1
|
Saadh MJ, Ahmed HH, Kareem RA, Jain V, Ballal S, Singh A, Sharma GC, Devi A, Nasirov A, Sameer HN, Yaseen A, Athab ZH, Adil M. In Silico design and molecular dynamics analysis of imidazole derivatives as selective cyclooxygenase-2 inhibitors. Comput Biol Chem 2025; 115:108341. [PMID: 39808951 DOI: 10.1016/j.compbiolchem.2025.108341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/08/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Cyclooxygenase-2 (COX-2), a key enzyme in the inflammatory pathway, is the target for various nonsteroidal anti-inflammatory drugs (NSAIDs) and selective inhibitors known as coxibs. This study focuses on the development of novel imidazole derivatives as COX-2 inhibitors, utilizing a Structure-Activity Relationship (SAR) approach to enhance binding affinity and selectivity. Molecular docking was performed using Autodock Vina, revealing binding energies of -6.928, -7.187, and -7.244 kJ/mol for compounds 5b, 5d, and 5e, respectively. Molecular dynamics simulations using GROMACS provided insights into the stability and conformational changes of the protein-ligand complexes. Key metrics such as RMSD, RMSF, Rg, SASA, and hydrogen bond analysis were employed to assess the interactions. The binding free energy of the inhibitors was estimated using the MMPBSA method, highlighting compound 5b (N-[(3-benzyl-2-methylsulfonylimidazol-4-yl)methyl]-4-methoxyaniline) with the lowest binding energy of -162.014 kcal/mol. ADMET analysis revealed that compound 5b exhibited the most favorable pharmacokinetic properties and safety profile. Overall, this investigation underscores the potential of these novel imidazole derivatives as effective COX-2 inhibitors, with compound 5b emerging as the most promising candidate for further development.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Vicky Jain
- Marwadi University Research Center, Department of Chemistry, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Abdulaziz Nasirov
- Department of Psychiatry, narcology and pediatric narcology, medical psychology and psychotherapy, Tashkent Pediatric Medical Institute, Bogishamol Street 223, Tashkent 100140, Uzbekistan
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
2
|
Rathod NV, Mishra S. Synthesis and Biological Evaluation of Bile Acid-Triclosan Conjugates: A Study on Antibacterial, Antibiofilm, and Molecular Docking. Bioconjug Chem 2025; 36:276-290. [PMID: 39841879 DOI: 10.1021/acs.bioconjchem.4c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/24/2025]
Abstract
This work describes the synthesis, characterization, and antibacterial properties of four bile acid-triclosan conjugates. The in vitro antibacterial activity of synthetic bile acid-triclosan conjugates was investigated against a panel of Gram-positive and Gram-negative bacteria. Conjugates 3 and 4 show high activity against Escherichia coli (ATCC25922), with IC50 values of 2.94 ± 0.7 and 1.51 ± 0.05 μM, respectively. Conjugate 4 demonstrated 9 times the activity of triclosan (6.77 μM) and 18 times the potency of kanamycin, a well-known antibiotic. Compound 3 showed higher potential activity against all evaluated strains, including Bacillus megaterium (IC50: 3.05 ± 0.02), Bacillus amyloquefaciens (IC50: 8.79 ± 0.01), Serratia marcescens (IC50: 6.77 ± 0.4), and E. coli (IC50: 1.51 ± 0.05 μM). These findings indicate that it has broad-spectrum antibacterial activity. Bile acid-triclosan conjugates prevent biofilms by up to 99% at low doses (conjugates 4; 4.16 ± 0.8 μM), compared to triclosan. Conjugate 5 was most potent against B. amyloquefaciens (IC50 = 5.23 ± 0.2 μM), while conjugate 4 was most effective against B. megaterium (IC50 = 4.16 ± 0.8 μM) in biofilm formation. These conjugates inhibit biofilm formation by limiting the extracellular polymeric substance generation. The in vitro antibacterial study revealed that bile acid-triclosan conjugates were more effective than the parent molecule triclosan at inhibiting bacterial growth and biofilm formation against both Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Neha V Rathod
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar 382426, India
| | - Satyendra Mishra
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar 382426, India
| |
Collapse
|
3
|
Salem MM, Gerges MN, Abd El Salam HA, Noser AA. New thiadiazolopyrimidine-ornamented pyrazolones as prospective anticancer candidates via suppressing VEGFR-2/PI3K/Akt signaling pathway: Synthesis, characterization, in-silico, and in-vitro studies. Int J Biol Macromol 2025; 289:138735. [PMID: 39689791 DOI: 10.1016/j.ijbiomac.2024.138735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024]
Abstract
New thiadiazolopyrimidine-ornamented pyrazolones (4a-8b) have been synthesized by a cyclocondensation reaction of 3a, b with different active methylene compounds. The structure of our products was confirmed via different physical and spectroscopic data. We assessed all newly thiadiazolopyrimidine-ornamented pyrazolones' potential to inhibit angiogenesis, metastasis, and cancer growth by utilizing in-silico investigations focused on the VEGFR-2 signaling pathway and elucidate their pharmacokinetic features using ADMET. Based on our results, compound 8b was chosen for in vitro evaluation since it had the highest binding energy of -10.252 kcal/mol using molecular docking. Compound 8b significantly damaged the T47D (IC50 = 33.01 ± 2.2 μM) cells, without any toxic effect on normal cells in comparison to chemotheraputic FDA approved drug cisplatin (Cis) (IC50 = 3.163 ± 1.7 μM). Additionally, compound 8b significantly suppressed the VEGFR-2 receptor protein that triggers the inhibition of PI3K/Akt genes which causes mitochondrial membrane malfunction resulting in Bax overexpression and Bcl-2 downregulation levels. Besides, compound 8b showed a notable decrease in the levels of nitric oxide (NO) production levels and arrested the cell cycle in the G0/G1 stage. These outcomes demonstrated that compound 8b adhered to Lipinski's rules and may serve as a potential candidate for future breast cancer treatments via obstructing the VEGFR-2/PI3K/Akt signaling pathway, which in turn prevents metastasis, angiogenesis, and proliferation.
Collapse
Affiliation(s)
- Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Marian N Gerges
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Hayam A Abd El Salam
- Green Chemistry Department, National Research Centre, Dokki, Giza 12622, Cairo, Egypt
| | - Ahmed A Noser
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
4
|
Eshal J, Tariq HZ, Li J, Aftab H, Şenol H, Taslimi P, Sadeghian N, Alharthy RD, Akram MS, Talib R, Shafiq Z. Synthesis, biological evaluation, and in silico studies of phenyl naphthalene-2-sulfonate derived thiosemicarbazones as potential carbonic anhydrase inhibitors. Bioorg Chem 2025; 155:108118. [PMID: 39793219 DOI: 10.1016/j.bioorg.2024.108118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/25/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025]
Abstract
A series of novel phenyl naphthalene-2-sulfonate-based thiosemicarbazones (5a-v) were synthesized and evaluated for their inhibitory activity against human carbonic anhydrases I and II (hCA I and hCA II). Compounds 5d and 5p demonstrated the highest inhibitory potency, with IC50 values of 4.32 ± 0.02 nM and 5.24 ± 0.03 nM for hCA I, and 3.89 ± 0.01 nM and 4.72 ± 0.01 nM for hCA II, respectively. Notably, compound 5d exhibited superior potency compared to the reference drug acetazolamide. The structure-activity relationship (SAR) analysis revealed that electron-withdrawing groups, particularly the dichlorophenyl group in 5d and 5p, enhanced inhibitory activity. Molecular docking and molecular dynamics simulations confirmed the high binding affinity of compound 5d, with docking scores of -9.7 kcal/mol for hCA I and -9.5 kcal/mol for hCA II. Stability in MD simulations further supported its potent inhibitory action. ADMET predictions suggested that compounds 5d and 5p have favorable pharmacokinetic profiles. In conclusion, phenyl naphthalene-2-sulfonate-based thiosemicarbazones, especially compound 5d, show strong potential as therapeutic agents targeting hCA I and hCA II.
Collapse
Affiliation(s)
- Javeria Eshal
- Institute of Chemical Sciences, Bahauddin Zakariya University, 60800 Multan, Pakistan
| | - Hafiza Zara Tariq
- School of Chemistry, Xi'an Jiaotong University, Xianning West Road, Xi'an 710049, China
| | - Jing Li
- School of Chemistry, Xi'an Jiaotong University, Xianning West Road, Xi'an 710049, China
| | - Hina Aftab
- Institute of Chemical Sciences, Bahauddin Zakariya University, 60800 Multan, Pakistan
| | - Halil Şenol
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 34093 Fatih, İstanbul, Turkey
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, 74110 Bartin, Turkey.
| | - Nastaran Sadeghian
- Department of Biotechnology, Faculty of Science, Bartin University, 74110 Bartin, Turkey
| | - Rima D Alharthy
- Department of Chemistry, Science & Arts College, Rabigh Branch, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| | - Muhammad Safwan Akram
- School of Science & Health, Teesside University, Middlesbrough TS1 3BA, UK; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington DL1 1HG, UK
| | - Rimsha Talib
- Institute of Chemical Sciences, Bahauddin Zakariya University, 60800 Multan, Pakistan
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, 60800 Multan, Pakistan.
| |
Collapse
|
5
|
Poustforoosh A. Optimizing kinase and PARP inhibitor combinations through machine learning and in silico approaches for targeted brain cancer therapy. Mol Divers 2025:10.1007/s11030-025-11114-9. [PMID: 39841319 DOI: 10.1007/s11030-025-11114-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/30/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
The drug combination is an attractive approach for cancer treatment. PARP and kinase inhibitors have recently been explored against cancer cells, but their combination has not been investigated comprehensively. In this study, we used various drug combination databases to build ML models for drug combinations against brain cancer cells. Some decision tree-based models were used for this purpose. The results were further evaluated using molecular docking and molecular dynamics (MD) simulation. The possibility of the hit drug combinations for crossing the Blood-brain barrier (BBB) was also examined. Based on the obtained results, the combination of niraparib, as the PARP inhibitor, and lapatinib, as the kinase inhibitor, exhibited more considerable outcomes with a remarkable model performance (accuracy of 0.915) and prediction confidence of 0.92. The protein tweety homolog 3 and BTB/POZ domain-containing protein 2 are the main targets of niraparib and lapatinib with - 10.2 and - 8.5 scores, respectively. Due to the outcomes, this drug combination can use the CAT1 transporter on the BBB surface and effectively cross the BBB. Based on the obtained results, niraparib-lapatinib can be a promising drug combination candidate for brain cancer treatment. This combination is worth to be examined by experimental investigation in vitro and in vivo.
Collapse
Affiliation(s)
- Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
6
|
Belal A, Abdou A, Miski SF, Ali MAM, Ghamry HI, Obaidullah AJ, Zaky MY, Hassan AHE, Roh EJ, Al-Karmalawy AA, Ibrahim MH. Exploring the potential of some natural indoles as antiviral agents: quantum chemical analysis, inverse molecular docking, and affinity calculations. Front Chem 2025; 12:1521298. [PMID: 39886558 PMCID: PMC11779707 DOI: 10.3389/fchem.2024.1521298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/01/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025] Open
Abstract
Human immunodeficiency virus (HIV) and hepatitis C virus (HCV) infections represent critical global health challenges due to the high morbidity and mortality associated with co-infections. HIV, the causative agent of acquired immunodeficiency syndrome (AIDS), infects 4,000 people daily, potentially leading to 1.2 million new cases by 2025, while HCV chronically affects 58 million people, causing cirrhosis and hepatocellular carcinoma. Indole-based compounds play a crucial role in antiviral drug development due to their "privileged scaffold" structure. This study investigates the antiviral potential of natural indoles, gardflorine A-C, derived from Gardneria multiflora Makino, a plant traditionally used to treat various ailments. We employed molecular docking, ADMET analysis, and computational techniques [frontier molecular orbital (FMO), natural bond orbital (NBO), and density functional theory (DFT)] to evaluate these compounds" potential as multi-target antiviral agents against HIV and HCV proteins.
Collapse
Affiliation(s)
- Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Aly Abdou
- Chemistry Department, Faculty of science, Sohag university, Sohag, Egypt
| | - Samar F. Miski
- Pharmacology and Toxicology Department, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Mohamed A. M. Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Heba I. Ghamry
- Nutrition and Food Science, Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmad J. Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Drug Exploration and Development Chair (DEDC), Department of Pharmaceutical Chemistry, College of Pharmacy, King Saudi University, Riyadh, Saudi Arabia
| | - Mohamed Y. Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, BeniSuef, Egypt
| | - Ahmed H. E. Hassan
- Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Eun Joo Roh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Ahmed A. Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad, Iraq
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| | - Mona H. Ibrahim
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
7
|
Rehman MU, He F, Shu X, Guo J, Liu Z, Cao S, Long S. Antibacterial and antifungal pyrazoles based on different construction strategies. Eur J Med Chem 2025; 282:117081. [PMID: 39608204 DOI: 10.1016/j.ejmech.2024.117081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/01/2024] [Revised: 11/05/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
The growing prevalence of microbial infections, and antimicrobial resistance (AMR) stemming from the overuse and misuse of antibiotics, call for novel therapeutic agents, particularly ones targeting resistant microbial strains. Scientists are striving to develop innovative agents to tackle the rising microbial infections and abate the risk of AMR. Pyrazole, a five-membered heterocyclic compound belonging to the azole family, is a versatile scaffold and serves as a core structure in many drugs with antimicrobial and other therapeutic effects. In this review, we have updated pyrazole-based antibacterial and antifungal agents mainly developed between 2016 and 2024, by combining with diverse pharmacophores such as coumarin, thiazole, oxadiazole, isoxazole, indole, etc. Meanwhile, the various strategies (molecular hybridization, bioisosterism, scaffold hopping, multicomponent reactions, and catalyst-free synthesis) for integrating different functional groups with the pyrazole ring are discussed. Additionally, structure-activity relationships of these pyrazole derivatives, i.e., how structural modifications impact their selectivity and therapeutic potential against bacterial and fungal strains, are highlighted. This review provides insights into designing next-generation antimicrobials to combat AMR, and offers valuable perspectives to the scientists working on heterocyclic compounds with diverse bioactivities.
Collapse
Affiliation(s)
- Muneeb Ur Rehman
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Hubei Engineering Research Center for Advanced Fine Chemicals, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, 206 1st Rd Optics Valley, East Lake New Technology Development District, Wuhan, Hubei, 430205, China
| | - Fang He
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Hubei Engineering Research Center for Advanced Fine Chemicals, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, 206 1st Rd Optics Valley, East Lake New Technology Development District, Wuhan, Hubei, 430205, China
| | - Xi Shu
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Hubei Engineering Research Center for Advanced Fine Chemicals, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, 206 1st Rd Optics Valley, East Lake New Technology Development District, Wuhan, Hubei, 430205, China
| | - Ju Guo
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Hubei Engineering Research Center for Advanced Fine Chemicals, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, 206 1st Rd Optics Valley, East Lake New Technology Development District, Wuhan, Hubei, 430205, China
| | - Ziwei Liu
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Hubei Engineering Research Center for Advanced Fine Chemicals, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, 206 1st Rd Optics Valley, East Lake New Technology Development District, Wuhan, Hubei, 430205, China
| | - Shuang Cao
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Hubei Engineering Research Center for Advanced Fine Chemicals, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, 206 1st Rd Optics Valley, East Lake New Technology Development District, Wuhan, Hubei, 430205, China.
| | - Sihui Long
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Hubei Engineering Research Center for Advanced Fine Chemicals, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, 206 1st Rd Optics Valley, East Lake New Technology Development District, Wuhan, Hubei, 430205, China.
| |
Collapse
|
8
|
Farooqi R, Ullah S, Khan A, Gurav SS, Mali SN, Aftab H, Al-Sadoon MK, Hsu MH, Taslimi P, Al-Harrasi A, Shafiq Z, Schenone S. Design, synthesis, in-vitro and in-silico studies of novel N-heterocycle based hydrazones as α-glucosidase inhibitors. Bioorg Chem 2025; 156:108155. [PMID: 39826499 DOI: 10.1016/j.bioorg.2025.108155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/17/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Diabetes mellitus has dominated the globe as a chronic health condition and has become a major global health concern. The inhibition of the key metabolic enzymes of carbohydrates digestion including α-amylase and α-glucosidase are the promising targets for the treatment of diabetes via delaying glucose absorption. Therefore, nitrogen containing saturated heterocycle (pyrrolidinyl, piperidinyl and N-methylpiperazinyl) based hydrazones derivatives 5-23 were synthesized through two step reactions and evaluated for their anti-diabetic potential. All compounds exhibited potent α-glucosidase inhibitory capability ranging (IC50 = 10.26-47.35 µM), as compared to acarbose (IC50 = 871.40 ± 1.24 µM). Interestingly these derivatives also exhibited significant inhibitory capability against α-amylase with IC50 values in the range 25.81-76.05 µM. Mechanistic study on the most potent compound indicated a competitive type of inhibition with a Ki value of 8.30 ± 0.0076 µM. Molecular docking was performed to predict binding interactions between receptor proteins and moiety. In QSAR analysis, through use of QSARINS different 1D and 2D descriptors were used to generate different models that enabled further identification of structural requirements that contributed to activity. pIC50 values were also predicted by QSAR model. Furthermore, in-silico ADMET and BOILED-egg model analysis showed that all analogues exhibited passive GI absorption, and all showed BBB penetration.
Collapse
Affiliation(s)
- Rehmatullah Farooqi
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800 Pakistan; National Changhua University of Education, Changhua 50007 Taiwan
| | - Saeed Ullah
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Oman; Department of Chemical and Biological Engineering, College of Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Shailesh S Gurav
- Department of Chemistry, VIVA College, Virar (W)-401303, Maharashtra, India
| | - Suraj N Mali
- School of Pharmacy, DY Patil Deemed to Be University, Navi Mumbai, India
| | - Hina Aftab
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800 Pakistan
| | - Mohammad Khalid Al-Sadoon
- Department of Zoology, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| | - Ming-Hua Hsu
- National Changhua University of Education, Changhua 50007 Taiwan
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, 74110 Bartin, Turkey
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Oman.
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800 Pakistan.
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, Genoa 16132, Italy.
| |
Collapse
|
9
|
Munir I, Batool Z, Khan F, Hussain J, Khan A, Mali SN, Radhakrishnan VV, Mathew B, Almutairi TM, Al-Harrasi A, Akram MS, Shafiq Z. Design, synthesis, in vitro, and in silico studies of novel isatin-hybrid hydrazones as potential triple-negative breast cancer agents. RSC Adv 2025; 15:948-965. [PMID: 39807200 PMCID: PMC11726183 DOI: 10.1039/d4ra07650h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/26/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Recent advances in cancer therapy have been made possible by monoclonal antibodies, domain antibodies, antibody drug conjugates, etc. The most impact has come from controlling cell cycle checkpoints through checkpoint inhibitors. This manuscript explores the potential of a series of novel N-benzyl isatin based hydrazones (5-25), which were synthesized and evaluated as anti-breast cancer agents. The synthesized hydrazones of N-benzyl isatin were screened in vitro against two cell lines, the MDA-MB-231 breast cancer cell line and the MCF-10A breast epithelial cell line. The results indicated that all compounds showed great potential against the triple-negative MDA-MB-231 breast cancer cell line. Compound 23 with nitro substitution at the 4th position of the phenyl ring exhibited significant antiproliferative potential for the MDA-MB-231 with an IC50 value of 15.8 ± 0.6 μM. Molecular dynamics and molecular docking simulations were performed to get a deeper understanding of the interactions between the synthesized compounds and cancer cells.
Collapse
Affiliation(s)
- Iqra Munir
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan-60800 Pakistan
| | - Zahra Batool
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan-60800 Pakistan
| | - Faizullah Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan KPK Pakistan
- Natural and Medical Sciences Research Centre, University of Nizwa P. O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Javid Hussain
- Department of Biological Sciences and Chemistry, University of Nizwa Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa P. O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
- Department of Chemical and Biological Engineering, College of Engineering, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Republic of Korea
| | - Suraj N Mali
- School of Pharmacy, DY Patil Deemed To Be University Navi Mumbai India
| | | | - Bijo Mathew
- Dept. Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham Cochin India
| | | | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa P. O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Muhammad Safwan Akram
- School of Science & Health, Teesside University Middlesbrough TS1 3BA UK
- National Horizons Centre, Teesside University 38 John Dixon Ln Darlington DL1 1HG UK
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan-60800 Pakistan
| |
Collapse
|
10
|
Khalid T, Malik A, Rasool N, Kanwal A, Nawaz H, Almas I. Cracking the code: the clinical and molecular impact of aminopyridines; a review (2019-2024). RSC Adv 2025; 15:688-711. [PMID: 39781020 PMCID: PMC11708541 DOI: 10.1039/d4ra07438f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/17/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Aminopyridines belong to a class of compounds that are monoamino and diamino derivatives of pyridine. They work primarily by blocking voltage-gated potassium channels in a dose-dependent manner. Essential heterocycles used extensively in synthetic, natural products, and medicinal chemistry are aminopyridine and its derivatives. A vast array of biological and pharmacological effects can result from the interaction of aminopyridine rings with different enzymes and receptors, due to their unique structural properties. Aminopyridine research is continually growing, and there are now greater expectations for how it may aid in the treatment of numerous disorders. This review article will serve as an innovative platform for researchers investigating aminopyridine compounds, intending thoroughly to examine both traditional and novel synthesis strategies in addition to investigating the various biological characteristics displayed by these adaptable heterocycles. We attempt to provide valuable insights that will contribute to further progress in the synthesis and utilization of aminopyridines in various fields.
Collapse
Affiliation(s)
- Tahira Khalid
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Ayesha Malik
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Nasir Rasool
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Aqsa Kanwal
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Hamna Nawaz
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Iffat Almas
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| |
Collapse
|
11
|
Abdelhalim WA, Rabee AR, Soliman SM, Hagar M, Moneer EA, Bakr BA, Barakat A, Haukka M, Rasheed HA. New formyl indole derivatives based on thiobarbituric acid and their nano-formulations; synthesis, characterization, parasitology and histopathology investigations. Sci Rep 2025; 15:299. [PMID: 39747136 PMCID: PMC11696224 DOI: 10.1038/s41598-024-81683-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/03/2024] [Accepted: 11/28/2024] [Indexed: 01/04/2025] Open
Abstract
New formyl indole derivatives based on thiobarbituric acid were designed for targeting parasitological applications. The new compounds (5-((1H-indol-3-yl)methylene)-2-thioxodihydropyrimidine-4,6(1H,5H)-dione (3a), and 5-((1-benzyl-1H-indol-3-yl)methylene)-2-thioxodihydropyrimidine-4,6(1H,5H)-dione (3b) were synthesized as thioxodihydropyrimidine derivatives via aldol condensation reaction. The structures of the synthesized compounds were confirmed based on their spectral data via FT-IR, 1H and 13C NMR spectral characterization. In addition, the structure of 3a is confirmed using X-ray crystallography. The synthesized compounds were prepared in nm scale via chitosan as a matrix, and their size was measured via scanning electronic microscope. Interestingly, the newly synthesized nano formulations show higher positive zeta potential (mV) values + 29.6 and + 26.1 for compounds NP-3a, and NP-3b; respectively. These compounds were tested for their parasitological activity. The results revealed that 3b had a great activity against cryptosporidium infection. Moreover, the nano formulation of compound 3b showed a significant reduction percent of oocyst count of cryptosporidium infected mice representing 66%. Furthermore, these compounds were screened by in-vitro hemolytic activity assay (IC50) values (cytotoxicity on RBCs) to assess their cytotoxic potentials and safety profiles.
Collapse
Affiliation(s)
- Walaa Ali Abdelhalim
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt
| | - Ahmed R Rabee
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt.
| | - Saied M Soliman
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt.
| | - Mohamed Hagar
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt.
| | - Esraa A Moneer
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21500, Egypt
| | - Basant A Bakr
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Assem Barakat
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Matti Haukka
- Department of Chemistry, University of Jyväskylä, P.O. Box 35, Jyväskylä, FI-40014, Finland
| | - Hanaa A Rasheed
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria, 21321, Egypt
| |
Collapse
|
12
|
Tahir S, Iqbal M, Shad S, Nisa S, Ibrar A, Nadeem A, Attia SM, Thebo KH, Ullah K. Biosynthesis of Zr-doped WO 3 nanoparticles: Evaluation of antibacterial, antioxidant, and enzymatic activities. Microb Pathog 2025; 198:107192. [PMID: 39622481 DOI: 10.1016/j.micpath.2024.107192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/15/2024]
Abstract
Herein, biocompatible pure tungsten oxide (WO3) and zirconium-doped tungsten oxide (Zr-doped WO3) nanoparticles (NPs) were prepared via a green approach from moringa plants with different doping concentrations (3, 5, and 7 %). The as-synthesized materials were morphologically and optically characterized using scanning electron microscopy (SEM), energy dispersive X-ray (EDX), X-ray diffraction (XRD), Fourier-transform infrared (FTIR), and ultraviolet-visible (UV-Vis) spectroscopy. The FTIR spectra clearly showed that two distinguishing bands at 603 and 674 cm-1 of WO3 were shifted to a higher wavenumber upon doping with zirconium. EDX analysis confirmed the successful synthesis of pure WO3 and Zr-doped WO3 by the green approach. The UV-Vis study exhibited that the bandgap of pure WO3 is blue-shifted upon Zr doping due to the Burstein-Moss effect. The XRD pattern revealed that the crystalline nature of WO3 is increased by increasing the Zr content. Further, the as-synthesized materials were evaluated for enzymatic, antibacterial, and antioxidant activities. The enzymatic results showed that 7 % of Zr-doped WO3 NPs have a higher activity for the α-amylase enzyme. Additionally, 7 % Zr-doped WO3 also showed better antioxidant activity, up to 85 % for free radical scavenging. The antibacterial performance of 7 % Zr-doped WO3 is higher as compared to other corresponding samples for different strains of bacteria. These results demonstrated that this facile and novel synthetic route will open a new door for designing an efficient nanomaterial for biomedical applications.
Collapse
Affiliation(s)
- Sana Tahir
- Department of Chemistry, Faculty of Physical and Applied Sciences, The University of Haripur, Haripur, 22620, KPK, Pakistan
| | - Muzaffar Iqbal
- Department of Chemistry, Faculty of Physical and Applied Sciences, The University of Haripur, Haripur, 22620, KPK, Pakistan.
| | - Salma Shad
- Department of Chemistry, Faculty of Physical and Applied Sciences, The University of Haripur, Haripur, 22620, KPK, Pakistan
| | - Sobia Nisa
- Department of Microbiology, Faculty of Biological and Biomedical Science, The University of Haripur, Haripur, 22620, KPK, Pakistan
| | - Aliya Ibrar
- Department of Chemistry, Faculty of Physical and Applied Sciences, The University of Haripur, Haripur, 22620, KPK, Pakistan.
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Khalid Hussain Thebo
- Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China.
| | - Kamran Ullah
- Department of Biology, Faculty of Biological and Biomedical Science, The University of Haripur, Haripur, 22620, KPK, Pakistan
| |
Collapse
|
13
|
Sujith P, Singh JR, Jayalakshmi S, Kandaswamy K, Shaik MR, Hussain SA, Kari ZA, Guru A. Metagenomics approaches in the discovery and development of new bioactive compound of 8-demethoxy-10-deoxysteffimycin from mangrove sediments. 3 Biotech 2024; 14:303. [PMID: 39564541 PMCID: PMC11570573 DOI: 10.1007/s13205-024-04137-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/05/2024] [Accepted: 10/19/2024] [Indexed: 11/21/2024] Open
Abstract
A metagenomic library consisting of 15,000 clones was constructed from the mangrove sediment. An antimicrobially active clone from the metagenomic library PS49 was identified by function- based screening. This paper presents the results of the biochemical characterization and metagenomic library screening of the marine-derived antibiotic, 8-demethoxy-10-deoxysteffimycin. Plasmid libraries were constructed, and clones were produced using a metagenomic approach. Out of 15,000 clones, 81 clones were screened for antimicrobial activity, and five potential clones were selected. The activity of one clone was characterized and named as PS49. The bioactive compounds from the selected clone were checked for antimicrobial, antioxidant, and anticancer activities. The clone PS49 was tested against various pathogens including bacteria and fungi and it showed inhibitory effects against all the tested pathogens. The antimicrobially active fractions were then crystallized and subjected to spectroscopic analysis such as FTIR, NMR and LC-MS analysis. The substance from clone PS49 has finally been recognized, and the compound from clone PS49 has been identified as 8-demethoxy-10-deoxysteffimycin. The substances isolated from the PS49 clone exhibited strong anticancer activity against skin cancer-cell lines SK-MEL2. The compounds showed a reduction in cell viability with an increase in the compound concentration. The compounds obtained from clone PS49 showed an IC50 value of 85 µg/ml.
Collapse
Affiliation(s)
- P Sujith
- Department of Microbiology, SRM Arts and Science College, Kattankulathur, Chengalpattu District, Tamil Nadu 603203 India
- C.A.S in Marine Biology, Annamalai University, Parangipettai, Tamil Nadu 608002 India
| | - J Rajesh Singh
- Department of Zoology, Annamalai University, Annamalai Nagar, Chidambaram, Tamil Nadu 608001 India
| | - S Jayalakshmi
- C.A.S in Marine Biology, Annamalai University, Parangipettai, Tamil Nadu 608002 India
| | - Karthikeyan Kandaswamy
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451 Saudi Arabia
| | - Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, P.O. Box - 2454, Riyadh, 11451 Saudi Arabia
| | - Zulhisyam Abdul Kari
- Department of Agricultural Sciences, Faculty of Agro‑Based Industry, Universiti Malaysia Kelantan, Jeli Campus, 17600 Jeli, Malaysia
- Advanced Livestock and Aquaculture Research Group, Faculty of Agro‑Based Industry, Universiti Malaysia Kelantan, Jeli Campus, 17600 Jeli, Malaysia
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
14
|
Abdalla Ali A, Mhamad SA, Hasan AH, Ahmad I, Abdullah SA, Jamil S, Patel H, Murugesan S, Jamalis J. Synthesis, biological evaluation and molecular modeling studies of modulated benzyloxychalcones as potential acetylcholinesterase inhibitors. J Biomol Struct Dyn 2024; 42:3604-3615. [PMID: 37293930 DOI: 10.1080/07391102.2023.2220032] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/16/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023]
Abstract
Acetylcholinesterase inhibitors (AChEIs) have become a significant target in the search for an efficient treatment of Alzheimer's disease. Chalcone-based compounds display a strong potency to hinder AChE. So, this study focused on the synthesis of a series of new chalcone derivatives with anti-cholinesterase potential and their structures were characterized based on spectroscopic methods including IR, 1H NMR, 13C NMR and HRMS. Chalcone derivatives were screened against AChE. Most of them exhibited potent inhibitory activity against AChE. Compound 11i showed the most potent activity toward acetylcholinesterase compared to the positive compound, Galantamine. Docking studies into the active site of the acetylcholinesterase enzyme ravealed the significant docking score of the synthesized compounds with docking score of -7.959 to -9.277 kcal/mol when compared to the co-crystallized ligand, Donepezil (-10.567 kcal/mol). The interaction's stability was further assessed using a conventional atomistic 100 ns dynamics simulation study, which revealed the conformational stability of representative compound 11i in the cavity of the acetylcholinesterase enzyme.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Arman Abdalla Ali
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Tafan Preparatory School, General Directorate of Education of Sulaimani, Sulaimani, Kurdistan, Iraq
| | - Shakhawan Ahmad Mhamad
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Department of Chemistry, College of Education, University of Sulaimani, Sulaimani, Kurdistan, Iraq
| | - Aso Hameed Hasan
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Department of Chemistry, College of Science, University of Garmian, Kalar, Kurdistan, Iraq
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Dhule, Maharashtra, India
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Siti Awanis Abdullah
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Shajarahtunnur Jamil
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Birla Institute of Technology & Science Pilani (BITS Pilani), Pilani, Rajasthan, India
| | - Joazaizulfazli Jamalis
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| |
Collapse
|
15
|
Recent advances on biologically active coumarin-based hybrid compounds. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/11/2023]
|
16
|
Hu CM, Luo YX, Wang WJ, Li JP, Li MY, Zhang YF, Xiao D, Lu L, Xiong Z, Feng N, Li C. Synthesis and Evaluation of Coumarin-Chalcone Derivatives as α-Glucosidase Inhibitors. Front Chem 2022; 10:926543. [PMID: 35832461 PMCID: PMC9271751 DOI: 10.3389/fchem.2022.926543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Coumarin and chalcone, two important kinds of natural product skeletons, both exhibit α-glucosidase inhibitory activity. In this work, coumarin-chalcone derivatives 3 (a∼v) were synthesized, and their α-glucosidase inhibitory activity was screened. The results showed that all synthetic derivatives (IC50: 24.09 ± 2.36 to 125.26 ± 1.18 μM) presented better α-glucosidase inhibitory activity than the parent compounds 3-acetylcoumarin (IC50: 1.5 × 105 μM) and the positive control acarbose (IC50: 259.90 ± 1.06 μM). Among them, compound 3t displayed the highest α-glucosidase inhibitory activity (IC50: 24.09 ± 2.36 μM), which was approximately 10 times stronger than that of acarbose. The kinetic assay of 3t (KI = 18.82 μM, KIS = 59.99 μM) revealed that these compounds inhibited α-glucosidase in a mixed-type manner. Molecular docking was used to simulate the interaction between α-glucosidase and compound 3t.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhuang Xiong
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| | - Na Feng
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| | - Chen Li
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| |
Collapse
|
17
|
Nasab NH, Azimian F, Kruger HG, Kim SJ. Coumarin‐Chalcones Generated from 3‐Acetylcoumarin as a Promising Agent: Synthesis and Pharmacological Properties. ChemistrySelect 2022. [DOI: 10.1002/slct.202200238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/05/2022]
Affiliation(s)
- Narges Hosseini Nasab
- Department of Biological Sciences Kongju National University Gongju, Chungnam 32588, Republic of Korea
| | - Fereshteh Azimian
- Department of Medicinal Chemistry School of Pharmacy Tabriz University of Medical Sciences Tabriz Iran
| | - Hendrik G. Kruger
- Catalysis and Peptide Research Unit School of Health Sciences University of KwaZulu-Natal Durban 4001 South Africa
| | - Song Ja Kim
- Department of Biological Sciences Kongju National University Gongju, Chungnam 32588, Republic of Korea
| |
Collapse
|
18
|
George G, Koyiparambath VP, Sukumaran S, Nair AS, Pappachan LK, Al-Sehemi AG, Kim H, Mathew B. Structural Modifications on Chalcone Framework for Developing New Class of Cholinesterase Inhibitors. Int J Mol Sci 2022; 23:ijms23063121. [PMID: 35328542 PMCID: PMC8953944 DOI: 10.3390/ijms23063121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/18/2022] Open
Abstract
Due to the multifaceted pharmacological activities of chalcones, these scaffolds have been considered one of the most privileged frameworks in the drug discovery process. Structurally, chalcones are α, β-unsaturated carbonyl functionalities with two aryl or heteroaryl units. Amongst the numerous pharmacological activities explored for chalcone derivatives, the development of novel chalcone analogs for the treatment of Alzheimer's disease (AD) is among the research topics of most interest. Chalcones possess numerous advantages, such as smaller molecular size, opportunities for further structural modification thereby altering the physicochemical properties, cost-effectiveness, and convenient synthetic methodology. The present review highlights the recent evidence of chalcones as a privileged structure in AD drug development processes. Different classes of chalcone-derived analogs are summarized for the easy understanding of the previously reported analogs as well as the importance of certain functionalities in exhibiting cholinesterase inhibition. In this way, this review will shed light on the medicinal chemistry fraternity for the design and development of novel promising chalcone candidates for the treatment of AD.
Collapse
Affiliation(s)
- Ginson George
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Vishal Payyalot Koyiparambath
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Sunitha Sukumaran
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Aathira Sujathan Nair
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Leena K. Pappachan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
| | - Abdullah G. Al-Sehemi
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Hoon Kim
- Department of Pharmacy, Sunchon National University, Suncheon 57922, Korea
- Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Korea
- Correspondence: (H.K.); (B.M.)
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682 041, India; (G.G.); (V.P.K.); (S.S.); (A.S.N.); (L.K.P.)
- Correspondence: (H.K.); (B.M.)
| |
Collapse
|
19
|
Verma A, Kumar Waiker D, Bhardwaj B, Saraf P, Shrivastava SK. The molecular mechanism, targets, and novel molecules in the treatment of Alzheimer's disease. Bioorg Chem 2021; 119:105562. [PMID: 34952243 DOI: 10.1016/j.bioorg.2021.105562] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2021] [Revised: 11/27/2021] [Accepted: 12/12/2021] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurological illness that causes dementia mainly in the elderly. The challenging obstacles related to AD has freaked global healthcare system to encourage scientists in developing novel therapeutic startegies to overcome with the fatal disease. The current treatment therapy of AD provides only symptomatic relief and to some extent disease-modifying effects. The current approach for AD treatment involves designing of cholinergic inhibitors, Aβ disaggregation inducing agents, tau inhibitors and several antioxidants. Hence, extensive research on AD therapy urgently requires a deep understanding of its pathophysiology and exploration of various chemical scaffolds to design and develop a potential drug candidate for the treatment. Various issues linked between disease and therapy need to be considered such as BBB penetration capability, clinical failure and multifaceted pathophisiology requires a proper attention to develop a lead candidate. This review article covers all probable mechanisms including one of the recent areas for investigation i.e., lipid dyshomeostasis, pathogenic involvement of P. gingivalis and neurovascular dysfunction, recently reported molecules and drugs under clinical investigations and approved by FDA for AD treatment. Our summarized information on AD will attract the researchers to understand and explore current status and structural modifications of the recently reported heterocyclic derivatives in drug development for AD therapy.
Collapse
Affiliation(s)
- Akash Verma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Digambar Kumar Waiker
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Bhagwati Bhardwaj
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Poorvi Saraf
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushant K Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India.
| |
Collapse
|
20
|
Synthesis and Spectral Characterisation of (E)-3-(3-(4 (Dimethylamino)Phenyl)Acrylo-yl)-4-Hydroxy-2H-Chromen-2-One and Their Antibacterial Activity and Acetylcholinesterase Inhibition. J CHEM-NY 2021. [DOI: 10.1155/2021/6101359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022] Open
Abstract
A new coumarin derivative, (E)-3-(3-(4-(dimethylamino) phenyl) acrylo-yl)-4-hydroxy-2H-chromen-2-one (3), was synthesized by the condensation of 3-acetyl-4-hydroxycoumarin (1) with 4-N,N-dimethylaminobenzaldehyde (2) in the presence of piperidine in ethanol. The structure of the synthesized compound was characterized using spectroscopic data (IR and 1H NMR) and elemental analysis. The antimicrobial properties and acetylcholinesterase inhibition activity (AChEI) of coumarin 3 were investigated, with the highest observed AChEI activity providing 48.25% inhibition. The electronic absorption and emission spectra revealed that 3 exists as two, main keto-enol tautomers. The ratios of these tautomers in both protic and aprotic solvents with different polarities and dielectric constants were calculated. The fluorescence of coumarin 3 was enhanced upon increasing the medium viscosity, which was due to the resultant molecular rigidity. This criterion was further investigated using DNA, whereby 3 showed enhanced fluorescence upon its uptake in DNA grooves and was therefore tested as a novel DNA fluorescent stain.
Collapse
|
21
|
Husain A, Balushi K A, Akhtar MJ, Khan SA. Coumarin linked heterocyclic hybrids: A promising approach to develop multi target drugs for Alzheimer's disease. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
|
22
|
Pawlaczyk M, Guć M, Schroeder G. Adsorption and selectivity studies of direct and magnetite-cored molecularly imprinted polymers (MIPs and magMIPs) towards chosen chalcones investigated with various analytical methods. RSC Adv 2021; 11:25334-25347. [PMID: 35478873 PMCID: PMC9036970 DOI: 10.1039/d1ra03391c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/30/2021] [Accepted: 07/15/2021] [Indexed: 11/21/2022] Open
Abstract
The following article presents a method for obtaining molecularly imprinted polymers (MIPs) dedicated to trans-chalcone (TC) and 2',4'-dihydroxy-3-methoxychalcone (DHMC). The synthetic protocol optimized with a choice of the most suitable functional monomer led to the synthesis of MIPs and their non-imprinted equivalents (NIP) performed via direct polymerization or on the surface of magnetite nanoparticles. The characterized materials were investigated for adsorption isotherms of TC and DHMC, which led to satisfactory values of maximal adsorption capacity, reaching 131.58 and 474.71 mg g-1, respectively. Moreover, all the polymers were studied for the adsorption kinetics and the selectivity towards four structurally different chalcones, which proved the proper selectiveness towards the template molecules. Also, the kinetic profiles of chalcones' adsorption on the synthesized MIPs showed a quasi-plateau reached already after 2 hours, indicating high sorption effectiveness. The studies involved the use of various analytical techniques, which afforded a comprehensive and reliable description of the materials' adsorption efficacy. It was found that the materials successfully bind the MIP-complementary analytes and also structurally similar chalcones, with slightly lower intensity.
Collapse
Affiliation(s)
- Mateusz Pawlaczyk
- Faculty of Chemistry, Adam Mickiewicz University in Poznań Uniwersytetu Poznańskiego 8 61-614 Poznań Poland +48 61 829 17 97
| | - Maria Guć
- Faculty of Chemistry, Adam Mickiewicz University in Poznań Uniwersytetu Poznańskiego 8 61-614 Poznań Poland +48 61 829 17 97
| | - Grzegorz Schroeder
- Faculty of Chemistry, Adam Mickiewicz University in Poznań Uniwersytetu Poznańskiego 8 61-614 Poznań Poland +48 61 829 17 97
| |
Collapse
|
23
|
Thapa P, Upadhyay SP, Suo WZ, Singh V, Gurung P, Lee ES, Sharma R, Sharma M. Chalcone and its analogs: Therapeutic and diagnostic applications in Alzheimer's disease. Bioorg Chem 2021; 108:104681. [PMID: 33571811 PMCID: PMC7928223 DOI: 10.1016/j.bioorg.2021.104681] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/23/2020] [Revised: 12/15/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
Chalcone [(E)-1,3-diphenyl-2-propene-1-one], a small molecule with α, β unsaturated carbonyl group is a precursor or component of many natural flavonoids and isoflavonoids. It is one of the privileged structures in medicinal chemistry. It possesses a wide range of biological activities encouraging many medicinal chemists to study this scaffold for its usefulness to oncology, infectious diseases, virology and neurodegenerative diseases including Alzheimer's disease (AD). Small molecular size, convenient and cost-effective synthesis, and flexibility for modifications to modulate lipophilicity suitable for blood brain barrier (BBB) permeability make chalcones a preferred candidate for their therapeutic and diagnostic potential in AD. This review summarizes and highlights the importance of chalcone and its analogs as single target small therapeutic agents, multi-target directed ligands (MTDLs) as well as molecular imaging agents for AD. The information summarized here will guide many medicinal chemist and researchers involved in drug discovery to consider chalcone as a potential scaffold for the development of anti-AD agents including theranostics.
Collapse
Affiliation(s)
- Pritam Thapa
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA.
| | - Sunil P Upadhyay
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
| | - William Z Suo
- Laboratory for Alzheimer's Disease & Aging Research, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
| | - Vikas Singh
- Division of Neurology, KCVA Medical Center, Kansas City, MO, USA
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, Iowa City, IA 52242, USA
| | - Eung Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Ram Sharma
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
| | - Mukut Sharma
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
| |
Collapse
|
24
|
Synthesis and neuroprotective effects of novel chalcone-triazole hybrids. Bioorg Chem 2020; 105:104384. [DOI: 10.1016/j.bioorg.2020.104384] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2020] [Revised: 09/16/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023]
|
25
|
Véras JH, do Vale CR, da Silva Lima DC, dos Anjos MM, Bernardes A, de Moraes Filho AV, e Silva CR, de Oliveira GR, Pérez CN, Chen-Chen L. Modulating effect of a hydroxychalcone and a novel coumarin–chalcone hybrid against mitomycin-induced genotoxicity in somatic cells of Drosophila melanogaster. Drug Chem Toxicol 2020; 45:775-784. [DOI: 10.1080/01480545.2020.1776314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jefferson Hollanda Véras
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| | - Camila Regina do Vale
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| | - Débora Cristina da Silva Lima
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| | | | - Aline Bernardes
- Chemistry Institute, Universidade Federal de Goiás, Goiânia, Brazil
| | - Aroldo Vieira de Moraes Filho
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| | - Carolina Ribeiro e Silva
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| | | | | | - Lee Chen-Chen
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia, Brazil
| |
Collapse
|
26
|
|
27
|
Design, synthesis, and in vitro evaluation of novel 1,3,4-oxadiazolecarbamothioate derivatives of Rivastigmine as selective inhibitors of BuChE. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02475-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2022]
|