1
|
Zala AR, Tiwari R, Naik HN, Ahmad I, Patel H, Jauhari S, Kumari P. Design and synthesis of pyrrolo[2,3-d]pyrimidine linked hybrids as α-amylase inhibitors: molecular docking, MD simulation, ADMET and antidiabetic screening. Mol Divers 2024; 28:1681-1695. [PMID: 37344700 DOI: 10.1007/s11030-023-10683-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
Novel pyrrolo[2,3-d]pyrimidine-based analogues were designed, synthesized, and evaluated for their ability to inhibit the α-amylase enzyme in order to treat diabetes. In vitro antidiabetic analysis demonstrated excellent antidiabetic action for compounds 5b, 6c, 7a, and 7b, with IC50 values in the 0.252-0.281 mM range. At a 200 μg/mL concentration, the exceptional percent inhibition values for compounds 5a, 5b, 5d, and 6a varied from 97.79 ± 2.86% to 85.56 ± 4.13% overperforming the standard (acarbose). Molecular docking of all compounds performed with Bacillus paralicheniformis α-amylase enzyme. The most active compounds via in vitro and non-toxic via in silico ADMET and molecular docking analysis, hybrids 6c, 7a, and 7b displayed binding affinity from - 8.2 and - 8.5 kcal/mol. Molecular dynamic simulations of most active compound 5b and 7a investigated into the active sites of the Bacillus paralicheniformis α-amylase enzyme for a 100-ns indicating the stability of hybrid-protein complex. Consistent RGyr values for the two complexes under study further suggest that the system's proteins are closely packed in the dynamic state. Synthesized analogs' in vitro biological assessments, ADMET, molecular docking, and MD modelling reveal that 5b, 6c, 7a, and 7b hybrid analogs may be employed in the development of future antidiabetic drugs.
Collapse
Affiliation(s)
- Ajayrajsinh R Zala
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, Gujarat, 395007, India
| | - Ramgopal Tiwari
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, Gujarat, 395007, India
| | - Hem N Naik
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, Gujarat, 395007, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, Maharashtra, 424002, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Smita Jauhari
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, Gujarat, 395007, India
| | - Premlata Kumari
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, Gujarat, 395007, India.
| |
Collapse
|
2
|
Chaudhary J, Sharma V, Jain A, Sharma D, Chopra B, Dhingra AK. A Profound Insight into the Structure-activity Relationship of Ubiquitous Scaffold Piperazine: An Explicative Review. Med Chem 2024; 20:17-29. [PMID: 37815177 DOI: 10.2174/0115734064244117230923172611] [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: 01/05/2023] [Revised: 08/02/2023] [Accepted: 08/23/2023] [Indexed: 10/11/2023]
Abstract
Despite extensive research in the field of drug discovery and development, still there is a need to develop novel molecular entities. Literature reveals a substantial heterocyclic nucleus named, piperazine, which shows an immense therapeutic voyage. For several decades, molecules having the piperazine nucleus have entered the market as a drug exhibiting biological potential. It was known to possess antipsychotic, antihistamine, antianginal, antidepressant, anticancer, antiviral, cardioprotective, and anti-inflammatory activity with a specific basis for structural activity relationship. Thus, it is regarded as a key structural feature in most of the already available therapeutic drugs in the market. Reports also suggest that the extensive utilization of these currently available drugs having a piperazine nucleus shows increasing tolerance significantly day by day. In addition to this, various other factors like solubility, low bioavailability, cost-effectiveness, and imbalance between pharmacokinetics and pharmacodynamics profile limit their utilization. Focusing on that issues, various structural modification studies were performed on the piperazine moiety to develop new derivatives/analogs to overcome the problems associated with available marketed drugs. Thus, this review article aims to gain insight into the number of structural modifications at the N-1 and N-4 positions of the piperazine scaffold. This SAR approach may prove to be the best way to overcome the above-discussed drawbacks and lead to the design of drug molecules with better efficacy and affinity. Hence, there is an urgent need to focus on the structural features of this scaffold which paves further work for deeper exploration and may help medicinal chemists as well as pharmaceutical industries.
Collapse
Affiliation(s)
- Jasmine Chaudhary
- Faculty of Pharmaceutical Sciences, M.M. College of Pharmacy, Maharishi Markandeshwer (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Vishal Sharma
- Faculty of Pharmaceutical Sciences, M.M. College of Pharmacy, Maharishi Markandeshwer (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Akash Jain
- Faculty of Pharmaceutical Sciences, M.M. College of Pharmacy, Maharishi Markandeshwer (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Diksha Sharma
- Research Scholar, Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, India
| | - Bhawna Chopra
- Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | | |
Collapse
|
3
|
de Oliveira LP, Florentino IF, Silva DPB, Pazini F, de Carvalho FS, Sanz G, Vaz BG, da Rocha FF, Fajemiroye JO, Ghedini PC, Lião LM, Menegatti R, Costa EA, de Oliveira TS. Anti-inflammatory, antinociceptive, and vasorelaxant effects of a new pyrazole compound 5-(1-(2-fluorophenyl)-1 H-pyrazol-4-yl)-1 H-tetrazole: role of NO/cGMP pathway and calcium channels. Can J Physiol Pharmacol 2023; 101:216-225. [PMID: 36866837 DOI: 10.1139/cjpp-2022-0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Molecular modification of compounds remains important strategy towards the discovery of new drugs. In this sense, this study presents a new pyrazole derivative 5-(1-(2-fluorophenyl)-1H-pyrazol-4-yl)-1H-tetrazole (LQFM039) and evaluated the anti-inflammatory, analgesic, and vasorelaxant effects of this compound as well the mechanisms of action involved in the pharmacological effects. For this, mice were orally treated with LQFM039 (17.5, 35, or 70 mg/kg) prior acetic acid-induced abdominal writhing, formalin, tail flick, and carrageenan-induced paw edema protocols. In addition, vascular reactivity protocols were made with aortic rings contraction with phenylephrine and stimulated with graded concentrations of LQFM039. Abdominal writhing and licking time in both neurogenic and inflammatory phases of formalin were reduced with LQFM039 without altering latency to nociceptive response in the tail flick test. Carrageenan-induced paw edema showed that LQFM039 reduces edema and cell migration. In addition, the mechanism of action of LQFM039 involves NO/cGMP pathway and calcium channels, since this new pyrazole derivate elicited concentration-dependent relaxation attenuated by Nω-nitro-l-arginine methyl ester and 1H-[1,2,4] oxadiazolo [4,3-alpha]quinoxalin-1-one, and blockade of CaCl2-induced contraction. Altogether, our finding suggests anti-inflammatory, antinociceptive, and vasorelaxant effect of this new pyrazole derivative with involvement of NO/cGMP pathway and calcium channels.
Collapse
Affiliation(s)
- Lanussy P de Oliveira
- Department of Pharmacology, ICB, Federal University of Goiás, Campus Samambaia, 74001-970, 314, Goiânia, GO, Brazil
| | - Iziara F Florentino
- Department of Pharmacology, ICB, Federal University of Goiás, Campus Samambaia, 74001-970, 314, Goiânia, GO, Brazil
| | - Daiany P B Silva
- Department of Pharmacology, ICB, Federal University of Goiás, Campus Samambaia, 74001-970, 314, Goiânia, GO, Brazil
| | - Francine Pazini
- Faculty of Pharmacy, Laboratory of Medicinal Pharmaceutical Chemistry, Federal University of Goiás, Goiânia, GO, Brazil
| | - Flávio S de Carvalho
- Chemistry Institute, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Germán Sanz
- Chemistry Institute, Laboratory of Chromatography and Mass Spectrometry-LaCEM, Federal University of Goiás, Goiânia, GO, Brazil
| | - Boniek G Vaz
- Chemistry Institute, Laboratory of Chromatography and Mass Spectrometry-LaCEM, Federal University of Goiás, Goiânia, GO, Brazil
| | - Fábio F da Rocha
- Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, Seropédica, RJ, Brazil
| | - James O Fajemiroye
- Department of Pharmacology, ICB, Federal University of Goiás, Campus Samambaia, 74001-970, 314, Goiânia, GO, Brazil
| | - Paulo C Ghedini
- Department of Pharmacology, ICB, Federal University of Goiás, Campus Samambaia, 74001-970, 314, Goiânia, GO, Brazil
| | - Luciano M Lião
- Chemistry Institute, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Ricardo Menegatti
- Faculty of Pharmacy, Laboratory of Medicinal Pharmaceutical Chemistry, Federal University of Goiás, Goiânia, GO, Brazil
| | - Elson A Costa
- Department of Pharmacology, ICB, Federal University of Goiás, Campus Samambaia, 74001-970, 314, Goiânia, GO, Brazil
| | - Thiago S de Oliveira
- Department of Pharmacology, ICB, Federal University of Goiás, Campus Samambaia, 74001-970, 314, Goiânia, GO, Brazil.,Department of Pharmacy, FCBS, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| |
Collapse
|
4
|
Martins AN, de Souza Almeida D, Florentino IF, da Silva Moreira LK, Turones LC, Batista DC, Machado LS, Vaz BG, Lião LM, de Almeida Ribeiro Oliveira G, Martins JLR, Fajemiroye JO, Menegatti R, Costa EA, da Silva DPB. Pharmacological evaluation of antinociceptive and anti-inflammatory activities of LQFM202: a new piperazine derivative. Inflammopharmacology 2023; 31:411-422. [PMID: 36443517 DOI: 10.1007/s10787-022-01103-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/12/2022] [Indexed: 11/30/2022]
Abstract
Advances have been made in the search for new multi-target modulators to control pain and inflammation. Therefore, compound 3,5-di-tert-butyl-4-hydroxyphenyl)(4-methylpiperazin-1-yl)methanone (LQFM202) was synthesised and evaluated. First, in vitro assays were performed for COX-1, COX-2, and 5-LOX enzymes. Subsequently, adult female Swiss albino mice treated orally with LQFM202 at doses of 25-200 mg/kg were subjected to acetic acid-induced writhing, formalin-induced pain, carrageenan-induced hyperalgesia, carrageenan- or zymosan-induced paw oedema, or pleurisy. LQFM202 inhibited COX-1, COX-2, and LOX-5 (IC50 = 3499 µM, 1565 µM, and 1343 µM, respectively). In acute animal models, LQFM202 (50, 100, or 200 mg/kg) decreased the amount of abdominal writhing (29%, 52% and 48%, respectively). Pain in the second phase of the formalin test was reduced by 46% with intermediate dose. LQFM202 (100 mg/kg) reduced the difference in nociceptive threshold in all 4 h evaluated (46%, 37%, 30%, and 26%, respectively). LQFM202 (50 mg/kg) decreased the carrageenan-oedema from the second hour (27%, 31% and 25%, respectively); however, LQFM202 (100 mg/kg) decreased the carrageenan-oedema in all hours evaluated (35%, 42%, 48% and 50%, respectively). When using zymosan, LQFM202 (50 mg/kg) decreased the oedema in all hours evaluated (33%, 32%, 31% and 20%, respectively). In the carrageenan-pleurisy test, LQFM202 (50 mg/kg) reduced significantly the number of polymorphonuclear cells (34%), the myeloperoxidase activity (53%), TNF-α levels (47%), and IL-1β levels (58.8%). When using zymosan, LQFM202 (50 mg/kg) reduced the number of polymorphonuclear and mononuclear cells (54% and 79%, respectively); and the myeloperoxidase activity (46%). These results suggest antinociceptive and anti-inflammatory effects of LQFM202.
Collapse
Affiliation(s)
- Aline N Martins
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Dionys de Souza Almeida
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Iziara F Florentino
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Lorrane K da Silva Moreira
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Larissa C Turones
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Daniel C Batista
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Lucas S Machado
- Chemistry Institute, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Boniek G Vaz
- Chemistry Institute, Laboratory of Chromatography and Mass Spectrometry, Federal University of Goiás, Goiânia, GO, Brazil
| | - Luciano M Lião
- Chemistry Institute, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | | | - José Luís Rodrigues Martins
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - James Oluwagbamigbe Fajemiroye
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Ricardo Menegatti
- Faculty of Pharmacy, Laboratory of Medicinal Pharmaceutical Chemistry, Federal University of Goiás, Goiânia, GO, Brazil
| | - Elson A Costa
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil
| | - Daiany P B da Silva
- Institute of Biological Sciences, Department of Pharmacology, Federal University of Goiás, Campus Samambaia, Goiânia, GO, Brazil.
| |
Collapse
|
5
|
Al-Ghorbani M, Gouda MA, Baashen M, Alharbi O, Almalki FA, Ranganatha LV. Piperazine Heterocycles as Potential Anticancer Agents: A Review. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02597-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Hanif M, Ameer N, Shehzad MA, Azeem M, Rana HL, Usman M. Improved anti-inflammatory effect of curcumin by designing self-emulsifying drug delivery system. Drug Dev Ind Pharm 2021; 47:1432-1438. [PMID: 34779318 DOI: 10.1080/03639045.2021.2001486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Purpose of present study was to prepare and evaluate self-emulsifying drug delivery system (SEDDS) of curcumin (Cur) to enhance its solubility and percentage release for the evaluation of anti-inflammatory effect. Curcumin loaded SEDDS formulation was prepared, and zones of self-emulsification were recognized by dilution method for the construction of phase diagram. Lauroglycol FCC, Tween 80 (surfactant), and Transcutol HP (co-surfactant) were selected based on their solubility and highest emulsion region in phase diagram. Thermodynamic stability of Cur-SEDDS was calculated through globule size, zeta potential, polydispersity index (PDI), viscosity and pH. Cur-SEDDS were also characterized by encapsulation efficiency (EE %), FT-IR, in vitro release, and in vivo anti-inflammatory effect. Results revealed that droplet size of Cur-SEDDS was 19.77 ± 0.03 nm with their PDI 0.22 ± 0.19, zeta potential -19.33 ± 0.94 and viscosity 25.68 ± 0.86 cp. EE % of Cur-SEDDS was found to be 94.99 ± 0.38%, percentage release 65.83% compared with pure curcumin powder. The designed formulation possesses significant anti-inflammatory activity in paw edema when compared with positive control in carrageenan induced rat paw edema assay. Newly developed Cur-SEDDS with enhanced curcumin solubility, percentage release and better anti-inflammatory action may be an alternative source of oral delivery of curcumin.
Collapse
Affiliation(s)
- Muhammad Hanif
- Department of Pharmaceutics Faculty of Pharmacy Bahauddin Zakariya University Multan
| | - Nabeela Ameer
- Department of Pharmaceutics Faculty of Pharmacy Bahauddin Zakariya University Multan
| | | | - Muhammad Azeem
- Department of Pharmaceutics Faculty of Pharmacy Bahauddin Zakariya University Multan
| | - Hafsa Latif Rana
- Department of Pharmaceutics Faculty of Pharmacy Bahauddin Zakariya University Multan
| | - Muhammad Usman
- Department of Pharmaceutics Faculty of Pharmacy Bahauddin Zakariya University Multan
| |
Collapse
|
7
|
Kadu R, Savani C, Roy H, Soni P, Singh AK, Vennapu DR, Singh VK. Impact of N-substituents on crystal packing of N-alkyl-N′-tosylpiperazines and development of new polymorph of tosylbis(2-(tosyloxy)ethyl)amine: Synthesis, DFT, photophysical, cytotoxic property. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
8
|
Approaches to the Synthesis of Dicarboxylic Derivatives of Bis(pyrazol-1-yl)alkanes. Molecules 2021; 26:molecules26020413. [PMID: 33466823 PMCID: PMC7829949 DOI: 10.3390/molecules26020413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
Carboxylation of bis(pyrazol-1-yl)alkanes by oxalyl chloride was studied. It was found that 4,4'-dicarboxylic derivatives of substrates with electron-donating methyl groups and short linkers (from one to three methylene groups) can be prepared using this method. Longer linkers lead to significantly lower product yields, which is probably due to instability of the intermediate acid chlorides that are initially formed in the reaction with oxalyl chloride. Thus, bis(pyrazol-1-yl)methane gave only monocarboxylic derivative even with a large excess of oxalyl chloride and prolonged reaction duration. An alternative approach involves the reaction of ethyl 4-pyrazolecarboxylates with dibromoalkanes in a superbasic medium (potassium hydroxide-dimethyl sulfoxide) and is suitable for the preparation of bis(4-carboxypyrazol-1-yl)alkanes with both short and long linkers independent of substitution in positions 3 and 5 of pyrazole rings. The obtained dicarboxylic acids are interesting as potential building blocks for metal-organic frameworks.
Collapse
|
9
|
Turones LC, Martins AN, Moreira LKDS, Fajemiroye JO, Costa EA. Development of pyrazole derivatives in the management of inflammation. Fundam Clin Pharmacol 2020; 35:217-234. [PMID: 33171533 DOI: 10.1111/fcp.12629] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 01/15/2023]
Abstract
The therapeutic limitations and poor management of inflammatory conditions are anticipated to impact patients negatively over the coming decades. Following the synthesis of the first pyrazole-antipyrine in 1887, several other derivatives have been screened for anti-inflammatory, analgesic, and antipyretic activities. Arguably, the pyrazole ring, as a major pharmacophore and central scaffold partly, defines the pharmacological profile of several derivatives. In this review, we explore the structural-activity relationship that accounts for the pharmacological profile of pyrazole derivatives and highlights future research perspectives capable of optimizing current advancement in the search for safe and efficacy anti-inflammatory drugs. The flourishing research into the pyrazole derivatives as drug candidates has advanced our understanding of inflammation-related diseases and treatment.
Collapse
Affiliation(s)
- Larissa Córdova Turones
- Laboratory of Pharmacology of Natural and Synthetic Products, Institute of Biological Sciences, Federal University of Goiás, Campus Samambaia, Goiânia, Goiás, 74001970, Brazil
| | - Aline Nazareth Martins
- Laboratory of Pharmacology of Natural and Synthetic Products, Institute of Biological Sciences, Federal University of Goiás, Campus Samambaia, Goiânia, Goiás, 74001970, Brazil
| | - Lorrane Kelle da Silva Moreira
- Laboratory of Pharmacology of Natural and Synthetic Products, Institute of Biological Sciences, Federal University of Goiás, Campus Samambaia, Goiânia, Goiás, 74001970, Brazil
| | - James Oluwagbamigbe Fajemiroye
- Laboratory of Pharmacology of Natural and Synthetic Products, Institute of Biological Sciences, Federal University of Goiás, Campus Samambaia, Goiânia, Goiás, 74001970, Brazil
| | - Elson Alves Costa
- Laboratory of Pharmacology of Natural and Synthetic Products, Institute of Biological Sciences, Federal University of Goiás, Campus Samambaia, Goiânia, Goiás, 74001970, Brazil
| |
Collapse
|
10
|
Anti-inflammatory and antinociceptive activity profile of a new lead compound - LQFM219. Int Immunopharmacol 2020; 88:106893. [PMID: 32892073 DOI: 10.1016/j.intimp.2020.106893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/31/2022]
Abstract
LQFM219 is a molecule designed from celecoxibe (COX-2 inhibitor) and darbufelone (inhibitor of COX-2 and 5-LOX) lead compounds through a molecular hybridisation strategy. Therefore, this work aimed to investigate the antinociceptive and anti-inflammatory activities of this new hybrid compound. The acute oral systemic toxicity of LQFM219 was evaluated via the neutral red uptake assay. Acetic acid-induced abdominal writhing and CFA-induced mechanical hyperalgesia were performed to evaluate the antinociceptive activity, and the anti-oedematogenic activity was studied by CFA-induced paw oedema and croton oil-induced ear oedema. Moreover, the acute anti-inflammatory activity was determined by carrageenan-induced pleurisy. In addition, cell migration, myeloperoxidase enzyme activity, and TNF-α and IL-1β levels were determined in pleural exudate. Moreover, a redox assay was conducted using electroanalytical and DPPH methods. The results demonstrated that LQFM219 was classified as GHS category 4, and it showed better free radical scavenger activity compared to BHT. Besides, LQFM219 decreased the number of writhings induced by acetic acid and the response to the mechanical stimulus in the CFA-induced mechanical hyperalgesia test. Furthermore, LQFM219 reduced oedema formation, cell migration, and IL-1β and TNF-α levels in the pleural cavity and inhibited myeloperoxidase enzyme activity. Thus, our study provides that the new pyrazole derivative, LQFM219, demonstrated low toxicity, antinociceptive and anti-inflammatory potential in vitro and in vivo.
Collapse
|
11
|
Molecular Insight into the Anti-Inflammatory Effects of the Curcumin Ester Prodrug Curcumin Diglutaric Acid In Vitro and In Vivo. Int J Mol Sci 2020; 21:ijms21165700. [PMID: 32784830 PMCID: PMC7461142 DOI: 10.3390/ijms21165700] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
Curcumin diglutaric acid (CurDG), an ester prodrug of curcumin, has the potential to be developed as an anti-inflammatory agent due to its improved solubility and stability. In this study, the anti-inflammatory effects of CurDG were evaluated. The effects of CurDG on inflammatory mediators were evaluated in LPS-stimulated RAW 264.7 macrophage cells. CurDG reduced the increased levels of NO, IL-6, and TNF- α, as well as iNOS and COX-2 expression in cells to a greater extent than those of curcumin, along with the potent inhibition of MAPK (ERK1/2, JNK, and p38) activity. The anti-inflammatory effects were assessed in vivo by employing a carrageenan-induced mouse paw edema model. Oral administration of CurDG demonstrated significant anti-inflammatory effects in a dose-dependent manner in mice. The effects were significantly higher compared to those of curcumin at the corresponding doses (p < 0.05). Moreover, 25 mg/kg curcumin did not exert a significant anti-inflammatory effect for the overall time course as indicated by the area under the curve data, while the equimolar dose of CurDG produced significant anti-inflammatory effects comparable with 50, 100, and 200 mg/kg curcumin (p < 0.05). Similarly, CurDG significantly reduced the proinflammatory cytokine expression in paw edema tissues compared to curcumin (p < 0.05). These results provide the first experimental evidence for CurDG as a promising anti-inflammatory agent.
Collapse
|
12
|
Sharma S, Kumar D, Singh G, Monga V, Kumar B. Recent advancements in the development of heterocyclic anti-inflammatory agents. Eur J Med Chem 2020; 200:112438. [DOI: 10.1016/j.ejmech.2020.112438] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023]
|
13
|
The peripheral corticotropin-releasing factor (CRF)-induced analgesic effect on somatic pain sensitivity in conscious rats: involving CRF, opioid and glucocorticoid receptors. Inflammopharmacology 2018; 26:305-318. [DOI: 10.1007/s10787-018-0445-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/23/2018] [Indexed: 12/30/2022]
|