1
|
Nagani A, Shah M, Patel S, Patel H, Parikh V, Patel A, Patel S, Patel K, Parmar H, Bhimani B, Yadav MR. Unveiling piperazine-quinoline hybrids as potential multi-target directed anti-Alzheimer's agents: design, synthesis and biological evaluation. Mol Divers 2025; 29:1453-1478. [PMID: 38990393 DOI: 10.1007/s11030-024-10927-4] [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: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Multi-target directed ligands (MTDLs) have recently been popularized due to their outstanding efficacy in combating the complicated features of Alzheimer's disease. This study details the synthesis of piperazine-quinoline-based MTDLs through a multicomponent Petasis reaction, targeting multiple factors such as AChE, BuChE, metal chelation to restore metal dyshomeostasis, and antioxidant activity. Some of the synthesized compounds exhibited notable inhibitory activity against AChE and BuChE enzymes at specific concentrations. Among the synthesized compounds compound (95) containing a 4-chloroaniline moiety and a 4-methoxybenzyl group displayed the most promising inhibitory activities against AChE (IC50 3.013 µM) and BuChE (IC50 = 3.144 µM). Compound (83) featuring 2-methoxyaniline and 4-fluorobenzyl substituents, exhibited the highest BuChE inhibition (IC50 1.888 µM). Notably, compound (79) demonstrated 93-times higher selectivity for BuChE over AChE. Molecular docking and molecular dynamics simulations were also performed to explore the binding modes and stability of these compounds with the AChE amd BuChE proteins. Further, kinetics study was performed against AChE for comounds (83 and 95) which indicated mixed inhibition of the enzyme by these compounds, Amongs the synthesized compounds, nine compounds were assessed for their antioxidant activity, displaying significant antioxidant properties with IC50 values ranging from 156 µM to 310 µM. Moreover, all the compounds demonstrated metal chelating tendency with Cu+2, Zn+2, Fe+2, Fe+3 and Al+3. This study provides insights into the design of novel MTDLs, highlighting compound (95) as a potential candidate for combating Alzheimer's disease.
Collapse
Affiliation(s)
- Afzal Nagani
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
- Research and Development Cell, Parul University, Vadodara, Gujarat, India
| | - Moksh Shah
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Salman Patel
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Harnisha Patel
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Vruti Parikh
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Ashish Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Anand, Gujarat, India
| | - Sagar Patel
- Sunnybrook Research Institute, Sunnybrook Health Science Centre, University of Toronto, Toronto, ON, Canada
| | - Kirti Patel
- Faculty of Pharmacy, The M.S University of Baroda, Vadodara, Gujarat, India
| | - Hardik Parmar
- Faculty of Pharmacy, The M.S University of Baroda, Vadodara, Gujarat, India
| | | | - Mange Ram Yadav
- Research and Development Cell, Parul University, Vadodara, Gujarat, India.
| |
Collapse
|
2
|
Ichale R, Kanhed AM, Vora A. Coumarin linked thiazole derivatives as potential α-glucosidase inhibitors to treat diabetes mellitus. Mol Divers 2024; 28:1239-1247. [PMID: 37118160 DOI: 10.1007/s11030-023-10652-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/14/2023] [Indexed: 04/30/2023]
Abstract
Diabetes is a leading cause of kidney failure, blindness, heart attacks and lower limb amputation. Prevalence of diabetes is rising globally. α-glucosidase is validated target for controlling hyperglycemia because of its role in catalysing hydrolysis of carbohydrates to glucose in GIT. In an attempt to find novel safe and effective α-glucosidase inhibitors, coumarin linked thiazole was identified as potential scaffold on the basis of its interactions with the active site of α-glucosidase studied in silico. A series of coumarin linked thiazole derivatives were synthesized and analyzed for α-glucosidase inhibitory potential in an in-vitro assay. The synthesized molecules showed potent inhibition of α-glucosidase with IC50 values ranging from 0.14 to 9.38 μM. The most potent compound 2-[(4-bromophenyl) amino)-N-(4- (2-oxo-2H-chromen-3-yl) thiazol-2-yl] acetamide was further docked with α-glucosidase and molecular dynamics studies were carried out for 100 ns which suggested the stability of protein and ligand in the protein active site over the simulation period and role of hydrophobic interactions slightly more than the electrostatic/polar interactions in ligand- receptor stability. In summary, our results demonstrate efficacy of coumarin-linked thiazole as potential leads for further optimization and development.
Collapse
Affiliation(s)
- Rushikesh Ichale
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM's NMIMS University, Vile Parle, Mumbai, 400056, India
| | - Ashish M Kanhed
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM's NMIMS University, Vile Parle, Mumbai, 400056, India.
| | - Amisha Vora
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM's NMIMS University, Vile Parle, Mumbai, 400056, India.
| |
Collapse
|
3
|
Afanasenko AM, Wu X, De Santi A, Elgaher WAM, Kany AM, Shafiei R, Schulze MS, Schulz TF, Haupenthal J, Hirsch AKH, Barta K. Clean Synthetic Strategies to Biologically Active Molecules from Lignin: A Green Path to Drug Discovery. Angew Chem Int Ed Engl 2024; 63:e202308131. [PMID: 37840425 DOI: 10.1002/anie.202308131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023]
Abstract
Deriving active pharmaceutical agents from renewable resources is crucial to increasing the economic feasibility of modern biorefineries and promises to alleviate critical supply-chain dependencies in pharma manufacturing. Our multidisciplinary approach combines research in lignin-first biorefining, sustainable catalysis, and alternative solvents with bioactivity screening, an in vivo efficacy study, and a structural-similarity search. The resulting sustainable path to novel anti-infective, anti-inflammatory, and anticancer molecules enabled the rapid identification of frontrunners for key therapeutic indications, including an anti-infective against the priority pathogen Streptococcus pneumoniae with efficacy in vivo and promising plasma and metabolic stability. Our catalytic methods provided straightforward access, inspired by the innate structural features of lignin, to synthetically challenging biologically active molecules with the core structure of dopamine, namely, tetrahydroisoquinolines, quinazolinones, 3-arylindoles and the natural product tetrahydropapaveroline. Our diverse array of atom-economic transformations produces only harmless side products and uses benign reaction media, such as tunable deep eutectic solvents for modulating reactivity in challenging cyclization steps.
Collapse
Affiliation(s)
- Anastasiia M Afanasenko
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747 AG, Groningen (the, Netherlands
| | - Xianyuan Wu
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747 AG, Groningen (the, Netherlands
| | - Alessandra De Santi
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747 AG, Groningen (the, Netherlands
| | - Walid A M Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Andreas M Kany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Roya Shafiei
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
| | | | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, 30625, Hannover, Germany
- Institute of Virology, Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
- Institute of Virology, Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany
| | - Katalin Barta
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747 AG, Groningen (the, Netherlands
- Institute for Chemistry, University of Graz, Heinrichstrasse 28/II, 8010, Graz, Austria
| |
Collapse
|
4
|
Plater MJ, Harrison WTA. Reactions of 4,5-difluoro-1,2-dinitrobenzene with amines in dimethylformamide or EtOH. JOURNAL OF CHEMICAL RESEARCH 2023. [DOI: 10.1177/17475198231154812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Substitution reactions of 1,2-difluoro-4,5-dinitrobenzene were explored in dimethylformamide alone and with KOH/H2O, Hünig’s base or Et3N and with Me2NHCl/Et3N in EtOH. The fluorine atoms were always displaced in preference to the nitro groups. Three compounds were prepared from these studies and were characterised by X-ray single crystal structure determinations.
Collapse
Affiliation(s)
- M John Plater
- Department of Chemistry, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
5
|
Patel KB, Patel DV, Patel NR, Kanhed AM, Teli DM, Gandhi B, Shah BS, Chaudhary BN, Prajapati NK, Patel KV, Yadav MR. Carbazole-based semicarbazones and hydrazones as multifunctional anti-Alzheimer agents. J Biomol Struct Dyn 2022; 40:10278-10299. [PMID: 34215173 DOI: 10.1080/07391102.2021.1942212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
With the aim to combat a multi-faceted neurodegenerative Alzheimer's disease (AD), a series of carbazole-based semicarbazide and hydrazide derivatives were designed, synthesized and assessed for their cholinesterase (ChE) inhibitory, antioxidant and biometal chelating activity. Among them, (E)-2-((9-ethyl-9H-carbazol-3-yl)methylene)-N-(pyridin-2-yl)hydrazinecarbothioamide (62) and (E)-2-((9-ethyl-9H-carbazol-3-yl)methylene)-N-(5-chloropyridin-2-yl)hydrazinecarbothioamide (63) emerged as the premier candidates with good ChE inhibitory activities (IC50 values of 1.37 µM and 1.18 µM for hAChE, IC50 values of 2.69 µM and 3.31 µM for EqBuChE, respectively). All the test compounds displayed excellent antioxidant activity (reduction percentage of DPPH values for compounds (62) and (63) were 85.67% and 84.49%, respectively at 100 µM concentration). Compounds (62) and (63) conferred specific copper ion chelating property in metal chelation study. Molecular docking studies of compounds (62) and (63) indicate strong interactions within the active sites of both the ChE enzymes. Besides that, these compounds also exhibited significant in silico drug-like pharmacokinetic properties. Thus, taken together, they can serve as a starting point in the designing of multifunctional ligands in pursuit of potential anti-AD agents that might further prevent the progression of ADs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kishan B Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Dushyant V Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Nirav R Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Ashish M Kanhed
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM's NMIMS University, Mumbai, India
| | - Divya M Teli
- Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Gujarat, India
| | - Bhumi Gandhi
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Bhavik S Shah
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Bharat N Chaudhary
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Navnit K Prajapati
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Kirti V Patel
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India.,Centre of Research for Development, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
6
|
Patel DV, Patel NR, Kanhed AM, Teli DM, Patel KB, Gandhi PM, Patel SP, Chaudhary BN, Shah DB, Prajapati NK, Patel KV, Yadav MR. Further Studies on Triazinoindoles as Potential Novel Multitarget-Directed Anti-Alzheimer's Agents. ACS Chem Neurosci 2020; 11:3557-3574. [PMID: 33073564 DOI: 10.1021/acschemneuro.0c00448] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The inadequate clinical efficacy of the present anti-Alzheimer's disease (AD) drugs and their low impact on the progression of Alzheimer's disease in patients have revised the research focus from single targets to multitarget-directed ligands. A novel series of substituted triazinoindole derivatives were obtained by introducing various substituents on the indole ring for the development of multitarget-directed ligands as anti-AD agents. The experimental data indicated that some of these compounds exhibited significant anti-AD properties. Among them, 8-(piperidin-1-yl)-N-(6-(pyrrolidin-1-yl)hexyl)-5H-[1,2,4]triazino[5,6-b]indol-3-amine (60), the most potent cholinesterase inhibitor (AChE, IC50 value of 0.32 μM; BuChE, IC50 value of 0.21 μM), was also found to possess significant self-mediated Aβ1-42 aggregation inhibitory activity (54% at 25 μM concentration). Additionally, compound 60 showed strong antioxidant activity. In the PAMPA assay, compound 60 exhibited blood-brain barrier penetrating ability. An acute toxicity study in rats demonstrated no sign of toxicity at doses up to 2000 mg/kg. Furthermore, compound 60 significantly restored the cognitive deficits in the scopolamine-induced mice model and Aβ1-42-induced rat model. In the in silico ADMET prediction studies, the compound satisfied all the parameters of CNS acting drugs. These results highlighted the potential of compound 60 to be a promising multitarget-directed ligand for the development of potential anti-AD drugs.
Collapse
Affiliation(s)
- Dushyant V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Nirav R. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Ashish M. Kanhed
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM’s NMIMS University, Vile Parle, Mumbai 400056, India
| | - Divya M. Teli
- Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380009 Gujarat, India
| | - Kishan B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Pallav M. Gandhi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Sagar P. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Bharat N. Chaudhary
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Dharti B. Shah
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Navnit K. Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Kirti V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390001 Gujarat, India
- Director (R & D), Centre of Research for Development, Parul University, Limbda, Waghodia Road, Vadodara, 391760 Gujarat, India
| |
Collapse
|
7
|
Kanhed AM, Patel DV, Patel NR, Sinha A, Thakor PS, Patel KB, Prajapati NK, Patel KV, Yadav MR. Indoloquinoxaline derivatives as promising multi-functional anti-Alzheimer agents. J Biomol Struct Dyn 2020; 40:2498-2515. [DOI: 10.1080/07391102.2020.1840441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ashish M. Kanhed
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKMs NMIMS University, Mumbai, India
| | - Dushyant V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Nirav R. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Anshuman Sinha
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Priyanka S. Thakor
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kishan B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Navnit K. Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kirti V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
8
|
Kumar V, Saha A, Roy K. In silico modeling for dual inhibition of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzymes in Alzheimer's disease. Comput Biol Chem 2020; 88:107355. [PMID: 32801088 DOI: 10.1016/j.compbiolchem.2020.107355] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 01/11/2023]
Abstract
In this research, we have implemented two-dimensional quantitative structure-activity relationship (2D-QSAR) modeling using two different datasets, namely, acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzyme inhibitors. A third dataset has been derived based on their selectivity and used for the development of partial least squares (PLS) based regression models. The developed models were extensively validated using various internal and external validation parameters. The features appearing in the model against AChE enzyme suggest that a small ring size, higher number of -CH2- groups, higher number of secondary aromatic amines and higher number of aromatic ketone groups may contribute to the inhibitory activity. The features obtained from the model against BuChE enzyme suggest that the sum of topological distances between two nitrogen atoms, higher number of fragments X-C(=X)-X, higher number of secondary aromatic amides, fragment R--CR-X may be more favorable for inhibition. The features obtained from selectivity based model suggest that the number of aromatic ethers, unsaturation content relative to the molecular size and molecular shape may be more specific for the inhibition of the AChE enzyme in comparison to the BuChE enzyme. Moreover, we have implemented the molecular docking studies using the most and least active molecules from the datasets in order to identify the binding pattern between ligand and target enzyme. The obtained information is then correlated with the essential structural features associated with the 2D-QSAR models.
Collapse
Affiliation(s)
- Vinay Kumar
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, 92 A P C Road, Kolkata 700 009, India
| | - Kunal Roy
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
9
|
Patel DV, Patel NR, Kanhed AM, Teli DM, Patel KB, Joshi PD, Patel SP, Gandhi PM, Chaudhary BN, Prajapati NK, Patel KV, Yadav MR. Novel carbazole-stilbene hybrids as multifunctional anti-Alzheimer agents. Bioorg Chem 2020; 101:103977. [PMID: 32485470 DOI: 10.1016/j.bioorg.2020.103977] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 01/03/2023]
Abstract
Molecules capable of engaging with multiple targets associated with pathological condition of Alzheimer's disease have proved to be potential anti-Alzheimer's agents. In our goal to develop multitarget-directed ligands for the treatment of Alzheimer's disease, a novel series of carbazole-based stilbene derivatives were designed by the fusion of carbazole ring with stilbene scaffold. The designed compounds were synthesized and evaluated for their anti-AD activities including cholinesterase inhibition, Aβ aggregation inhibition, antioxidant and metal chelation properties. Amongst them, (E)-1-(4-(2-(9-ethyl-9H-carbazol-3-yl)vinyl)phenyl)-3-(2-(pyrrolidin-1-yl)ethyl)thiourea (50) appeared to be the best candidate with good inhibitory activities against AChE (IC50 value of 2.64 μM) and BuChE (IC50 value of 1.29 μM), and significant inhibition of self-mediated Aβ1-42 aggregation (51.29% at 25 μM concentration). The metal chelation study showed that compound (50) possessed specific copper ion chelating property. Additionally, compound (50) exhibited moderate antioxidant activity. To understand the binding mode of 50, molecular docking studies were performed, and the results indicated strong non-covalent interactions of 50 with the enzymes in the active sites of AChE, BuChE as well as of the Aβ1-42 peptide. Additionally, it showed promising in silico ADMET properties. Putting together, these findings evidently showed compound (50) as a potential multitarget-directed ligand in the course of developing novel anti-AD drugs.
Collapse
Affiliation(s)
- Dushyant V Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Nirav R Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Ashish M Kanhed
- Shobhaben Pratapbhai Patel - School of Pharmacy & Technology Management, SVKM's NMIMS University, Vile Parle, Mumbai 400056, India
| | - Divya M Teli
- Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Kishan B Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Prashant D Joshi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Sagar P Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Pallav M Gandhi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Bharat N Chaudhary
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Navnit K Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Kirti V Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, Gujarat, India.
| |
Collapse
|
10
|
Mozaffarnia S, Teimuri-Mofrad R, Rashidi MR. Design, synthesis and biological evaluation of 2,3-dihydro-5,6-dimethoxy-1H-inden-1-one and piperazinium salt hybrid derivatives as hAChE and hBuChE enzyme inhibitors. Eur J Med Chem 2020; 191:112140. [PMID: 32088494 DOI: 10.1016/j.ejmech.2020.112140] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/09/2020] [Accepted: 02/09/2020] [Indexed: 12/15/2022]
Abstract
2,3-Dihydro-5,6-dimethoxy-2-[4-(4-alkyl-4-methylpiperazinium-1-yl)benzylidine]-1H-inden-1-one halide salt derivatives as a novel donepezil hybrid analogs with the property of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzyme inhibition were designed and synthesized via N-alkylation reaction of 2,3-dihydro-5,6-dimethoxy-2-[4-(4-methylpiperazin-1-yl)benzylidene]-1H-inden-1-one with some alkyl halides. Biological tests demonstrated that most of the synthesized compounds have moderate to good inhibitory activities effect on cholinesterase enzymes. Among them, 10e showed the best profile as a selected compound for inhibition of hAChE (IC50 = 0.32) and hBuChE (IC50 = 0.43 μM) enzymes. Kinetic analysis and molecular docking led to a better understanding of this compound. Kinetic studies disclosed that 10e inhibited acetylcholinesterase in mixed-type and butyrylcholinesterase in non-competitive type. The toxicity results showed that 10e is less toxic than donepezil and has better inhibitory activity against hBuChE when compared to donepezil or Galantamine. Other performed experiments revealed that 10e has an anti-β amyloid effect which is capable of reducing ROS, LDH and MDA also possing positive effect on TAC. On the other hand, it has shown a good anti-inflammation effect.
Collapse
Affiliation(s)
- Sakineh Mozaffarnia
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Teimuri-Mofrad
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.
| | - Mohammad-Reza Rashidi
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Thapa P, Palacios PM, Tran T, Pierce BS, Foss FW. 1,2-Disubstituted Benzimidazoles by the Iron Catalyzed Cross-Dehydrogenative Coupling of Isomeric o-Phenylenediamine Substrates. J Org Chem 2020; 85:1991-2009. [PMID: 31928002 DOI: 10.1021/acs.joc.9b02714] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Benzimidazoles are common in nature, medicines, and materials. Numerous strategies for preparing 2-arylbenzimidazoles exist. In this work, 1,2-disubstituted benzimidazoles were prepared from various mono- and disubstituted ortho-phenylenediamines (OPD) by iron-catalyzed oxidative coupling. Specifically, O2 and FeCl3·6H2O catalyzed the cross-dehydrogenative coupling and aromatization of diarylmethyl and dialkyl benzimidazole precursors. N,N'-Disubstituted-OPD substrates were significantly more reactive than their N,N-disubstituted isomers, which appears to be relative to their propensity for complexation and charge transfer with Fe3+. The reaction also converted N-monosubstituted OPD substrates to 2-substituted benzimidazoles; however, electron-poor substrates produce 1,2-disubstituted benzimidazoles by intermolecular imino-transfer. Kinetic, reagent, and spectroscopic (UV-vis and EPR) studies suggest a mechanism involving metal-substrate complexation, charge transfer, and aerobic turnover, involving high-valent Fe(IV) intermediates. Overall, comparative strategies for the relatively sustainable and efficient synthesis of 1,2-disubstituted benzimidazoles are demonstrated.
Collapse
Affiliation(s)
- Pawan Thapa
- Department of Chemistry and Biochemistry , The University of Texas Arlington , Arlington , Texas 76019-0065 , United States
| | - Philip M Palacios
- Department of Chemistry and Biochemistry , The University of Texas Arlington , Arlington , Texas 76019-0065 , United States
| | - Tam Tran
- Department of Chemistry and Biochemistry , The University of Texas Arlington , Arlington , Texas 76019-0065 , United States
| | - Brad S Pierce
- Department of Chemistry and Biochemistry , The University of Alabama , Tuscaloosa , Alabama 35487 , United States
| | - Frank W Foss
- Department of Chemistry and Biochemistry , The University of Texas Arlington , Arlington , Texas 76019-0065 , United States
| |
Collapse
|
12
|
Suthprasertporn N, Suwanna N, Thangnipon W. Protective effects of diarylpropionitrile against hydrogen peroxide-induced damage in human neuroblastoma SH-SY5Y cells. Drug Chem Toxicol 2019; 45:44-51. [PMID: 31495239 DOI: 10.1080/01480545.2019.1658768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oxidative stress is implicated in pathogenesis of neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. The study demonstrates diarylpropionitrile (DPN), an antioxidant selective agonist of estrogen receptor β, protected human neuroblastoma SH-SY5Y cells against H2O2-induced toxicity by attenuating production of reactive oxygen species, apoptosis, autophagy, NF-κB activation, MAPK p38, JNK and ERK 1/2 signaling pathways, and β-site amyloid precursor protein cleaving enzyme level, but, interestingly, stimulating Akt pathway. These findings indicate the important potential of DPN to ameliorate oxidative stress-associated damage in neurodegenerative disorders.
Collapse
Affiliation(s)
- Nopparat Suthprasertporn
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University , Salaya , Nakhonpathom , 73170 , Thailand
| | - Nirut Suwanna
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University , Kamphaeng Saen , Nakhonpathom , 73140 , Thailand
| | - Wipawan Thangnipon
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University , Salaya , Nakhonpathom , 73170 , Thailand
| |
Collapse
|
13
|
Patel DV, Patel NR, Kanhed AM, Patel SP, Sinha A, Kansara DD, Mecwan AR, Patel SB, Upadhyay PN, Patel KB, Shah DB, Prajapati NK, Murumkar PR, Patel KV, Yadav MR. Novel Multitarget Directed Triazinoindole Derivatives as Anti-Alzheimer Agents. ACS Chem Neurosci 2019; 10:3635-3661. [PMID: 31310717 DOI: 10.1021/acschemneuro.9b00226] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The multifaceted nature of Alzheimer's disease (AD) demands treatment with multitarget-directed ligands (MTDLs) to confront the key pathological aberrations. A novel series of triazinoindole derivatives were designed and synthesized. In vitro studies revealed that all the compounds showed moderate to good anticholinesterase activity; the most active compound 23e showed an IC50 value of 0.56 ± 0.02 μM for AChE and an IC50 value of 1.17 ± 0.09 μM for BuChE. These derivatives are also endowed with potent antioxidant activity. To understand the plausible binding mode of the compound 23e, molecular docking studies and molecular dynamics simulation studies were performed, and the results indicated significant interactions of 23e within the active sites of AChE as well as BuChE. Compound 23e successfully diminished H2O2-induced oxidative stress in SH-SY5Y cells and displayed excellent neuroprotective activity against H2O2 as well as Aβ-induced toxicity in SH-SY5Y cells in a concentration dependent manner. Furthermore, it did not show any significant toxicity in neuronal SH-SY5Y cells in the cytotoxicity assay. Compound 23e did not show any acute toxicity in rats at doses up to 2000 mg/kg, and it significantly reversed scopolamine-induced memory deficit in mice model. Additionally, compound 23e showed notable in silico ADMET properties. Taken collectively, these findings project compound 23e as a potential balanced MTDL in the evolution process of novel anti-AD drugs.
Collapse
Affiliation(s)
- Dushyant V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Nirav R. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Ashish M. Kanhed
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Sagar P. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Anshuman Sinha
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Deep D. Kansara
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Annie R. Mecwan
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Sarvangee B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Pragnesh N. Upadhyay
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Kishan B. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Dharti B. Shah
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Navnit K. Prajapati
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Prashant R. Murumkar
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Kirti V. Patel
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara-390001 Gujarat, India
| |
Collapse
|
14
|
Puangmalai N, Thangnipon W, Soi-Ampornkul R, Suwanna N, Tuchinda P, Nobsathian S. Neuroprotection of N-benzylcinnamide on scopolamine-induced cholinergic dysfunction in human SH-SY5Y neuroblastoma cells. Neural Regen Res 2017; 12:1492-1498. [PMID: 29089996 PMCID: PMC5649471 DOI: 10.4103/1673-5374.215262] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease, a progressive neurodegenerative disease, affects learning and memory resulting from cholinergic dysfunction. Scopolamine has been employed to induce Alzheimer's disease-like pathology in vivo and in vitro through alteration of cholinergic system. N-benzylcinnamide (PT-3), purified from Piper submultinerve, has been shown to exhibit neuroprotective properties against amyloid-β-induced neuronal toxicity in rat cortical primary cell culture and to improve spatial learning and memory of aged rats through alleviating oxidative stress. We proposed a hypothesis that PT3 has a neuroprotective effect against scopolamine-induced cholinergic dysfunction. PT-3 (125–200 nM) pretreatment was performed in human neuroblastoma SH-SY5Y cell line following scopolamine induction. PT-3 (125–200 nM) inhibited scopolamine (2 mM)-induced generation of reactive oxygen species, cellular apoptosis, upregulation of acetylcholinesterase activity, downregulation of choline acetyltransferase level, and activation of p38 and JNK signalling pathways. These findings revealed the underlying mechanisms of scopolamine-induced Alzheimer's disease-like cellular dysfunctions, which provide evidence for developing drugs for the treatment of this debilitating disease.
Collapse
Affiliation(s)
- Nicha Puangmalai
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, Thailand
| | - Wipawan Thangnipon
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, Thailand
| | - Rungtip Soi-Ampornkul
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nirut Suwanna
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Kampaeng Saen, Nakhonpathom, Thailand
| | | | - Saksit Nobsathian
- Nakhon Sawan Campus, Mahidol University, Phayuhakiri, Nakhon Sawan, Thailand
| |
Collapse
|
15
|
Yao D, Wang J, Wang G, Jiang Y, Shang L, Zhao Y, Huang J, Yang S, Wang J, Yu Y. Design, synthesis and biological evaluation of coumarin derivatives as novel acetylcholinesterase inhibitors that attenuate H2O2-induced apoptosis in SH-SY5Y cells. Bioorg Chem 2016; 68:112-23. [DOI: 10.1016/j.bioorg.2016.07.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/20/2016] [Accepted: 07/25/2016] [Indexed: 01/04/2023]
|
16
|
Gurung AB, Aguan K, Mitra S, Bhattacharjee A. Identification of molecular descriptors for design of novel Isoalloxazine derivatives as potential Acetylcholinesterase inhibitors against Alzheimer's disease. J Biomol Struct Dyn 2016; 35:1729-1742. [PMID: 27410776 DOI: 10.1080/07391102.2016.1192485] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In Alzheimer's disease (AD), the level of Acetylcholine (ACh) neurotransmitter is reduced. Since Acetylcholinesterase (AChE) cleaves ACh, inhibitors of AChE are very much sought after for AD treatment. The side effects of current inhibitors necessitate development of newer AChE inhibitors. Isoalloxazine derivatives have proved to be promising (AChE) inhibitors. However, their structure-activity relationship studies have not been reported till date. In the present work, various quantitative structure-activity relationship (QSAR) building methods such as multiple linear regression (MLR), partial least squares ,and principal component regression were employed to derive 3D-QSAR models using steric and electrostatic field descriptors. Statistically significant model was obtained using MLR coupled with stepwise selection method having r2 = .9405, cross validated r2 (q2) = .6683, and a high predictability (pred_r2 = .6206 and standard error, pred_r2se = .2491). Steric and electrostatic contribution plot revealed three electrostatic fields E_496, E_386 and E_577 and one steric field S_60 contributing towards biological activity. A ligand-based 3D-pharmacophore model was generated consisting of eight pharmacophore features. Isoalloxazine derivatives were docked against human AChE, which revealed critical residues implicated in hydrogen bonds as well as hydrophobic interactions. The binding modes of docked complexes (AChE_IA1 and AChE_IA14) were validated by molecular dynamics simulation which showed their stable trajectories in terms of root mean square deviation and molecular mechanics/Poisson-Boltzmann surface area binding free energy analysis revealed key residues contributing significantly to overall binding energy. The present study may be useful in the design of more potent Isoalloxazine derivatives as AChE inhibitors.
Collapse
Affiliation(s)
- Arun Bahadur Gurung
- a Computational Biology Laboratory, Department of Biotechnology and Bioinformatics , North-Eastern Hill University , Shillong 793022 , India
| | - Kripamoy Aguan
- b Molecular Biology Laboratory, Department of Biotechnology and Bioinformatics , North-Eastern Hill University , Shillong 793022 , India
| | - Sivaprasad Mitra
- c Center for Advanced Studies in Chemistry , North-Eastern Hill University , Shillong 793022 , India
| | - Atanu Bhattacharjee
- a Computational Biology Laboratory, Department of Biotechnology and Bioinformatics , North-Eastern Hill University , Shillong 793022 , India
| |
Collapse
|
17
|
Shidore M, Machhi J, Shingala K, Murumkar P, Sharma MK, Agrawal N, Tripathi A, Parikh Z, Pillai P, Yadav MR. Benzylpiperidine-Linked Diarylthiazoles as Potential Anti-Alzheimer’s Agents: Synthesis and Biological Evaluation. J Med Chem 2016; 59:5823-46. [DOI: 10.1021/acs.jmedchem.6b00426] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Mahesh Shidore
- Faculty
of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390 001, India
| | - Jatin Machhi
- Faculty
of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390 001, India
| | - Kaushik Shingala
- Faculty
of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390 001, India
| | - Prashant Murumkar
- Faculty
of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390 001, India
| | - Mayank Kumar Sharma
- Faculty
of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390 001, India
| | - Neetesh Agrawal
- Faculty
of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390 001, India
| | - Ashutosh Tripathi
- Zoology
Department, Faculty of Science, The Maharaja Sayajirao University of Baroda Vadodara, 390 001, India
| | - Zalak Parikh
- Zoology
Department, Faculty of Science, The Maharaja Sayajirao University of Baroda Vadodara, 390 001, India
| | - Prakash Pillai
- Zoology
Department, Faculty of Science, The Maharaja Sayajirao University of Baroda Vadodara, 390 001, India
| | - Mange Ram Yadav
- Faculty
of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, 390 001, India
| |
Collapse
|
18
|
Machhi J, Sinha A, Patel P, Kanhed AM, Upadhyay P, Tripathi A, Parikh ZS, Chruvattil R, Pillai PP, Gupta S, Patel K, Giridhar R, Yadav MR. Neuroprotective Potential of Novel Multi-Targeted Isoalloxazine Derivatives in Rodent Models of Alzheimer's Disease Through Activation of Canonical Wnt/β-Catenin Signalling Pathway. Neurotox Res 2016; 29:495-513. [PMID: 26797524 DOI: 10.1007/s12640-016-9598-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/06/2016] [Accepted: 01/08/2016] [Indexed: 01/04/2023]
Abstract
Previous reports suggest that Alzheimer's disease is protected by cholinesterase inhibitors. We synthesized some isoalloxazine derivatives and evaluated them using in vitro cholinesterase inhibition assay. Two of the compounds (7m and 7q) were figured out as potent cholinesterase inhibitors. They further showed anti-Aβ aggregatory activity in the in vitro assay. The current study deals with the evaluation of neuroprotective potentials of the potent compounds (7m and 7q) using different in vitro and in vivo experiments. The compounds were first assessed for their tendency to cross blood-brain barrier using in vitro permeation assay. They were evaluated using scopolamine-induced amnesic mice model. Additionally, ROS scavenging and anti-apoptotic properties of 7m and 7q were established against Aβ1-42-induced toxicity in rat hippocampal neuronal cells. 7m and 7q were also evaluated using Aβ1-42-induced Alzheimer's rat model. Lastly, their involvement in Wnt/β-catenin pathway was also demonstrated. The results indicated good CNS penetration for 7m and 7q. The neuroprotective effects of 7m and 7q were evidenced by improved cognitive ability in both scopolamine and Aβ1-42-induced Alzheimer's-like condition in rodents. The in vivo results also confirmed their anti-cholinesterase and anti-oxidant potential. Immunoblot results showed that treatment with 7m and 7q decreased Aβ1-42, p-tau, cleaved caspase-3, and cleaved PARP levels in Aβ1-42-induced Alzheimer's rat brain. Additionally, immunoblot results demonstrated that 7m and 7q activated the Wnt/β-catenin pathway as evidenced by increased p-GSK-3, β-catenin, and neuroD1 levels in Aβ1-42-induced Alzheimer's rat brain. These findings have shown that isoalloxazine derivatives (7m and 7q) could be the potential leads for developing effective drugs for the treatment of AD.
Collapse
Affiliation(s)
- Jatin Machhi
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India
| | - Anshuman Sinha
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India
| | - Pratik Patel
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India
| | - Ashish M Kanhed
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India
| | - Pragnesh Upadhyay
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India
| | - Ashutosh Tripathi
- Division of Neurobiology, Department of Zoology, Faculty of Science, The M. S. University of Baroda, Vadodara, Gujarat, 390002, India
| | - Zalak S Parikh
- Division of Neurobiology, Department of Zoology, Faculty of Science, The M. S. University of Baroda, Vadodara, Gujarat, 390002, India
| | - Ragitha Chruvattil
- Department of Biochemistry, Faculty of Science, The M. S. University of Baroda, Vadodara, Gujarat, 390002, India
| | - Prakash P Pillai
- Division of Neurobiology, Department of Zoology, Faculty of Science, The M. S. University of Baroda, Vadodara, Gujarat, 390002, India
| | - Sarita Gupta
- Department of Biochemistry, Faculty of Science, The M. S. University of Baroda, Vadodara, Gujarat, 390002, India
| | - Kirti Patel
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India
| | - Rajani Giridhar
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan, The M. S. University of Baroda, Vadodara, Gujarat, 390001, India.
| |
Collapse
|