1
|
Huang L, Li Y, Pang J, Lv L, Zhou J, Liang L, He X, Li J, Xu W, Yang R. Isolation and Characterization of Antimicrobial Metabolites from the Sophora tonkinensis-Associated Fungus Penicillium sp. GDGJ-N37. Molecules 2024; 29:348. [PMID: 38257261 PMCID: PMC10818566 DOI: 10.3390/molecules29020348] [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: 12/01/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Chemical investigation of Penicillium sp. GDGJ-N37, a Sophora tonkinensis-associated fungus, yielded two new azaphilone derivatives, N-isoamylsclerotiorinamine (1) and 7-methoxyl-N-isoamylsclerotiorinamine (2), and four known azaphilones (3-6), together with two new chromone derivatives, penithochromones X and Y (7 and 8). Their structures were elucidated based on spectroscopic data, CD spectrum, and semi-synthesis. Sclerotioramine (3) showed significant antibacterial activities against B. subtilis and S. dysentery, and it also showed most potent anti-plant pathogenic fungi activities against P. theae, C. miyabeanus, and E. turcicum.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Weifeng Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; (L.H.); (Y.L.); (J.P.); (L.L.); (J.Z.); (L.L.); (X.H.); (J.L.)
| | - Ruiyun Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; (L.H.); (Y.L.); (J.P.); (L.L.); (J.Z.); (L.L.); (X.H.); (J.L.)
| |
Collapse
|
2
|
Ayyolath A, Kallingal A, Kundil VT, Suresh AM, Jayadevi Variyar E. Investigating the disease-modifying properties of sclerotiorin in Alzheimer's therapy using acetylcholinesterase inhibition. Chem Biol Drug Des 2023; 102:292-302. [PMID: 37076430 DOI: 10.1111/cbdd.14244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/27/2023] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder caused due to the damage and loss of neurons in specific brain regions. It is the most common form of dementia observed in older people. The symptoms start with memory loss and gradually cause the inability to speak and do day-to-day activities. The cost of caring for those affected individuals is huge and is probably beyond most developing countries capability. Current pharmacotherapy for AD includes compounds that aim to increase neurotransmitters at nerve endings. This can be achieved by cholinergic neurotransmission through inhibition of the cholinesterase enzyme. The current research aims to find natural substances that can be used as drugs to treat AD. The present work identifies and explains compounds with considerable Acetylcholinesterase (AChE) inhibitory activities. The pigment was extracted from the Penicillium mallochii ARA1 (MT373688.1) strain using ethyl acetate, and the active compound was identified using chromatographic techniques followed by structural confirmation with NMR. AChE inhibition experiments, enzyme kinetics, and molecular dynamics simulation studies were done to explain the pharmacological and pharmacodynamic properties. We identified that the compound sclerotiorin in the pigment has AChE inhibitory activity. The compound is stable and can bind to the enzyme non-competitively. Sclerotiorin obeys all the drug-likeliness parameters and can be developed as a promising drug in treating AD.
Collapse
Affiliation(s)
- Aravind Ayyolath
- Laboratory of Bacterial Genetics, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Anoop Kallingal
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Varun Thachan Kundil
- Department of Biotechnology and Microbiology, School of Life Science, Kannur University, Palayad, Kerala, India
| | - Akshay Maniyeri Suresh
- Laboratory of Bacterial Genetics, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - E Jayadevi Variyar
- Department of Biotechnology and Microbiology, School of Life Science, Kannur University, Palayad, Kerala, India
| |
Collapse
|
3
|
Design, synthesis, molecular docking studies and biological evaluation of thiazole carboxamide derivatives as COX inhibitors. BMC Chem 2023; 17:11. [PMID: 36879343 PMCID: PMC9987136 DOI: 10.1186/s13065-023-00924-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/25/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs (NSAIDs) have been the most commonly used class of medications worldwide for the last three decades. OBJECTIVES This study aimed to design and synthesize a novel series of methoxyphenyl thiazole carboxamide derivatives and evaluate their cyclooxygenase (COX) suppressant and cytotoxic properties. METHODS The synthesized compounds were characterized using 1H, 13C-NMR, IR, and HRMS spectrum analysis and were evaluated for their selectivity towards COX-1 and COX-2 using an in vitro COX inhibition assay kit. Besides, their cytotoxicity was evaluated using the Sulforhodamine B (SRB) assay. Moreover, molecular docking studies were conducted to identify the possible binding patterns of these compounds within both COX-1 and COX-2 isozymes, utilizing human X-ray crystal structures. The density functional theory (DFT) analysis was used to evaluate compound chemical reactivity, which was determined by calculating the frontier orbital energy of both HOMO and LUMO orbitals, as well as the HOMO-LUMO energy gap. Finally, the QiKProp module was used for ADME-T analysis. RESULTS The results revealed that all synthesized molecules have potent inhibitory activities against COX enzymes. The percentage of inhibitory activities at 5 µM concentration against the COX2 enzyme was in the range of 53.9-81.5%, while the percentage against the COX-1 enzyme was 14.7-74.8%. That means almost all of our compounds have selective inhibition activities against the COX-2 enzyme, and the most selective compound was 2f, with selectivity ratio (SR) value of 3.67 at 5 µM concentration, which has a bulky group of trimethoxy on the phenyl ring that could not bind well with the COX-1 enzyme. Compound 2h was the most potent, with an inhibitory activity percentage at 5 µM concentration of 81.5 and 58.2% against COX-2 and COX-1, respectively. The cytotoxicity of these compounds was evaluated against three cancer cell lines: Huh7, MCF-7, and HCT116, and negligible or very weak activities were observed for all of these compounds except compound 2f, which showed moderate activities with IC50 values of 17.47 and 14.57 µM against Huh7 and HCT116 cancer cell lines, respectively. Analysis of the molecular docking suggests 2d, 2e, 2f, and 2i molecules were bound to COX-2 isozyme favorably over COX-1 enzyme, and their interaction behaviors within COX-1 and COX-2 isozymes were comparable to celecoxib, as an ideal selective COX-2 drug, which explained their high potency and COX-2 selectivity. The molecular docking scores and expected affinity using the MM-GBSA approach were consistent with the recorded biological activity. The calculated global reactivity descriptors, such as HOMO and LUMO energies and the HOMO-LUMO gaps, confirmed the key structural features required to achieve favorable binding interactions and thus improve affinity. The in silico ADME-T studies asserted the druggability of molecules and have the potential to become lead molecules in the drug discovery process. CONCLUSION In general, the series of the synthesized compounds had a strong effect on both enzymes (COX-1 and COX-2) and the trimethoxy compound 2f was more selective than the other compounds.
Collapse
|
4
|
Sousa TF, de Araújo Júnior MB, Peres EG, Souza MP, da Silva FMA, de Medeiros LS, de Souza ADL, de Souza AQL, Yamagishi MEB, da Silva GF, Koolen HHF, De Queiroz MV. Discovery of dual PKS involved in sclerotiorin biosynthesis in Penicillium meliponae using genome mining and gene knockout. Arch Microbiol 2023; 205:75. [PMID: 36708387 DOI: 10.1007/s00203-023-03414-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/29/2023]
Abstract
Fungi of the genus Penicillium section Sclerotiora have as their main characteristic the presence of orange-pigmented mycelium, which is associated with sclerotiorin, a chlorinated secondary metabolite of the azaphilone subclass of polyketides. Sclerotiorin presents anti-diabetes, antioxidant, anti-inflammatory, anti-Alzheimer, antiviral, and antimicrobial activities, which has always attracted the attention of researchers worldwide. During our ongoing search for azaphilone-producing Amazonian fungi, the strain of Penicillium MMSRG-058 was isolated as an endophyte from the roots of Duguetia stelechantha and showed great capacity for producing sclerotiorin-like metabolites. Using multilocus phylogeny, this strain was identified as Penicillium meliponae. Moreover, based on the genome mining of this strain through the reverse approach, a cluster of putative biosynthetic genes (BGC) responsible for the biosynthesis of sclerotiorin-like metabolites (scl cluster) was identified. The knockout of the sclA (highly reducing PKS) and sclI (non-reducing PKS) genes resulted in mutants with loss of mycelial pigmentation and terminated the biosynthesis of sclerotiorin-like metabolites: geumsanol B, chlorogeumsanol B, 7-deacetylisochromophilone VI, isochromophilone VI, ochrephilone, isorotiorin, and sclerotiorin. Based on these results, a biosynthetic pathway was proposed considering the homology of BGC scl genes with the azaphilone BGCs that have already been functionally characterized.
Collapse
Affiliation(s)
- Thiago F Sousa
- Grupo de Pesquisas em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus, 690065-130, Brazil.,Embrapa Amazônia Ocidental, Manaus, 69010-970, Brazil.,Laboratório de Genética Molecular e de Microrganismos, Universidade Federal de Viçosa, Viçosa, 36570-900, Brazil
| | - Moysés B de Araújo Júnior
- Grupo de Pesquisas em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus, 690065-130, Brazil.,Instituto de Ciências Exatas e Tecnologia, Universidade Federal do Amazonas, Itacoatiara, 69103-128, Brazil
| | - Eldrinei G Peres
- Grupo de Pesquisas em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus, 690065-130, Brazil.,Departamento de Química, Universidade Federal do Amazonas, Manaus, 69067-005, Brazil
| | - Mayane P Souza
- Departamento de Química, Universidade Federal do Amazonas, Manaus, 69067-005, Brazil
| | - Felipe M A da Silva
- Departamento de Química, Universidade Federal do Amazonas, Manaus, 69067-005, Brazil
| | - Lívia S de Medeiros
- Instituto de Ciências Ambientais Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, 09972-270, Brazil
| | - Afonso D L de Souza
- Departamento de Química, Universidade Federal do Amazonas, Manaus, 69067-005, Brazil
| | - Antonia Q L de Souza
- Faculdade de Ciências Agrárias, Universidade Federal do Amazonas, Manaus, 69067-005, Brazil
| | | | | | - Hector H F Koolen
- Grupo de Pesquisas em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus, 690065-130, Brazil
| | - Marisa V De Queiroz
- Laboratório de Genética Molecular e de Microrganismos, Universidade Federal de Viçosa, Viçosa, 36570-900, Brazil.
| |
Collapse
|
5
|
Rashed SA, Hammad SF, Eldakak MM, Khalil IA, Osman A. Assessment of the Anticancer Potentials of the Free and Metal-Organic Framework (UiO-66) - Delivered Phycocyanobilin. J Pharm Sci 2023; 112:213-224. [PMID: 36087776 DOI: 10.1016/j.xphs.2022.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022]
Abstract
Phycocyanin (C-PC) is a constitutive chromoprotein of Arthrospira platensis, which exhibits promising efficacy against different types of cancer. In this study, we cleaved C-PC's chromophore phycocyanobilin (PCB) and demonstrated its ability as an anti-cancer drug for Colorectal cancer (CRC). PCB displayed an anti-cancer effect for CRC (HT-29) cells with IC50 of 108 µg/ml. Assessing the transcripts levels of some biomarkers revealed that the PCB caused an upregulation in the anti-metastatic gene NME1 level and downregulation of the COX-2 level. The flow cytometric results showed the effect of PCB on the arrest of the cell cycle's G1 phase. In addition, we successfully synthesized the UiO-66 (Zr-MOF). We incorporated the PCB into UiO-66 nanoparticles with a loading percentage of 46 %. Assessment of the cytotoxic effects of UiO-66@PCB showed a 2-fold improvement in the IC50 compared to the free PCB. In conclusion, we have shown that PCB displayed a promising potential as an anti-cancer agent. Yet, it is considered a safe and natural substance that can help to mitigate cancer spread and symptoms. In the meantime, UiO-66 can be used as a safe nano-delivery tool for PCB.
Collapse
Affiliation(s)
- Suzan A Rashed
- Biotechnology Program, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, Borg El-Arab, Egypt; Botany and Microbiology Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Sherif F Hammad
- Biotechnology Program, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, Borg El-Arab, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Moustafa M Eldakak
- Genetics Department, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Islam A Khalil
- Pharmaceutics Department, Faculty of Pharmacy and Drug Manufacturing, Misr University for Science and Technology, 6 October, Egypt
| | - Ahmed Osman
- Biotechnology Program, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, Borg El-Arab, Egypt; Department of Biochemistry, Faculty of Science, Ain shams University, Cairo, Egypt
| |
Collapse
|
6
|
Mohsin NUA, Aslam S, Ahmad M, Irfan M, Al-Hussain SA, Zaki MEA. Cyclooxygenase-2 (COX-2) as a Target of Anticancer Agents: A Review of Novel Synthesized Scaffolds Having Anticancer and COX-2 Inhibitory Potentialities. Pharmaceuticals (Basel) 2022; 15:ph15121471. [PMID: 36558921 PMCID: PMC9783503 DOI: 10.3390/ph15121471] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a serious threat to human beings and is the second-largest cause of death all over the globe. Chemotherapy is one of the most common treatments for cancer; however, drug resistance and severe adverse effects are major problems associated with anticancer therapy. New compounds with multi-target inhibitory properties are targeted to surmount these challenges. Cyclooxygenase-2 (COX-2) is overexpressed in cancers of the pancreas, breast, colorectal, stomach, and lung carcinoma. Therefore, COX-2 is considered a significant target for the synthesis of new anticancer agents. This review discusses the biological activity of recently prepared dual anticancer and COX-2 inhibitory agents. The most important intermolecular interactions with the COX-2 enzyme have also been presented. Analysis of these agents in the active area of the COX-2 enzyme could guide the introduction of new lead compounds with extreme selectivity and minor side effects.
Collapse
Affiliation(s)
- Noor ul Amin Mohsin
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sana Aslam
- Department of Chemistry, Government College Women University, Faisalabad 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.A.); (M.E.A.Z.)
| | - Muhammad Irfan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
- Correspondence: (M.A.); (M.E.A.Z.)
| |
Collapse
|
7
|
Wang K, Chen YF, Yang YCSH, Huang HM, Lee SY, Shih YJ, Li ZL, Whang-Peng J, Lin HY, Davis PJ. The power of heteronemin in cancers. J Biomed Sci 2022; 29:41. [PMID: 35705962 PMCID: PMC9202199 DOI: 10.1186/s12929-022-00816-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Heteronemin (Haimian jing) is a sesterterpenoid-type natural marine product that is isolated from sponges and has anticancer properties. It inhibits cancer cell proliferation via different mechanisms, such as reactive oxygen species (ROS) production, cell cycle arrest, apoptosis as well as proliferative gene changes in various types of cancers. Recently, the novel structure and bioactivity evaluation of heteronemin has received extensive attention. Hormones control physiological activities regularly, however, they may also affect several abnormalities such as cancer. L-Thyroxine (T4), steroid hormones, and epidermal growth factor (EGF) up-regulate the accumulation of checkpoint programmed death-ligand 1 (PD-L1) and promote inflammation in cancer cells. Heteronemin suppresses PD-L1 expression and reduces the PD-L1-induced proliferative effect. In the current review, we evaluated research and evidence regarding the antitumor effects of heteronemin and the antagonizing effects of non-peptide hormones and growth factors on heteronemin-induced anti-cancer properties and utilized computational molecular modeling to explain how these ligands interacted with the integrin αvβ3 receptors. On the other hand, thyroid hormone deaminated analogue, tetraiodothyroacetic acid (tetrac), modulates signal pathways and inhibits cancer growth and metastasis. The combination of heteronemin and tetrac derivatives has been demonstrated to compensate for anti-proliferation in cancer cells under different circumstances. Overall, this review outlines the potential of heteronemin in managing different types of cancers that may lead to its clinical development as an anticancer agent.
Collapse
Affiliation(s)
- Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, 250 Wuxing Street, Taipei 110, Taipei, 11031, Taiwan
| | - Yi-Fong Chen
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, 11031, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Sheng-Yang Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ya-Jung Shih
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, 250 Wuxing Street, Taipei 110, Taipei, 11031, Taiwan.,Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Zi-Lin Li
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, 250 Wuxing Street, Taipei 110, Taipei, 11031, Taiwan.,Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Jacqueline Whang-Peng
- Cancer Center, Wan Fang Hospital, Taipei Medical University, No. 111, Section 3, Xinglong Road, Wenshan District, Taipei City, 116, Taipei, 11031, Taiwan.
| | - Hung-Yun Lin
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan. .,Cancer Center, Wan Fang Hospital, Taipei Medical University, No. 111, Section 3, Xinglong Road, Wenshan District, Taipei City, 116, Taipei, 11031, Taiwan. .,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan. .,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan. .,Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, 12144, USA.
| | - Paul J Davis
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, 12144, USA.,Department of Medicine, Albany Medical College, Albany, NY12144, USA
| |
Collapse
|
8
|
Ahmadi M, Bekeschus S, Weltmann KD, von Woedtke T, Wende K. Non-steroidal anti-inflammatory drugs: recent advances in the use of synthetic COX-2 inhibitors. RSC Med Chem 2022; 13:471-496. [PMID: 35685617 PMCID: PMC9132194 DOI: 10.1039/d1md00280e] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/30/2021] [Indexed: 12/24/2022] Open
Abstract
Cyclooxygenase (COX) enzymes comprise COX-1 and COX-2 isoforms and are responsible for prostaglandin production. Prostaglandins have critical roles in the inflammation pathway and must be controlled by administration of selective nonsteroidal anti-inflammatory drugs (NSAIDs). Selective COX-2 inhibitors have been among the most used NSAIDs during the ongoing coronavirus 2019 pandemic because they reduce pain and protect against inflammation-related diseases. In this framework, the mechanism of action of both COX isoforms (particularly COX-2) as inflammation mediators must be reviewed. Moreover, proinflammatory cytokines such as tumor necrosis factor-α and interleukin (IL)-6, IL-1β, and IL-8 must be highlighted due to their major participation in upregulation of the inflammatory reaction. Structural and functional analyses of selective COX-2 inhibitors within the active-site cavity of COXs could enable introduction of lead structures with higher selectivity and potency against inflammation with fewer adverse effects. This review focuses on the biological activity of recently discovered synthetic COX-2, dual COX-2/lipoxygenase, and COX-2/soluble epoxide hydrolase hybrid inhibitors based primarily on the active motifs of related US Food and Drug Administration-approved drugs. These new agents could provide several advantages with regard to anti-inflammatory activity, gastrointestinal protection, and a safer profile compared with those of the NSAIDs celecoxib, valdecoxib, and rofecoxib.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Leibniz Institute for Plasma Science and Technology (INP Greifswald), Center for Innovation Competence (ZIK) plasmatis Felix-Hausdorff-Straße 2 17489 Greifswald Germany
| | - Sander Bekeschus
- Leibniz Institute for Plasma Science and Technology (INP Greifswald), Center for Innovation Competence (ZIK) plasmatis Felix-Hausdorff-Straße 2 17489 Greifswald Germany
| | - Klaus-Dieter Weltmann
- Leibniz Institute for Plasma Science and Technology (INP Greifswald), Center for Innovation Competence (ZIK) plasmatis Felix-Hausdorff-Straße 2 17489 Greifswald Germany
- Leibniz Institute for Plasma Science and Technology (INP Greifswald) Felix-Hausdorff-Straße 2 17489 Greifswald Germany
| | - Thomas von Woedtke
- Leibniz Institute for Plasma Science and Technology (INP Greifswald), Center for Innovation Competence (ZIK) plasmatis Felix-Hausdorff-Straße 2 17489 Greifswald Germany
- Leibniz Institute for Plasma Science and Technology (INP Greifswald) Felix-Hausdorff-Straße 2 17489 Greifswald Germany
- University Medicine Greifswald, Institute for Hygiene and Environmental Medicine Walther-Rathenau-Straße 49A 17489 Germany
| | - Kristian Wende
- Leibniz Institute for Plasma Science and Technology (INP Greifswald), Center for Innovation Competence (ZIK) plasmatis Felix-Hausdorff-Straße 2 17489 Greifswald Germany
| |
Collapse
|
9
|
Semisynthesis and biological evaluation of (+)-sclerotiorin derivatives as antitumor agents for the treatment of hepatocellular carcinoma. Eur J Med Chem 2022; 232:114166. [DOI: 10.1016/j.ejmech.2022.114166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 11/22/2022]
|
10
|
Srour AM, Panda SS, Mostafa A, Fayad W, El-Manawaty MA, A. F. Soliman A, Moatasim Y, El Taweel A, Abdelhameed MF, Bekheit MS, Ali MA, Girgis AS. Synthesis of aspirin-curcumin mimic conjugates of potential antitumor and anti-SARS-CoV-2 properties. Bioorg Chem 2021; 117:105466. [PMID: 34775204 PMCID: PMC8566089 DOI: 10.1016/j.bioorg.2021.105466] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/31/2021] [Indexed: 12/16/2022]
Abstract
Series of piperidone-salicylate conjugates were synthesized through the reaction of 3E,5E-bis(arylidene)-4-piperidones with the appropriate acid chloride of acetylsalicylate in the presence of triethylamine. All the synthesized conjugates reveal antiproliferative properties against A431 (squamous skin) cancer cell line with potency higher than that of 5-fluorouracil. Many of the synthesized agents also exhibit promising antiproliferative properties against HCT116 (colon) cancer cell line, of which 5o and 5c are the most effective with 12.9, 9.8 folds potency compared with Sunitinib. Promising activity is also shown against MCF7 (breast) cancer cell line with 1.19, 1.12 folds relative to 5-fluorouracil. PI-flow cytometry of compound 5c supports the arrest of cell cycle at G1-phase. However, compound 5o and Sunitinib arrest the cell cycle at S-phase. The synthesized conjugates can be considered as multi-targeted tyrosine kinase inhibitors due to the promising properties against VEGFR-2 and EGFR in MCF7 and HCT116. CDOCKER studies support the EGFR inhibitory properties. Compounds 5p and 5i possessing thienylidene heterocycle are anti-SARS-CoV-2 with high therapeutic indices. Many of the synthesized agents show enhanced COX-1/2 properties than aspirin with better selectivity index towards COX-2 relative to COX-1. The possible applicability of the potent candidates discovered as antitumor and anti-SARS-CoV-2 is supported by the safe profile against normal (non-cancer, RPE1 and VERO-E6) cells.
Collapse
Affiliation(s)
- Aladdin M. Srour
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Siva S. Panda
- Department of Chemistry & Physics, Augusta University, Augusta, GA 30912, US
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - May A. El-Manawaty
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Ahmed A. F. Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Ahmed El Taweel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | | | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt,Corresponding author
| |
Collapse
|
11
|
Secondary Metabolites with α-Glucosidase Inhibitory Activity from Mangrove Endophytic Fungus Talaromyces sp. CY-3. Mar Drugs 2021; 19:md19090492. [PMID: 34564154 PMCID: PMC8465095 DOI: 10.3390/md19090492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
Eight new compounds, including two sambutoxin derivatives (1-2), two highly oxygenated cyclopentenones (7-8), four highly oxygenated cyclohexenones (9-12), together with four known sambutoxin derivatives (3-6), were isolated from semimangrove endophytic fungus Talaromyces sp. CY-3, under the guidance of molecular networking. The structures of new isolates were elucidated by analysis of detailed spectroscopic data, ECD spectra, chemical hydrolysis, 13C NMR calculation, and DP4+ analysis. In bioassays, compounds 1-5 displayed better α-glucosidase inhibitory activity than the positive control 1-deoxynojirimycin (IC50 = 80.8 ± 0.3 μM), and the IC50 value was in the range of 12.6 ± 0.9 to 57.3 ± 1.3 μM.
Collapse
|