1
|
Owczarzy A, Kulig K, Morak-Młodawska B, Jeleń M, Muhammetoglu T, Rogóż W, Maciążek-Jurczyk M. Human Serum Albumin and Human Serum Albumin Nanoparticles as Carriers of 10-(2'-Pyrimidyl)-3,6-diazaphenothiazine: In Vitro Spectroscopic Studies. Molecules 2025; 30:315. [PMID: 39860185 PMCID: PMC11767950 DOI: 10.3390/molecules30020315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Human serum albumin (HSA) plays a fundamental role in the human body, including the transport of exogenous and endogenous substances. HSA is also a biopolymer with a great medical and pharmaceutical potential. Due to nontoxicity and biocompatibility, this protein can be used as a nanocarrier. 10-(2'-Pyrimidyl)-3,6-diazaphenothiazine (10-Pyr-3,6-DAPT) is a phenothiazine showing high anticancer potential in vitro against glioma, melanoma and breast cancer cells. Additionally, this compound is characterized by selectivity of action towards MCF-7 breast cancer and has low cytotoxicity towards normal cells. Considering the promising pharmacological potential of this compound and using spectroscopic techniques, HSA and human serum albumin nanoparticles (HSA-NP) were tested as carriers of this molecule. Based on the obtained data and the appropriate mathematical models (Stern-Volmer and Klotz models), it can be concluded that 10-Pyr-3,6-DAPT probably forms a weak (Ka = (5.24 ± 0.57) × 104 and Ka = (4.67 ± 0.59) × 104) for excitation wavelengths λex 275 nm and λex 295 nm, respectively) static complex (kq > 1010) with HSA (at Sudlow site II (subdomain IIIA), and the phenomenon of it having both strong therapeutic and toxic effects is possible. High encapsulation efficiency of 10-Pyr-3,6-DAPT into the HSA-NPs was obtained, and the changes in albumin secondary structure due to the presence of 10-Pyr-3,6-DAPT were registered. Based on the data presented, it can be concluded that due to the high toxic effects of 10-Pyr-3,6-DAPT, a better carrier may be HSA-NPs.
Collapse
Affiliation(s)
- Aleksandra Owczarzy
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| | - Karolina Kulig
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-005 Katowice, Poland; (B.M.-M.); (M.J.)
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-005 Katowice, Poland; (B.M.-M.); (M.J.)
| | | | - Wojciech Rogóż
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| | - Małgorzata Maciążek-Jurczyk
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| |
Collapse
|
2
|
Kumar G, Pandey DM, Ghosh M, Dall'Acqua S, Gupta R, Tiwari NP, Siddique UM, Vishwakrama L, Guleri SK, Lal UR, Dubey S. Karanjin, A Promising Bioactive Compound Possessing Anti-cancer Activity against Experimental Model of Non-small Cell Lung Cancer Cells. Anticancer Agents Med Chem 2024; 24:317-333. [PMID: 37936467 DOI: 10.2174/0118715206255557231024095245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/07/2023] [Accepted: 09/25/2023] [Indexed: 11/09/2023]
Abstract
AIMS The aim of this study is to isolate the Millettia pinnata (Karanj) leaf extract for pure compound with anticancer properties and to study the molecular target of the isolates in non-small cell lung cancer cell lines. BACKGROUND In our earlier research Millettia pinnata leaf extract has demonstrated potential anticancer activities. Thus, in pursuit of the bioactive compounds, the most potential active extract from our previous study was purified. Furthermore, the anticancer properties of the isolated compound karanjin was studied and aimed for apoptosis and restraining growth. METHODS A novel method was developed through column chromatography for isolation and purification of the compound karanjin from leaf chloroform extract. The purified component was then characterised using FTIR, mass spectrometry, and NMR. An MTT-based cytotoxicity assay was used to analyse cell cytotoxicity, whereas fluorescence staining was used for apoptosis and reactive oxygen species inhibition quantification. Furthermore, the real-time PCR assay was used to determine the molecular mechanism of action in cells causing cytotoxicity induced by karanjin dosing. RESULTS The anticancer activity of karanjin in A549 cell line exhibited prominent activity revealing IC50 value of 4.85 μM. Conferring the predicted molecular pathway study, karanjin restrains the proliferation of cancer cells through apoptosis, which is controlled by extrinsic pathway proteins FAS/FADD/Caspases 8/3/9. Downregulation of KRAS and dependent gene expression also stopped cell proliferation. CONCLUSION Karanjin has been identified as a compound with potential effect in non-small cell lung cancer cells. Molecular mechanism for apoptosis and inhibition of reactive oxygen species induced through H2O2 were observed, concluding karanjin have medicinal and antioxidant properties.
Collapse
Affiliation(s)
- Gourav Kumar
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
- Birsa Munda Government Medical College, Shahdol, India
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Dev Mani Pandey
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Rashmi Gupta
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Nishi Prakash Tiwari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Usman Mohd Siddique
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | | | | | - Uma Ranjan Lal
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
- Department of Natural Products, NIPER Ahmedabad, India
| | - Supriya Dubey
- Department of Chemistry, Kanya Gurukul Campus, Gurukul Kangri (Deemed to be University), Haridwar, India
| |
Collapse
|
3
|
Martula E, Morak-Młodawska B, Jeleń M, Okechukwu PN, Balachandran A, Tehirunavukarasu P, Anamalay K, Ulaganathan V. Synthesis and Structural Characterization of Novel Dimers of Dipyridothiazine as Promising Antiproliferative Agents. Molecules 2023; 28:7662. [PMID: 38005384 PMCID: PMC10674446 DOI: 10.3390/molecules28227662] [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: 11/02/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Many new isomeric dipyridothiazine dimers have been presented as molecules with anticancer potential. These compounds were obtained in efficient syntheses of 1,6-, 1,8-, 2,7- and 3,6-diazaphenothiazines with selected alkylaromatic linkers. The structures of these compounds has been proven with two-dimensional spectroscopic techniques (COSY, NOESY, HSQC and HMBC) and high-resolution mass spectrometry (HRMS). In silico analyses of probable molecular targets were performed using the Way2Drug server. All new dimers were tested for anticancer activity against breast cancer line MCF7 and colon cancer line SW480. Cytotoxicity was assessed on normal L6 muscle cells. The tested dimers had high anticancer potential expressed as IC50 and the selectivity index SI. The most active derivative, 4c, showed an IC50 activity of less than 1 µM and an SI selectivity index higher than 100. Moreover, the compounds were characterized by low toxicity towards normal cells, simultaneously indicating a high cytostatic potential.
Collapse
Affiliation(s)
- Emilia Martula
- Doctoral School of The Medical University of Silesia, 40-055 Katowice, Poland;
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland;
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland;
| | - Patrick N. Okechukwu
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Abbirami Balachandran
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Prethika Tehirunavukarasu
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Kirthani Anamalay
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Vaidehi Ulaganathan
- Department of Food Science and Nutrition, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
4
|
Hahn V. Potential of the enzyme laccase for the synthesis and derivatization of antimicrobial compounds. World J Microbiol Biotechnol 2023; 39:107. [PMID: 36854853 PMCID: PMC9974771 DOI: 10.1007/s11274-023-03539-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/01/2023] [Indexed: 03/02/2023]
Abstract
Laccases [E.C. 1.10.3.2, benzenediol:dioxygen oxidoreductase] can oxidize phenolic substances, e.g. di- and polyphenols, hydroxylated biaryls, aminophenols or aryldiamines. This large substrate spectrum is the basis for various reaction possibilities, which include depolymerization and polymerization reactions, but also the coupling of different substance classes. To catalyze these reactions, laccases demand only atmospheric oxygen and no depletive cofactors. The utilization of mild and environmentally friendly reaction conditions such as room temperature, atmospheric pressure, and the avoidance of organic solvents makes the laccase-mediated reaction a valuable tool in green chemistry for the synthesis of biologically active compounds such as antimicrobial substances. In particular, the production of novel antibiotics becomes vital due to the evolution of antibiotic resistances amongst bacteria and fungi. Therefore, laccase-mediated homo- and heteromolecular coupling reactions result in derivatized or newly synthesized antibiotics. The coupling or derivatization of biologically active compounds or its basic structures may allow the development of novel pharmaceuticals, as well as the improvement of efficacy or tolerability of an already applied drug. Furthermore, by the laccase-mediated coupling of two different active substances a synergistic effect may be possible. However, the coupling of compounds that have no described efficacy can lead to biologically active substances by means of laccase. The review summarizes laccase-mediated reactions for the synthesis of antimicrobial compounds valuable for medical purposes. In particular, reactions with two different reaction partners were shown in detail. In addition, studies with in vitro and in vivo experimental data for the confirmation of the antibacterial and/or antifungal efficacy of the products, synthesized with laccase, were of special interest. Analyses of the structure-activity relationship confirm the great potential of the novel compounds. These substances may represent not only a value for pharmaceutical and chemical industry, but also for other industries due to a possible functionalization of surfaces such as wood or textiles.
Collapse
Affiliation(s)
- Veronika Hahn
- Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
- Institute for Microbiology, University of Greifswald, Felix-Hausdorff-Str. 8, 17489, Greifswald, Germany.
| |
Collapse
|
5
|
Bera M, Das M, Dolai M, Laha S, Islam MM, Samanta BC, Das A, Choudhuri I, Bhattacharyya N, Maity T. DNA/Protein Binding and Apoptotic-Induced Anticancer Property of a First Time Reported Quercetin-Iron(III) Complex Having a Secondary Anionic Residue: A Combined Experimental and Theoretical Approach. ACS OMEGA 2023; 8:636-647. [PMID: 36643564 PMCID: PMC9835804 DOI: 10.1021/acsomega.2c05790] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
A new quercetin-based iron(III) cationic complex [Fe(Qr)Cl(H2O)(MeO)] (complex 1) is created in the current study by condensation of quercetin with ferric chloride in the presence of Et3N. Comprehensive spectroscopic analysis and conductometric measurement are used to pinpoint complex 1. The generated complex's +3-oxidation state has been verified by electron paramagnetic resonance (EPR) research. Density functional theory analysis was used to structurally optimize the structure of complex 1. Before biomedical use, a variety of biophysical studies are implemented to evaluate the binding capacity of complex 1 with DNA and human serum albumin (HSA) protein. The findings of the electronic titration between complex 1 and DNA, as well as the stunning fall in the fluorescence intensities of the HSA and EtBr-DNA/DAPI-DNA domain after complex 1 is gradually added, give us confidence that complex 1 has a strong affinity for both macromolecules. It is interesting to note that the displacement experiment confirms partial intercalation as well as the groove binding mechanism of the title complex with DNA. The time-dependent fluorescence analysis indicates that after interaction with complex 1, HSA will exhibit static quenching. The thermodynamic parameter values in the HSA-complex 1 interaction provide evidence for the hydrophobicity-induced pathway leading to spontaneous protein-complex 1 interaction. The two macromolecules' configurations are verified to be preserved when they are associated with complex 1, and this is done via circular dichroism spectral titration. The molecular docking investigation, which is a theoretical experiment, provides complete support for the experimental findings. The potential of the investigated complex to be an anticancer drug has been examined by employing the MTT assay technique, which is carried out on HeLa cancer cell lines and HEK-293 normal cell lines. The MTT assay results validate the ability of complex 1 to display significant anticancer properties. Finally, by using the AO/PI staining approach, the apoptotic-induced cell-killing mechanism as well as the detection of cell morphological changes has been confirmed.
Collapse
Affiliation(s)
- Manjushree Bera
- Department
of Nutrition, Prabhat Kumar College, Contai, Purba Medinipur, Contai721404, India
| | - Manik Das
- Department
of Chemistry, Prabhat Kumar College, Contai, Purba Medinipur, Contai721404, India
| | - Malay Dolai
- Department
of Chemistry, Prabhat Kumar College, Contai, Purba Medinipur, Contai721404, India
| | - Soumik Laha
- IICB,
Kolkata, Kolkata, 700032West Bengal, India
| | - Md Maidul Islam
- Department
of Chemistry, Aliah University, Kolkata700064, India
| | - Bidhan Chandra Samanta
- Department
of Chemistry, Mugberia Gangadhar Mahavidyalaya, Purba Medinipur, Contai721425, India
| | - Arindam Das
- Department
of Chemistry, Jadavpur University, Kolkata700032, India
| | | | | | - Tithi Maity
- Department
of Chemistry, Prabhat Kumar College, Contai, Purba Medinipur, Contai721404, India
| |
Collapse
|
6
|
Lubeluzole Repositioning as Chemosensitizing Agent on Multidrug-Resistant Human Ovarian A2780/DX3 Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227870. [PMID: 36431971 PMCID: PMC9695310 DOI: 10.3390/molecules27227870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/19/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
In a previous paper, we demonstrated the synergistic action of the anti-ischemic lubeluzole (Lube S) on the cytotoxic activity of doxorubicin (Dox) and paclitaxel in human ovarian cancer A2780 and lung cancer A549 cells. In the present paper, we extended in vitro the study to the multi-drug-resistant A2780/DX3 cell line to verify the hypothesis that the Dox and Lube S drug association may potentiate the antitumor activity of this anticancer compound also in the context of drug resistance. We also evaluated some possible mechanisms underlying this activity. We analyzed the antiproliferative activity in different cancer cell lines. Furthermore, apoptosis, Dox accumulation, MDR1 downregulation, ROS, and NO production in A2780/DX3 cells were also evaluated. Our results confirm that Lube S improves Dox antiproliferative and apoptotic activities through different mechanisms of action, all of which may contribute to the final antitumor effect. Moderate stereoselectivity was found, with Lube S significantly more effective than its enantiomer (Lube R) and the corresponding racemate (Lube S/R). Docking simulation studies on the ABCB1 Cryo-EM structure supported the hypothesis that Lube S forms a stable MDR1-Dox-Lube S complex, which hampers the protein transmembrane domain flipping and blocks the efflux of Dox from resistant A2780/DX3 cells. In conclusion, our in vitro studies reinforce our previous hypothesis for repositioning the anti-ischemic Lube S as a potentiating agent in anticancer chemotherapy.
Collapse
|
7
|
Tri- and Pentacyclic Azaphenothiazine as Pro-Apoptotic Agents in Lung Carcinoma with a Protective Potential to Healthy Cell Lines. Molecules 2022; 27:molecules27165255. [PMID: 36014495 PMCID: PMC9413739 DOI: 10.3390/molecules27165255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022] Open
Abstract
The phenothiazine derivatives, tricyclic 10H-3,6-diazaphenothiazine (DPT-1) and pentacyclic 7-(3′-dimethylaminopropyl)diquinothiazine (DPT-2), have recently been shown to exhibit promising anticancer activities in vitro. In this report, we demonstrated that DPT-1 and DPT-2 could be pro-apoptotic agents in lung carcinoma, the human lung carcinoma A549 and non-small lung carcinoma H1299, in the range of IC50 = 1.52–12.89 µM, with a protective potential to healthy cell lines BEAS-2B and NHDF. The compounds showed higher activity in the range of the tested concentrations and low cytotoxicity in relation to normal healthy cells than doxorubicin, used as the reference drug. The cytostatic potential of DPT-1 and DPT-2 was demonstrated with the use of MTT assay. Cell cycle analysis via flow cytometry using Annexin-V assay showed the pro-apoptotic and pro-necrotic role of the studied diazaphenothiazines in the cell cycle. DPT-1 and DPT-2 initiated a biological response in the investigated cancer models with a different mechanism and at a different rate. Based on these findings, it can be concluded that DPT-1 and DPT-2 have potential as chemotherapeutic agents.
Collapse
|
8
|
Mor S, Sindhu S, Khatri M, Punia R, Sandhu H, Sindhu J, Jakhar K. Antimicrobial evaluation and QSAR studies of 3,6-disubstituted-11H-benzo[5,6][1,4]thiazino[3,4-a]isoindol-11-ones. EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY REPORTS 2022; 5:100050. [DOI: 10.1016/j.ejmcr.2022.100050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Das M, Mukherjee S, Islam MM, Choudhuri I, Bhattacharyya N, Samanta BC, Dutta B, Maity T. Response of Ancillary Azide Ligand in Designing a 1D Copper(II) Polymeric Complex along with the Introduction of High DNA- and HAS-Binding Efficacy, Leading to Impressive Anticancer Activity: A Compact Experimental and Theoretical Approach. ACS OMEGA 2022; 7:23276-23288. [PMID: 35847281 PMCID: PMC9281303 DOI: 10.1021/acsomega.2c01403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A new versatile azide-bridged polymeric Cu(II) complex, namely, [Cu(L)(μ1,3-N3)]∞ (1), was synthesized utilizing an N,N,O-donor piperidine-based Schiff base ligand (E)-4-bromo-2-((2-(-1-yl)imino)methyl)phenol (HL), obtained via the condensation reaction of 1-(2-aminoethyl) piperidine and 5-bromo salicylaldehyde. The single-crystal X-ray diffraction analysis reveals that complex 1 consists of an end-to-end azido-bridged polymeric network, which is further rationalized with the help of a density functional theory (DFT) study. After routine characterization with a range of physicochemical studies, complex 1 is exploited to evaluate its biomedical potential. Initially, theoretical inspection with the help of a molecular docking study indicated the ability of complex 1 to effectively bind with macromolecules such as DNA and the human serum albumin (HSA) protein. The theoretical aspect was further verified by adopting several spectroscopic techniques. The electronic absorption spectroscopic analysis indicates a remarkable binding efficiency of Complex 1 with both DNA and HSA. The notable fluorescence intensity reduction of the ethidium bromide (EtBr)-DNA adduct, 4',6-diamidino-2-phenylindole (DAPI)-DNA adduct, and HSA after the gradual addition of complex 1 authenticates its promising binding potential with the macromolecules. The retention of the canonical B form of DNA and α form of HSA during the association of complex 1 was confirmed by implementing a circular dichroism spectral study. The association ability of complex 1 with macromolecules further inspired us to inspect its impact on different cell lines such as HeLa (cervical cancer cell), PA1 (ovarian cancer cell), and HEK (normal cell). The dose-dependent and time-dependent in vitro 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay suggests an effective antiproliferative property of complex 1 with low toxicity toward the normal cell line. Finally, the anticancer activity of complex 1 toward carcinoma cell lines was analyzed by nuclear and cellular staining techniques, unveiling the cell death mechanism.
Collapse
Affiliation(s)
- Manik Das
- Department
of Chemistry, Prabhat Kumar College, Contai, Purba Medinipur, Contai 721404, India
| | - Somali Mukherjee
- School
of Chemical Sciences, Indian Association
for the Cultivation of Science, Jadavpur, Kolkata 700032, West Bengal, India
| | - Md. Maidul Islam
- Department
of Chemistry, Aliah University, Kolkata 700064, India
| | - Indranil Choudhuri
- Department
of Chemistry, Panskura Banamali College, Panskura 721152, India
| | | | - Bidhan Chandra Samanta
- Department
of Chemistry, Mugberia Gangadhar Mahavidyalaya, Purba Medinipur 721425, India
| | - Basudeb Dutta
- Department
of Chemical Science, IISER Kolkata, Mohanpur, Kolkata 741246, India
| | - Tithi Maity
- Department
of Chemistry, Prabhat Kumar College, Contai, Purba Medinipur, Contai 721404, India
| |
Collapse
|
10
|
Laxmikeshav K, Kumari P, Shankaraiah N. Expedition of sulfur-containing heterocyclic derivatives as cytotoxic agents in medicinal chemistry: A decade update. Med Res Rev 2021; 42:513-575. [PMID: 34453452 DOI: 10.1002/med.21852] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 04/20/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
This review article proposes a comprehensive report of the design strategies engaged in the development of various sulfur-bearing cytotoxic agents. The outcomes of various studies depict that the sulfur heterocyclic framework is a fundamental structure in diverse synthetic analogs representing a myriad scope of therapeutic activities. A number of five-, six- and seven-membered sulfur-containing heterocyclic scaffolds, such as thiazoles, thiadiazoles, thiazolidinediones, thiophenes, thiopyrans, benzothiazoles, benzothiophenes, thienopyrimidines, simple and modified phenothiazines, and thiazepines have been discussed. The subsequent studies of the derivatives unveiled their cytotoxic effects through multiple mechanisms (viz. inhibition of tyrosine kinases, topoisomerase I and II, tubulin, COX, DNA synthesis, and PI3K/Akt and Raf/MEK/ERK signaling pathways), and several others. Thus, our concise illustration explains the design strategy and anticancer potential of these five- and six-membered sulfur-containing heterocyclic molecules along with a brief outline on seven-membered sulfur heterocycles. The thorough assessment of antiproliferative activities with the reference drug allows a proficient assessment of the structure-activity relationships (SARs) of the diversely synthesized molecules of the series.
Collapse
Affiliation(s)
- Kritika Laxmikeshav
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pooja Kumari
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
11
|
The Influence of Oxidative Stress on Serum Albumin Structure as a Carrier of Selected Diazaphenothiazine with Potential Anticancer Activity. Pharmaceuticals (Basel) 2021; 14:ph14030285. [PMID: 33806875 PMCID: PMC8005128 DOI: 10.3390/ph14030285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/05/2021] [Accepted: 03/18/2021] [Indexed: 12/20/2022] Open
Abstract
Albumin is one of the most important proteins in human blood. Among its multiple functions, drug binding is crucial in terms of drug distribution in human body. This protein undergoes many modifications that are certain to influence protein activity and affect its structure. One such reaction is albumin oxidation. Chloramine T is a strong oxidant. Solutions of human serum albumin, both non-modified and modified by chloramine T, were examined with the use of fluorescence, absorption and circular dichroism (CD) spectroscopy. 10H-3,6-diazaphenothiazine (DAPT) has anticancer activity and it has been studied for the first time in terms of binding with human serum albumin—its potential as a transporting protein. Using fluorescence spectroscopy, in the presence of dansylated amino acids, dansyl-l-glutamine (dGlu), dansyl-l-proline (dPro), DAPT binding with two main albumin sites—in subdomain IIA and IIIA—has been evaluated. Based on the conducted data, in order to measure the stability of DAPT complexes with human (HSA) and oxidized (oHSA) serum albumin, association constant (Ka) for ligand-HSA and ligand-oHSA complexes were calculated. It has been presumed that oxidation is not an important issue in terms of 10H-3,6-diazaphenothiazine binding to albumin. It means that the distribution of this substance is similar regardless of changes in albumin structure caused by oxidation, natural occurring in the organism.
Collapse
|
12
|
Morak-Młodawska B, Jeleń M, Pluta K. Phenothiazines Modified with the Pyridine Ring as Promising Anticancer Agents. Life (Basel) 2021; 11:206. [PMID: 33807874 PMCID: PMC8001302 DOI: 10.3390/life11030206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/29/2022] Open
Abstract
Azaphenothiazines are the largest and most perspective group of modified phenothiazines, and they exhibit variety of biological activities. The review sums up the current knowledge on the anticancer activity of isomeric pyridobenzothiazines and dipyridothiazines, which are modified azaphenothiazines with one and two pyridine rings, respectively, against 10 types of cancer cell lines. Some 10-substituted dipyridothiazines and even 10-unsubstituted parent compounds, such as 10H-1,9-diazaphenothiazine and 10H-3,6-diazaphenothiazine, exhibited very potent action with the IC50 values less than 1 µg/mL and 1 µM against selected cancer cell lines. The strength of the anticancer action depends both on the tricyclic ring scaffolds and the substituents at the thiazine nitrogen atom. The review discusses the kind of the substituents, nature of tricyclic ring scaffolds with the location of the azine nitrogen atoms, the types of the cancer cell lines, and the mechanism of action.
Collapse
Affiliation(s)
- Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.J.); (K.P.)
| | | | | |
Collapse
|
13
|
Ren G, Hao X, Yang S, Chen J, Qiu G, Ang KP, Mohd Tamrin MI. 10H-3,6-Diazaphenothiazines triggered the mitochondrial-dependent and cell death receptor-dependent apoptosis pathways and further increased the chemosensitivity of MCF-7 breast cancer cells via inhibition of AKT1 pathways. J Biochem Mol Toxicol 2020; 34:e22544. [PMID: 32619082 DOI: 10.1002/jbt.22544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 11/11/2022]
Abstract
Breast cancer is one of the leading causes of death in cancer categories, followed by lung, colorectal, and ovarian among the female gender across the world. 10H-3,6-diazaphenothiazine (PTZ) is a thiazine derivative compound that exhibits many pharmacological activities. Herein, we proceed to investigate the pharmacological activities of PTZ toward breast cancer MCF-7 cells as a representative in vitro breast cancer cell model. The PTZ exhibited a proliferation inhibition (IC50 = 0.895 µM) toward MCF-7 cells. Further, cell cycle analysis illustrated that the S-phase checkpoint was activated to achieve proliferation inhibition. In vitro cytotoxicity test on three normal cell lines (HEK293 normal kidney cells, MCF-10A normal breast cells, and H9C2 normal heart cells) demonstrated that PTZ was more potent toward cancer cells. Increase in the levels of reactive oxygen species results in polarization of mitochondrial membrane potential (ΔΨm), together with suppression of mitochondrial thioredoxin reductase enzymatic activity suggested that PTZ induced oxidative damages toward mitochondria and contributed to improved drug efficacy toward treatment. The RT2 PCR Profiler Array (human apoptosis pathways) proved that PTZ induced cell death via mitochondria-dependent and cell death receptor-dependent pathways, through a series of modulation of caspases, and the respective morphology of apoptosis was observed. Mechanistic studies of apoptosis suggested that PTZ inhibited AKT1 pathways resulting in enhanced drug efficacy despite it preventing invasion of cancer cells. These results showed the effectiveness of PTZ in initiation of apoptosis, programmed cell death, toward highly chemoresistant MCF-7 cells, thus suggesting its potential as a chemotherapeutic drug.
Collapse
Affiliation(s)
- Guanghui Ren
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Xiaoyan Hao
- Department of Thyroid and Breast Surgery, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Shuyi Yang
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Jun Chen
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Guobin Qiu
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Kok Pian Ang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mohd Islahuddin Mohd Tamrin
- Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
14
|
Evaluation of the Lipophilicity of New Anticancer 1,2,3-Triazole-Dipyridothiazine Hybrids Using RP TLC and Different Computational Methods. Processes (Basel) 2020. [DOI: 10.3390/pr8070858] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Two new anticancer-active 1,2,3-triazole-dipyridothiazine hybrids were evaluated for their lipophilicity using thin-layer chromatography (TLC) and computational methods. The experimental lipophilicity was evaluated with mobile phases (mixtures of TRIS buffer and acetone), exploiting a linear correlation between the retention parameter (RM) and the volume of acetone. The relative lipophilicity parameter (RM0) was obtained by extrapolation to 0% acetone concentration. This parameter was intercorrelated with a specific hydrophobic surface area (b) revealing two congeneric subgroups: hybrids of 1,2,3-triazole-2,7-diazaphenothiazines and 1,2,3-triazole-3,6-diazaphenothiazines. The parameter RM0 was converted into the absolute lipophilicity parameter logPTLC using a calibration curve prepared on the basis of compounds of known logP values. Triazole–dipyridothiazine hybrids turned out to be medium lipophilic with logPTLC values of 1.232–2.979. The chromatographically established parameter logPTLC was compared to the calculated lipophilic parameter logPcalcd obtained with various algorithms. The lipophilicity was correlated with molecular descriptors and ADME properties. The new triazole–dipyridothiazine hybrids followed Lipinski’s rule of five. The lipophilicity of these hybrids was dependent on the substituents attached to the triazole ring and the location of the azine nitrogen atoms.
Collapse
|
15
|
Hahn V, Mikolasch A, Weitemeyer J, Petters S, Davids T, Lalk M, Lackmann JW, Schauer F. Ring-Closure Mechanisms Mediated by Laccase to Synthesize Phenothiazines, Phenoxazines, and Phenazines. ACS OMEGA 2020; 5:14324-14339. [PMID: 32596570 PMCID: PMC7315418 DOI: 10.1021/acsomega.0c00719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 05/31/2023]
Abstract
The green and environmentally friendly synthesis of highly valuable organic substances is one possibility for the utilization of laccases (EC 1.10.3.2). As reactants for the herein described syntheses, different o-substituted arylamines or arylthiols and 2,5-dihydroxybenzoic acid and its derivatives were used. In this way, the formation of phenothiazines, phenoxazines, and phenazines was achieved in aqueous solution mediated by the laccase of Pycnoporus cinnabarinus in the presence of oxygen. Two types of phenothiazines (3-hydroxy- and 3-oxo-phenothiazines) formed in one reaction assay were described for the first time. The cyclization reactions yielded C-N, C-S, or C-O bonds. The syntheses were investigated with regard to the substitution pattern of the reaction partners. Differences in C-S and C-N bond formations without cyclization are discussed.
Collapse
Affiliation(s)
- Veronika Hahn
- Institut
für Mikrobiologie, Universität
Greifswald, Friedrich-Ludwig-Jahn Str. 15, 17487 Greifswald, Germany
- Leibniz-Institut
für Plasmaforschung und Technologie e.V. (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Annett Mikolasch
- Institut
für Mikrobiologie, Universität
Greifswald, Friedrich-Ludwig-Jahn Str. 15, 17487 Greifswald, Germany
| | - Josephine Weitemeyer
- Institut
für Mikrobiologie, Universität
Greifswald, Friedrich-Ludwig-Jahn Str. 15, 17487 Greifswald, Germany
| | - Sebastian Petters
- Institut
für Mikrobiologie, Universität
Greifswald, Friedrich-Ludwig-Jahn Str. 15, 17487 Greifswald, Germany
| | - Timo Davids
- Institut
für Mikrobiologie, Universität
Greifswald, Friedrich-Ludwig-Jahn Str. 15, 17487 Greifswald, Germany
| | - Michael Lalk
- Institut
für Biochemie, Universität
Greifswald, Felix-Hausdorff-Str. 4, 17487 Greifswald, Germany
| | - Jan-Wilm Lackmann
- Leibniz-Institut
für Plasmaforschung und Technologie e.V. (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Frieder Schauer
- Institut
für Mikrobiologie, Universität
Greifswald, Friedrich-Ludwig-Jahn Str. 15, 17487 Greifswald, Germany
| |
Collapse
|
16
|
Lin K, Rong Y, Chen D, Zhao Z, Bo H, Qiao A, Hao X, Wang J. Combination of Ruthenium Complex and Doxorubicin Synergistically Inhibits Cancer Cell Growth by Down-Regulating PI3K/AKT Signaling Pathway. Front Oncol 2020; 10:141. [PMID: 32133289 PMCID: PMC7041628 DOI: 10.3389/fonc.2020.00141] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 01/21/2023] Open
Abstract
Combinational use of drugs has been a common strategy in cancer treatment because of synergistic advantages in reducing dose and toxicity, minimizing or delaying drug resistance. To improve the efficacy of chemotherapy, various potential combinations have been investigated. Ruthenium complex is considered a potential alternative of the platinum-based drugs due to its significant efficacy and safety. Previously, we reported that ruthenium(II) complex (Δ-Ru1) has great anticancer potential and minor toxicity toward normal tissues. However, the therapeutic efficacy and mechanism of action of ruthenium(II) complex combined with other anticancer drugs is still unknown. Here, we investigated the combinational effect of Δ-Ru1 and doxorubicin in different cancer cells. The data assessed by Chou-Talalay method showed significant synergism in MCF-7 cells. Furthermore, the results in antiproliferation efficacy indicated that the combination showed strong cytotoxicity and increasing apoptosis of MCF-7 cells in 2D and 3D multicellular tumor spheroids (MCTSs). Significant inhibition of MCF-7 cells accompanied with increased ROS generation was observed. Furthermore, the expression of PI3K/AKT was significantly down-regulated, while the expression of PTEN was strongly up-regulated in cells treated with combination of Δ-Ru1 and doxorubicin. The expression of NF-κB and XIAP decreased while the expression of P53 increased and associated with apoptosis. These findings suggest that the combination of ruthenium complex and doxorubicin has a significant synergistic effect by down-regulating the PI3K/AKT signaling pathway in MCF-7 cells. This study may trigger more research in ruthenium complex and combination therapy that will be able to provide opportunities for developing better therapeutics for cancer treatment.
Collapse
Affiliation(s)
- Ke Lin
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Rong
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dan Chen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zizhuo Zhao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaben Bo
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Aimin Qiao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaojuan Hao
- Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
| | - Jinquan Wang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
17
|
Morak-Młodawska B, Pluta K, Latocha M, Jeleń M, Kuśmierz D, Suwińska K, Shkurenko A, Czuba Z, Jurzak M. 10 H-1,9-diazaphenothiazine and its 10-derivatives: synthesis, characterisation and biological evaluation as potential anticancer agents. J Enzyme Inhib Med Chem 2019; 34:1298-1306. [PMID: 31307242 PMCID: PMC6691808 DOI: 10.1080/14756366.2019.1639695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 02/07/2023] Open
Abstract
10H-1,9-diazaphenothiazine was obtained in the sulphurisation reaction of diphenylamine with elemental sulphur and transformed into new 10-substituted derivatives, containing alkyl and dialkylaminoalkyl groups at the thiazine nitrogen atom. The 1,9-diazaphenothiazine ring system was identified with advanced 1H and 13C NMR techniques (COSY, NOESY, HSQC and HMBC) and confirmed by X-ray diffraction analysis of the methyl derivative. The compounds exhibited significant anticancer activities against the human glioblastoma SNB-19, melanoma C-32 and breast cancer MDA-MB-231 cell lines. The most active 1,9-diazaphenothiazines were the derivatives with the propynyl and N, N-diethylaminoethyl groups being more potent than cisplatin. For those two compounds, the expression of H3, TP53, CDKN1A, BCL-2 and BAX genes was detected by the RT-QPCR method. The proteome profiling study showed the most probable compound action on SNB-19 cells through the intrinsic mitochondrial pathway of apoptosis. The 1,9-diazaphenotiazine system seems to be more potent than known isomeric ones (1,6-diaza-, 1,8-diaza-, 2,7-diaza- and 3,6-diazaphenothiazine).
Collapse
Affiliation(s)
- Beata Morak-Młodawska
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Krystian Pluta
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Małgorzata Latocha
- Department of Cell Biology, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Małgorzata Jeleń
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Dariusz Kuśmierz
- Department of Cell Biology, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| | - Kinga Suwińska
- Faculty of Mathematics and Natural Sciences, Cardinal Stefan Wyszyński University, Warszawa, Poland
- A. M. Butlerov Institute of Chemistry, Kazan Federal University, Kazan, Russia
| | - Aleksander Shkurenko
- Division of Physical Functional Materials Design, Discovery & Development Research Group (FMD3), Sciences and Engineering Advanced Membranes & Porous Materials (AMPM), King Abdullah University of Science and Technology (KAU ST), Thuwal, Kingdom of Saudi Arabia
| | - Zenon Czuba
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Magdalena Jurzak
- Department of Cell Biology, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Sosnowiec, Poland
| |
Collapse
|
18
|
Morak-Młodawska B, Pluta K, Latocha M, Jeleń M, Kuśmierz D. Design, Synthesis, and Structural Characterization of Novel Diazaphenothiazines with 1,2,3-Triazole Substituents as Promising Antiproliferative Agents. Molecules 2019; 24:molecules24234388. [PMID: 31801304 PMCID: PMC6930555 DOI: 10.3390/molecules24234388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/19/2019] [Accepted: 11/27/2019] [Indexed: 01/07/2023] Open
Abstract
A series of novel 1,2,3-triazole-diazphenothiazine hybrids was designed, synthesized, and evaluated for anticancer activity against four selected human tumor cell lines (SNB-19, Caco-2, A549, and MDA-MB231). The majority of the synthesized compounds exhibited significant potent activity against the investigated cell lines. Among them, compounds 1d and 4c showed excellent broad spectrum anticancer activity, with IC50 values ranging from 0.25 to 4.66 μM and 0.25 to 6.25 μM, respectively. The most promising compound 1d, possessing low cytotoxicity against normal human fibroblasts NHFF, was used for gene expression analysis using reverse transcription–quantitative real-time PCR (RT–qPCR). The expression of H3, TP53, CDKN1A, BCL-2, and BAX genes revealed that these compounds inhibited the proliferation in all cells (H3) and activated mitochondrial events of apoptosis (BAX/BCL-2).
Collapse
Affiliation(s)
- Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (K.P.); (M.J.)
- Correspondence: ; Tel.: +48-32-364-16-04
| | - Krystian Pluta
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (K.P.); (M.J.)
| | - Małgorzata Latocha
- Department of Cell Biology, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (M.L.); (D.K.)
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (K.P.); (M.J.)
| | - Dariusz Kuśmierz
- Department of Cell Biology, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (M.L.); (D.K.)
| |
Collapse
|
19
|
Mirza-Aghazadeh-Attari M, Ostadian C, Saei AA, Mihanfar A, Darband SG, Sadighparvar S, Kaviani M, Samadi Kafil H, Yousefi B, Majidinia M. DNA damage response and repair in ovarian cancer: Potential targets for therapeutic strategies. DNA Repair (Amst) 2019; 80:59-84. [PMID: 31279973 DOI: 10.1016/j.dnarep.2019.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 06/01/2019] [Accepted: 06/15/2019] [Indexed: 12/24/2022]
Abstract
Ovarian cancer is among the most lethal gynecologic malignancies with a poor survival prognosis. The current therapeutic strategies involve surgery and chemotherapy. Research is now focused on novel agents especially those targeting DNA damage response (DDR) pathways. Understanding the DDR process in ovarian cancer necessitates having a detailed knowledge on a series of signaling mediators at the cellular and molecular levels. The complexity of the DDR process in ovarian cancer and how this process works in metastatic conditions is comprehensively reviewed. For evaluating the efficacy of therapeutic agents targeting DNA damage in ovarian cancer, we will discuss the components of this system including DDR sensors, DDR transducers, DDR mediators, and DDR effectors. The constituent pathways include DNA repair machinery, cell cycle checkpoints, and apoptotic pathways. We also will assess the potential of active mediators involved in the DDR process such as therapeutic and prognostic candidates that may facilitate future studies.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Caspian Ostadian
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Amir Ata Saei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Ainaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saber Ghazizadeh Darband
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden; Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Shirin Sadighparvar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Kaviani
- School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada
| | | | - Bahman Yousefi
- Molecular MedicineResearch Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
20
|
Mor S, Sindhu S. Convenient and efficient synthesis of novel 11 H-benzo[5,6][1,4]thiazino[3,4- a]isoindol-11-ones derived from 2-bromo-(2/3-substitutedphenyl)-1 H-indene-1,3(2 H)-diones. RSC Adv 2019; 9:12784-12792. [PMID: 35557558 PMCID: PMC9092632 DOI: 10.1039/c9ra02403d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 04/16/2019] [Indexed: 11/21/2022] Open
Abstract
An unprecedented formation of 11H-benzo[5,6][1,4]thiazino[3,4-a]isoindol-11-ones through a one-step reaction of differently substituted 2-aminobenzenethiols and 2-bromo-(2/3-substitutedphenyl)-1H-indene-1,3(2H)-diones in freshly dried ethanol under reflux conditions has been investigated. This unique transformation probably occurs through an initial nucleophilic substitution followed by ring opening and subsequent intramolecular cyclization. The structures of all the synthesized benzo[1,4]thiazino isoindolinones were established by FTIR, 1H NMR, 13C NMR, HRMS, and X-ray crystallographic analysis. This approach was found to be simple and convenient and provides several advantages such as substantial atom economy, short reaction time and operational simplicity.
Collapse
Affiliation(s)
- Satbir Mor
- Department of Chemistry, Guru Jambheshwar University of Science & Technology Hisar-125001 Haryana India +91-1662-276240 +91-1662-263397
| | - Suchita Sindhu
- Department of Chemistry, Guru Jambheshwar University of Science & Technology Hisar-125001 Haryana India +91-1662-276240 +91-1662-263397
| |
Collapse
|
21
|
Morak-Młodawska B, Pluta K, Latocha M, Jeleń M, Kuśmierz D. Synthesis, Anticancer Activity, and Apoptosis Induction of Novel 3,6-Diazaphenothiazines. Molecules 2019; 24:molecules24020267. [PMID: 30642021 PMCID: PMC6359424 DOI: 10.3390/molecules24020267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 02/03/2023] Open
Abstract
New 10-substituted derivatives of 3,6-diazaphenothiazine, containing the triple bond linker terminated with tertiary cyclic and acyclic amine groups, were synthesized and screened for their anticancer action. The compounds exhibited varied anticancer activities against human glioblastoma SNB-19, melanoma C-32, and breast cancer MDA-MB231 cell lines, depending on the nature of the substituents. The most active 3,6-diazaphenothiazine, 4, was the derivative with the N,N-diethylamino-2-butynyl substituent against glioblastoma SNB-19, and was ten times more potent than cisplatin. For this compound, the expression of H3, TP53, CDKN1A, BCL-2, and BAX genes was detected by the RT-qPCR method. The gene expression ratio BAX/BCL-2 indicated the induction of mitochondrial apoptosis in cancer cell lines. The transformation of the propynyl substituent into amino-2-butynyl can be a method applicable to the search for more anticancer-active azaphenothiazines.
Collapse
Affiliation(s)
- Beata Morak-Młodawska
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland.
| | - Krystian Pluta
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland.
| | - Małgorzata Latocha
- Department of Cell Biology, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland.
| | - Małgorzata Jeleń
- Department of Organic Chemistry, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland.
| | - Dariusz Kuśmierz
- Department of Cell Biology, School of Pharmacy with the Division of Laboratory Medicine, The Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland.
| |
Collapse
|