1
|
Mushtaq A, Irfan M, Haq AU, Mansha A, Khan SG, Zahoor AF, Parveen B, Irfan A, Kotwica-Mojzych K, Glowacka M, Mojzych M. Novel transition metal-free synthetic protocols toward the construction of 2,3-dihydrobenzofurans: a recent update. Front Chem 2024; 12:1470861. [PMID: 39734577 PMCID: PMC11672212 DOI: 10.3389/fchem.2024.1470861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/31/2024] [Indexed: 12/31/2024] Open
Abstract
2,3-Dihydrobenzofurans are noteworthy scaffolds in organic and medicinal chemistry, constituting the structural framework of many of the varied medicinally active organic compounds. Moreover, a diverse variety of biologically potent natural products also contain this heterocyclic nucleus. Reflecting on the wide biological substantiality of dihydrobenzofurans, several innovative and facile synthetic developments are evolving to achieve these heterocycles. This review summarizes the transition-metal-free, efficient, and novel synthetic pathways toward constructing the dihydrobenzofuran nucleus established after 2020.
Collapse
Affiliation(s)
- Aqsa Mushtaq
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Irfan
- Department of Pharmaceutics, Government College University, Faisalabad, Pakistan
| | - Atta ul Haq
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Asim Mansha
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Samreen Gul Khan
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Bushra Parveen
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Katarzyna Kotwica-Mojzych
- Department of Basic Sciences, Department of Histology, Embriology and Cytophysiology, Medical University of Lublin, Lublin, Poland
| | - Mariola Glowacka
- Faculty of Health Sciences Collegium Medicum, The Mazovian Academy in Plock, Płock, Poland
| | - Mariusz Mojzych
- Faculty of Health Sciences Collegium Medicum, The Mazovian Academy in Plock, Płock, Poland
| |
Collapse
|
2
|
Mushtaq A, Ahmad MN, Zahoor AF, Kamal S, Ali KG, Javid J, Parveen B, Nazeer U, Bhat MA. Design, CTAB-catalyzed ultrasound-assisted synthesis and tyrosinase inhibition potential of naphthofuran-triazole conjugates. RSC Adv 2024; 14:37521-37538. [PMID: 39582937 PMCID: PMC11583631 DOI: 10.1039/d4ra05649c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/10/2024] [Indexed: 11/26/2024] Open
Abstract
The development of novel and efficient tyrosinase inhibitors is a critical necessity of agricultural, cosmetic and medicinal chemistry. Bearing in mind the therapeutical potential of naphthofuran-containing organic compounds, we carried out the CTAB-catalyzed ultrasound-assisted synthesis of a library of novel naphthofuran-triazole joined N-aryl/alkyl acetamides 20(a-j) in 74-92% yield, which were further assessed for their tyrosinase inhibitory potential by taking kojic acid and ascorbic acid as standard inhibitors. The tyrosinase inhibitory assay demonstrated the promising tyrosinase inhibiting tendency of all prepared derivatives 20(a-h) as they all were found to be more efficient in comparison to the standard kojic acid. Similarly, most of the derivatives also exhibited tyrosinase inhibition potency in juxtaposition to ascorbic acid. More specifically, among the catalog of compounds, 20f and 20i exhibited potent inhibition results with IC50 = 0.51 ± 0.12 and 1.99 ± 0.07, respectively. Overall, 20f was shown to be the most efficacious tyrosinase inhibitor, owing to the presence of an electronegative group, i.e., 2-chloro substitution on the phenyl ring. The tyrosinase inhibition activity results of 20f and 20i were further supplemented with molecular docking analysis to validate experimental studies. In silico modelling findings revealed their significant interactions with the tyrosinase protein (PDB ID: 5OAE), thereby illustrating the efficient docking score of -7.10 kcal mol-1 and -6.95 kcal mol-1 in comparison to kojic acid (-5.03 kcal mol-1).
Collapse
Affiliation(s)
- Aqsa Mushtaq
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Mirza Nadeem Ahmad
- Department of Applied Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Shagufta Kamal
- Department of Biochemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Kulsoom Ghulam Ali
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Jamila Javid
- Department of Chemistry, University of Sialkot Sialkot Pakistan
| | - Bushra Parveen
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Usman Nazeer
- Department of Chemistry, University of Houston 3585 Cullen Boulevard Texas 77204-5003 USA
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University Riyadh 11451 Saudi Arabia
| |
Collapse
|
3
|
Almas I, Malik A, Rasool N, Kanwal A, Khalid T, Nawaz H. Microwave-assisted protocol towards synthesis of heterocyclic molecules: a comparative analysis with conventional synthetic methodologies (years 2019-2023): a review. Mol Divers 2024:10.1007/s11030-024-10981-y. [PMID: 39302538 DOI: 10.1007/s11030-024-10981-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
Microwave-assisted protocols have become extensively accepted across various scientific and technological domains because of their numerous advantages, shorter reaction times, higher yields, and often milder reaction conditions. In this review, we focus on the synthesis of N, O, and S-containing heterocyclic structural cores, crucial in the development of pharmaceuticals, agrochemicals, and materials science following through conventional and microwave method via eliminating the side products and enhances the product yield that is nowadays the biggest barrier for a synthetic chemist. The major findings emphasizes the substantial advantages of microwave-assisted techniques over conventional synthetic protocols. This comparative study underscores the potential of microwave-assisted techniques to revolutionize heterocyclic compound synthesis, providing insights into optimizing reaction conditions and expanding the scope of chemical synthesis in industrial applications.
Collapse
Affiliation(s)
- Iffat Almas
- 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
| | - Tahira Khalid
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Hamna Nawaz
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| |
Collapse
|
4
|
Shu P, You G, Li W, Chen Y, Chu Z, Qin D, Wang Y, Zhou H, Zhao L. Cefmetazole sodium as an allosteric effector that regulates the oxygen supply efficiency of adult hemoglobin. J Biomol Struct Dyn 2024; 42:7442-7456. [PMID: 37555593 DOI: 10.1080/07391102.2023.2245043] [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: 03/15/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Allosteric effectors play an important role in regulating the oxygen supply efficiency of hemoglobin for blood storage and disease treatment. However, allosteric effectors that are approved by the US FDA are limited. In this study, cefmetazole sodium (CS) was found to bind adult hemoglobin (HbA) from FDA library (1338 compounds) using surface plasmon resonance imaging high-throughput screening. Using surface plasmon resonance (SPR), the interaction between CS and HbA was verified. The oxygen dissociation curve of HbA after CS interaction showed a significant increase in P50 and theoretical oxygen-release capacity. Acid-base sensitivity (SI) exhibited a decreasing trend, although not significantly different. An oxygen dissociation assay indicated that CS accelerated HbA deoxygenation. Microfluidic modulated spectroscopy showed that CS changed the ratio of the alpha-helix to the beta-sheet of HbA. Molecular docking suggested CS bound to HbA's β-chains via hydrogen bonds, with key amino acids being N282, K225, H545, K625, K675, and V544.The results of molecular dynamics simulations (MD) revealed a stable orientation of the HbA-CS complex. CS did not significantly affect the P50 of bovine hemoglobin, possibly due to the lack of Valβ1 and Hisβ2, indicating that these were the crucial amino acids involved in HbA's oxygen affinity. Competition between the 2,3-Diphosphoglycerate (2,3-DPG) and CS in the HbA interaction was also determined by SPR, molecular docking and MD. In summary, CS could interact with HbA and regulate the oxygen supply efficiency via forming stable hydrogen bonds with the β-chains of HbA, and showed competition with 2,3-DPG.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Peilin Shu
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Guoxing You
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Weidan Li
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Yuzhi Chen
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Zongtang Chu
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Dong Qin
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Ying Wang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Lian Zhao
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| |
Collapse
|
5
|
Mushtaq A, Zahoor AF, Ahmad S, Saif MJ, ul Haq A, Khan SG, Al-Mutairi AA, Irfan A, Al-Hussain SA, Zaki MEA. A Comprehensive Review on Benzofuran Synthesis Featuring Innovative and Catalytic Strategies. ACS OMEGA 2024; 9:20728-20752. [PMID: 38764672 PMCID: PMC11097366 DOI: 10.1021/acsomega.4c02677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/21/2024]
Abstract
Benzofurans have intrigued both pharmaceutical researchers and chemists owing to the medicinal usage of their derivatives against copious disease-causing agents (i.e., bacteria, viruses, and tumors). These heterocyclic scaffolds are pervasively encountered in a number of natural products and drugs. The ever-increasing utilization of benzofuran derivatives as pharmaceutical agents persuaded the chemists to devise novel and facile methodological approaches to assemble the biologically potent benzofuran nucleus. This review summarizes the current developments regarding the innovative synthetic routes and catalytic strategies to procure the synthesis of benzofuran heterocycles with their corresponding mechanistic details, reported by several research groups during 2021-2023.
Collapse
Affiliation(s)
- Aqsa Mushtaq
- Department
of Chemistry, Government College University
Faisalabad, 38000 Faisalabad, Pakistan
| | - Ameer Fawad Zahoor
- Department
of Chemistry, Government College University
Faisalabad, 38000 Faisalabad, Pakistan
| | - Sajjad Ahmad
- Department
of Chemistry, University of Engineering
and Technology Lahore, Faisalabad Campus, 38000 Faisalabad, Pakistan
| | - Muhammad Jawwad Saif
- Department
of Applied Chemistry, Government College
University Faisalabad, 38000 Faisalabad, Pakistan
| | - Atta ul Haq
- Department
of Chemistry, Government College University
Faisalabad, 38000 Faisalabad, Pakistan
| | - Samreen Gul Khan
- Department
of Chemistry, Government College University
Faisalabad, 38000 Faisalabad, Pakistan
| | - Aamal A. Al-Mutairi
- Department
of Chemistry, College of Science, Imam Mohammad
Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Ali Irfan
- Department
of Chemistry, Government College University
Faisalabad, 38000 Faisalabad, Pakistan
| | - Sami A. Al-Hussain
- Department
of Chemistry, College of Science, Imam Mohammad
Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Magdi E. A. Zaki
- Department
of Chemistry, College of Science, Imam Mohammad
Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| |
Collapse
|
6
|
Saeed S, Saif MJ, Zahoor AF, Tabassum H, Kamal S, Faisal S, Ashraf R, Khan SG, Nazeer U, Irfan A, Bhat MA. Discovery of novel 1,2,4-triazole tethered β-hydroxy sulfides as bacterial tyrosinase inhibitors: synthesis and biophysical evaluation through in vitro and in silico approaches. RSC Adv 2024; 14:15419-15430. [PMID: 38741974 PMCID: PMC11089527 DOI: 10.1039/d4ra01252f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
In this study, a series of 1,2,4-triazole-tethered β-hydroxy sulfide scaffolds 11a-h was synthesized in good to remarkable yields (69-90%) through the thiolysis of oxiranes by the thiols in aqueous basic catalytic conditions. The synthesized 1,2,4-triazole-tethered β-hydroxy sulfides were screened against bacterial tyrosinase enzyme, and Gram-positive and Gram-negative bacterial cultures i.e., (S. aureus) Staphylococcus aureus & (E. coli) Escherichia coli. Among the synthesized derivatives, the molecules 11a (IC50 = 7.67 ± 1.00 μM), 11c (IC50 = 4.52 ± 0.09 μM), 11d (IC50 = 6.60 ± 1.25 μM), and 11f (IC50 = 5.93 ± 0.50 μM) displayed the better tyrosinase inhibitory activity in comparison to reference drugs ascorbic acid (IC50 = 11.5 ± 1.00 μM) and kojic acid (IC50 = 30.34 ± 0.75 μM). The molecule benzofuran-triazol-propan-2-ol 11c proved to be the most potent bacterial tyrosinase inhibitory agent with a minimum IC50 of 4.52 ± 0.09 μM, as compared to other synthesized counterparts and both standards (kojic acid and ascorbic acid). The compound diphenyl-triazol-propan-2-ol 11a and benzofuran-triazole-propan-2-ol 11c showed comparable anti-bacterial chemotherapeutic efficacy with minimum inhibitory concentrations (MIC = 2.0 ± 2.25 mg mL-1 and 2.5 ± 0.00 mg mL-1, respectively) against S. aureus bacterial strain in comparison with standard antibiotic penicillin (MIC = 2.2 ± 1.15 mg mL-1). Furthermore, among the synthesized derivatives, only compound 11c demonstrated better anti-bacterial activity (MIC = 10 ± 0.40 mg mL-1) against E. coli, which was slightly less than the standard antibiotic i.e., penicillin (MIC = 2.4 ± 1.00 mg mL-1). The compound 11c demonstrated a better binding score (-7.08 kcal mol-1) than ascorbic acid (-5.59 kcal mol-1) and kojic acid (-5.78 kcal mol-1). Molecular docking studies also validate the in vitro anti-tyrosinase assay results; therefore, the molecule 11c can be the lead bacterial tyrosinase inhibitor as well as the antibacterial agent against both types of bacterial strains after suitable structural modifications.
Collapse
Affiliation(s)
- Sadaf Saeed
- Department of Chemistry, Government College University Faisalabad 38000-Faisalabad Pakistan
| | - Muhammad Jawwad Saif
- Department of Applied Chemistry, Government College University Faisalabad 38000-Faisalabad Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad 38000-Faisalabad Pakistan
| | - Hina Tabassum
- London Metropolitan University 166-220 Holloway Road London N7 8DB UK
| | - Shagufta Kamal
- Department of Biochemistry, Government College University Faisalabad 38000-Faisalabad Pakistan
| | - Shah Faisal
- Department of Chemistry, Islamia College University Peshawar Peshawar 25120 Pakistan
| | - Rabia Ashraf
- Department of Chemistry, Government College University Faisalabad 38000-Faisalabad Pakistan
| | - Samreen Gul Khan
- Department of Chemistry, Government College University Faisalabad 38000-Faisalabad Pakistan
| | - Usman Nazeer
- Department of Chemistry, University of Houston 3585 Cullen Boulevard Texas 77204-5003 USA
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad 38000-Faisalabad Pakistan
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University Riyadh 11451 Saudi Arabia
| |
Collapse
|
7
|
Alam MM, Alsenani NI, Abdelhamid AA, Ahmad A, Baothman OA, Hosawi SA, Altayeb H, Nadeem MS, Ahmad V, Nazreen S, Elhenawy AA. New paracetamol hybrids as anticancer and COX-2 inhibitors: Synthesis, biological evaluation and docking studies. Arch Pharm (Weinheim) 2024; 357:e2300340. [PMID: 37880869 DOI: 10.1002/ardp.202300340] [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] [Scholar Register] [Received: 06/23/2023] [Revised: 08/31/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023]
Abstract
Drug repurposing is an emerging field in drug development that has provided many successful drugs. In the current study, paracetamol, a known antipyretic and analgesic agent, was chemically modified to generate paracetamol derivatives as anticancer and anticyclooxygenase-2 (COX-2) agents. Compound 11 bearing a fluoro group was the best cytotoxic candidate with half-maximal inhibitory concentration (IC50 ) values ranging from 1.51 to 6.31 μM and anti-COX-2 activity with IC50 = 0.29 μM, compared to the standard drugs, doxorubicin and celecoxib. The cell cycle and apoptosis studies revealed that compound 11 possesses the ability to induce cell cycle arrest in the S phase and apoptosis in colon Huh-7 cells. These results were strongly supported by docking studies, which showed strong interactions with the amino acids of the COX-2 protein, and in silico pharmacokinetic predictions were found to be favorable for these newly synthesized paracetamol derivatives. It can be concluded that compound 11 could block cell growth and proliferation by inhibiting the COX-2 enzyme in cancer therapy.
Collapse
Affiliation(s)
- Mohammad Mahboob Alam
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Nawaf I Alsenani
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Antar A Abdelhamid
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Abrar Ahmad
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Othman A Baothman
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Salman A Hosawi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hisham Altayeb
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohammad Shahid Nadeem
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Varish Ahmad
- Department of Health Information Technology, Faculty of Applied Studies, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Syed Nazreen
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Ahmed A Elhenawy
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
8
|
Lv L, Shi Y, Deng Z, Xu J, Ye Z, He J, Chen G, Yu X, Wu J, Huang X, Li G. A polymeric nanocarrier that eradicates breast cancer stem cells and delivers chemotherapeutic drugs. Biomater Res 2023; 27:133. [PMID: 38102651 PMCID: PMC10722842 DOI: 10.1186/s40824-023-00465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Drug nanocarriers can markedly reduce the toxicities and side effects of encapsulated chemotherapeutic drugs in the clinic. However, these drug nanocarriers have little effect on eradicating breast cancer stem cells (BCSCs). Although compounds that can inhibit BCSCs have been reported, these compounds are difficult to use as carriers for the widespread delivery of conventional chemotherapeutic drugs. METHODS Herein, we synthesize a polymeric nanocarrier, hyaluronic acid-block-poly (curcumin-dithiodipropionic acid) (HA-b-PCDA), and explore the use of HA-b-PCDA to simultaneously deliver chemotherapeutic drugs and eradicate BCSCs. RESULTS Based on molecular docking and molecular dynamics studies, HA-b-PCDA delivers 35 clinical chemotherapeutic drugs. To further verify the drug deliver ability of HA-b-PCDA, doxorubicin, paclitaxel, docetaxel, gemcitabine and camptothecin are employed as model drugs to prepare nanoparticles. These drug-loaded HA-b-PCDA nanoparticles significantly inhibit the proliferation and stemness of BCSC-enriched 4T1 mammospheres. Moreover, doxorubicin-loaded HA-b-PCDA nanoparticles efficiently inhibit tumor growth and eradicate approximately 95% of BCSCs fraction in vivo. Finally, HA-b-PCDA eradicates BCSCs by activating Hippo and inhibiting the JAK2/STAT3 pathway. CONCLUSION HA-b-PCDA is a polymeric nanocarrier that eradicates BCSCs and potentially delivers numerous clinical chemotherapeutic drugs.
Collapse
Affiliation(s)
- Li Lv
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Yonghui Shi
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Zhicheng Deng
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, Guangdong, 516600, China
| | - Jiajia Xu
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zicong Ye
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Jianxiong He
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Guanghui Chen
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Xiaoxia Yu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Junyan Wu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
| | - Xingzhen Huang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Guocheng Li
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, Guangdong, 516600, China.
| |
Collapse
|
9
|
Zahoor AF, Hafeez F, Mansha A, Kamal S, Anjum MN, Raza Z, Khan SG, Javid J, Irfan A, Bhat MA. Bacterial Tyrosinase Inhibition, Hemolytic and Thrombolytic Screening, and In Silico Modeling of Rationally Designed Tosyl Piperazine-Engrafted Dithiocarbamate Derivatives. Biomedicines 2023; 11:2739. [PMID: 37893112 PMCID: PMC10603954 DOI: 10.3390/biomedicines11102739] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/03/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Piperazine is a privileged moiety that is a structural part of many clinical drugs. Piperazine-based scaffolds have attracted the attention of pharmaceutical and medicinal scientists to develop novel, efficient therapeutic agents owing to their significant and promising biological profile. In the current study, an ecofriendly ultrasonic-assisted synthetic approach was applied to achieve a novel series of 1-tosyl piperazine dithiocarbamate acetamide hybrids 4a-4j, which was evaluated for in vitro tyrosinase inhibition and thrombolytic and hemolytic cytotoxic activities. Among all the piperazine-based dithiocarbamate acetamide target molecules 4a-4j, the structural analogs 4d displayed excellent tyrosinase inhibition efficacy (IC50 = 6.88 ± 0.11 µM) which was better than the reference standard drugs kojic acid (30.34 ± 0.75 µM) and ascorbic acid (11.5 ± 1.00 µM), respectively, which was further confirmed by in silico induced-fit docking (IFD) simulation Good tyrosinase activities were exhibited by 4g (IC50 = 7.24 ± 0.15 µM), 4b (IC50 = 8.01 ± 0.11 µM) and 4c (IC50 = 8.1 ± 0.30 µM) dithiocarbamate acetamides, which were also better tyrosinase inhibitors than the reference drugs but were less active than the 4d structural hybrid. All the derivatives are less toxic, having values in the 0.29 ± 0.01% to 15.6 ± 0.5% range. The scaffold 4b demonstrated better hemolytic potential (0.29 ± 0.01%), while a remarkably high thrombolytic chemotherapeutic potential was displayed by analog 4e (67.3 ± 0.2%).
Collapse
Affiliation(s)
- Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan; (A.F.Z.); (F.H.); (S.G.K.); (A.I.)
| | - Freeha Hafeez
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan; (A.F.Z.); (F.H.); (S.G.K.); (A.I.)
- Department of Chemistry, Riphah International University Faisalabad, Faisalabad 38000, Pakistan
| | - Asim Mansha
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan; (A.F.Z.); (F.H.); (S.G.K.); (A.I.)
| | - Shagufta Kamal
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Muhammad Naveed Anjum
- Department of Applied Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Zohaib Raza
- Department of Chemistry, School of Physical Sciences, University of Adelaide, Adelaide, SA 5000, Australia;
| | - Samreen Gul Khan
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan; (A.F.Z.); (F.H.); (S.G.K.); (A.I.)
| | - Jamila Javid
- Department of Chemistry, University of Sialkot, Sialkot 51310, Pakistan;
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan; (A.F.Z.); (F.H.); (S.G.K.); (A.I.)
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
10
|
Fatima M, Aslam S, Zafar AM, Irfan A, Khan MA, Ashraf M, Faisal S, Noreen S, Shazly GA, Shah BR, Bin Jardan YA. Exploring the Synthetic Chemistry of Phenyl-3-(5-aryl-2-furyl)- 2-propen-1-ones as Urease Inhibitors: Mechanistic Approach through Urease Inhibition, Molecular Docking and Structure-Activity Relationship. Biomedicines 2023; 11:2428. [PMID: 37760869 PMCID: PMC10525509 DOI: 10.3390/biomedicines11092428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Furan chalcone scaffolds belong to the most privileged and promising oxygen-containing heterocyclic class of compounds, which have a wide spectrum of therapeutic applications in the field of pharmaceutics, pharmacology, and medicinal chemistry. This research described the synthesis of a series of twelve novel and seven reported furan chalcone (conventional synthetic approach) analogues 4a-s through the application of microwave-assisted synthetic methodology and evaluated for therapeutic inhibition potential against bacterial urease enzyme. In the first step, a series of nineteen substituted 5-aryl-2-furan-2-carbaldehyde derivatives 3a-s were achieved in moderate to good yields (40-70%). These substituted 5-aryl-2-furan-2-carbaldehyde derivatives 3a-s were condensed with acetophenone via Claisen-Schmidt condensation to furnish 19 substituted furan chalcone scaffolds 4a-s in excellent yields (85-92%) in microwave-assisted synthetic approach, while in conventional methodology, these furan chalcone 4a-s were furnished in good yield (65-90%). Furan chalcone structural motifs 4a-s were characterized through elemental analysis and spectroscopic techniques. These nineteen (19)-afforded furan chalcones 4a-s were screened for urease inhibitory chemotherapeutic efficacy and most of the furan chalcones displayed promising urease inhibition activity. The most active urease inhibitors were 1-phenyl-3-[5-(2',5'-dichlorophenyl)-2-furyl]-2-propen-1-one 4h with an IC50 value of 16.13 ± 2.45 μM, and 1-phenyl- 3-[5-(2'-chlorophenyl)-2-furyl] -2-propen-1-one 4s with an IC50 value of 18.75 ± 0.85 μM in comparison with reference drug thiourea (IC50 = 21.25 ± 0.15 μM). These furan chalcone derivatives 4h and 4s are more efficient urease inhibitors than reference drug thiourea. Structure-activity relationship (SAR) revealed that the 2,5-dichloro 4h and 2-chloro 4s moiety containing furan chalcone derivatives may be considered as potential lead reagents for urease inhibition. The in silico molecular docking study results are in agreement with the experimental biological findings. The results of this study may be helpful in the future drug discovery and designing of novel efficient urease inhibitory agents from this biologically active class of furan chalcones.
Collapse
Affiliation(s)
- Miraj Fatima
- Department of Chemistry, The Women University, Multan 66000, Pakistan
| | - Samina Aslam
- Department of Chemistry, The Women University, Multan 66000, Pakistan
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Ansa Madeeha Zafar
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Chemistry, Government Sadiq Women University, Bahawalpur 63100, Pakistan
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Misbahul Ain Khan
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Muhammad Ashraf
- Department of Biotechnology and Biochemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Shah Faisal
- Department of Chemistry, Islamia College University Peshawar, Peshawar 25120, Pakistan
| | - Sobia Noreen
- Institute of Chemistry, University of Sargodha, Sargodha 40100, Pakistan
| | - Gamal A. Shazly
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Bakht Ramin Shah
- Skin Barrier Research Group, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
11
|
Saeed S, Zahoor AF, Kamal S, Raza Z, Bhat MA. Unfolding the Antibacterial Activity and Acetylcholinesterase Inhibition Potential of Benzofuran-Triazole Hybrids: Synthesis, Antibacterial, Acetylcholinesterase Inhibition, and Molecular Docking Studies. Molecules 2023; 28:6007. [PMID: 37630258 PMCID: PMC10459521 DOI: 10.3390/molecules28166007] [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] [Scholar Register] [Received: 06/08/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 08/27/2023] Open
Abstract
In this study, a series of novel benzofuran-based 1,2,4-triazole derivatives (10a-e) were synthesized and evaluated for their inhibitory potential against acetylcholinesterase (AChE) and bacterial strains (E. coli and B. subtilis). Preliminary results revealed that almost all assayed compounds displayed promising efficacy against AChE, while compound 10d was found to be a highly potent inhibitor of AChE. Similarly, these 5-bromobenzofuran-triazoles 10a-e were screened against B. subtilis QB-928 and E. coli AB-274 to evaluate their antibacterial potential in comparison to the standard antibacterial drug penicillin. Compound 10b was found to be the most active among all screened scaffolds, with an MIC value of 1.25 ± 0.60 µg/mL against B. subtilis, having comparable therapeutic efficacy to the standard drug penicillin (1 ± 1.50 µg/mL). Compound 10a displayed excellent antibacterial therapeutic efficacy against the E. coli strain with comparable MIC of 1.80 ± 0.25 µg/mL to that of the commercial drug penicillin (2.4 ± 1.00 µg/mL). Both the benzofuran-triazole molecules 10a and 10b showed a larger zone of inhibition. Moreover, IFD simulation highlighted compound 10d as a novel lead anticholinesterase scaffold conforming to block entrance, limiting the swinging gate, and disrupting the catalytic triad of AChE, and further supported its significant AChE inhibition with an IC50 value of 0.55 ± 1.00 µM. Therefore, compound 10d might be a promising candidate for further development in Alzheimer's disease treatment, and compounds 10a and 10b may be lead antibacterial agents.
Collapse
Affiliation(s)
- Sadaf Saeed
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Shagufta Kamal
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Zohaib Raza
- Department of Chemistry, School of Physical Sciences, University of Adelaide, Adelaide, SA 5000, Australia;
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
12
|
Ahmad R, Khan M, Alam A, Elhenawy AA, Qadeer A, AlAsmari AF, Alharbi M, Alasmari F, Ahmad M. Synthesis, molecular structure and urease inhibitory activity of novel bis-Schiff bases of benzyl phenyl ketone: A combined theoretical and experimental approach. Saudi Pharm J 2023; 31:101688. [PMID: 37457366 PMCID: PMC10345485 DOI: 10.1016/j.jsps.2023.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Background Urease belongs to the family of amid hydrolases with two nickel atoms in their core structure. On the basis of literature survey, this research work is mainly focused on the study of bis-Schiff base derivatives of benzyl phenyl ketone nucleus. Objective Synthesis of benzyl phenyl ketone based bis-Schiff bases in search of potent urease inhibitors. Method In the current work, bis-Schiff bases were synthesized through two steps reaction by reacting benzyl phenyl ketone with excess of hydrazine hydrate in ethanol solvent in the first step to get the desired hydrazone. In last, different substituted aromatic aldehydes were refluxed in catalytic amount of acetic acid with the desired hydrazone to obtain bis-Schiff base derivatives in tremendous yields. Using various spectroscopic techniques including FTIR, HR-ESI-MS, and 1H NMR spectroscopy were used to clarify the structures of the created bis-Schiff base derivatives. Results The prepared compounds were finally screened for their in-vitro urease inhibition activity. All the synthesized derivatives (3-9) showed excellent to less inhibitory activity when compared with standard thiourea (IC50 = 21.15 ± 0.32 µM). Compounds 3 (IC50 = 22.21 ± 0.42 µM), 4 (IC50 = 26.11 ± 0.22 µM) and 6 (IC50 = 28.11 ± 0.22 µM) were found the most active urease inhibitors near to standard thiourea among the synthesized series. Similarly, compound 5 having IC50 value of 34.32 ± 0.65 µM showed significant inhibitory activity against urease enzyme. Furthermore, three compounds 7, 8, and 9 exhibited less activity with IC50 values of 45.91 ± 0.14, 47.91 ± 0.14, and 48.33 ± 0.72 µM respectively. DFT used to calculate frontier molecular orbitals including; HOMO and LUMO to indicate the charge transfer from molecule to biological transfer, and MEP map to indicate the chemically reactive zone suitable for drug action. The electron localization function (ELF), non-bonding orbitals, AIM charges are also calculated. The docking study contributed to the analysis of urease protein binding.
Collapse
Affiliation(s)
- Rashid Ahmad
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
- Department of Chemistry, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Momin Khan
- Department of Chemistry, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Aftab Alam
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| | - Ahmed A. Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Abdul Qadeer
- Key Laboratory of Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Manzoor Ahmad
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
13
|
Irfan A, Faisal S, Zahoor AF, Noreen R, Al-Hussain SA, Tuzun B, Javaid R, Elhenawy AA, Zaki MEA, Ahmad S, Abdellattif MH. In Silico Development of Novel Benzofuran-1,3,4-Oxadiazoles as Lead Inhibitors of M. tuberculosis Polyketide Synthase 13. Pharmaceuticals (Basel) 2023; 16:829. [PMID: 37375776 DOI: 10.3390/ph16060829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Benzofuran and 1,3,4-oxadiazole are privileged and versatile heterocyclic pharmacophores which display a broad spectrum of biological and pharmacological therapeutic potential against a wide variety of diseases. This article reports in silico CADD (computer-aided drug design) and molecular hybridization approaches for the evaluation of the chemotherapeutic efficacy of 16 S-linked N-phenyl acetamide moiety containing benzofuran-1,3,4-oxadiazole scaffolds BF1-BF16. This virtual screening was carried out to discover and assess the chemotherapeutic efficacy of BF1-BF16 structural motifs as Mycobacterium tuberculosis polyketide synthase 13 (Mtb Pks13) enzyme inhibitors. The CADD study results revealed that the benzofuran clubbed oxadiazole derivatives BF3, BF4, and BF8 showed excellent and remarkably significant binding energies against the Mtb Pks13 enzyme comparable with the standard benzofuran-based TAM-16 inhibitor. The best binding affinity scores were displayed by 1,3,4-oxadiazoles-based benzofuran scaffolds BF3 (-14.23 kcal/mol), BF4 (-14.82 kcal/mol), and BF8 (-14.11 kcal/mol), in comparison to the binding affinity score of the standard reference TAM-16 drug (-14.61 kcal/mol). 2,5-Dimethoxy moiety-based bromobenzofuran-oxadiazole derivative BF4 demonstrated the highest binding affinity score amongst the screened compounds, and was higher than the reference Pks13 inhibitor TAM-16 drug. The bindings of these three leads BF3, BF4, and BF8 were further confirmed by the MM-PBSA investigations in which they also exhibited strong bindings with the Pks13 of Mtb. Moreover, the stability analysis of these benzofuran-1,3,4-oxadiazoles in the active sites of the Pks13 enzyme was achieved through molecular dynamic (MD) simulations at 250 ns virtual simulation time, which indicated that these three in silico predicted bio-potent benzofuran tethered oxadiazole molecules BF3, BF4, and BF8 demonstrated stability with the active site of the Pks13 enzyme.
Collapse
Affiliation(s)
- Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Shah Faisal
- Department of Chemistry, Islamia College University Peshawar, Peshawar 25120, Pakistan
| | - Ameer Fawad Zahoor
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Razia Noreen
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Sami A Al-Hussain
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13623, Saudi Arabia
| | - Burak Tuzun
- Plant and Animal Production Department, Technical Sciences Vocational School of Sivas, Sivas Cumhuriyet University, Sivas 58140, Turkey
| | - Rakshanda Javaid
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt
- Chemistry Department, Faculty of Science and Art, AlBaha University, Mukhwah, Al Bahah 65731, Saudi Arabia
| | - Magdi E A Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13623, Saudi Arabia
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar 25000, Pakistan
| | - Magda H Abdellattif
- Department of Chemistry, College of Science, Taif University, Taif 21944, Saudi Arabia
| |
Collapse
|