1
|
Abera B, Abdissa N, Endale M, Melaku Y, Shenkute K, Ensermu U, Hunsen M, Rentsch D, Eswaramoorthy R. Evaluation of the Antibacterial and Antioxidant Properties of Chemical Constituents of the Roots of Woodfordia uniflora: An Integrated Approach of Experimental and Computational Study. Biochem Res Int 2024; 2024:1322756. [PMID: 39659700 PMCID: PMC11631344 DOI: 10.1155/bri/1322756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 09/13/2024] [Accepted: 10/04/2024] [Indexed: 12/12/2024] Open
Abstract
Woodfordia uniflora is a medicinal plant used for the treatment of malaria, toothache, and stomach problems. The root parts of the plant are also used for healing liver disorders. Silica gel chromatography separation of CH2Cl2/MeOH (1:1) and MeOH extracts of roots of W. uniflora result in the isolation of three compounds, namely, bergenin (1), β-sitosterol (2), and epigallocatechin 3-gallate (3), reported herein for the first time from the plant. The structure of the isolated compounds was elucidated using NMR (1D and 2D) techniques. Disk diffusion and DPPH assay were used to evaluate the antibacterial and antioxidant activities, respectively. Molecular docking was done by the AutoDock Vina 4.2 program. The pharmacokinetics and toxicity profile of compounds were predicted by Swiss ADME and Pro Tox II online servers. GC-MS analysis roots of W. uniflora result in the identification of five compounds, of which palmitic acid (34.9%) was the major constituent. The antibacterial activity result indicated that the oil extract had promising activity against P. aeruginosa, E. coli, S. pyogenes, and S. aureus with IZ of 14.3 ± 0.81, 15.0 ± 0.0, 15.6 ± 0.47, and 17.6 ± 0.47 mm, respectively, at 5 mg/mL, compared to ciprofloxacin (1Z 27-30.0 ± 0.0 mm) at 30 μg/mL. MeOH and CH2Cl2/MeOH (1:1) extract showed inhibition against E. coli (IZ of 13.6 ± 0.47 mm) and P. aeruginosa (IZ of 10.0 ± 0.0 mm), respectively, at 200 mg/mL. Bergenin (1) and β-sitosterol (2) also displayed maximum inhibition of E. coil (IZ of 11.6 ± 0.47) and S. aureus (11.0 ± 0.0 mm), respectively, at 5 mg/mL. The antioxidant activity results showed that CH2Cl2/MeOH (1:1) and MeOH extracts, bergenin (1), and compound 3 displayed potent scavenging DPPH radical with a percentage of inhibition of 76.8 ± 0.12, 77.8 ± 0.08, 71.4 ± 0.08, and 91.2 ± 0.16, respectively, compared to ascorbic acid (93.2% ± 0.04%) at 100 μg/mL. The molecular docking analysis showed that all compounds (1-3) exhibited minimum binding energy toward PDB ID: 1HD2 (-5.2 to -6.3 kcal/mol), compared to ascorbic acid (-5.6 kcal/mol), and toward PDB ID: 1DNU (-8.0 to -10.7 kcal/mol) receptors, compared to ascorbic acid (-5.7 kcal/mol). Toward the PDB ID: 4FM9 receptor, β-sitosterol (2) and compound 3 exhibited the best binding free energy of -9.1 and -9.8 kcal·mol, respectively, compared to vosaroxin (-7.8 kcal/mol). The drug-likeness analysis result indicated that bergenin (1) and β-sitosterol (2) obeyed four and five criteria of Lipinski's rule, respectively, and are more likely to be administered orally. The in silico toxicity analysis showed none of the compounds would be cytotoxic, mutagenic, or hepatotoxic. The in vitro antioxidant and antibacterial results supported by in silico analysis demonstrated that the roots of W. uniflora have the potential to be therapeutic agents for bacterial infections and free radical-inducing diseases.
Collapse
Affiliation(s)
- Bihon Abera
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Negera Abdissa
- Traditional and Modern Medicine Research and Development Directorate, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Milkyas Endale
- Traditional and Modern Medicine Research and Development Directorate, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Yadessa Melaku
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Kebede Shenkute
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Urgessa Ensermu
- Department of Applied Biology, School of Applied Natural Science, Adama Science and Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Mo Hunsen
- Department of Chemistry, Kenyon College, Gambier, Ohio 43022, USA
| | - Daniel Rentsch
- Laboratory for Functional Polymers, Empa, Swiss Federal Laboratories for Materials Science and Technology, Überlandstrasse 129, Dübendorf 8600, Switzerland
| | - Rajalakshmanan Eswaramoorthy
- Department of Biomaterials, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| |
Collapse
|
2
|
Shaldam MA, Mousa MHA, Tawfik HO, El-Dessouki AM, Sharaky M, Saleh MM, Alzahrani AYA, Moussa SB, Al-Karmalawy AA. Muti-target rationale design of novel substituted N-phenyl-2-((6-phenylpyridazin-3-yl)thio)acetamide candidates as telomerase/JAK1/STAT3/TLR4 inhibitors: In vitro and in vivo investigations. Bioorg Chem 2024; 153:107843. [PMID: 39332072 DOI: 10.1016/j.bioorg.2024.107843] [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: 06/11/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
In this work, additional effort was applied to design new BIBR1532-based analogues with potential inhibitory activity against telomerase and acting as multitarget antitumor candidates to overcome the resistance problem. Therefore, novel substituted N-phenyl-2-((6-phenylpyridazin-3-yl)thio)acetamide candidates (4a-n) were synthesized. Applying the lead optimization strategy of the previously designed compound 8e; compound 4l showed an improved telomerase inhibition of 64.95 % and a superior growth inhibition of 79 % suggesting its potential use as a successful "multitarget-directed drug" for cancer therapy. Accordingly, compound 4l was further selected to evaluate its additional JAK1/STAT3/TLR4 inhibitory potentials. Compound 4l represented a very promising JAK1 inhibitory potential with a 0.46-fold change, compared to that of pacritinib reference standard (0.33-fold change). Besides, it showed a superior STAT3-inhibitory potential with a 0.22-fold change compared to sorafenib (0.33-fold change). Additionally, compound 4l downregulated TLR4 protein expression by 0.81-fold change compared to that of resatorvid (0.29-fold change). Also, molecular docking was performed to investigate the binding mode and affinity of the superior candidate 4l towards the four target receptors (telomerase, JAK1, STAT3, and TLR4). Furthermore, the therapeutic potential of compound 4l as an antitumor agent was additionally explored through in vivo studies involving female mice implanted with Solid Ehrlich Carcinoma (SEC). Remarkably, compound 4l led to prominent reductions in tumor size and mass. Concurrent enhancements in biochemical, hematologic, histopathologic, and immunohistochemical parameters further confirmed the suppression of angiogenesis and inflammation, elucidating additional mechanisms by which compound 4l exerts its anticancer effects.
Collapse
Affiliation(s)
- Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt.
| | - Mai H A Mousa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo 11786, Egypt.
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt.
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt.
| | - Mohamed M Saleh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | | | - Sana Ben Moussa
- Department of Chemistry, Faculty of Science and Arts, King Khalid University, Mohail Assir 61421, Saudi Arabia
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Mashreq, Baghdad 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt.
| |
Collapse
|
3
|
Althikrallah HA, Shaaban S, Elmaaty AA, Ba-Ghazal H, Almarri MN, Sharaky M, Alnajjar R, Al-Karmalawy AA. Investigating the anti-inflammatory potential of N-amidic acid organoselenium candidates: biological assessments, molecular docking, and molecular dynamics simulations. RSC Adv 2024; 14:31990-32000. [PMID: 39391620 PMCID: PMC11463133 DOI: 10.1039/d4ra04762a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Inflammation is a complex process with many contributing factors, and it often causes pain. The pathophysiology of pain involves the release of inflammatory mediators that initiate pain sensation, as well as edema and other inflammation hallmarks. Selenium-containing compounds (OSe) are very promising for developing new medicines because they can treat many different diseases. In this study, we estimated the anti-inflammatory properties of maleanilic and succinanilic acids containing selenium (OSe). These molecules were designed by combining different strategies to enhance their anti-inflammatory properties. Hence, the anti-inflammatory impacts of compounds 8, 9, 10, and 11 were pursued using inflammatory markers COX-2, IL-1β, and IL-6. Notably, it was revealed that compounds 8, 9, 10, and 11 downregulated COX-2, IL-1β, and IL-6 by (2.01, 1.63, 2.26, and 2.05), (1.42, 1.64, 1.93, and 2.59), and (1.67, 2.54, 2.22, and 4.06)-fold changes, respectively. Moreover, molecular docking studies were conducted on compounds 8, 9, 10, and 11 to pursue their binding affinities for the COX-2 enzyme. Notably, very promising binding scores of compounds 8, 9, 10, and 11 towards the binding site of the COX-2 receptor were attained. Additionally, more accurate molecular dynamics simulations were performed for 200 ns for the docked complexes of compounds 8, 9, 10, and 11 to confirm the molecular docking findings, which ignore the protein's flexibility. Therefore, the exact stability of the N-amidic acids OSe compounds 8, 9, 10, and 11 towards the binding pocket of the COX-2 enzyme was examined and explained as well. Also, the MM-GBSA binding energy was calculated for equilibrated MD trajectory, and 200 snapshots were selected with a 50 ps interval for further analysis. Accordingly, the investigated compounds can be treated as prominent lead anti-inflammatory candidates for further optimization.
Collapse
Affiliation(s)
- Hanan A Althikrallah
- Department of Chemistry, College of Science, King Faisal University Al-Ahsa 31982 Saudi Arabia
| | - Saad Shaaban
- Department of Chemistry, College of Science, King Faisal University Al-Ahsa 31982 Saudi Arabia
- Department of Chemistry, Faculty of Science, Mansoura University 35516 Mansoura Egypt
| | - Ayman Abo Elmaaty
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University Port Said 42511 Egypt
| | - Hussein Ba-Ghazal
- Department of Chemistry, College of Science, King Faisal University Al-Ahsa 31982 Saudi Arabia
| | - Mohammed N Almarri
- Department of Chemistry, College of Science, King Faisal University Al-Ahsa 31982 Saudi Arabia
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University Cairo Egypt
| | - Radwan Alnajjar
- CADD Unit, Faculty of Pharmacy, Libyan International Medical University Benghazi Libya
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq Baghdad 10023 Iraq
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt New Damietta 34518 Egypt
| |
Collapse
|
4
|
Tesfaye D, Endale M, Ramachandran VP, Getaneh E, Amenu G, Guta L, Demissie TB, Ombito JO, Eswaramoorthy R, Melaku Y. Antibacterial and Cytotoxicity of Extracts and Isolated Compounds from Artemisia abyssinica: A Combined Experimental and Computational Study. ACS OMEGA 2024; 9:31508-31520. [PMID: 39072116 PMCID: PMC11270564 DOI: 10.1021/acsomega.4c01096] [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: 02/07/2024] [Revised: 06/11/2024] [Accepted: 06/19/2024] [Indexed: 07/30/2024]
Abstract
Artemisia abyssinica is a widely cultivated hedge plant in Ethiopia. Traditionally, they have been used to treat a variety of health conditions, including intestinal problems, infectious diseases, tonsillitis, and leishmaniasis. Silica gel chromatographic separation of the methanol and ethyl acetate extracts of the leaves, roots, and stem barks of A. abyssinica led to the isolation of 12 compounds, labeled as 1-12. Among these, compounds 1, 3, 4, 5, and 7-11 are reported as new to the genus Artemisia. The extracts and isolated compounds from A. abyssinica were evaluated for their in vitro antibacterial activity against four bacterial strains: Streptococcus pyogenes, Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli, using the disc diffusion assay. All of the extracts displayed weak antibacterial activity, with inhibition zone diameters (IZDs) ranging from 6.10 ± 0.3 to 9.30 ± 0.20 mm. The isolated compounds, on the other hand, exhibited weak to moderate antibacterial activity, with IZDs ranging from 6.00 ± 0.300 to 13.50 ± 0.50 mm. The most potent antibacterial activity was observed for compound 6, which showed an IZD of 13.30 ± 0.50 mm against E. coli and 13.50 ± 0.50 mm against P. aeruginosa. This activity was comparable to that of the positive control ceftriaxone, which had IZDs of 14.1 ± 0.3 and 13.8 ± 0.5 mm against E. coli and P. aeruginosa, respectively. The in silico molecular docking analysis against DNA gyrase B revealed that compound 5 showed a higher binding affinity (-6.9 kcal/mol), followed by compound 10 (-6.7 kcal/mol) and compound 12 (-6.3 kcal/mol), whereas ciprofloxacin showed -7.3 kcal/mol. The binding affinities of compounds 5, 11, 10, and 9 were found to be -5.0, -4.3, -4.2, and -4.0 kcal/mol against S. aureus Pyruvate kinase, respectively, whereas ciprofloxacin showed a binding affinity of -4.9 kcal/mol, suggesting that compound 5 had a better binding affinity compared with ciprofloxacin. The effect of extracts of A. abyssinica was evaluated for cytotoxic activity against the breast cancer cell line (MCF-7) by the MTT assay. The extracts induced a decrease in cell viability and exerted a cytotoxic effect at a concentration of 20 μg/mL. The highest percent cell viability was observed for the methanol extract of the stem (92.9%), whereas the least was observed for the methanol extract of the root (34.5%). The result of the latter was significant compared with the positive control. The binding affinities of the isolated compounds were also assessed against human topoisomerase inhibitors IIβ. Results showed that compound 5 showed a binding affinity of -6.0 kcal/mol, followed by 11 (-5.4 kcal/mol), 10 (-5.0 kcal/mol), and 11 (-4.9 kcal/mol). Similar to ciprofloxacin, compounds 4, 5, 6, 9, 10, and 12 comply with Lipinski's rule of five. Overall, the comprehensive investigation of the chemical constituents and their biological activities reinforces the traditional medicinal applications of A. abyssinica and warrants further exploration of this plant as a source of novel therapeutic agents.
Collapse
Affiliation(s)
- Dawit Tesfaye
- Department
of Applied Chemistry, Adama Science and
Technology University, P.O. Box 1888, Adama, Ethiopia
| | - Milkyas Endale
- Traditional
and Modern Medicine Research and Development Directorate, Armauer Hansen Research Institute, P.O. Box 1242, Addis Ababa 1165, Ethiopia
| | | | - Emebet Getaneh
- Department
of Applied Biology, Adama Science and Technology
University, P.O. Box 1888, Adama 311-2118 Ethiopia
| | - Guta Amenu
- Department
of Applied Biology, Adama Science and Technology
University, P.O. Box 1888, Adama 311-2118 Ethiopia
| | - Leta Guta
- Department
of Applied Biology, Adama Science and Technology
University, P.O. Box 1888, Adama 311-2118 Ethiopia
| | - Taye B. Demissie
- Department
of Chemistry, University of Botswana, Gaborone P/Bag 00704, Botswana
| | - Japheth O. Ombito
- Department
of Chemistry, University of Botswana, Gaborone P/Bag 00704, Botswana
| | - Rajalakshmanan Eswaramoorthy
- Department
of Biomaterials, Saveetha Dental College and Hospitals, Saveetha Institute
of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600 077, India
| | - Yadessa Melaku
- Department
of Applied Chemistry, Adama Science and
Technology University, P.O. Box 1888, Adama, Ethiopia
| |
Collapse
|
5
|
Khanal M, Acharya A, Maharjan R, Gyawali K, Adhikari R, Mulmi DD, Lamichhane TR, Lamichhane HP. Identification of potent inhibitors of HDAC2 from herbal products for the treatment of colon cancer: Molecular docking, molecular dynamics simulation, MM/GBSA calculations, DFT studies, and pharmacokinetic analysis. PLoS One 2024; 19:e0307501. [PMID: 39037973 PMCID: PMC11262678 DOI: 10.1371/journal.pone.0307501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/06/2024] [Indexed: 07/24/2024] Open
Abstract
The histone deacetylase 2 (HDAC2), an enzyme involved in gene regulation, is a potent drug target for the treatment of colon cancer. Phytocompounds having anticancer properties show the ability to interact with HDAC2 enzyme. Among the compounds, docking scores of caffeic acid (CA) and p-coumaric acid (pCA) with HDAC2 showed good binding efficacy of -5.46 kcal/mol and -5.16 kcal/mol, respectively, with small inhibition constants. The higher binding efficacy of CA compared to pCA can be credited to the presence of an extra oxygen atom in the CA molecule, which forms an additional hydrogen bond with Tyr297. The HDAC2 in complex with these molecules was found to be stable by analyzing RMSD, RMSF, Rg, and SASA values obtained through MD simulations. Furthermore, CA and pCA exhibited low MM/GBSA free energies of -16.32 ± 2.62 kcal/mol and -17.01 ± 2.87 kcal/mol, respectively. The HOMO and LUMO energy gaps, dipole moments, global reactivity descriptor values, and MEP surfaces showed the reactivity of the molecules. The favourable physicochemical and pharmacokinetic properties, along with absence of toxicity of the molecules determined using ADMET analysis, suggested both the acids to be regarded as effective drugs in the treatment of colon cancer.
Collapse
Affiliation(s)
- Madan Khanal
- Central Department of Physics, Tribhuvan University, Kathmandu, Nepal
| | - Arjun Acharya
- Central Department of Physics, Tribhuvan University, Kathmandu, Nepal
| | - Rajesh Maharjan
- Central Department of Physics, Tribhuvan University, Kathmandu, Nepal
| | - Kalpana Gyawali
- Central Department of Physics, Tribhuvan University, Kathmandu, Nepal
| | - Rameshwar Adhikari
- Central Department of Chemistry, Tribhuvan University, Kathmandu, Nepal
- Research Center for Applied Science and Technology (RECAST), Tribhuvan University, Kathmandu, Nepal
| | - Deependra Das Mulmi
- Nanomaterials Research Laboratory, Nepal Academy of Science and Technology (NAST), Lalitpur, Nepal
| | | | | |
Collapse
|
6
|
Abdel-Motaal M, Aldakhili DA, Abo Elmaaty A, Sharaky M, Mourad MAE, Alzahrani AYA, Mohamed NA, Al-Karmalawy AA. Design and synthesis of novel tetrabromophthalimide derivatives as potential tubulin inhibitors endowed with apoptotic induction for cancer treatment. Drug Dev Res 2024; 85:e22197. [PMID: 38751223 DOI: 10.1002/ddr.22197] [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: 02/14/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
Although various approaches exist for treating cancer, chemotherapy continues to hold a prominent role in the management of this disease. Besides, microtubules serve as a vital component of the cellular skeleton, playing a pivotal role in the process of cell division making it an attractive target for cancer treatment. Hence, the scope of this work was adapted to design and synthesize new anti-tubulin tetrabromophthalimide hybrids (3-17) with colchicine binding site (CBS) inhibitory potential. The conducted in vitro studies showed that compound 16 displayed the lowest IC50 values (11.46 µM) at the FaDu cancer cell lines, whereas compound 17 exhibited the lowest IC50 value (13.62 µM) at the PC3 cancer cell line. However, compound 7b exhibited the lowest IC50 value (11.45 µM) at the MDA-MB-468 cancer cell line. Moreover, compound 17 was observed to be the superior antitumor candidate against all three tested cancer cell lines (MDA-MB-468, PC3, and FaDu) with IC50 values of 17.22, 13.15, and 13.62 µM, respectively. In addition, compound 17 showed a well-established upregulation of apoptotic markers (Caspases 3, 7, 8, and 9, Bax, and P53). Moreover, compound 17 induced downregulation of the antiapoptotic markers (MMP2, MMP9, and BCL-2). Furthermore, the colchicine binding site inhibition assay showed that compounds 15a and 17 exhibited particularly significant inhibitory potentials, with IC50 values of 23.07 and 4.25 µM, respectively, compared to colchicine, which had an IC50 value of 3.89 µM. Additionally, cell cycle analysis was conducted, showing that compound 17 could prompt cell cycle arrest at both the G0-G1 and G2-M phases. On the other hand, a molecular docking approach was applied to investigate the binding interactions of the examined candidates compared to colchicine towards CBS of the β-tubulin subunit. Thus, the synthesized tetrabromophthalimide hybrids can be regarded as outstanding anticancer candidates with significant apoptotic activity.
Collapse
Affiliation(s)
- Marwa Abdel-Motaal
- Department of Chemistry, College of Science, Qassim University, Buraydah, Saudi Arabia
- Department of Chemistry, Organic Chemistry Division, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Dalal A Aldakhili
- Department of Chemistry, College of Science, Qassim University, Buraydah, Saudi Arabia
| | - Ayman Abo Elmaaty
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, Cairo, Egypt
- Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October, Egypt
| | - Mai A E Mourad
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Abdullah Y A Alzahrani
- Department of Chemistry, Faculty of Science and Arts, King Khalid University, Mohail Assir, Saudi Arabia
| | - Nadia A Mohamed
- Department of Chemistry, College of Science, Qassim University, Buraydah, Saudi Arabia
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| |
Collapse
|
7
|
Eid NM, Al-Karmalawy AA, Eldebss TMA, Elhakim HKA. Investigating the Promising Anticancer Activity of Cetuximab and Fenbendazole Combination as Dual CBS and VEGFR-2 Inhibitors and Endowed with Apoptotic Potential. Chem Biodivers 2024; 21:e202302081. [PMID: 38318954 DOI: 10.1002/cbdv.202302081] [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: 12/22/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/07/2024]
Abstract
In this work, the cytotoxicity of monoclonal antibody (Cetuximab, Ce) and Fenbendazole (Fen), as well as their combination therapy were tested with the MTT assay. On the other side, Ce, Fen, and a combination between them were subjected to a colchicine-tubulin binding test, which was conducted and compared to Colchicine as a reference standard. Besides, Ce, Fen, and the combination of them were tested against the VEGFR-2 target receptor, compared to Sorafenib as the standard medication. Moreover, the qRT-PCR technique was used to investigate the levels of apoptotic genes (p53 and Bax) and anti-apoptotic gene (Bcl-2) as well. Also, the effect of Ce, Fen, and the combination of them on the level of ROS was studied. Furthermore, the cell cycle analysis and Annexin V apoptosis assay were carried out for Ce, Fen, and a combination of them. In addition, the molecular docking studies were used to describe the molecular levels of interactions for both (Fen and colchicine) or (Fen and sorafenib) within the binding pockets of the colchicine binding site (CBS) and vascular endothelial growth factor-2 receptor (VEGFR-2), respectively.
Collapse
Affiliation(s)
- Norhan M Eid
- Biochemistry Division, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt
| | - Taha M A Eldebss
- Chemistry Division, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Heba K A Elhakim
- Biochemistry Division, Faculty of Science, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
8
|
Assefa T, Tesso H, Ramachandran VP, Guta L, Demissie TB, Ombito JO, Eswaramoorthy R, Melaku Y. In Silico Molecular Docking Analysis, Cytotoxicity, and Antibacterial Activities of Constituents of Fruits of Cucumis dipsaceus. ACS OMEGA 2024; 9:1945-1955. [PMID: 38222496 PMCID: PMC10785779 DOI: 10.1021/acsomega.3c08866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/16/2024]
Abstract
Cucumis dipsaceus (Cucurbitaceae) is a plant traditionally used against diarrhea, teeth-ach, wounds, stomach ache, meningitis, and cancer. The extracts of C. dipsaceus after silica gel column chromatography gave nine compounds identified using spectroscopic methods such as hexacosane (1), octadecane (2), 17-(-5-ethyl-2,6-dihydroxy-6-methylhept-3-en-2-yl)-9-(hydroxymethyl)-13-methylcyclopenta[α]phenanthren-3-ol (3), erythrodiol (4), (9,12)-propyl icosa-9,12-dienoate (5), α-spinasterol (6), 16-dehydroxycucurbitacin (7), cucurbitacin D (8), and 23,24-dihydroisocucurbitacin D (9). Compounds 3 and 4 are new to the genus Cucumis. α-Spinasterol showed better inhibition zone diameter = 13.67 ± 0.57, 15.00 ± 0.10, and 13.33 ± 0.57 mm against Escherichia coli, Pseudomonas aeruginosa, and Streptococcus pyogenes compared with the other tested samples. α-Spinasterol (-8.0 kcal/mol) and 3 (-7.6 kcal/mol) displayed high binding affinity against DNA Gyrase compared to ciprofloxacin (-7.3 kcal/mol). α-Spinasterol and 16-dehydroxycucurbitacin showed better binding affinity against protein kinase. The cytotoxicity results revealed that the EtOAc extract showed the highest potency with IC50 = 16.05 μg/mL. 16-Dehydroxycucurbitacin showed a higher binding affinity (-7.7 kcal/mol) against human topoisomerase IIβ than etoposide. The cytotoxicity and antibacterial activities and in silico molecular docking analysis displayed by the constituents corroborate the traditional use of the plant against bacteria and cancer.
Collapse
Affiliation(s)
- Teshale Assefa
- Department
of Applied Chemistry, Adama Science and
Technology University, P.O. Box, Adama 1888, Ethiopia
| | - Hailemichael Tesso
- Department
of Applied Chemistry, Adama Science and
Technology University, P.O. Box, Adama 1888, Ethiopia
| | | | - Leta Guta
- Department
of Applied Biology, Adama Science and Technology
University, P.O. Box, Adama 1888, Ethiopia
| | - Taye B. Demissie
- Department
of Chemistry, University of Botswana, Gaborone P/Bag 00704, Botswana
| | - Japheth O. Ombito
- Department
of Chemistry, University of Botswana, Gaborone P/Bag 00704, Botswana
| | - Rajalakshmanan Eswaramoorthy
- Department
of Biomaterials, S Aveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences
(SIMATS), Saveetha University, Chennai 600 077, India
| | - Yadessa Melaku
- Department
of Applied Chemistry, Adama Science and
Technology University, P.O. Box, Adama 1888, Ethiopia
| |
Collapse
|
9
|
He S, Ye X, Dou L, Sakurai T. FIAMol-AB: A feature fusion and attention-based deep learning method for enhanced antibiotic discovery. Comput Biol Med 2024; 168:107762. [PMID: 38056212 DOI: 10.1016/j.compbiomed.2023.107762] [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: 07/29/2023] [Revised: 10/31/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023]
Abstract
Antibiotic resistance continues to be a growing concern for global health, accentuating the need for novel antibiotic discoveries. Traditional methodologies in this field have relied heavily on extensive experimental screening, which is often time-consuming and costly. Contrastly, computer-assisted drug screening offers rapid, cost-effective solutions. In this work, we propose FIAMol-AB, a deep learning model that combines graph neural networks, text convolutional networks and molecular fingerprint techniques. This method also combines an attention mechanism to fuse multiple forms of information within the model. The experiments show that FIAMol-AB may offer potential advantages in antibiotic discovery tasks over some existing methods. We conducted some analysis based on our model's results, which help highlight the potential significance of certain features in the model's predictive performance. Compared to different models, ours demonstrate promising results, indicating potential robustness and versatility. This suggests that by integrating multi-view information and attention mechanisms, FIAMol-AB might better learn complex molecular structures, potentially improving the precision and efficiency of antibiotic discovery. We hope our FIAMol-AB can be used as a useful method in the ongoing fight against antibiotic resistance.
Collapse
Affiliation(s)
- Shida He
- Department of Computer Science, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Xiucai Ye
- Department of Computer Science, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Lijun Dou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland, OH, 44106, USA
| | - Tetsuya Sakurai
- Department of Computer Science, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
10
|
Aktaş E, Saygılı İ, Kahveci E, Tekbıyık Z, Özgentürk NÖ. Bioinformatic investigation of Nipah virus surface protein mutations: Molecular docking with Ephrin B2 receptor, molecular dynamics simulation, and structural impact analysis. Microbiol Immunol 2023; 67:501-513. [PMID: 37812043 DOI: 10.1111/1348-0421.13098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023]
Abstract
The SARS-CoV-2 outbreak resulted in significant challenges and loss of life. The Nipah virus, known for its high infectivity and severity, was designated an emergency concern by the World Health Organization. To understand its mutations, the Nipah virus proteins were analyzed extensively, with a focus on the essential G and F proteins responsible for viral entry into host cells. Our bioinformatics analysis unveiled multiple mutations, including simultaneous mutations within a single sequence. Notably, the G273S mutation in the F protein was identified as a potential cause of structural damage, which carries significant implications for vaccine development. Comparing the docking scores of G and F proteins with the Ephrin B2 receptor, it was found that the Y228H mutation in the G protein and the D252G mutation in the F protein likely affect virus entry into host cells. Moreover, our investigation into stability and deformability highlighted the impact of the Y228H mutation in the G protein complex. Molecular dynamics simulations revealed increased flexibility and conformational changes in the G protein complex with the Y228H mutation compared with the known complex. Furthermore, evaluating the root mean square deviation variation demonstrated greater dynamic behavior in the G protein complex and the Ephrin B2 receptor complex. This comprehensive study provides valuable insights into Nipah virus mutations, their significance for vaccine development, and the importance of understanding protein complex behavior in drug discovery. The identified mutations, especially G273S and Y228H, hold crucial implications for future research and potential interventions against the Nipah virus.
Collapse
Affiliation(s)
- Emre Aktaş
- Faculty of Art and Science, Molecular Biology and Genetics, Yıldız Technical University, Istanbul, Turkey
| | - İrem Saygılı
- Faculty of Art and Science, Molecular Biology and Genetics, Yıldız Technical University, Istanbul, Turkey
| | - Elif Kahveci
- Faculty of Art and Science, Molecular Biology and Genetics, Yıldız Technical University, Istanbul, Turkey
| | - Zeynep Tekbıyık
- Faculty of Art and Science, Molecular Biology and Genetics, Yıldız Technical University, Istanbul, Turkey
| | - Nehir Özdemir Özgentürk
- Faculty of Art and Science, Molecular Biology and Genetics, Yıldız Technical University, Istanbul, Turkey
| |
Collapse
|
11
|
Al-Karmalawy AA, Rashed M, Sharaky M, Abulkhair HS, Hammouda MM, Tawfik HO, Shaldam MA. Novel fused imidazotriazines acting as promising top. II inhibitors and apoptotic inducers with greater selectivity against head and neck tumors: Design, synthesis, and biological assessments. Eur J Med Chem 2023; 259:115661. [PMID: 37482023 DOI: 10.1016/j.ejmech.2023.115661] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/08/2023] [Accepted: 07/16/2023] [Indexed: 07/25/2023]
Abstract
Although the great effectiveness of doxorubicin (Dox) in the treatment of many types of tumors, it showed limited effectiveness against the head and neck squamous cell carcinoma (HNSCC) subtype which is attributed to its reported multiple drug resistance (MDR). In the current study, we considered the essential pharmacophoric features of Dox as an effective Top. II inhibitor and sought to develop a novel set of imidazo[1,2-a] [1,3,5]triazin-2-amines (2a-2p) as a suggested anticancer option that could intercalate the DNA base pairs. We evaluated the % inhibition of the newly synthesized compounds on thirteen cancer cell lines and the analysis of structure-activity relationships revealed that the human head and neck cancer cell line (HNO97) was the most sensitive to their growth inhibition effect. Then, the IC50 values were recorded against the most sensitive cancer cell lines (HNO97, MDA-MB-231, and HEPG2), and compared to the normal cell line OEC (human oral epithelial cells). Compounds 2f and 2g showed very strong activities against HNO97 with IC50 values of (4 ± 1 and 3 ± 1.5 μg/mL), respectively, compared to that of Dox (9 ± 1.6 μg/mL). Next, a quantitative determination of human DNA Top. II concentrations in the most sensitive cell line (HNO97) were recorded for the most active anticancer derivatives. Again, compound 2f showed a superior Top. II inhibition with 87.86% compared to that of Dox (86.44%), while compound 2g achieved an inhibition of 81.37% which was close to the effect of Dox. To further investigate their effects on cell cycle progression and apoptosis induction in HNO97 cells, both 2f and 2g were selected for analysis. Both candidates arrested cell cycle progression at both the S and G2-M phases, as well as increased the early and late apoptosis phase ratios. Besides, both 2f and 2g were subjected to protein expression analysis of apoptosis-related genes (p53, BAX, IL-6, and BCL2). Moreover, the antioxidant effect of 2f and 2g was evaluated by measuring GSH, MDA, and NO markers in HNO97 cells. Furthermore, molecular docking for the newly designed tricyclic derivatives against both the Top. II and DNA double helix was carried out.
Collapse
Affiliation(s)
- Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt.
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, 11884, Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University - Egypt, International Coastal Road, New Damietta, 34518, Egypt
| | - Mohamed M Hammouda
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia; Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| |
Collapse
|
12
|
Tarín-Pelló A, Suay-García B, Forés-Martos J, Falcó A, Pérez-Gracia MT. Computer-aided drug repurposing to tackle antibiotic resistance based on topological data analysis. Comput Biol Med 2023; 166:107496. [PMID: 37793206 DOI: 10.1016/j.compbiomed.2023.107496] [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: 04/23/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
The progressive emergence of antimicrobial resistance has become a global health problem in need of rapid solution. Research into new antimicrobial drugs is imperative. Drug repositioning, together with computational mathematical prediction models, could be a fast and efficient method of searching for new antibiotics. The aim of this study was to identify compounds with potential antimicrobial capacity against Escherichia coli from US Food and Drug Administration-approved drugs, and the similarity between known drug targets and E. coli proteins using a topological structure-activity data analysis model. This model has been shown to identify molecules with known antibiotic capacity, such as carbapenems and cephalosporins, as well as new molecules that could act as antimicrobials. Topological similarities were also found between E. coli proteins and proteins from different bacterial species such as Mycobacterium tuberculosis, Pseudomonas aeruginosa and Salmonella Typhimurium, which could imply that the selected molecules have a broader spectrum than expected. These molecules include antitumor drugs, antihistamines, lipid-lowering agents, hypoglycemic agents, antidepressants, nucleotides, and nucleosides, among others. The results presented in this study prove the ability of computational mathematical prediction models to predict molecules with potential antimicrobial capacity and/or possible new pharmacological targets of interest in the design of new antibiotics and in the better understanding of antimicrobial resistance.
Collapse
Affiliation(s)
- Antonio Tarín-Pelló
- Área de Microbiología, Departamento de Farmacia, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud Universidad Cardenal Herrera-CEU, CEU Universities, C/ Santiago Ramón y Cajal, 46115, Alfara del Patriarca, Valencia, Spain
| | - Beatriz Suay-García
- ESI International Chair@CEU-UCH, Departamento de Matemáticas, Física y Ciencias Tecnológicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/ San Bartolomé 55, 46115, Alfara del Patriarca, Valencia, Spain
| | - Jaume Forés-Martos
- ESI International Chair@CEU-UCH, Departamento de Matemáticas, Física y Ciencias Tecnológicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/ San Bartolomé 55, 46115, Alfara del Patriarca, Valencia, Spain
| | - Antonio Falcó
- ESI International Chair@CEU-UCH, Departamento de Matemáticas, Física y Ciencias Tecnológicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/ San Bartolomé 55, 46115, Alfara del Patriarca, Valencia, Spain
| | - María-Teresa Pérez-Gracia
- Área de Microbiología, Departamento de Farmacia, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud Universidad Cardenal Herrera-CEU, CEU Universities, C/ Santiago Ramón y Cajal, 46115, Alfara del Patriarca, Valencia, Spain.
| |
Collapse
|
13
|
Shalaby AS, Eid HH, El-Shiekh RA, Youssef FS, Al-Karmalawy AA, Farag NA, Sleem AA, Morsy FA, Ibrahim KM, Tadros SH. A Comparative GC/MS Analysis of Citrus Essential Oils: Unveiling the Potential Benefits of Herb-Drug Interactions in Preventing Paracetamol-Induced Hepatotoxicity. Chem Biodivers 2023; 20:e202300778. [PMID: 37599265 DOI: 10.1002/cbdv.202300778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023]
Abstract
Our study aimed to test the potential of Citrus oils in protecting against paracetamol (PAR)-induced hepatotoxicity. The essential oils of Pineapple sweet orange (OO), Murcott mandarin (MO), Red grapefruit (GO), and Oval kumquat (KO) were investigated using gas chromatography coupled with mass spectrometry (GC/MS). Twenty-seven compounds were identified, with monoterpene hydrocarbons being abundant class. d-Limonene had the highest percentage (92.98 %, 92.82 %, 89.75 %, and 94.46 % in OO, MO, GO, and KO, respectively). Hierarchical cluster analysis (HCA) and principal components analysis (PCA) revealed that octanal, linalool, germacrene D, and d-limonene were the principal discriminatory metabolites that segregated the samples into three distinct clusters. In vitro antioxidant capacities were ranged from 1.2-12.27, 1.79-5.91, and 235.05-585.28 μM Trolox eq/mg oil for 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic (ABTS), ferric-reducing antioxidant power (FRAP), and oxygen radical absorbance capacity (ORAC), respectively. In vivo, citrus oils exhibited a significant reduction in alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), and nitric oxide (NO). Additionally, there was an increase in glutathione reductase (GSH), and the liver architecture was nearly normal. Molecular docking revealed that d-limonene exhibited a good inhibitory interaction with cytochrome P450 (CYP450) isoforms 1A2, 3A4, and 2E1, with binding energies of -6.17, -4.51, and -5.61 kcal/mol, respectively.
Collapse
Affiliation(s)
- Aya S Shalaby
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Hanaa H Eid
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Riham A El-Shiekh
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Fadia S Youssef
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abbasia, Cairo, 11566, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12566, Egypt
| | - Nahla A Farag
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt
| | - Amany A Sleem
- Pharmacology Department, National Research Center, Dokki, Cairo, 12622, Egypt
| | - Fatma Adly Morsy
- Pathology Department, National Research Center, Dokki, Cairo, 12622, Egypt
| | - Khaled M Ibrahim
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Soad H Tadros
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
14
|
El-Kalyoubi S, Khalifa MM, Abo-Elfadl MT, El-Sayed AA, Elkamhawy A, Lee K, Al-Karmalawy AA. Design and synthesis of new spirooxindole candidates and their selenium nanoparticles as potential dual Topo I/II inhibitors, DNA intercalators, and apoptotic inducers. J Enzyme Inhib Med Chem 2023; 38:2242714. [PMID: 37592917 PMCID: PMC10444021 DOI: 10.1080/14756366.2023.2242714] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/15/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
A new wave of dual Topo I/II inhibitors was designed and synthesised via the hybridisation of spirooxindoles and pyrimidines. In situ selenium nanoparticles (SeNPs) for some derivatives were synthesised. The targets and the SeNP derivatives were examined for their cytotoxicity towards five cancer cell lines. The inhibitory potencies of the best members against Topo I and Topo II were also assayed besides their DNA intercalation abilities. Compound 7d NPs exhibited the best inhibition against Topo I and Topo II enzymes with IC50 of 0.042 and 1.172 μM, respectively. The ability of compound 7d NPs to arrest the cell cycle and induce apoptosis was investigated. It arrested the cell cycle in the A549 cell at the S phase and prompted apoptosis by 41.02% vs. 23.81% in the control. In silico studies were then performed to study the possible binding interactions between the designed members and the target proteins.
Collapse
Affiliation(s)
- Samar El-Kalyoubi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Mohamed M. Khalifa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mahmoud T. Abo-Elfadl
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt
| | - Ahmed A. El-Sayed
- Photochemistry Department, Chemical Industries Research Institute, National Research Centre, Giza, Egypt
| | - Ahmed Elkamhawy
- College of Pharmacy, BK21 FOUR Team and Integrated Research Institute for Drug Development, Dongguk University—Seoul, Goyang, Republic of Korea
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Kyeong Lee
- College of Pharmacy, BK21 FOUR Team and Integrated Research Institute for Drug Development, Dongguk University—Seoul, Goyang, Republic of Korea
| | - Ahmed A. Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, Egypt
| |
Collapse
|
15
|
Eissa KI, Kamel MM, Mohamed LW, Doghish AS, Alnajjar R, Al-Karmalawy AA, Kassab AE. Design, synthesis, and biological evaluation of thienopyrimidine derivatives as multifunctional agents against Alzheimer's disease. Drug Dev Res 2023; 84:937-961. [PMID: 37067008 DOI: 10.1002/ddr.22064] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/24/2023] [Accepted: 04/01/2023] [Indexed: 04/18/2023]
Abstract
A series of 12 S-substituted tetrahydrobenzothienopyrimidines were designed and synthesized based on the donepezil scaffold. All the newly synthesized compounds were evaluated for their acetylcholinesterase (AChE) inhibitory activity and the most active compounds were tested for their butyrylcholinesterase (BuChE) inhibitory activity. Moreover, all the synthesized compounds were evaluated for their inhibitory effects against Aβ aggregation and antioxidant activity using the oxygen radical absorbance capacity method. Compounds 4b, 6b, and 8b displayed the most prominent AChE inhibitory action comparable to donepezil. Compound 6b showed the greatest AChE inhibitory action (IC50 = 0.07 ± 0.003 µM) and the most potent BuChE inhibitory action (IC50 = 0.059 ± 0.004 µM). Furthermore, the three compounds exhibited significant antioxidant activity. Compounds 6b and 8b exerted more inhibitory action on Aβ aggregation than donepezil. The cytotoxic activity of compounds 4b, 6b, and 8b against the WI-38 cell line in comparison with donepezil was examined using 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide assay. The results revealed that compounds 6b and 8b were less cytotixic than donepezil, while compound 4b showed nonsignificant cytotoxicity compared to donepezil. For more insights about the binding patterns of the most promising compounds (4b, 6b, and 8b) with the AChE at molecular levels; molecular docking and molecular dynamics simulations were performed. The density functional theory calculations and absorption, distribution, metabolism, excretion and toxicity properties were described as well. The results highlighted compound 6b, which incorporates a phenylpiperazine moiety coupled to a thienopyrimidone scaffold via two-atom spacer, to be a promising multifunctional therapeutic agent for the treatment of Alzheimer's disease. It is a potent dual AChE and BuChE inhibitor. Furthermore, it had stronger Aβ aggregation inhibitory action than donepezil. Additionally, compound 6b exerted significant antioxidant activity.
Collapse
Affiliation(s)
- Kholoud I Eissa
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mona M Kamel
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Lamia W Mohamed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Radwan Alnajjar
- Department of Chemistry, Faculty of Science, University of Benghazi, Benghazi, Libya
- Faculty of Pharmacy, Libyan International Medical University, Benghazi, Libya
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
16
|
Ghamry HI, Belal A, El-Ashrey MK, Tawfik HO, Alsantali RI, Obaidullah AJ, El-Mansi AA, Abdelrahman D. Evaluating the ability of some natural phenolic acids to target the main protease and AAK1 in SARS COV-2. Sci Rep 2023; 13:7357. [PMID: 37147518 PMCID: PMC10162004 DOI: 10.1038/s41598-023-34189-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
Researchers are constantly searching for drugs to combat the coronavirus pandemic caused by SARS-CoV-2, which has lasted for over two years. Natural compounds such as phenolic acids are being tested against Mpro and AAK1, which are key players in the SARS-CoV-2 life cycle. This research work aims to study the ability of a panel of natural phenolic acids to inhibit the virus's multiplication directly through Mpro and indirectly by affecting the adaptor-associated protein kinase-1 (AAK1). Pharmacophore mapping, molecular docking, and dynamic studies were conducted over 50 ns and 100 ns on a panel of 39 natural phenolic acids. Rosmarinic acid (16) on the Mpro receptor (- 16.33 kcal/mol) and tannic acid (17) on the AAK1 receptor (- 17.15 kcal/mol) exhibited the best docking energy against both receptors. These favourable docking score values were found to be superior to those of the co-crystallized ligands. Preclinical and clinical research is required before using them simultaneously to halt the COVID-19 life cycle in a synergistic manner.
Collapse
Affiliation(s)
- Heba I Ghamry
- Nutrition and Food Sciences, Department of Home Economics, College of Home Economics, King Khalid University, P.O. Box 960, Abha, 61421, Saudi Arabia
| | - Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| | - Mohamed Kandeel El-Ashrey
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr Elini St., Cairo, 11562, Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, King Salman International University, Ras-Sedr, South Sinai, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Reem I Alsantali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Ahmed A El-Mansi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Doaa Abdelrahman
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|