conditions. Pretreatment of conventional rats with intragastric kanamycin sulfate, an aminoglycoside antibiotic, attenuated indomethacin enteropathy in a dose-dependent fashion. Interestingly, when lipopolysaccharide was injected intraperitoneally in kanamycin-pretreated rats, it fully restored enteropathy in these rats in a dose-dependent manner. We confirmed that kanamycin decreased the number of gram-negative bacteria and endotoxin concentration of the small intestine in a dose-dependent fashion. These results indicate that indomethacin enteropathy is bacteria dependent and does not require a T cell function. Synergy between indomethacin and bacterial lipopolysaccharide may play a major role in this enteropathy.
Collapse
Affiliation(s)
- H Koga
- Second Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | |
Collapse
|
47
|
Abstract
A growing body of experimental evidence indicates that leukocyte-endothelial cell interactions may play an important role in the pathogenesis of NSAID-induced gastropathy. Using a newly described, dual radiolabeled monoclonal antibody technique to quantify adhesion molecule surface expression in vivo, we have demonstrated increases in surface expression of ICAM-1 and P-selectin in the gastric mucosa after oral administration of indomethacin. We have also found that CD18-, ICAM-1-, or P-selectin-deficient mice are less sensitive to the ulcerogenic effects of orally administered indomethacin. Although there is virtually no information regarding the regulation of expression of endothelial cell adhesion molecules (ECAMs) in experimental NSAID-induced gastropathy, the nuclear transcription factor KB (NFKB) may represent a potential modulator of transcriptional activation of ECAM expression. We have demonstrated that two structurally distinct yet highly selective proteasome inhibitors (MG341, lactacystin) inhibit tumor necrosis factor (TNF)-induced NFKB activation as well as ECAM expression in human endothelial cells in vitro. In addition, we found that these proteasome inhibitors significantly reduced indomethacin-induced gastric mucosal injury as well as gastric mucosal ICAM-1 expression in the rat in vivo. We conclude from these studies that indomethacin activates NFKB (possibly via TNF synthesis) in gastric microvascular endothelial cells, thereby enhancing surface expression of ICAM-1 which binds the CD18 on polymorphonuclear leukocytes (PMNs). These adherent PMNs are then believed to mediate endothelial and/or epithelial cell injury either directly or indirectly.
Collapse
Affiliation(s)
- Z Morise
- Department of Molecular and Cellular Physiology, Louisiana State University Medical Center, Shreveport 71130, USA
| | | |
Collapse
|
48
|
Uçar A, Sak SD, Melli M. Effects of chronic treatment with indomethacin at clinically relevant doses on intestinal tissue 6-keto prostaglandin F1 alpha and leukotriene B4 level in relation to gastroenteropathy. Inflammation 1998; 22:243-52. [PMID: 9604712 DOI: 10.1023/a:1022354614385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study investigated the effects of indomethacin at clinically relevant doses and its chronic usage on intestinal pathology, survival time and intestinal tissue 6-keto prostaglandin F1 alpha and leukotriene B4 level in rats during various periods with different doses. Indomethacin was administered ranging from 0.625 to 5 mg/kg. When used in doses of 0.625 and 1.25 mg/kg, indomethacin caused no apparent intestinal lesions or death during a treatment period of 30 days. On the other hand, all rats died in 7 days when 5 mg/kg of indomethacin was given. Mortality rate reached 53.3% in seven days in the group where 3.75 mg/kg indomethacin was given. The minimal dose of indomethacin, which induced intestinal ulcer and death, was 2.5 mg/kg. The main pathological findings were intestinal ulcers, but no macroscopic and microscopic changes were observed in the stomach. Intestinal tissue 6-keto prostaglandin F1 alpha and leukotriene B4 levels were quantified by enzyme immunoassay after homogenisation and extraction of tissue. In dose-dependent studies, only the dose of indomethacin, 3.75 mg/kg, significantly inhibited intestinal tissue 6-keto prostaglandin F1 alpha levels during seven days application period (197.39 +/- 24.26 vs 383.66 +/- 46.68 ng/g tissue, treatment vs control). 2.5 mg/kg of indomethacin caused no intestinal ulceration on 4th day, however, it significantly inhibited intestinal tissue 6-keto prostaglandin F1 alpha levels on 4th day in time-dependent studies (190.3 +/- 26.62 vs 383.66 +/- 46.68 ng/g tissue, treatment vs control). Neither dose-dependent nor time-dependent indomethacin administration changed intestinal tissue leukotriene B4 level. The results of this study indicated that indomethacin produced enteropathy rather than gastropathy when used chronically in clinically relevant doses in rats. Inhibition of prostaglandin synthesis, which was estimated by quantification of intestinal tissue 6-keto prostaglandin F1 alpha level, seemed not to be a prerequisite for its enteropathic effect.
Collapse
Affiliation(s)
- A Uçar
- Department of Pharmacology, Medical Faculty of Ankara University, Turkey
| | | | | |
Collapse
|
49
|
Kirchner T, Aparicio B, Argentieri DC, Lau CY, Ritchie DM. Effects of tepoxalin, a dual inhibitor of cyclooxygenase/5-lipoxygenase, on events associated with NSAID-induced gastrointestinal inflammation. Prostaglandins Leukot Essent Fatty Acids 1997; 56:417-23. [PMID: 9223651 DOI: 10.1016/s0952-3278(97)90593-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Prostaglandins and thromboxanes are products of arachidonic acid metabolism via the cyclooxygenase (CO) enzyme and are responsible for the pain and swelling common to sites of inflammation. Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit the production of these substances and are used in the treatment of inflammatory diseases such as arthritis. However, one of the major side-effects of NSAID therapy is gastric ulceration. It is possible that inhibition of prostaglandin production and a related increase in the formation of leukotrienes via the 5-lipoxygenase (5-LO) enzymatic pathway are responsible for attracting inflammatory cells, causing local sites of inflammation and producing ulceration. To determine the effects of 5-LO inhibition on this hypothesis, studies were performed in rats to evaluate the effects of tepoxalin, a dual CO/LO inhibitor on leukotriene B4 levels in gastric mucosa and neutrophil adhesion in mesenteric venules. In rats, chronic oral administration of an NSAID, indomethacin (2 mg/kg daily over 4 days), resulted in 40% mortality, accompanied by intestinal adhesions and perforations when evaluated 24 h after the fourth dose of drug. Additionally, neutrophil adhesion was increased in the mesenteric venules and cell infiltration was evident in the mesenteric interstitium. These gastrointestinal side-effects were inhibited in a separate group of rats administered tepoxalin (20 mg/kg, p.o) 30 min prior to each daily indomethacin treatment. Further studies were performed to determine tepoxalin's effects on early events associated with NSAID-induced gastrointestinal inflammation, including neutrophil adhesion, lipid peroxide generation and LTB4 production. Indomethacin (100 mg/kg, p.o.) produced elevated levels of LTB4 in rat gastric mucosa 90 min after administration. Additionally, neutrophil adhesion in mesenteric venules was increased at this dose and with the administration of another NSAID, naproxen. No generation of lipid peroxides was evident in the gastric mucosa at this timepoint. Tepoxalin (up to 400 mg/kg, p.o.) did not have an effects on gastric mucosal LTB4 generation and lipid peroxide levels. A decrease in neutrophil adhesion was observed at the highest dose. In another study, pretreatment with tepoxalin (ED50=7.5 mg/kg, p.o.) or the selective 5-LO inhibitor zileuton (100 mg/kg, p.o.) prevented the increases in gastric mucosal LTB4 levels and neutrophil adhesion induced by indomethacin (100 mg/kg, p.o.). These data suggest that LO inhibition may play a vital role in the prevention of NSAID-induced gastric inflammation, providing insight into the lack of ulcerogenicity with tepoxalin and new approaches to anti-inflammatory therapy which may prevent gastric side effects.
Collapse
Affiliation(s)
- T Kirchner
- Department of Immunopharmacology, The R.W. Johnson Pharmaceutical Research Institute, Raritan, NJ 08869, USA.
| | | | | | | | | |
Collapse
|
50
|
Affiliation(s)
- C Denzlinger
- Medizinische Klinik III, Klinikum Grosshadern, Ludwig-Maximilians Universität München, Germany
| |
Collapse
|