1
|
Farghadani R, Lim HY, Abdulla MA, Rajarajeswaran J. Novel indole Schiff base β-diiminato compound as an anti-cancer agent against triple-negative breast cancer: In vitro anticancer activity evaluation and in vivo acute toxicity study. Bioorg Chem 2024; 152:107730. [PMID: 39216194 DOI: 10.1016/j.bioorg.2024.107730] [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: 05/28/2024] [Revised: 08/04/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer is the most prevalent cancer among women globally, with triple-negative breast cancer (TNBC) associated with poor prognosis and low five-year survival rates. Schiff base compounds, known for their extensive pharmacological activities, have garnered significant attention in cancer drug research. This study aimed to evaluate the anticancer potential of a novel β-diiminato compound and elucidate its mechanism of action. The compound's effect on cell viability was assessed using MTT assays in breast cancer cell lines including MCF-7 and MDA-MB-231. Cytotoxic effects were further analyzed using trypan blue exclusion and lactate dehydrogenase (LDH) release assays. In order to assess the mechanism of inhibitory activity and mode of cell death induced by this compound, flow cytometry of cell cycle distribution and apoptosis analysis were carried out. Apoptosis incidence was initially assessed through cell and nuclear morphological changes (Hoechst 33342/Propidium iodide (PI) staining) and further confirmed by Annexin V/PI staining and flow cytometry analysis. In addition, the effect of this compound on the disruption of mitochondrial membrane potential (MMP) and generation of the reactive oxygen species (ROS) was determined using the JC-1 indicator and DCFDA dye, respectively. The results demonstrated that the 24 h treatment with β-diiminato compound significantly suppressed the viability of MDA-MB-231 and MCF-7 cancer cells in a dose-dependent manner with the IC50 value of 2.41 ± 0.29 and 3.51 ± 0.14, respectively. The cytotoxic effect of the compound was further confirmed with a dose-dependent increase in the number of dead cells and enhanced LDH level in the culture medium. This compound exerted its anti-proliferative effect by G2/M phase cell growth arrest in MDA-MB-231 breast cancer cells and induced apoptosis-mediated cell death, which involved characteristic changes in cell and nuclear morphology, phosphatidylserine externalization, mitochondrial membrane depolarization, and increased ROS level. Neither hepatotoxicity nor nephrotoxicity was detected in the biochemical and histopathological analysis confirming the safety characterization of this compound usage. Therefore, the results significantly confirmed the potential anticancer activity of a novel β-diiminato compound, as evidenced by the induction of cell cycle arrest and apoptosis, which might be driven by the ROS‑mediated mitochondrial death pathway. This compound can be a promising candidate for future anticancer drug design and TNBC treatment, and further preclinical and clinical studies are warranted.
Collapse
Affiliation(s)
- Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Han Yin Lim
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Jayakumar Rajarajeswaran
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India.
| |
Collapse
|
2
|
Şahin S, Can NN. A Schiff Base with Polymorphic Structure ( Z′ = 2): Investigations with Computational Techniques and in Silico Predictions. Polycycl Aromat Compd 2023. [DOI: 10.1080/10406638.2022.2161585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Songül Şahin
- Department of Chemistry, Faculty of Art and Sciences, Ondokuz Mayis University, Samsun, Turkey
| | - Nisa Nur Can
- Department of Neuroscience, Institute of Health Sciences, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
3
|
Qi J, Luo Y, Zhou Q, Su G, Zhang X, Nie X, Lv M, Li W. Synthesis, structure and anticancer studies of Cu(Ⅱ) and Ni(Ⅱ) complexes based on 2-hydroxy-1-naphthaldehyde-4-aminoantipyrine Schiff-base. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
4
|
Chang LL, Yang J, Lai SQ, Liu XR, Yang ZW, Zhao SS. Synthesis, crystal structures and CT-DNA/BSA binding properties of Co(III) and Cu(II) complexes with bipyridine Schiff base ligand. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2021.120751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
5
|
Zhang Y, Zhou T, Li J, Xu N, Cai M, Zhang H, Zhao Q, Wang S. Au Catalyzing Control Release NO in vivo and Tumor Growth-Inhibiting Effect in Chemo-Photothermal Combination Therapy. Int J Nanomedicine 2021; 16:2501-2513. [PMID: 33824588 PMCID: PMC8018432 DOI: 10.2147/ijn.s270466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/27/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Aim to obtain a NO donor that can control released NO in vivo with the high efficacy of tumor suppression and targeting, a nanoplatform consisting of FA-Fe3O4@mSiO2-Au/DOX was constructed. METHODS In vitro, the nanoplatform catalyzed NO's release with the maximum value of 4.91 μM within 60 min at 43°C pH=5.0, which was increased by 1.14 times when the temperature was 37°C. In vivo, 11.7 μg Au in the tumor tissue was found to catalyze S-nitrosoglutathione continuously, and 54 μM NO was checked out in the urine. RESULTS AND DISCUSSION The high concentration of NO was found to increase the apoptotic rate and to reduce tumor proliferation. In the chemo-photothermal combination therapy, the tumor inhibition rate was increased up to 94.3%, and Au's contribution from catalyzing NO release NO was 8.17%.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Tianfu Zhou
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Jian Li
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Nuo Xu
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Mingze Cai
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Hong Zhang
- Van ’T Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, 1098 XH, the Netherlands
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| |
Collapse
|
6
|
Mukhtar SS, Hassan AS, Morsy NM, Hafez TS, Saleh FM, Hassaneen HM. Design, synthesis, molecular prediction and biological evaluation of pyrazole-azomethine conjugates as antimicrobial agents. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1894338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Shorouk S. Mukhtar
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Dokki, Egypt
| | - Ashraf S. Hassan
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Dokki, Egypt
| | - Nesrin M. Morsy
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Dokki, Egypt
| | - Taghrid S. Hafez
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Dokki, Egypt
| | - Fatma M. Saleh
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Hamdi M. Hassaneen
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
7
|
New cytotoxic zinc(II) and copper(II) complexes of Schiff base ligands derived from homopiperonylamine and halogenated salicylaldehyde. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2020.120171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
8
|
Narayanan J, Hernández JG, Aguilar CAH, Rodríguez MM, Cerda SDG. Glutamine chelation governs the selective inhibition of Staphylococcus aureus and Salmonella typhi growth by cis-dichloro-bis(8-quinolinolato)zirconium(IV): Theory and experiment. Eur J Pharm Sci 2020; 151:105427. [PMID: 32544422 DOI: 10.1016/j.ejps.2020.105427] [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: 03/31/2020] [Revised: 05/25/2020] [Accepted: 06/11/2020] [Indexed: 10/24/2022]
Abstract
Quinolone-based Schiff base zirconium(IV) complex was studied as potential bacterial inhibitor against Gram-positive Staphylococcus aureus and Gram-negative Salmonella typhi growth, showing that the interaction of the complex with L-glutamine which presents in the membrane of wall leads cell death, and the mode of bacterial interaction was analyzed theoretically by DFT. Furthermore, the interaction of different amino acid residues L-alanine, D-alanine, L-lysine and D-glutamine with the metal complex through UV-vis docking studies was conducted observing that D-glutamine interacts efficiently among other amino acid residues. This observation is consistent with the interaction of the metal complex that was effective when participating in an insight of the peptidoglycan cell wall since the binding nature of glutamine potentially inhibits these microorganisms.
Collapse
Affiliation(s)
- Jayanthi Narayanan
- Division de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad Politécnica, Tultitlan, Estado de México, CP 54910, México.
| | - José Guadalupe Hernández
- Centro Tecnológico, Facultad de Estudios Superiores (FES-Aragón), Universidad Nacional Autónoma de México (UNAM), Estado de México, CP 57130, México
| | - Carlos Alberto Huerta Aguilar
- Division de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad Politécnica, Tultitlan, Estado de México, CP 54910, México
| | - Miguel Morales Rodríguez
- Division de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad Politécnica, Tultitlan, Estado de México, CP 54910, México
| | - Susana Dianey Gallegos Cerda
- Division de Ingeniería en Nanotecnología, Universidad Politécnica del Valle de México, Av. Mexiquense s/n esquina Av. Universidad Politécnica, Tultitlan, Estado de México, CP 54910, México
| |
Collapse
|
9
|
Serra M, Columbano A, Ammarah U, Mazzone M, Menga A. Understanding Metal Dynamics Between Cancer Cells and Macrophages: Competition or Synergism? Front Oncol 2020; 10:646. [PMID: 32426284 PMCID: PMC7203474 DOI: 10.3389/fonc.2020.00646] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
Metal ions, such as selenium, copper, zinc, and iron are naturally present in the environment (air, drinking water, and food) and are vital for cellular functions at chemical, molecular, and biological levels. These trace elements are involved in various biochemical reactions by acting as cofactors for many enzymes and control important biological processes by binding to the receptors and transcription factors. Moreover, they are essential for the stabilization of the cellular structures and for the maintenance of genome stability. A body of preclinical and clinical evidence indicates that dysregulation of metal homeostasis, both at intracellular and tissue level, contributes to the pathogenesis of many different types of cancer. These trace minerals play a crucial role in preventing or accelerating neoplastic cell transformation and in modulating the inflammatory and pro-tumorigenic response in immune cells, such as macrophages, by controlling a plethora of metabolic reactions. In this context, macrophages and cancer cells interact in different manners and some of these interactions are modulated by availability of metals. The current review discusses the new findings and focuses on the involvement of these micronutrients in metabolic and cellular signaling mechanisms that influence macrophage functions, onset of cancer and its progression. An improved understanding of "metallic" cross-talk between macrophages and cancer cells may pave the way for innovative pharmaceutical or dietary interventions in order to restore the balance of these trace elements and also strengthen the chemotherapeutic treatment.
Collapse
Affiliation(s)
- Marina Serra
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Amedeo Columbano
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ummi Ammarah
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| | - Alessio Menga
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| |
Collapse
|
10
|
Tan XJ, Wang D, Hei XM, Yang FC, Zhu YL, Xing DX, Ma JP. Synthesis, crystal structures, antiproliferative activities and reverse docking studies of eight novel Schiff bases derived from benzil. ACTA CRYSTALLOGRAPHICA SECTION C-STRUCTURAL CHEMISTRY 2020; 76:44-63. [PMID: 31919307 DOI: 10.1107/s2053229619015687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/19/2019] [Indexed: 12/15/2022]
Abstract
Eight novel Schiff bases derived from benzil dihydrazone (BDH) or benzil monohydrazone (BMH) and four fused-ring carbonyl compounds (3-formylindole, FI; 3-acetylindole, AI; 3-formyl-1-methylindole, MFI; 1-formylnaphthalene, FN) were synthesized and characterized by elemental analysis, ESI-QTOF-MS, 1H and 13C NMR spectroscopy, as well as single-crystal X-ray diffraction. They are (1Z,2Z)-1,2-bis{(E)-[(1H-indol-3-yl)methylidene]hydrazinylidene}-1,2-diphenylethane (BDHFI), C32H24N6, (1Z,2Z)-1,2-bis{(E)-[1-(1H-indol-3-yl)ethylidene]hydrazinylidene}-1,2-diphenylethane (BDHAI), C34H28N6, (1Z,2Z)-1,2-bis{(E)-[(1-methyl-1H-indol-3-yl)methylidene]hydrazinylidene}-1,2-diphenylethane (BMHMFI) acetonitrile hemisolvate, C34H28N6·0.5CH3CN, (1Z,2Z)-1,2-bis{(E)-[(naphthalen-1-yl)methylidene]hydrazinylidene}-1,2-diphenylethane (BDHFN), C36H26N4, (Z)-2-{(E)-[(1H-indol-3-yl)methylidene]hydrazinylidene}-1,2-diphenylethanone (BMHFI), C23H17N3O, (Z)-2-{(E)-[1-(1H-indol-3-yl)ethylidene]hydrazinylidene}-1,2-diphenylethanone (BMHAI), C24H19N3O, (Z)-2-{(E)-[(1-methyl-1H-indol-3-yl)methylidene]hydrazinylidene}-1,2-diphenylethanone (BMHMFI), C24H19N3O, and (Z)-2-{(E)-[(naphthalen-1-yl)methylidene]hydrazinylidene}-1,2-diphenylethanone (BMHFN) C25H18N2O. Moreover, the in vitro cytotoxicity of the eight title compounds was evaluated against two tumour cell lines (A549 human lung cancer and 4T1 mouse breast cancer) and two normal cell lines (MRC-5 normal lung cells and NIH 3T3 fibroblasts) by MTT assay. The results indicate that four (BDHMFI, BDHFN, BMHMFI and BMHFN) are inactive and the other four (BDHFI, BDHAI, BMHFI and BMHAI) show severe toxicities against human A549 and mouse 4T1 cells, similar to the standard cisplatin. All the compounds exhibited weaker cytotoxicity against normal cells than cancer cells. The Swiss Target Prediction web server was applied for the prediction of protein targets. After analyzing the differences in frequency hits between these active and inactive Schiff bases, 18 probable targets were selected for reverse docking with the Surflex-dock function in SYBYL-X 2.0 software. Three target proteins, i.e. human ether-á-go-go-related (hERG) potassium channel, the inhibitor of apoptosis protein 3 and serine/threonine-protein kinase PIM1, were chosen as the targets. Finally, the ligand-based structure-activity relationships were analyzed based on the putative protein target (hERG) docking results, which will be used to design and synthesize novel hERG ion channel inhibitors.
Collapse
Affiliation(s)
- Xue Jie Tan
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province 250353, People's Republic of China
| | - Di Wang
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province 250353, People's Republic of China
| | - Xiao Ming Hei
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province 250353, People's Republic of China
| | - Feng Cun Yang
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province 250353, People's Republic of China
| | - Ya Ling Zhu
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province 250353, People's Republic of China
| | - Dian Xiang Xing
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong Province 250353, People's Republic of China
| | - Jian Ping Ma
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, People's Republic of China
| |
Collapse
|
11
|
Gupta SD, Swapanthi PS, Bhagya D, Federicci F, Mazaira GI, Galigniana MD, Subrahmanyam CVS, Gowrishankar NL, Raghavendra NM. Rational Identification of Hsp90 Inhibitors as Anticancer Lead Molecules by Structure Based Drug Designing Approach. Anticancer Agents Med Chem 2019; 20:369-385. [PMID: 31713499 DOI: 10.2174/1871520619666191111152050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 08/28/2019] [Accepted: 09/18/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Heat shock protein 90 (Hsp90) is an encouraging anticancer target for the development of clinically significant molecules. Schiff bases play a crucial role in anticancer research because of their ease of synthesis and excellent antiproliferative effect against multiple cancer cell lines. Therefore, we started our research work with the discovery of resorcinol/4-chloro resorcinol derived Schiff bases as Hsp90 inhibitors, which resulted in the discovery of a viable anticancer lead molecule. OBJECTIVE The objective of the study is to discover more promising lead molecules using our previously established drug discovery program, wherein the rational drug design is achieved by molecular docking studies. METHODS The docking studies were carried out by using Surflex Geom X programme of Sybyl X-1.2 version software. The molecules with good docking scores were synthesized and their structures were confirmed by IR, 1H NMR and mass spectral analysis. Subsequently, the molecules were evaluated for their potential to attenuate Hsp90 ATPase activity by Malachite green assay. The anticancer effect of the molecules was examined on PC3 prostate cancer cell lines by utilizing 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay methodology. RESULTS Schiff bases 11, 12, 20, 23 and 27 exhibiting IC50 value below 1μM and 15μM, in malachite green assay and MTT assay, respectively, emerged as viable lead molecules for future optimization. CONCLUSION The research work will pave the way for the rational development of cost-effective Schiff bases as Hsp90 inhibitors as the method employed for the synthesis of the molecules is simple, economic and facile.
Collapse
Affiliation(s)
- Sayan D Gupta
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India.,R&D centre, Department of Pharmaceutical Sciences, Jawaharlal Nehru Technological University, Hyderabad, India
| | - Pappu S Swapanthi
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - Deshetti Bhagya
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - Fernando Federicci
- Department of Biological Chemistry, Faculty of Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Gisela I Mazaira
- Department of Biological Chemistry, Faculty of Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Mario D Galigniana
- Department of Biological Chemistry, Faculty of Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Experimental Biology and Medicine-CONICET, Buenos Aires, Argentina
| | - Chavali V S Subrahmanyam
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | | | - Nulgumnalli M Raghavendra
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| |
Collapse
|
12
|
Farghadani R, Seifaddinipour M, Rajarajeswaran J, Abdulla MA, Mohd Hashim NB, Khaing SL, Salehen NB. In vivo acute toxicity evaluation and in vitro molecular mechanism study of antiproliferative activity of a novel indole Schiff base β-diiminato manganese III complex in hormone-dependent and triple negative breast cancer cells. PeerJ 2019; 7:e7686. [PMID: 31608167 PMCID: PMC6786247 DOI: 10.7717/peerj.7686] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most frequently diagnosed cancer among women worldwide. Recently, increasing attention has been paid to the anticancer effects of transition metal complexes of indole Schiff bases. β-diiminato ManganeseIII complex has shown promising cell cycle arrest and apoptosis induction against MCF-7 and MDA-MB-231 breast cancer cells. In this study, time- and dose- dependent inhibitory activity were evaluated using MTT assay after 48 h and 72 h exposure time. In addition, median effect analysis was conducted according to Chou-Talalay method to investigate whether MnIII complex has synergistic effect in combination with chemotherapeutic drugs on inhibiting breast cancer cell growth. The molecular mechanisms underlying its potent antiproliferative effect was determined through bioluminescent caspase-3/7, -8 and -9 activity assays and quantitative expression analysis of cell cycle- and apoptosis-related genes. Furthermore, safety evaluation of MnIII complex was assessed through the acute oral toxicity test in in vivo model. The MTT assay results revealed that it potently reduced the viability of MCF-7 (IC50 of 0.63 ± 0.07 µg/mL for 48 h and 0.39 ± 0.08 µg/mL for 72 h) and MDA-MB-231 (1.17 ± 0.06 µg/mL for 48 h, 1.03 ± 0.15 µg/mL for 72 h) cells in dose- and time-dependent manner. Combination treatment also enhanced the cytotoxic effects of doxorubicin but not tamoxifen on inhibiting breast cancer cell growth. The involvement of intrinsic and extrinsic pathway in apoptosis induction was exhibited through the increased activity of caspase-9 and caspase-8, respectively, leading to enhanced downstream executioner caspase-3/7 activity in treated MCF-7 and MDA-MB-231 cells. In addition, gene expression analysis revealed that MnIII complex exerts its antiproliferative effect via up-and down-regulation of p21 and cyclin D1, respectively, along with increased expression of Bax/Bcl-2 ratio, TNF-α, initiator caspase-8 and -10 and effector caspase-3 in MCF-7 and MDA-MB-231 cells. However, the results did not show increased caspase-8 activity in treated MCF-7 cells. Furthermore, in vivo acute oral toxicity test revealed no signs of toxicity and mortality in treated animal models compared to the control group. Collectively, the promising inhibitory effect and molecular and mechanistic evidence of antiproliferative activity of MnIII complex and its safety characterization have demonstrated that it may have therapeutic value in breast cancer treatment worthy of further investigation and development.
Collapse
Affiliation(s)
- Reyhaneh Farghadani
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Maryam Seifaddinipour
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Jayakumar Rajarajeswaran
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Si Lay Khaing
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nur'ain Binti Salehen
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Petanidis S, Kioseoglou E, Salifoglou A. Metallodrugs in Targeted Cancer Therapeutics: Aiming at Chemoresistance- related Patterns and Immunosuppressive Tumor Networks. Curr Med Chem 2019; 26:607-623. [DOI: 10.2174/0929867324666171116125908] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022]
Abstract
Tumor cell chemoresistance is a major challenge in cancer therapeutics. Major
select metal-based drugs are potent anticancer mediators yet they exhibit adverse sideeffects
and are efficient against limited types of malignancies. A need, therefore, arises
for novel metallodrugs with improved efficacy and decreased toxicity. Enhancement of
antitumor drugs based on anticancer metals is currently a very active research field, with
considerable efforts having been made toward elucidating the mechanisms of immune action
of complex metalloforms and optimizing their immunoregulatory bioactivity through
appropriate synthetic structural modification(s) and encapsulation in suitable nanocarriers,
thereby enhancing their selectivity, specificity, stability, and bioactivity. In that respect,
comprehending the molecular factors involved in drug resistance and immune response
may help us develop new approaches toward more promising chemotherapies, reducing
the rate of relapse and overcoming chemoresistance. In this review, a) molecular immunerelated
mechanisms in the tumor microenvironment, responsible for lower drug sensitivity
and tumor relapse, along with b) strategies for reversing drug resistance and targeting
immunosuppressive tumor networks, while concurrently optimizing the design of complex
metalloforms bearing anti-tumor activity, are discussed in an effort to identify and
overcome chemoresistance mechanisms for effective tumor immunotherapeutic approaches.
Collapse
Affiliation(s)
- Savvas Petanidis
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Efrosini Kioseoglou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Athanasios Salifoglou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| |
Collapse
|
14
|
Biswas N, Saha S, Khanra S, Sarkar A, Prasad Mandal D, Bhattacharjee S, Chaudhuri A, Chakraborty S, Roy Choudhury C. Example of two novel thiocyanato bridged copper (II) complexes derived from substituted thiosemicarbazone ligand: structural elucidation, DNA/albumin binding, biological profile analysis, and molecular docking study. J Biomol Struct Dyn 2018; 37:2801-2822. [PMID: 30101692 DOI: 10.1080/07391102.2018.1503564] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Two novel copper (II) substituted thiosemicarbazone Schiff base complexes [Cu(L1)(µ-SCN)]n(NO3)2 (1) and [Cu2(µ-SCN)(SCN)(L2)2](NO3) (2) have been synthesized by condensing substituted thiosemicarbazides like 4-methyl-3-thiosemicarbazide or 4-ethyl-3-thiosemicarbazide with 2-acetylpyridine. Both the metal complexes 1 and 2 are characterized using different spectroscopic techniques like IR, UV-Vis, ESR spectroscopy followed by elemental analysis, cyclic voltammetric measurement and single crystal X-ray structure analysis. X-ray crystal structure analysis reveal that complex 1 is polymeric while complex 2 is dimeric in nature. The coordination geometry around Cu(II) are square pyramidal in which thiosemicarbazone Schiff base ligand coordinate to the central Cu(II) atom in tridentate fashion. The prominent interaction patterns of 1 and 2 with CT-DNA were examined by employing electronic absorption and emission spectral titrations, cyclic voltammetry and viscosity measurements. All the results show that CT-DNA binds with both copper (II) complexes 1 and 2. Furthermore, protein binding ability in vitro of complexes 1 and 2 with both BSA and HSA were carried out using multispectroscopic techniques and a static quenching pattern was observed in both cases. Molecular docking study was employed to ascertain the exact mechanism of action of 1 and 2 with DNA and protein molecules (BSA and HSA). In vitro cytotoxicity activity of complexes 1 and 2 toward AGS and A549 was evaluated using MTT assay which demonstrates that both complexes 1 and 2 have superior prospectus to act as anticancer agents. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Niladri Biswas
- a Department of Chemistry , West Bengal State University , Barasat , India
| | - Sandeepta Saha
- a Department of Chemistry , West Bengal State University , Barasat , India.,b Sripur High School , Madhyamgram , India
| | - Sumit Khanra
- c Department of Chemistry , Indian Institute of Science Education and Research , Kolkata Mohanpur , India
| | - Arnab Sarkar
- d Department of Zoology , West Bengal State University , Barasat , India
| | - Deba Prasad Mandal
- d Department of Zoology , West Bengal State University , Barasat , India
| | | | - Ankur Chaudhuri
- e Department of Microbiology , West Bengal State University , Barasat , India
| | - Sibani Chakraborty
- e Department of Microbiology , West Bengal State University , Barasat , India
| | | |
Collapse
|
15
|
Sıdır YG, Aslan C, Berber H, Sıdır İ. The electronic structure, solvatochromism, and electric dipole moments of new Schiff base derivatives using absorbance and fluorescence spectra. Struct Chem 2018. [DOI: 10.1007/s11224-018-1228-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
16
|
Prasher P, Sharma M. Medicinal chemistry of acridine and its analogues. MEDCHEMCOMM 2018; 9:1589-1618. [PMID: 30429967 PMCID: PMC6195008 DOI: 10.1039/c8md00384j] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 08/14/2018] [Indexed: 02/01/2023]
Abstract
'Acridine' along with its functional analogue 'Acridone' is the most privileged pharmacophore in medicinal chemistry with diverse applications ranging from DNA intercalators, endonuclease mimics, ratiometric selective ion sensors, and P-glycoprotein inhibitors in countering the multi-drug resistance, enzyme inhibitors, and reversals of neurodegenerative disorders. Their interaction with DNA and ability of selectively identifying numerous biologically useful ions has cemented exploitability of the acridone nucleus in modern day therapeutics. Additionally, most derivatives and salts of acridine are planar, crystalline, and stable displaying a strong fluorescence which, when coupled with their marked bio selectivity and low cytotoxicity, enables the studying and monitoring of several biochemical, metabolic, and pharmacological processes. In this review, a detailed picture covering the important therapeutic aspects of the acridone nucleus and its functional analogues is discussed.
Collapse
Affiliation(s)
- Parteek Prasher
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
- Department of Chemistry , University of Petroleum & Energy Studies , Dehradun 248007 , India . ;
| | - Mousmee Sharma
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India
| |
Collapse
|
17
|
Hu K, Liu C, Li J, Liang F. Copper(ii) complexes based on quinoline-derived Schiff-base ligands: synthesis, characterization, HSA/DNA binding ability, and anticancer activity. MEDCHEMCOMM 2018; 9:1663-1672. [PMID: 30429971 DOI: 10.1039/c8md00223a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/01/2018] [Indexed: 12/20/2022]
Abstract
Three copper(ii) complexes, [Cu(L1)(NO3)2] (C1), [Cu(L2)Cl2] (C2) and [Cu(L2)SO4]2·H2O (C3), were designed and synthesized by the reaction of Cu(NO3)2·3H2O, CuCl2·2H2O and CuSO4·5H2O with a quinoline-derived Schiff base ligand, L1 or L2, prepared by the condensation of quinoline-8-carbaldehyde with 4-aminobenzoic acid methyl ester or 4-aminobenzoic acid ethyl ester (benzocaine). The efficient bindings of the C1-C3 complexes with human serum albumin (HSA) and calf thymus DNA (CT-DNA) were analyzed by spectroscopy and molecular docking. These complexes could significantly quench the fluorescence of HSA through the static quenching process, and hydrophobic interactions with HSA through the sub-domain IIA and IIIA cavities. The complexes bind to DNA via the intercalative mode and they fit well into the curved contour of the DNA target in the minor groove region. Furthermore, the interaction abilities of the Cu(ii) complexes with HSA/DNA were greater as compared to their corresponding ligands. Interestingly, C1-C3, particularly C3, exhibited more cytotoxicity toward HeLa cells compared to normal HL-7702 cells and three other tumor cell lines (Hep-G2, NCI-H460, and MGC80-3). Their cytotoxicity toward the HeLa cell lines was 1.9-3.5-fold more potent than cisplatin. Further studies indicated that these complexes arrested the cell cycle in the G0/G1 phase and promoted tumor cell apoptosis via a reactive oxygen species (ROS)-mediated mitochondrial pathway.
Collapse
Affiliation(s)
- Kun Hu
- State Key Laboratory Cultivation Base for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmacy , Guangxi Normal University , 15 Yucai Road , Guilin 541004 , P. R. China . ;
| | - Chensi Liu
- State Key Laboratory Cultivation Base for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmacy , Guangxi Normal University , 15 Yucai Road , Guilin 541004 , P. R. China . ;
| | - Jingui Li
- State Key Laboratory Cultivation Base for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmacy , Guangxi Normal University , 15 Yucai Road , Guilin 541004 , P. R. China . ;
| | - Fupei Liang
- State Key Laboratory Cultivation Base for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmacy , Guangxi Normal University , 15 Yucai Road , Guilin 541004 , P. R. China . ; .,Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials , College of Chemistry and Bioengineering , Guilin University of Technology , Guilin 541004 , China
| |
Collapse
|
18
|
Ameri A, Khodarahmi G, Forootanfar H, Hassanzadeh F, Hakimelahi GH. Hybrid Pharmacophore Design, Molecular Docking, Synthesis, and Biological Evaluation of Novel Aldimine-Type Schiff
Base Derivatives as Tubulin Polymerization Inhibitor. Chem Biodivers 2018; 15:e1700518. [DOI: 10.1002/cbdv.201700518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 12/29/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Alieh Ameri
- Department of Medicinal Chemistry; Faculty of Pharmacy; Kerman University of Medical Sciences; Kerman Iran
| | - Ghadamali Khodarahmi
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
| | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology; Faculty of Pharmacy; Kerman University of Medical Sciences; Kerman Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
| | - Gholam-Hosein Hakimelahi
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
- Institute of Chemistry; Academia Sinica; Nankang Taipei Taiwan
| |
Collapse
|
19
|
Banerjee A, Banerjee K, Sinha A, Das S, Majumder S, Majumdar S, Choudhuri SK. A zinc Schiff base complex inhibits cancer progression both in vivo and in vitro by inducing apoptosis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 56:383-392. [PMID: 29145169 DOI: 10.1016/j.etap.2017.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 05/25/2023]
Abstract
Cancer chemotherapy suffers from selectivity and undesired toxicity of the drugs. Since zinc is a biocompatible tracer element and cysteine derivatives are used in cancer chemoprevention, we intend to develop a complex of zinc and cysteine-derivatives as potent, non-toxic anticancer agents. Herein, we synthesized and characterized cysteine based ligand, 2-[(2-Hydroxy-3-methoxy-benzylidene)-amino]-3-mercapto-propionic acid and its Zn-complex, which are found to be non-toxic towards normal human PBMC. Data also revealed that only Zn-complex exhibited remarkable apoptosis in drug-sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 cancer cells as assessed by MTT, Cell cycle and AnnexinV binding assay. Moreover, Zn-complex altered ROS and GSH level of the respective cell lines. Finally, treatment of Zn-complex in Swiss albino mice did not show any systemic toxicity in preliminary trials in normal mice and remarkably increased the life-span of EAC bearing mice. In conclusion, the synthesized Zn-complex may be developed for efficacious, multidrug resistance reversal, non-toxic chemotherapeutic agents in future.
Collapse
Affiliation(s)
- Arpita Banerjee
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India; Department of Chemistry, Rammohan College, 102/1, Raja Rammohan Sarani, College Street, Kolkata, West Bengal 700009, India
| | - Kaushik Banerjee
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Abhinaba Sinha
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Satyajit Das
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Saikat Majumder
- Division of Molecular Medicine, Bose Institute, P 1/12, C. I. T. Road, Scheme - VIIM, Kolkata, 700054, West Bengal, India
| | - Subrata Majumdar
- Division of Molecular Medicine, Bose Institute, P 1/12, C. I. T. Road, Scheme - VIIM, Kolkata, 700054, West Bengal, India
| | - Soumitra Kumar Choudhuri
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India.
| |
Collapse
|
20
|
Banerjee K, Biswas MK, Choudhuri SK. A newly synthesized nickel chelate can selectively target and overcome multidrug resistance in cancer through redox imbalance both in vivo and in vitro. J Biol Inorg Chem 2017; 22:1223-1249. [PMID: 29063196 DOI: 10.1007/s00775-017-1498-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/06/2017] [Indexed: 01/24/2023]
Abstract
Induction of undesired toxicity and emergence of multidrug resistance (MDR) are the major obstacles for cancer treatment. Moreover, aggressive cancers are less sensitive towards existing chemotherapeutics. Therefore, selective targeting of cancers without inducing undesired side effects and designing proper strategies to overcome MDR has utmost importance in modern chemotherapy. Previously we revealed the anticancer properties of some transition metal chelates of Schiff base, but the effectiveness of nickel complex is still unrevealed. Herein, we synthesized and characterized a Schiff base nickel chelate, nickel-(II) N-(2-hydroxyacetophenone) glycinate (NiNG), through different spectroscopic means. NiNG proves to be a broad spectrum anticancer agent with considerable efficacy to overcome MDR in cancer. Antiproliferative effects of NiNG was evaluated using drug-resistant (CEM/ADR5000; NIH-MDR-G185; EAC/Dox), drug-sensitive aggressive (Hct116; CCRF-CEM; EAC/S) and normal (NIH-3T3) cells that reveal the selective nature of NiNG towards drug resistant and sensitive cancer cells without inducing any significant toxicity in normal cells. Moreover, NiNG involves reactive oxygen species (ROS)-mediated redox imbalance for induction of caspase 3-dependent apoptosis in aggressive drug-sensitive Hct116 and drug-resistant NIH-MDR-G185 cells through disruption of mitochondrial membrane potential. Moreover, intraperitoneal (i.p.) application of NiNG at non-toxic doses caused significant increase in the life-span of Swiss albino mice bearing sensitive and doxorubicin-resistant subline of Ehrlich ascites carcinoma cells. It is noteworthy that, in vitro NiNG can only overcome P-glycoprotein-mediated MDR while in vivo NiNG can overcome MRP1-mediated MDR in cancer. Therefore, NiNG has therapeutic potential to target and overcome MDR in cancer.
Collapse
Affiliation(s)
- Kaushik Banerjee
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, India
| | - Manas Kumar Biswas
- Department of Chemistry, Ramakrishna Mission Residential College, Kolkata, India
| | - Soumitra Kumar Choudhuri
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, India.
| |
Collapse
|
21
|
Sinha A, Banerjee K, Banerjee A, Sarkar A, Ahir M, Adhikary A, Chatterjee M, Choudhuri SK. Induction of apoptosis in human colorectal cancer cell line, HCT-116 by a vanadium- Schiff base complex. Biomed Pharmacother 2017; 92:509-518. [PMID: 28575808 DOI: 10.1016/j.biopha.2017.05.108] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 05/20/2017] [Accepted: 05/22/2017] [Indexed: 01/17/2023] Open
Abstract
Vanadium compounds are well known for their therapeutic interventions against several diseases. Various biochemical attributes of vanadium complexes inspired us to evaluate the cancer cell killing efficacy of the vanadium complex, viz., vanadyl N-(2-hydroxyacetophenone) glycinate [VO(NG)2]. Previously we showed that VO(NG)2 is an effective anticancer agent in in vitro and in vivo cancer models and imposed miniscule side effects. Herein we report that VO(NG)2 is significantly cytotoxic to various cancer cell lines. Furthermore, this redox active vanadyl complex altered the redox homeostatsis of many human cancer cell lines significantly. VO(NG)2 actuates programmed cell death in human colorectal carcinoma cells(HCT-116) through mitochondrial outer membrane permeabilization but in caspase independent manner, possibly by altering cellular redox status and by inflicting DNA damage. Thus, the present work is an attempt to provide many evidences regarding the potent and selective chemotherapeutic efficacy of the novel VO(NG)2.
Collapse
Affiliation(s)
- Abhinaba Sinha
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata 700 026, India
| | - Kaushik Banerjee
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata 700 026, India
| | - Arpita Banerjee
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata 700 026, India
| | - Avijit Sarkar
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244, A.J.C. Bose Road, Kolkata 700020, India
| | - Manisha Ahir
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, India
| | - Arghya Adhikary
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, India
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244, A.J.C. Bose Road, Kolkata 700020, India
| | - Soumitra Kumar Choudhuri
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata 700 026, India.
| |
Collapse
|
22
|
Chigurupati S, Shaikh SA, Mohammad JI, Selvarajan KK, Nemala AR, Khaw CH, Teoh CF, Kee TH. In vitro antioxidant and in vivo antidepressant activity of green synthesized azomethine derivatives of cinnamaldehyde. Indian J Pharmacol 2017; 49:229-235. [PMID: 29033482 PMCID: PMC5637133 DOI: 10.4103/ijp.ijp_293_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 05/19/2017] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVES In this study, three (CS-1 to CS-3) azomethine derivatives of cinnamaldehyde were green synthesized, characterized, and their antioxidant and antidepressant activities were explored. MATERIALS AND METHODS The antioxidant effect of these compounds was initially performed in vitro using 1,1-diphenyl-2-picrylhydrazyl (DPPH) and 2,2-azinobis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) assay methods before subjecting them to in vivo experiments. Compounds showing potent antioxidant activity (CS-1 and CS-2) were investigated further for their antidepressant activity using the forced swim test (FST) and tail suspension test (TST). Ascorbic acid (AA) and fluoxetine (20 mg/kg, p.o) were used as reference drugs for comparison in the antioxidant and antidepressant experiments, respectively. RESULTS It was observed that CS-2 and CS-3 exhibited highest DPPH (half maximal inhibitory concentration [IC50]: 16.22 and 25.18 μg/mL) and ABTS (IC50: 17.2 and 28.86 μg/mL) radical scavenging activity, respectively, compared to AA (IC50: 15.73 and 16.79 μg/mL) and therefore, both CS-2 and CS-3 were tested for their antidepressant effect using FST and TST as experimental models. Pretreatment of CS-2 and CS-3 (20 mg/kg) for 10 days considerably decreased the immobility time in both the FST and TST models. CONCLUSION The antioxidant and antidepressant effect of CS-2 and CS-3 may be attributed to the presence of azomethine linkage in the molecule.
Collapse
Affiliation(s)
- Sridevi Chigurupati
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah, Malaysia
| | - Sohrab Akhtar Shaikh
- Department of Pharmacology, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah, Malaysia
| | | | | | - Appala Raju Nemala
- Department of Pharmaceutical Chemistry, Sultan Ul Uloom College of Pharmacy, Hyderabad, Telangana, India
| | - Chu How Khaw
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah, Malaysia
| | - Chun Foo Teoh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah, Malaysia
| | - Ting Hei Kee
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah, Malaysia
| |
Collapse
|
23
|
Banerjee K, Das S, Majumder S, Majumdar S, Biswas J, Choudhuri SK. Modulation of cell death in human colorectal and breast cancer cells through a manganese chelate by involving GSH with intracellular p53 status. Mol Cell Biochem 2016; 427:35-58. [PMID: 28012015 DOI: 10.1007/s11010-016-2896-6] [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: 08/31/2016] [Accepted: 12/02/2016] [Indexed: 10/20/2022]
Abstract
Chemotherapy is central to current treatment modality especially for advanced and metastatic colorectal and breast cancers. Targeting the key molecular events of the neoplastic cells may open a possibility to treat cancer. Although some improvements in understanding of colorectal and breast cancer treatment have been recorded, the involvement of glutathione (GSH) and dependency of p53 status on the modulation of GSH-mediated treatment efficacy have been largely overlooked. Herein, we tried to decipher the underlying mechanism of the action of Mn-N-(2-hydroxyacetophenone) glycinate (MnNG) against differential p53 status bearing Hct116, MCF-7, and MDA-MB-468 cells on the backdrop of intracellular GSH level and reveal the role of p53 status in modulating GSH-dependant abrogation of MnNG-induced apoptosis in these cancer cells. Present study discloses that MnNG targets specifically wild-type-p53 expressing Hct116 and MCF-7 cells by significantly depleting both cytosolic, mitochondrial GSH, and modulating nuclear GSH through Glutathione reductase and Glutamate-cysteine ligase depletion that may in turn induce p53-mediated intrinsic apoptosis in them. Thus GSH addition abrogates p53-mediated apoptosis in wild-type-p53 expressing cells. GSH addition also overrides MnNG-induced modulation of phase II detoxifying parameters in them. However, GSH addition partially replenishes the down-regulated or modulated GSH pool in cytosol, mitochondria, and nucleus, and relatively abrogates MnNG-induced intrinsic apoptosis in p53-mutated MDA-MB-468 cells. On the contrary, although MnNG induces significant cell death in p53-null Hct116 cells, GSH addition fails to negate MnNG-induced cell death. Thus p53 status with intracellular GSH is critical for the modulation of MnNG-induced apoptosis.
Collapse
Affiliation(s)
- Kaushik Banerjee
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, India
| | - Satyajit Das
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, India
| | - Saikat Majumder
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Jaydip Biswas
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, India
| | - Soumitra Kumar Choudhuri
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, India.
| |
Collapse
|
24
|
Banerjee K, Das S, Sarkar A, Chatterjee M, Biswas J, Choudhuri SK. A copper chelate induces apoptosis and overcomes multidrug resistance in T-cell acute lymphoblastic leukemia through redox imbalance and inhibition of EGFR/PI3K/Akt expression. Biomed Pharmacother 2016; 84:71-92. [DOI: 10.1016/j.biopha.2016.08.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/10/2016] [Accepted: 08/24/2016] [Indexed: 01/25/2023] Open
|
25
|
Shakir M, Hanif S, Alam MF, Younus H. Molecular hybridization approach of bio-potent CuII/ZnII complexes derived from N, O donor bidentate imine scaffolds: Synthesis, spectral, human serum albumin binding, antioxidant and antibacterial studies. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 165:96-114. [DOI: 10.1016/j.jphotobiol.2016.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/06/2016] [Indexed: 11/24/2022]
|
26
|
Banerjee K, Basu S, Das S, Sinha A, Biswas MK, Choudhuri SK. Induction of intrinsic and extrinsic apoptosis through oxidative stress in drug-resistant cancer by a newly synthesized Schiff base copper chelate. Free Radic Res 2016; 50:426-46. [PMID: 26733073 DOI: 10.3109/10715762.2015.1136062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Multidrug resistance (MDR) in cancer represents a variety of strategies employed by tumor cells to evade the beneficial cytotoxic effects of structurally different anticancer drugs and thus confers impediments to the successful treatment of cancers. Efflux of drugs by MDR protein-1, functional P-glycoprotein and elevated level of reduced glutathione confer resistance to cell death or apoptosis and thus provide a possible therapeutic target for overcoming MDR in cancer. Previously, we reported that a Schiff base ligand, potassium-N-(2-hydroxy 3-methoxy-benzaldehyde)-alaninate (PHMBA) overcomes MDR in both in vivo and in vitro by targeting intrinsic apoptotic/necrotic pathway through induction of reactive oxygen species (ROS). The present study describes the synthesis and spectroscopic characterization of a copper chelate of Schiff base, viz., copper (II)-N-(2-hydroxy-3-methoxy-benzaldehyde)-alaninate (CuPHMBA) and the underlying mechanism of cell death induced by CuPHMBA in vitro. CuPHMBA kills both the drug-resistant and sensitive cell types irrespective of their drug resistance phenotype. The cell death induced by CuPHMBA follows apoptotic pathway and moreover, the cell death is associated with intrinsic mitochondrial and extrinsic receptor-mediated pathways. Oxidative stress plays a pivotal role in the process as proved by the fact that antioxidant enzyme; polyethylene glycol conjugated-catalase completely blocked CuPHMBA-induced ROS generation and abrogated cell death. To summarize, the present work provides a compelling rationale for the future clinical use of CuPHMBA, a redox active copper chelate in the treatment of cancer patients, irrespective of their drug-resistance status.
Collapse
Affiliation(s)
- Kaushik Banerjee
- a Department of In Vitro Carcinogenesis and Cellular Chemotherapy , Chittaranjan National Cancer Institute , Kolkata , West Bengal , India
| | - Soumya Basu
- a Department of In Vitro Carcinogenesis and Cellular Chemotherapy , Chittaranjan National Cancer Institute , Kolkata , West Bengal , India
| | - Satyajit Das
- a Department of In Vitro Carcinogenesis and Cellular Chemotherapy , Chittaranjan National Cancer Institute , Kolkata , West Bengal , India
| | - Abhinaba Sinha
- a Department of In Vitro Carcinogenesis and Cellular Chemotherapy , Chittaranjan National Cancer Institute , Kolkata , West Bengal , India
| | - Manas Kumar Biswas
- b Department of Chemistry , Ramakrishna Mission Residential College , Kolkata , West Bengal , India
| | - Soumitra Kumar Choudhuri
- a Department of In Vitro Carcinogenesis and Cellular Chemotherapy , Chittaranjan National Cancer Institute , Kolkata , West Bengal , India
| |
Collapse
|
27
|
Tektas O, Akkemik E, Baykara H. Investigation of the Effect of Some Optically Active Imine Compounds on the Enzyme Activities of hCA-I and hCA-II under In Vitro Conditions: An Experimental and Theoretical Study. J Biochem Mol Toxicol 2016; 30:277-86. [DOI: 10.1002/jbt.21788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/06/2015] [Accepted: 12/15/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Osman Tektas
- Department of Chemistry, Faculty of Arts and Sciences; Siirt University; 56100 Siirt Turkey
| | - Ebru Akkemik
- Faculty of Engineering and Architecture, Food Engineering; Siirt University; 56100 Siirt Turkey
| | - Haci Baykara
- Department of Chemistry, Faculty of Arts and Sciences; Siirt University; 56100 Siirt Turkey
- Center of Nanotechnology Research and Development (CIDNA), Facultad de Ingeniería Mecánica y Ciencias de la Producción; Escuela Superior Politécnica del Litoral, ESPOL; Campus Gustavo Galindo Km 30.5 Vía Perimetral Guayaquil Ecuador
| |
Collapse
|
28
|
Barta Holló B, Magyari J, Armaković S, Bogdanović GA, Rodić MV, Armaković SJ, Molnár J, Spengler G, Leovac VM, Mészáros Szécsényi K. Coordination compounds of a hydrazone derivative with Co(iii), Ni(ii), Cu(ii) and Zn(ii): synthesis, characterization, reactivity assessment and biological evaluation. NEW J CHEM 2016. [DOI: 10.1039/c6nj00560h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increasing inhibitory effect of Zn-Hp2DAP < Hp2DAP < Co-Hp2DAP on ABC transporter MDR1 drug efflux pump.
Collapse
|
29
|
Lian WJ, Wang XT, Xie CZ, Tian H, Song XQ, Pan HT, Qiao X, Xu JY. Mixed-ligand copper(ii) Schiff base complexes: the role of the co-ligand in DNA binding, DNA cleavage, protein binding and cytotoxicity. Dalton Trans 2016; 45:9073-87. [DOI: 10.1039/c6dt00461j] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Four novel mixed-ligand copper(ii) Schiff base complexes were synthesized and characterized. The biological features of the complexes and how acetic auxiliary ligands manipulate these features were investigated.
Collapse
Affiliation(s)
- Wen-Jing Lian
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| | - Xin-Tian Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| | - Cheng-Zhi Xie
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| | - He Tian
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| | - Xue-Qing Song
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| | - He-Ting Pan
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics
- School of Pharmacy
- Tianjin Medical University
- Tianjin 300070
- P.R. China
| |
Collapse
|
30
|
Szwed M, Wrona D, Kania KD, Koceva-Chyla A, Marczak A. Doxorubicin-transferrin conjugate triggers pro-oxidative disorders in solid tumor cells. Toxicol In Vitro 2015; 31:60-71. [PMID: 26607004 DOI: 10.1016/j.tiv.2015.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/03/2015] [Accepted: 11/16/2015] [Indexed: 12/26/2022]
Abstract
The formation of reactive oxygen species (ROS) is a widely accepted mechanism of doxorubicin (DOX) toxicity toward cancer cells. However, little is known about the potential of new systems, designed for more efficient and targeted doxorubicin delivery (i.e. protein conjugates, polymeric micelles, liposomes, monoclonal antibodies), to induce oxidative stress (OS) in tumors and hematological malignancies. Therefore, the objective of our study was to determine the relation between the toxicity of doxorubicin-transferring (DOX-TRF) conjugate and its capability to generate oxidative/nitrosative stress in solid tumor cells. Our research proves that DOX-TRF conjugate displays higher cytotoxicity towards lung adenocarcinoma epithelial (A549) and hepatocellular carcinoma (HepG2) cell lines than the reference free drug (DOX) and induces more extensive OS, characterized by a significant decrease in the total cellular antioxidant capacity, glutathione level and amount of -SH groups and an increase in hydroperoxide content. The intracellular redox imbalance was accompanied by changes in the transcription of genes encoding key antioxidant enzymes engaged in the sustaining of cellular redox homeostasis: superoxide dismutase (SOD), catalase (CAT), glutathione transferase (GST) and glutathione peroxidase (GP).
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Dominika Wrona
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Katarzyna D Kania
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, PAS, Lodowa St. 106, 93-232 Lodz, Poland.
| | - Aneta Koceva-Chyla
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| | - Agnieszka Marczak
- Department of Thermobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St. 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
31
|
Dutta Gupta S, Revathi B, Mazaira GI, Galigniana MD, Subrahmanyam CVS, Gowrishankar NL, Raghavendra NM. 2,4-dihydroxy benzaldehyde derived Schiff bases as small molecule Hsp90 inhibitors: rational identification of a new anticancer lead. Bioorg Chem 2015; 59:97-105. [PMID: 25727264 DOI: 10.1016/j.bioorg.2015.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/31/2015] [Accepted: 02/08/2015] [Indexed: 11/18/2022]
Abstract
Hsp90 is a molecular chaperone that heals diverse array of biomolecules ranging from multiple oncogenic proteins to the ones responsible for development of resistance to chemotherapeutic agents. Moreover they are over-expressed in cancer cells as a complex with co-chaperones and under-expressed in normal cells as a single free entity. Hence inhibitors of Hsp90 will be more effective and selective in destroying cancer cells with minimum chances of acquiring resistance to them. In continuation of our goal to rationally develop effective small molecule azomethines against Hsp90, we designed few more compounds belonging to the class of 2,4-dihydroxy benzaldehyde derived imines (1-13) with our validated docking protocol. The molecules exhibiting good docking score were synthesized and their structures were confirmed by IR, (1)H NMR and mass spectral analysis. Subsequently, they were evaluated for their potential to suppress Hsp90 ATPase activity by Malachite green assay. The antiproliferative effect of the molecules were examined on PC3 prostate cancer cell lines by adopting 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay methodology. Finally, schiff base 13 emerged as the lead molecule for future design and development of Hsp90 inhibitors as anticancer agents.
Collapse
Affiliation(s)
- Sayan Dutta Gupta
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India; R&D centre, Department of Pharmaceutical Sciences, Jawaharlal Nehru Technological University, Hyderabad, India.
| | - B Revathi
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - Gisela I Mazaira
- Department of Biological Chemistry, Faculty of Natural Sciences, University of Buenos Aires, Argentina
| | - Mario D Galigniana
- Department of Biological Chemistry, Faculty of Natural Sciences, University of Buenos Aires, Argentina; Institute of Experimental Biology and Medicine-CONICET, Argentina
| | - C V S Subrahmanyam
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - N L Gowrishankar
- Swami Vivekananda Institute of Pharmaceutical Sciences, Nalgonda, Andhra Pradesh, India
| | - N M Raghavendra
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| |
Collapse
|
32
|
Roseiro APS, Adão P, Galvão AM, Costa Pessoa J, Botelho do Rego AM, Carvalho MFNN. Oxygen activation by copper camphor complexes. Inorg Chem Front 2015. [DOI: 10.1039/c5qi00064e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In situreduction of CuCl2enables formation of Cu(i) complexes prone to activate molecular oxygen from air towards catalytic oxidation of 1,3-dicarbonyls.
Collapse
Affiliation(s)
- Alexandra P. S. Roseiro
- Centro de Química Estrutural
- Instituto Superior Técnico
- Universidade de Lisboa
- 1049-001 Lisboa
- Portugal
| | - Pedro Adão
- Centro de Química Estrutural
- Instituto Superior Técnico
- Universidade de Lisboa
- 1049-001 Lisboa
- Portugal
| | - Adelino M. Galvão
- Centro de Química Estrutural
- Instituto Superior Técnico
- Universidade de Lisboa
- 1049-001 Lisboa
- Portugal
| | - João Costa Pessoa
- Centro de Química Estrutural
- Instituto Superior Técnico
- Universidade de Lisboa
- 1049-001 Lisboa
- Portugal
| | - Ana M. Botelho do Rego
- Centro de Química-Física Molecular (CQFM) and Institute of Nanoscience and Nanotechnology (IN)
- Instituto Superior Técnico
- Universidade de Lisboa
- 1049-001 Lisboa
- Portugal
| | | |
Collapse
|
33
|
Synthesis, characterization and biological evaluation of a novel vanadium complex as a possible anticancer agent. J Organomet Chem 2014. [DOI: 10.1016/j.jorganchem.2014.08.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Halabi MF, Shakir RM, Bardi DA, Al-Wajeeh NS, Ablat A, Hassandarvish P, Hajrezaie M, Norazit A, Abdulla MA. Gastroprotective activity of ethyl-4-[(3,5-di-tert-butyl-2-hydroxybenzylidene) amino]benzoate against ethanol-induced gastric mucosal ulcer in rats. PLoS One 2014; 9:e95908. [PMID: 24800807 PMCID: PMC4011731 DOI: 10.1371/journal.pone.0095908] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 04/01/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The study was carried out to determine the cytotoxic, antioxidant and gastro-protective effect of ethyl-4-[(3,5-di-tert-butyl-2-hydroxybenzylid ene)amino] benzoate (ETHAB) in rats. METHODOLOGY/PRINCIPAL FINDINGS The cytotoxic effect of ETHAB was assessed using a MTT cleavage assay on a WRL68 cell line, while its antioxidant activity was evaluated in vitro. In the anti-ulcer study, rats were divided into six groups. Group 1 and group 2 received 10% Tween 20 (vehicle). Group 3 received 20 mg/kg Omeprazole. Groups 4, 5 and 6 received ETHAB at doses of 5, 10, and 20 mg/kg, respectively. After an hour, group 1 received the vehicle. Groups 2-6 received absolute ethanol to induce gastric mucosal lesions. In the WRL68 cell line, an IC50 of more than 100 µg/mL was observed. ETHAB results showed antioxidant activity in the DPPH, FRAP, nitric oxide and metal chelating assays. There was no acute toxicity even at the highest dosage (1000 mg/kg). Microscopy showed that rats pretreated with ETHAB revealed protection of gastric mucosa as ascertained by significant increases in superoxide dismutase (SOD), pH level, mucus secretion, reduced gastric lesions, malondialdehyde (MDA) level and remarkable flattened gastric mucosa. Histologically, pretreatment with ETHAB resulted in comparatively better gastric protection, due to reduction of submucosal edema with leucocyte infiltration. PAS staining showed increased intensity in uptake of Alcian blue. In terms of immunohistochemistry, ETHAB showed down-expression of Bax proteins and over-expression of Hsp70 proteins. CONCLUSION/SIGNIFICANCE The gastroprotective effect of ETHAB may be attributed to antioxidant activity, increased gastric wall mucus, pH level of gastric contents, SOD activity, decrease in MDA level, ulcer area, flattening of gastric mucosa, reduction of edema and leucocyte infiltration of the submucosal layer, increased PAS staining, up-regulation of Hsp70 protein and suppressed expression of Bax.
Collapse
Affiliation(s)
- Mohammed Farouq Halabi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Al-Moalim Mohamed Awad Center for Scientific Miracles of Prophetic Medicine, College of Medicine, Taibah University, Al-Madinah Al-Munawarah, Saudi Arabia
| | - Raied Mustafa Shakir
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- Department. of Chemistry, Ibn Al-Haitham, University of Baghdad. Baghdad, Iraq
| | - Daleya Abdulaziz Bardi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nahla Saeed Al-Wajeeh
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Abdulwali Ablat
- Institute of Biological Science, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Pouya Hassandarvish
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Maryam Hajrezaie
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Anwar Norazit
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
35
|
Chakraborty P, Das S, Banerjee K, Sinha A, Roy S, Chatterjee M, Choudhuri SK. A copper chelate selectively triggers apoptosis in myeloid-derived suppressor cells in a drug-resistant tumor model and enhances antitumor immune response. Immunopharmacol Immunotoxicol 2014; 36:165-75. [PMID: 24611750 DOI: 10.3109/08923973.2014.897727] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs), one of the major orchestrators of immunosuppressive network are present in the tumor microenvironment suppress antitumor immunity by subverting Th1 response in tumor site and considered as a great obstacle for advancement of different cancer immunotherapeutic protocols. Till date, various pharmacological approaches have been explored to modulate the suppressive functions of MDSCs in vivo. The present study describes our endeavor to explore a possibility of eradicating MDSCs by the application of a copper chelate, namely copper N-(2-hydroxy acetophenone) glycinate (CuNG), previously found to be a potential immunomodulator that can elicit antitumorogenic Th1 response in doxorubicin-resistant EAC (EAC/Dox) bearing mice. Herein, we demonstrated that CuNG treatment could reduce Gr-1+CD11b+ MDSC accumulation in ascitic fluid and spleen of EAC/Dox tumor model. Furthermore, we found that CuNG mediated reduction in MDSCs is associated with induction of Th1 response and reduction in Treg cells. Moreover, we observed that CuNG could deplete MDSCs by inducing Fas-FasL mediated apoptotic cell death where death receptor Fas expression is enhanced in MDSCs and FasL is provided by activated T cells. However, MDSC expansion from bone marrow cells and their differentiation was not affected by CuNG. Altogether, these findings suggest that the immunomodulatory property of CuNG is attributed to, at least in part, by its selective cytotoxic action on MDSCs. So, this preclinical study unveils a new mechanism of regulating MDSC levels in drug-resistant cancer model and holds promise of translating the findings into clinical settings.
Collapse
Affiliation(s)
- Paramita Chakraborty
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute , Kolkata, West Bengal , India and
| | | | | | | | | | | | | |
Collapse
|