1
|
Mittal R, Sharma S, Mittal A, Kushwah AS. Novel Dual COX-2/5-LOX Inhibitory Activity by Chalcone Derivatives: A Safe and Efficacious Anti-inflammatory Agent. Antiinflamm Antiallergy Agents Med Chem 2024; 23:174-186. [PMID: 38939991 DOI: 10.2174/0118715230301176240605072113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/10/2024] [Accepted: 05/03/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Non-communicable diseases are chronic systemic inflammation in humans that occurs because of enhanced inflammatory mediators of the arachidonic acid cascade. We aimed to explore whether the lead chalcone compounds could exhibit anti-inflammatory activity via dual blockage of COX-2/5-LOX enzymes and their regulatory mechanism. METHODS RAW 264.7 macrophages were collected from NCC, Pune, for in-vitro experiments. The IC50 values of chalcone compounds C45 and C64 were calculated. RAW 264.7 macrophages were treated with C45 and C64 (10%, 5%, 2.5%, 0.125%, and 0.0625% concentration). The cell viability was carried out with an MTT assay. The COX-1, COX-2, 5-LOX, PGE2, and LTB4 levels were detected by ELISA-based kits. The in-vivo evaluation was carried out in Male Wistar rats (250-300 g, 7-8 weeks old) with acute and chronic anti-inflammatory models and histopathological studies on the stomach, liver, and kidney. RESULTS The present study described the in-vitro and in-vivo biological evaluation of dual COX-2/5-LOX inhibitors in chalcone derivatives (C45 and C64) compounds showed the most effective COX-2 and 5-LOX inhibition with IC50 values 0.092 and 0.136 μM respectively. Simultaneously, compound C64 showed comparable selectivity towards COX-2 with a Selectivity Index (SI) of 68.43 compared to etoricoxib, with an SI of 89.32. In-vivo carrageenaninduced rat paw oedema activity, the compound C64 showed a significant reduction in oedema with 78.28% compared to indomethacin with 88.07% inhibition. Furthermore, cotton pelletinduced granuloma activity revealed that compound C64 significantly reduced 32.85% compared with standard 40.13% granuloma inhibition. CONCLUSION The chalcone compound C64, (E)-1-(4-Amino-2-hydroxyphenyl)-3-(3,4,5-trimethoxyphenyl)- prop-2-en-1-one was proved to be a potent and novel Dual COX-2/5-LOX inhibitor with improved gastric safety profiling.
Collapse
Affiliation(s)
- Roopal Mittal
- Department of Pharmacy, IK Gujral Punjab Technical University, Jalandhar Punjab, 144601, India
- R.K.S.D. College of Pharmacy, Kaithal Haryana, 136027, India
| | - Shailesh Sharma
- Department of Pharmaceutics, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela Ropar, Punjab, 140111, India
| | - Amit Mittal
- Department of Pharmaceutical Chemistry, School of Pharmacy, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, India
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Desh Bhagat University, Amloh Road, Mandi Gobindgarh, Fatehgarh Sahib, Punjab, 147301, India
| | - Ajay Singh Kushwah
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela Ropar, Punjab, 140111, India
| |
Collapse
|
2
|
Vishwakarma RK, Negi DS, Negi A. Abortitristoside A and desrhamnosylverbanscoside: the potential COX-2 inhibitor from the leaves of Nyctanthes arbor-tristis as anti-inflammatory agents based on the in vitro assay, molecular docking and ADMET prediction. CHEMICAL PAPERS 2023. [DOI: 10.1007/s11696-023-02686-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
3
|
Noori S, Ghorbani-Vaghei R, Azadbakht R, Karamshahi Z, Koolivand M. Graphene-oxide/schiff base N2O4 ligand-palladium: A new catalyst for the synthesis of furan derivatives. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
4
|
Shwetha B, Sudhanva MS, Jagadeesha GS, Thimmegowda NR, Hamse VK, Sridhar BT, Thimmaiah KN, Ananda Kumar CS, Shobith R, Rangappa KS. Furan-2-carboxamide derivative, a novel microtubule stabilizing agent induces mitotic arrest and potentiates apoptosis in cancer cells. Bioorg Chem 2021; 108:104586. [PMID: 33607574 DOI: 10.1016/j.bioorg.2020.104586] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/12/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022]
Abstract
The vital role played by microtubules in the cell division process, marks them as a potential druggable target to decimate cancer. A novel furan-2-carboxamide based small molecule, is a selective microtubule stabilizing agent (MSA) with IC50 ranging from 4 µM to 8 µM in different cancer cell lines. Inhibition of tubulin polymerization or stabilization of tubulin polymers abrogates chromosomal segregation during cell division, results in cell cycle arrest and leads to cell death due to the delayed repair mechanism. A novel furan-2-carboxamide based small molecule exhibited potent anti-proliferative and anti-metastatic property In-Vitro against the panel of cancer cells. Annexin V-FITC/PI, double staining reveals potent cytotoxic effect of SH09 against HeLa cells. FACS analysis displays induction of G2/M arrest and accumulation of subG1 population of cells upon treatment with SH09. Molecular docking study unveils SH09 binding affinity to the Taxol binding pocket of tubulin proteins and MM-GBSA also confirms strong binding energies of SH09 with tubulin proteins.
Collapse
Affiliation(s)
- B Shwetha
- Department of Nanotechnology, CPGS, Visvesvaraya Technological University, Muddenahalli, Karnataka 562101, India
| | - M Srinivasa Sudhanva
- Adichunchanagiri Institute for Molecular Medicine, AIMS, Adichunchanagiri University, BG Nagara 571448, Karnataka 02, India; Faculty of Natural Sciences, Adichunchanagiri University, BG Nagara 571448, Karnataka, India
| | - G S Jagadeesha
- Department of Chemistry, Govt. S. K. S. J. Technological Institute (Affiliated to Visvesvaraya Technological University), K R Circle, Bangalore, Karnataka 560001, India
| | - N R Thimmegowda
- Department of Chemistry, Govt. S. K. S. J. Technological Institute (Affiliated to Visvesvaraya Technological University), K R Circle, Bangalore, Karnataka 560001, India
| | - Vivek K Hamse
- Faculty of Natural Sciences, Adichunchanagiri University, BG Nagara 571448, Karnataka, India
| | - B T Sridhar
- Department of Chemistry, Maharani's Science College for Women, Palace Road, Bangalore, Karnataka 560001, India
| | - K N Thimmaiah
- Division of Natural Science Northwest Mississippi Community College, University of Mississippi Campus, Desoto Centre, Southaven, MS 38671, USA
| | - C S Ananda Kumar
- Department of Nanotechnology, CPGS, Visvesvaraya Technological University, Muddenahalli, Karnataka 562101, India; Centre for Material Science, University of Mysore, Mysore, Karnataka 570006, India.
| | - Rangappa Shobith
- Adichunchanagiri Institute for Molecular Medicine, AIMS, Adichunchanagiri University, BG Nagara 571448, Karnataka 02, India.
| | - K S Rangappa
- Institution of Excellence, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India
| |
Collapse
|
5
|
P JJ, S L M. Novel approach of multi-targeted thiazoles and thiazolidenes toward anti-inflammatory and anticancer therapy—dual inhibition of COX-2 and 5-LOX enzymes. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02655-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
6
|
Jacob P J, Manju SL. Identification and development of thiazole leads as COX-2/5-LOX inhibitors through in-vitro and in-vivo biological evaluation for anti-inflammatory activity. Bioorg Chem 2020; 100:103882. [PMID: 32361295 DOI: 10.1016/j.bioorg.2020.103882] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/13/2020] [Accepted: 04/22/2020] [Indexed: 12/22/2022]
Abstract
Treatment of inflammation using NSAIDs is coupled with a risk of severe gastric adverse events. Development of dual COX-2/5-LOX inhibitors turns out to be an imperative area devoted to safer NSAIDs. A series of thiourea, thiazole, and thiazolidene derivatives were synthesized by green synthetic approach and COX-1, COX-2 and 5-LOX inhibition screening resulted in the identification of a potent compound 6l with IC50 of 5.55 µM, 0.09 µM, and 0.38 µM respectively. Compound 6l made significant decrease (60.82%) in the carrageenan-induced edema in male Wistar rats. qRT-PCR analysis and determination of PGE2 and LTB4 in the rat paw tissues indicated that this thiazole based dual inhibitor significantly reduced the expression of COX-2 and 5-LOX genes besides the marked reduction in both PGE2 and LTB4 levels. The gastric safety profiling revealed an enhanced gastrointestinal safety of the compound 6l on histopathological examination. Molecular docking studies at COX-2 and 5-LOX active sites were consistent with biological studies by significant protein-ligand interaction. Besides, results of in-vitro PGE2 and LTB4 studies on RAW 264.7 cells as well as antioxidant studies were parallel to the dual inhibitory activity. The present investigations identify a promising lead having anti-inflammatory potential with an improved gastric safety profile.
Collapse
Affiliation(s)
- Jaismy Jacob P
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| | - S L Manju
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
7
|
Wang S, Han X, Yang Y, Zhou C, Luo D, He W, Zhu Q, Xu Y. Discovery of deoxylimonin δ-lactam derivative with favorable anti-inflammation and antinociception efficacy from chemical modified limonin/deoxylimonin analogs. Bioorg Chem 2020; 100:103886. [PMID: 32371249 DOI: 10.1016/j.bioorg.2020.103886] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 11/30/2022]
Abstract
Chemical modifications on the A ring of limonin (1) and deoxylimonin (2) afforded 28 structural characterized derivatives, which were firstly subjected to preliminary in vivo analgesic and anti-inflammatory screen by mice model. The most promising candidate, deoxylimonin analog II-B-2 (70 mg/kg) with 3,4-dimethoxyphenylethyl moiety substitued δ-lactam in the A ring, exhibited better analgesic activity than aspirin (200 mg/kg) and stronger anti-inflammatory efficacy than naproxen (150 mg/kg). Further in vivo evaluation confirmed its advantage over limonin and showed dose-response dependent manner, and follow-up research suggested that the anti-inflammatory effect of compound II-B-2 may be attributed to the downregulation of cyclooxygenase 2 expression and the suppression of prostaglandin E2 formation.
Collapse
Affiliation(s)
- Shaochi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xueqing Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yun Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Chen Zhou
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Danmeng Luo
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wensong He
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Qihua Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Yungen Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
8
|
Karamshahi Z, Ghorbani‐Vaghei R. Efficient synthesis of multiply substituted furans using BF@Propyl/dopamine/Pd as a green catalyst. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.5530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Zahra Karamshahi
- Department of Organic Chemistry, Faculty of ChemistryBu‐Ali Sina University Hamedan 6517838683 Iran
| | - Ramin Ghorbani‐Vaghei
- Department of Organic Chemistry, Faculty of ChemistryBu‐Ali Sina University Hamedan 6517838683 Iran
| |
Collapse
|
9
|
Safer anti-inflammatory therapy through dual COX-2/5-LOX inhibitors: A structure-based approach. Eur J Pharm Sci 2018; 121:356-381. [PMID: 29883727 DOI: 10.1016/j.ejps.2018.06.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/02/2018] [Accepted: 06/04/2018] [Indexed: 12/22/2022]
Abstract
Inflammatory mediators of the arachidonic acid cascade from cyclooxygenase (COX) and lipoxygenase (LOX) pathways are primarily responsible for many diseases in human beings. Chronic inflammation is associated with the pathogenesis and progression of cancer, arthritis, autoimmune, cardiovascular and neurological diseases. Traditional non-steroidal anti-inflammatory agents (tNSAIDs) inhibit cyclooxygenase pathway non-selectively and produce gastric mucosal damage due to COX-1 inhibition and allergic reactions and bronchospasm resulting from increased leukotriene levels. 'Coxibs' which are selective COX-2 inhibitors cause adverse cardiovascular events. Inhibition of any of these biosynthetic pathways could switch the metabolism to the other, which can lead to fatal side effects. Hence, there is undoubtedly an urgent need for new anti-inflammatory agents having dual mechanism that prevent release of both prostaglandins and leukotrienes. Though several molecules have been synthesized with this objective, their unfavourable toxicity profile prevented them from being used in clinics. Here, this integrative review attempts to identify the promising pharmacophore that serves as dual inhibitors of COX-2/5-LOX enzymes with improved safety profile. A better acquaintance of structural features that balance safety and efficacy of dual inhibitors would be a different approach to the process of understanding and interpreting the designing of novel anti-inflammatory agents.
Collapse
|
10
|
Discovery and structure-activity relationship studies of 2-benzylidene-2,3-dihydro-1 H -inden-1-one and benzofuran-3(2 H )-one derivatives as a novel class of potential therapeutics for inflammatory bowel disease. Eur J Med Chem 2017. [DOI: 10.1016/j.ejmech.2017.06.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
11
|
Vinayagam J, Gajbhiye RL, Mandal L, Arumugam M, Achari A, Jaisankar P. Substituted furans as potent lipoxygenase inhibitors: Synthesis, in vitro and molecular docking studies. Bioorg Chem 2017; 71:97-101. [PMID: 28143657 DOI: 10.1016/j.bioorg.2017.01.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/29/2016] [Accepted: 01/20/2017] [Indexed: 11/18/2022]
Abstract
A number of 2-methyl-4-(2-oxo-2-phenyl-ethyl)-5-phenyl-furan-3-carboxylic acid alkyl ester derivatives (3a-j) were synthesized and evaluated for their in vitro inhibitory activity on soybean lipoxygenase enzyme. Among the screened compounds, 5-(4-bromo-phenyl)-4-[2-(4-bromo-phenyl)-2-oxo-ethyl]-2-methyl-furan-3-carboxylic acid methyl ester (3g) has been found to exhibit potent inhibitory activity with IC5012.8μM using nordihydroguaiaretic acid (NDGA) as standard. Molecular modeling was employed for better understanding of the binding between compounds and soybean lipoxygenase enzyme. The predicted binding energy values correlated well with the observed in vitro data.
Collapse
Affiliation(s)
- Jayaraman Vinayagam
- Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR - Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, India
| | - Rahul L Gajbhiye
- Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR - Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, India
| | - Likhit Mandal
- Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR - Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, India
| | - Meyyappan Arumugam
- School of Environmental Studies, Jadavpur University, Kolkata 700 032, India
| | - Anushree Achari
- Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR - Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, India
| | - Parasuraman Jaisankar
- Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR - Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, India.
| |
Collapse
|
12
|
Pereira-Leite C, Nunes C, Jamal SK, Cuccovia IM, Reis S. Nonsteroidal Anti-Inflammatory Therapy: A Journey Toward Safety. Med Res Rev 2016; 37:802-859. [PMID: 28005273 DOI: 10.1002/med.21424] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 01/01/2023]
Abstract
The efficacy of nonsteroidal anti-inflammatory drugs (NSAIDs) against inflammation, pain, and fever has been supporting their worldwide use in the treatment of painful conditions and chronic inflammatory diseases until today. However, the long-term therapy with NSAIDs was soon associated with high incidences of adverse events in the gastrointestinal tract. Therefore, the search for novel drugs with improved safety has begun with COX-2 selective inhibitors (coxibs) being straightaway developed and commercialized. Nevertheless, the excitement has fast turned to disappointment when diverse coxibs were withdrawn from the market due to cardiovascular toxicity. Such events have once again triggered the emergence of different strategies to overcome NSAIDs toxicity. Here, an integrative review is provided to address the breakthroughs of two main approaches: (i) the association of NSAIDs with protective mediators and (ii) the design of novel compounds to target downstream and/or multiple enzymes of the arachidonic acid cascade. To date, just one phosphatidylcholine-associated NSAID has already been approved for commercialization. Nevertheless, the preclinical and clinical data obtained so far indicate that both strategies may improve the safety of nonsteroidal anti-inflammatory therapy.
Collapse
Affiliation(s)
- Catarina Pereira-Leite
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Cláudia Nunes
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Sarah K Jamal
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Iolanda M Cuccovia
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Salette Reis
- UCIBIO, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
13
|
Teixeira A, Cox RC, Egmond MR. Furan fatty acids efficiently rescue brain cells from cell death induced by oxidative stress. Food Funct 2014; 4:1209-15. [PMID: 23719714 DOI: 10.1039/c3fo60094g] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Treatment of rat brain C6 astroglioma cells with furan fatty acid F6 prior to exposure to hydrogen peroxide shows a strong protective effect of F6 against cell death resulting from oxidative stress. This protective effect is obtained only for F6 administered as a free fatty acid and with an intact furan ring. It is proposed that brain cells are rescued by F6 scavenging radicals elicited by lipid peroxidation within the cell membrane. Oxidative processes outside the cell membrane, such as protein carbonylation, are not affected by F6. Furan fatty acids such as those present in fish oils and marine organisms are likely beneficial for consumption in reducing the risk of diseases that have been implicated to arise from oxidative stress, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Antoinette Teixeira
- Department of Membrane Biochemistry and Biophysics, Bijvoet Center, University of Utrecht, Padualaan 8, 3584CH Utrecht, The Netherlands
| | | | | |
Collapse
|
14
|
Park SY, Ku SK, Lee ES, Kim JA. 1,3-Diphenylpropenone ameliorates TNBS-induced rat colitis through suppression of NF-κB activation and IL-8 induction. Chem Biol Interact 2012; 196:39-49. [PMID: 22410118 DOI: 10.1016/j.cbi.2012.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 02/16/2012] [Accepted: 02/25/2012] [Indexed: 01/24/2023]
Abstract
In the present study, we examined whether newly synthesized phenylpropenone derivatives, by inhibiting NF-κB activity, would inhibit IL-8 expression, inflammation and abnormal angiogenesis, resulting in amelioration of disease conditions. The phenylpropenone derivatives inhibited NF-κB transcriptional activity, which correlated with their suppressive activity against TNF-α-induced adhesion of U937 human monocytic cells to HT-29 human colonic epithelial cells, an in vitro model of IBD. Among the derivatives, 1,3-diphenylpropenone (DPhP) was most efficacious, and it significantly suppressed TNF-α-induced production of IL-8 which is a proinflammatory and proangiogenic cytokine. The anti-inflammatory activity of DPhP was also confirmed in the trinitrobenzene sulfonic acid (TNBS)-induced rat colitis model. DPhP was protective against the TNBS-induced inflammatory responses, which included weight loss, increased myeloperoxidase activity and mucosal damage. In the colon tissue, DPhP inhibited TNBS-induced NF-κB nuclear translocation, IL-8 and TNF-α expressions, and abnormal angiogenesis. In addition, DPhP also suppressed IL-8-induced angiogenesis, which was revealed by an in vivo assay using chick chorioallantoic membrane. Furthermore, the level of IL-6, a pleiotropic cytokine which is implicated in the pathogenesis of IBD and colitis-associated cancer, was suppressed by DPhP in rat colon tissue and serum. In conclusion, the results suggest that DPhP is a potential dual-acting IBD drug candidate targeting both inflammation and abnormal angiogenesis, possibly through the NF-κB and IL-8 signaling pathway.
Collapse
Affiliation(s)
- Su-Young Park
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, South Korea
| | | | | | | |
Collapse
|
15
|
Lee JS, Kang Y, Kim JT, Thapa D, Lee ES, Kim JA. The anti-angiogenic and anti-tumor activity of synthetic phenylpropenone derivatives is mediated through the inhibition of receptor tyrosine kinases. Eur J Pharmacol 2011; 677:22-30. [PMID: 22200628 DOI: 10.1016/j.ejphar.2011.12.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 12/02/2011] [Accepted: 12/09/2011] [Indexed: 12/31/2022]
Abstract
Abnormal angiogenesis plays a critical role in the pathogenesis of various diseases such as cancer and chronic inflammation. A variety of pro-angiogenic factors, including vascular endothelial growth factor (VEGF), exert their action through endothelial receptor tyrosine kinases (RTKs). The synthetic phenylpropenone derivatives, used in this study were the following: 1,3-diphenyl-propenone (DPhP), 3-phenyl-1-thiophen-2-yl-propenone (PhT2P), 3-phenyl-1-thiophen-3-yl-propenone (PhT3P) and 1-furan-2-yl-3-phenyl-propenone (FPhP). These derivatives were screened for their inhibitory effect on VEGF-induced angiogenesis in vitro using HUVECs and in vivo using chick chorioallantoic membrane (CAM). The order of anti-angiogenic activity was DPhP>FPhP>PhT3P>PhT2P. The most effective compound DPhP, also known as chalcone, showed weak VEGF receptor tyrosine kinase activity compared with the specific inhibitor, SU4312 (3-[[4-(dimethylamino)phenyl]methylene]-1,3-dihydro-2H-indol-2-one). However, DPhP also inhibited several other receptor tyrosine kinases including Tie-2, epithermal growth factor (EGF) receptor, EphB2, fibroblast growth factor (FGF) receptor 3 and insulin-like growth factor-1 (IGF-1) receptor, as revealed by a receptor tyrosine kinase array assay. In addition, the down-stream signaling, including ERK phosphorylation and NF-κB activation, after receptor activation was significantly inhibited by DPhP. Furthermore, in the HT29 human colon cancer cell-inoculated CAM assay, the tumor growth and tumor-induced angiogenesis was significantly inhibited by DPhP (10μg/ml). These results suggest that the simple flavonoid, DPhP (chalcone), has valuable potential as an antiangiogenic and anti-cancer agent, and its action is mediated through the inhibition of multi-target RTKs including VEGF receptor 2.
Collapse
Affiliation(s)
- Jong-Suk Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, South Korea
| | | | | | | | | | | |
Collapse
|
16
|
Jeon TW, Kim CH, Lee SK, Shin S, Choi JH, Kang WK, Kim SH, Kang MJ, Lee ES, Jeong TC. Hepatotoxic Effects of 1-Furan-2-yl-3-pyridin-2-yl-propenone, a New Anti-Inflammatory Agent, in Mice. Biomol Ther (Seoul) 2009. [DOI: 10.4062/biomolther.2009.17.3.318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
17
|
Park BC, Park SY, Lee JS, Mousa SA, Kim JT, Kwak MK, Kang KW, Lee ES, Choi HG, Yong CS, Kim JA. The anti-angiogenic effects of 1-furan-2-yl-3-pyridin-2-yl-propenone are mediated through the suppression of both VEGF production and VEGF-induced signaling. Vascul Pharmacol 2009; 50:123-31. [DOI: 10.1016/j.vph.2008.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 10/27/2008] [Accepted: 11/14/2008] [Indexed: 11/12/2022]
|
18
|
Manandhar S, You A, Lee ES, Kim JA, Kwak MK. Activation of the Nrf2-antioxidant system by a novel cyclooxygenase-2 inhibitor furan-2-yl-3-pyridin-2-yl-propenone: implication in anti-inflammatory function by Nrf2 activator. J Pharm Pharmacol 2008; 60:879-87. [PMID: 18549674 DOI: 10.1211/jpp.60.7.0009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Furan-2-yl-3-pyridin-2-yl-propenone (FPP-3) is a novel synthetic compound and has demonstrated anti-inflammatory activity by inhibiting cyclooxygenase-2 (COX-2). It is widely accepted that reactive oxygen species (ROS) generated by activated inflammatory cells can exacerbate inflammation. In this study, the potential antioxidative efficacy of FPP-3 has been investigated in murine cells. FPP-3 increased the expression of multiple antioxidative enzymes, including NAD(P)H:quinone oxidoreductase 1 (Nqo1), gamma-glutamylcysteine ligase (GCL) and heme oxygenase-1 (HO-1), by facilitating the nuclear translocation of nuclear factor-erythroid 2-p45-related factor 2 (Nrf2). Inducibility of antioxidant proteins such as HO-1 were lost in nrf2-deficient murine fibroblasts. As a result of enhanced cellular antioxidative capacity, elevation of NF-kappaB-driven reporter gene expression by lipopolysaccharide was attenuated by FPP-3 treatment in murine fibroblasts. Furthermore, FPP-3 treatment inhibited UVA-mediated induction of COX-2 in murine keratinocytes. Our current study suggests that FPP-3, which has been developed as a novel COX-2 inhibitor, has antioxidative properties by activating the Nrf2-ARE pathway. The dual function of this compound may provide a better strategy to block/attenuate the inflammation process and to alleviate ROS-associated inflammatory complications.
Collapse
Affiliation(s)
- Sarala Manandhar
- College of Pharmacy, Yeungnam University, 214-1 Dae-dong, Gyeongsan-si, Gyeongsangbuk-do 712-749, South Korea
| | | | | | | | | |
Collapse
|
19
|
Lee SK, Kim JH, Seo YM, Kim HCH, Kang MJ, Jeong HG, Lee ES, Jeong TC. In vitro characterization of the enzymes involved in the metabolism of 1-furan-2-yl-3-pyridin-2-yl-propenone, an anti-inflammatory propenone compound. Arch Pharm Res 2008; 31:764-70. [PMID: 18563359 DOI: 10.1007/s12272-001-1224-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 03/04/2008] [Accepted: 04/10/2008] [Indexed: 11/28/2022]
Abstract
Carbonyl reduction is a significant step in the phase I biotransformation of a great variety of aromatic, alicyclic and aliphatic carbonyl compounds. 1-Furan-2-yl-3-pyridin-2-yl-propenone (FPP-3) has been shown to have anti-inflammatory activity as it inhibits the production of nitric oxide and tumor necrosis factor-beta. In the present study, the metabolic fate and possible involvement of 11beta-hydroxysteroid dehydrogenase (11beta-HSD) and carbonyl reductase (CBR) in the metabolism of FPP-3 were investigated in rat liver subcellular fractions. When FPP-3 was incubated with rat liver subcellular fractions in the presence of beta-NADPH, two major peaks were detected by reduction on the propenone: M1 (1-furan-2-yl-3-pyridin-2-yl-propan-1-one) and M2 (1-furan-2-yl-3-pyridin-2-yl-propan-1-ol). Inhibitors of CBR, such as quercitrin, ethacrynic acid and menadione, significantly increased the formation of M1, but effectively inhibited the formation of M2 in subcellular fractions. Meanwhile, 18beta-glycyrrhetinic acid, a selective inhibitor of 11beta-HSD, marginally inhibited the reduction of FPP-3 in microsomes. A good correlation was observed between the formation of M2 and CBR activity with either 4-pyridine carboxaldehyde (r=0.72) or D,L-glyceraldehyde (r=0.63) as substrates in the cytosol. These results indicated that FPP-3 might be metabolized by cytosolic CBR and uncharacterized microsomal reductase(s) in rat liver.
Collapse
Affiliation(s)
- Sang Kyu Lee
- Doping Control Center, Korea Institute of Science and Technology, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Hwang YP, Han EH, Choi JH, Kim HG, Lee KJ, Jeong TC, Lee ES, Jeong HG. Chemopreventive effects of Furan-2-yl-3-pyridin-2-yl-propenone against 7,12-dimethylbenz[a]anthracene-inducible genotoxicity. Toxicol Appl Pharmacol 2008; 228:343-50. [DOI: 10.1016/j.taap.2007.12.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Revised: 12/13/2007] [Accepted: 12/14/2007] [Indexed: 10/22/2022]
|
21
|
Lee SK, Jeong HG, Lee ES, Jeong TC. Metabolism of FPP-3, an anti-inflammatory propenone compound, in rat by liquid chromatography-electrospray ionization tandem mass spectrometry. Biol Pharm Bull 2007; 30:967-71. [PMID: 17473444 DOI: 10.1248/bpb.30.967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
1-Furan-2-yl-3-pyridin-2-yl-propenone (FPP-3) is an anti-inflammatory agent with a propenone moiety. Following a single intravenous injection of male Sprague-Dawley rats with 4 mg/kg of FPP-3, three different metabolites of FPP-3 were identified as M1 (1-furan-2-yl-3-pyridin-2-yl-propan-1-one), M2 (1-furan-2-yl-3-pyridin-2-yl-propan-1-ol) and M3 (a glucuronide conjugate of M2) in rat urine by a liquid chromatography-electrospray tandem mass spectrometry. The structures of M1 and M2 were the same as observed previously following the incubation of rat liver microsomes with FPP-3 in the presence of NADPH. Although all metabolites of FPP-3 were identified in rat urine, only M1 and M2 were observed in the bile and feces. In addition, FPP-3 and its metabolites were mostly excreted into the urine. The M3 was identified as a glucuronide conjugate of M2 because of the addition of 176 Da from the protonated molecular ion of M2 in MS(2) and because of the production of free M2 following an incubation of urine with beta-glucuronidase. From these studies, a possible metabolic fate of FPP-3 could be proposed in vivo.
Collapse
Affiliation(s)
- Sang Kyu Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, Korea
| | | | | | | |
Collapse
|
22
|
Park BC, Thapa D, Lee YS, Kwak MK, Lee ES, Choi HG, Yong CS, Kim JA. 1-furan-2-yl-3-pyridin-2-yl-propenone inhibits the invasion and migration of HT1080 human fibrosarcoma cells through the inhibition of proMMP-2 activation and down regulation of MMP-9 and MT1-MMP. Eur J Pharmacol 2007; 567:193-7. [PMID: 17507007 DOI: 10.1016/j.ejphar.2007.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2006] [Revised: 03/28/2007] [Accepted: 04/01/2007] [Indexed: 12/13/2022]
Abstract
Matrix metalloproteinases (MMPs) play important roles in solid tumor invasion and migration. In this study, we showed that 1-furan-2-yl-3-pyridin-2-yl-propenone (FPP-3) dose-dependently inhibited HT1080 cell invasion and migration, and decreased MMP-2 and MMP-9 activities. Furthermore, FPP-3 reduced MMP-2 expression at protein and mRNA levels, and suppressed 12-O-tetradecanoylphorbol-13-acetate (TPA)-enhanced expression of MT1-MMP without changing tissue inhibitors of metalloproteinase (TIMP)-2 level. FPP-3 also suppressed TPA-induced increases in MMP-9 protein and mRNA levels, but did not alter TIMP-1 level. Our results suggest that FFP-3 may be a valuable anti-invasive drug candidate for cancer therapy by suppressing MMP-2, MMP-9, and MT1-MMP.
Collapse
Affiliation(s)
- Byung Chul Park
- College of Pharmacy, Yeungnam University, 214-1 Dae-dong, Gyeongsan 712-749, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Lee SK, Jeon TW, Basnet A, Jeong HG, Lee ES, Jeong TC. Identification of 1-furan-2-yl-3-pyridin-2-yl-propenone, an anti-inflammatory agent, and its metabolites in rat liver subcellular fractions. Arch Pharm Res 2006; 29:984-9. [PMID: 17146967 DOI: 10.1007/bf02969282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1-Furan-2-yl-3-pyridin-2-yl-propenone (FPP-3) has been characterized to have an anti-inflammatory activity through the inhibition of the production of nitric oxide and tumor necrosis factor-alpha. In the present studies, the phase 1 metabolism of FPP-3 was investigated in rat liver microsomes and cytosols. When FPP-3 was incubated with rat liver microsomes and cytosols in the presence of NADPH, 2 major peaks were detected on a liquid chromatography/electrospray ionization-mass spectrometry. Two metabolites (i.e., M1 and M2) were characterized as reduced forms on propenone: M1 (1-furan-2-yl-3-pyridin-2-yl-propan-1-one) was the initial metabolite and M2 (1-furan-2-yl-3-pyridin-2-yl-propan-l-ol) was a secondary alcohol believed to be formed from M1.
Collapse
Affiliation(s)
- Sang Kyu Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Korea
| | | | | | | | | | | |
Collapse
|
24
|
Lee SK, Jeon TW, Basnet A, Lee ES, Jeong TC. Determination of 1-furan-2-yl-3-pyridin-2-yl-propenone, an anti-inflammatory propenone compound, by high performance liquid chromatography with ultraviolet spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2006; 837:108-11. [PMID: 16687255 DOI: 10.1016/j.jchromb.2006.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 04/04/2006] [Accepted: 04/10/2006] [Indexed: 11/19/2022]
Abstract
1-Furan-2-yl-3-pyridin-2-yl-propenone (FPP-3) has recently been synthesized and characterized to have an anti-inflammatory activity. In the present study, pharmacokinetic parameters for FPP-3 and its metabolites were determined at the same time by using high-performance liquid chromatography-ultraviolet spectrometry. Two metabolites were detected in sera when FPP-3 was administered intravenously to male SD rats. The linearity of FPP-3, M1 (1-furan-2-yl-3-pyridin-2-yl-propan-1-one) and M2 (1-furan-2-yl-3-pyridin-2-yl-propan-1-ol) was confirmed in the concentration ranges of 0.5-20, 0.101-4.04 and 1.04-20.4 microg/ml, respectively. The lower limits of quantitation of FPP-3, M1 and M2 were 0.5, 0.1 and 1.0 microg/ml, respectively. The intra- and inter-day precision and accuracy over the concentration range of target compounds were within 13.5 and 14.2%, respectively. The half-lives of FPP-3, M1 and M2 were 16.3, 27.7 and 22.1 min, respectively.
Collapse
Affiliation(s)
- Sang Kyu Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, South Korea
| | | | | | | | | |
Collapse
|