1
|
Hosseini-Hashemi Z, Eslami Moghadam M, Mirzaei M, Notash B. Biological Activity of Two Anticancer Pt Complexes with a Cyclohexylglycine Ligand against a Colon Cancer Cell Line: Theoretical and Experimental Study. ACS OMEGA 2022; 7:39794-39811. [PMID: 36385884 PMCID: PMC9648137 DOI: 10.1021/acsomega.2c03776] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/26/2022] [Indexed: 05/30/2023]
Abstract
Because of their extraordinary ability to disrupt the natural structure of nucleic acids, metal complexes could be used in cancer therapy. In this study, cyclohexylglycine (HL) as a ligand and two new Pt complexes, [Pt(NH3)2(L)]NO3 (1) and [Pt(bipy)(L)]NO3 (2), were synthesized and characterized by elemental analysis, LC-MS, UV-vis spectrometry, FT-IR, 1H NMR spectroscopy, 13C NMR spectroscopy, 195Pt NMR spectroscopy, HPLC analysis, and single-crystal X-ray diffraction. Complex 2 crystallized in the orthorhombic Pbca space group, and density functional theory (DFT) was used to describe its structural parameters were described in detail. These complexes can be classified as oral medications and drug-like molecules based on a comparison of their absorption, distribution, metabolism, and excretion assessment. Quantum chemical descriptors (QCDs) were determined using DFT calculations to predict the tendency of DNA to approach these complexes. During the determination of the function of the metallodrug in DNA binding, the fluorescence data indicated that static quenching took place for all ligands and complexes with higher DNA binding affinity. CD and isothermal absorption studies indicate the presence of electrostatic and groove binding for the amine derivative and that DNA binds with the bipy moiety via groove binding. Furthermore, the interaction modes were determined using molecular docking to investigate the binding of these compounds with the target DNA molecule. According to docking investigations, binding energies of -5.7, -11.56, and -10.00 kcal/mol for HL and complexes 1 and 2, respectively, indicate partially electrostatic and groove binding. The anticancer activities of the Pt(II) complexes were tested against the HCT116 human colon cancer cell line, with IC50 values of 35.51 and 51.33 μM for 1 and 2, respectively, after 72 h. These values show that the inhibitory effect of complex 1 was better than those of 2 and carboplatin (IC50 = 51.94 μM).
Collapse
Affiliation(s)
- Zahra Hosseini-Hashemi
- Department
of Chemistry, Faculty of Science, Ferdowsi
University of Mashhad, Mashhad, Razavi Khorasan9177948974, Iran
| | | | - Masoud Mirzaei
- Department
of Chemistry, Faculty of Science, Ferdowsi
University of Mashhad, Mashhad, Razavi Khorasan9177948974, Iran
- Khorasan
Science and Technology Park (KSTP), 12th km of Mashhad-Quchan Road, Mashhad, Razavi Khorasan9185173911, Iran
| | - Behrouz Notash
- Department
of Inorganic Chemistry and Catalysis, Shahid
Beheshti University, Tehran, Tehran1983969411, Iran
| |
Collapse
|
2
|
Ghorbani F, Ghalandari B, Liu Z, Li D, Yu B. Injectable light-assisted thermo-responsive methylcellulose-sodium humate hydrogel proposed for photothermal ablation and localized delivery of cisplatin. Front Bioeng Biotechnol 2022; 10:967438. [PMID: 36003535 PMCID: PMC9395131 DOI: 10.3389/fbioe.2022.967438] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/06/2022] [Indexed: 11/20/2022] Open
Abstract
This study aimed to develop injectable light-assisted thermo-responsive methylcellulose hydrogels filled with sodium humate, which were proposed for photothermal ablation and localized cisplatin delivery. Sodium humate converts light energy from laser beams into thermal energy, which causes methylcellulose to gel, thereby controlling the release of chemotherapy agents. Meanwhile, light emission causes to the photothermal ablation of tumor cells. For determining the optimal production conditions, different concentrations of sodium humate and light emission times were investigated. Results show that hydrogel uniformity is highly dependent on variables. An increase in sodium humate concentration and emission time resulted in a slight reduction in swelling ratio and an increase in durability. According to the simulation conditions, the cisplatin release profile was consistent with a non-Fickian mechanism with a predominant erosion contribution. In conjugation with increasing light emission time and sodium humate content, the storage modulus and viscosity increased, demonstrating hydrogel’s sol-gel transition and long-lasting durability. The intrinsic fluorescence spectroscopy study revealed that the hydrogel-model protein complex empowered hydrogel bio-performance. Laser emission and cisplatin release synergistically reduced the number of viable osteosarcoma cell lines, suggesting the possibility of tumor ablation. This study describes the potential of simultaneous photothermal therapy and chemotherapy in osteosarcoma treatment, laying the groundwork for future preclinical and clinical trials.
Collapse
Affiliation(s)
- Farnaz Ghorbani
- Department of Orthopedics, Shanghai Pudong New Area People’s Hospital, Shanghai, China
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zichen Liu
- School of Materials Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Dejian Li
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Baoqing Yu
- Department of Orthopedics, Shanghai Pudong New Area People’s Hospital, Shanghai, China
- *Correspondence: Baoqing Yu,
| |
Collapse
|
3
|
Evaluation of Gastroprotective Activity of Licorice and Turmeric Rhizome Aqueous Extract against Ethanol-Induced Gastric Injury in Male Wistar Rats. MEDICAL LABORATORY JOURNAL 2022. [DOI: 10.52547/mlj.16.4.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
4
|
Silibinin exerts anti-cancer activity on human ovarian cancer cells by increasing apoptosis and inhibiting epithelial-mesenchymal transition (EMT). Gene 2022; 823:146275. [PMID: 35189245 DOI: 10.1016/j.gene.2022.146275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 01/13/2022] [Accepted: 02/03/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND Silibinin, the principal flavonoid derived from milk thistle seeds, has been demonstrated to have strong inhibitory effects against human malignancies. The inhibitory function of silibinin on ovarian cancer, however, is not fully identified. In this essay, both in vivo and in vitro investigations were conducted to survey the silibinin's blocking effects on ovarian cancer. METHODS The impacts of silibinin on two ovarian cancer cell lines, SKOV-3 and A2870, were determined by evaluating cell viability, migration, invasion, and apoptosis. Q-RT-PCR and western blotting techniques were carried out to explore the protein levels of signaling pathway markers. A mouse xenograft model was utilized to determine the silibinin efficacy in inhibiting tumor growth. RESULTS After cell treatment with silibinin, cell viability, migration, and invasion were appreciably inhibited in cancer cell lines, but cell apoptosis was promoted. Also, silibinin reversed the epithelial-mesenchymal transition (EMT) mechanism by inducing E-cadherin expression and reducing N-cadherin and vimentin expression, suppressing the levels of regulators related to EMT such as Snail, Slug, and ZEB1 transcription factors, and also decreasing PI3K/AKT, Smad2/3, and β-catenin intermediate molecules in vitro. Silibinin effectively ameliorated tumor growth in vivo. CONCLUSION silibinin could be considered a potent agent against ovarian cancer based on the results.
Collapse
|
5
|
Ghalandari B, Asadollahi K, Ghorbani F, Ghalehbaghi S, Rafiee S, Komeili A, Kamrava SK. Determinants of gold nanoparticle interactions with Proteins: Off-Target effect study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 269:120736. [PMID: 34923375 DOI: 10.1016/j.saa.2021.120736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Photothermal therapy is one of the promising approaches toward cancer treatment. To date, several compounds have been developed for this application, among which nanoparticles are attracting ever-increasing attention. One of the obstacles in developing efficient photothermal nanoparticle agents is their off-target effect which is mainly mediated via non-specific interactions with proteins. Such interaction not only reduces the bioavailability of the agent but also will cause protein aggregation that can be lethal. So, gaining knowledge on the mechanisms mediating such interactions will facilitate development of more effective agents. Our last studies showed the mechanism of action of two modified gold nanoparticles, folic acid functionalized gold nanoparticles (FA-AuNPs) and gold shelled Fe3O4 nanoparticles (AuFeNPs), as photothermal agents. In the current work, we focus on the interaction of these two NPs with human serum albumin (HSA) and human hemoglobin (Hb) as model proteins. The complex formation between NPs and proteins was investigated by fluorescence spectroscopy, dynamic light scattering and circular dichroism. Our data distinguishes the very distinct mode of interaction of charged and neutral NPs with proteins. While the interaction of neutral AuFeNP to proteins is protein dependent, charged nanoparticles FA-AuNP interact indistinguishably with all proteins via electrostatic interactions. Moreover, complexes obtained from FA-AuNPs with proteins are more stable than that of AuFeNP. However, the secondary structure content of proteins in the presence of NPs indicates the insignificant effect of NPs on the secondary structure of these proteins. Our data propose that the charge functionalization of the NPs is an effective way for modulating the interaction of nanoparticles with proteins.
Collapse
Affiliation(s)
- Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Kazem Asadollahi
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Farnaz Ghorbani
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China
| | - Suzan Ghalehbaghi
- Medical Engineering Department, South Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Saharnaz Rafiee
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ali Komeili
- Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Kamran Kamrava
- Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
6
|
Ghorbani F, Ghalandari B, Liu C. A Facile Method to Synthesize 3D Pomegranate-like Polydopamine Microspheres. Front Bioeng Biotechnol 2022; 9:737074. [PMID: 34993182 PMCID: PMC8724573 DOI: 10.3389/fbioe.2021.737074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/25/2021] [Indexed: 12/18/2022] Open
Abstract
Nanospheres have found versatile applications in the biomedical field; however, their possible harmful effects on immune and inflammatory systems are also a crucial concern. Inspired by a pomegranate structure, we demonstrated a novel structure for the nanostructured microspheres to overcome the drawbacks of nanospheres without compromising their merits. In this study, 3D pomegranate-like polydopamine microspheres (PDAMS) were synthesized by self-oxidative polymerization of dopamine hydrochloride. Herein, controlling the pH during polymerization led to synthesizing homogeneous agglomerated nano-sized spheres (400–2000 nm) and finally forming tunable and monodisperse micron-sized particles (21 µm) with uniform spherical shape porous microstructure. PDAMS interaction with the potential targets, Bone morphogenetic protein-2 (BMP2), Decorin, and Matrilin-1, was investigated via molecular calculations. Theoretical energy analysis revealed that PDAMS interaction with BMP2, Decorin, and Matrilin-1 is spontaneous, so that a protein layer formation on the PDAMS surface suggests application in bone and cartilage repair. It was also observed that PDAMS presented in-vitro degradation within 4 weeks. Here, disappearance of the UV-VIS spectrum peak at 280 nm is accompanied by the degradation of catechol groups. Pomegranate-like PDAMS support the biomimetic formation of hydroxyapatite-like layers, making them appropriate candidates for hard tissue applications. Herein, the appearance of peaks in XRD spectrum at 31.37, 39.57, 45.21, and 50.13° attributed to hydroxyapatite-like layers formation. All these results demonstrated that self-oxidative polymerization under a controllable pH can be a green and straightforward technique for preparing the pomegranate-like PDAMS and providing an innovative basis for further pre-clinical and clinical investigations.
Collapse
Affiliation(s)
- Farnaz Ghorbani
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chaozong Liu
- Institute of Orthopaedic and Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| |
Collapse
|
7
|
Investigation of Decitabine Effects on HDAC3 and HDAC7 mRNA Expression in NALM-6 and HL-60 Cancer Cell Lines. Rep Biochem Mol Biol 2022; 10:420-428. [PMID: 34981019 DOI: 10.52547/rbmb.10.3.420] [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: 03/23/2021] [Accepted: 04/06/2021] [Indexed: 11/18/2022]
Abstract
Background Decitabine is a potent anticancer hypomethylating agent and changes the gene expression through the gene's promoter demethylation and also independently from DNA demethylation. So, the present study was designed to distinguish whether Decitabine, in addition to inhibitory effects on DNA methyltransferase, can change HDAC3 and HDAC7 mRNA expression in NALM-6 and HL-60 cancer cell lines. Methods HL-60, NALM-6, and normal cells were cultured, and the Decitabine treatment dose was obtained (1 µM) through the MTT assay. Finally, HDAC3 and HDAC7 mRNA expression were measured by Real-Time PCR in HL-60 and NALM-6 cancerous cells before and after treatment. Furthermore, HDAC3 and HDAC7 mRNA expression in untreated HL-60 and NALM-6 cancerous cells were compared to normal cells. Results Our results revealed that the expression of HDAC3 and HDAC7 in HL-60 and NALM-6 cells increases as compared to normal cells. After treatment of HL-60 and NALM-6 cells with Decitabine, HDAC3, and HDAC7 mRNA expression were decreased significantly. Conclusion Our data confirmed that the effects of Decitabine are not limited to direct hypomethylation of DNMTs, but it can indirectly affect other epigenetic factors, such as HDACs activity, through converging pathways.
Collapse
|
8
|
Yu Y, Ghalandari B, Shen G, Wang L, Liu X, Wang A, Li S, Xie H, Ding X. Poly (N-vinylpyrrolidone) modification mitigates plasma protein corona formation on phosphomolybdate-based nanoparticles. J Nanobiotechnology 2021; 19:445. [PMID: 34949196 PMCID: PMC8697440 DOI: 10.1186/s12951-021-01140-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/15/2021] [Indexed: 01/08/2023] Open
Abstract
Phosphomolybdate-based nanoparticles (PMo12-based NPs) have been commonly applied in nanomedicine. However, upon contact with biofluids, proteins are quickly adsorbed onto the NPs surface to form a protein corona, which induces the opsonization and facilitates the rapid clearance of the NPs by macrophage uptake. Herein, we introduce a family of structurally homologous PMo12-based NPs (CDS-PMo12@PVPx(x = 0 ~ 1) NPs) capping diverse content of zwitterionic polymer poly (N-vinylpyrrolidone) (PVP) to regulate the protein corona formation on PMo12-based NPs. The fluorescence quenching data indicate that the introduction of PVP effectively reduces the number of binding sites of proteins on PMo12-based NPs. Molecular docking simulations results show that the contact surface area and binding energy of proteins to CDS-PMo12@PVP1 NPs are smaller than the CDS-PMo12@PVP0 NPs. The liquid chromatography-tandem mass spectrometry (LC–MS/MS) is further applied to analyze and quantify the compositions of the human plasma corona formation on CDS-PMo12@PVPx(x = 0 ~ 1) NPs. The number of plasma protein groups adsorption on CDS-PMo12@PVP1 NPs, compared to CDS-PMo12@PVP0 NPs, decreases from 372 to 271. In addition, 76 differentially adsorption proteins are identified between CDS-PMo12@PVP0 and CDS-PMo12@PVP1 NPs, in which apolipoprotein is up-regulated in CDS-PMo12@PVP1 NPs. The apolipoprotein adsorption onto the NPs is proposed to have dysoponic activity and enhance the circulation time of NPs. Our findings demonstrate that PVP grafting on PMo12-based NPs is a promising strategy to improve the anti-biofouling property for PMo12-based nanodrug design. ![]()
Collapse
Affiliation(s)
- Youyi Yu
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Guangxia Shen
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Liping Wang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiao Liu
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Aiting Wang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Sijie Li
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Haiyang Xie
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
9
|
Panji M, Behmard V, Zare Z, Malekpour M, Nejadbiglari H, Yavari S, Nayerpour Dizaj T, Safaeian A, Maleki N, Abbasi M, Abazari O, Shabanzadeh M, Khanicheragh P. Suppressing effects of green tea extract and Epigallocatechin-3-gallate (EGCG) on TGF-β- induced Epithelial-to-mesenchymal transition via ROS/Smad signaling in human cervical cancer cells. Gene 2021; 794:145774. [PMID: 34126197 DOI: 10.1016/j.gene.2021.145774] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/29/2021] [Accepted: 06/09/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Transforming growth factor-β (TGF-β)-induced Epithelial-to-mesenchymal transition (EMT) process is a fundamental target for preventing cervical cancer cells' progression and invasion. Green tea and its principal active substance, Epigallocatechin-3-gallate (EGCG), demonstrate anti-tumor activities in various tumor cells. METHODS The cell viability of two cervical cancer cell lines, Hela and SiHa, in the experimental groups was examined employing the MTT method, and ROS generation was probed applying 2',7'-dichlorofluorescein diacetate-based assay. The Smad signaling and EMT process was evaluated utilizing western blot analysis and quantitative real-time polymerase chain reaction (qRT-PCR). Chromatin immunoprecipitation (ChIP) and Smad binding element (SBE)-luciferase assays were employed to measure Smad-DNA interaction and Smad transcriptional activity, respectively. RESULTS EGCG (0-100 μmol/L) and green tea extract (0-250 μg/ml) suppressed the viability of cancer cells in a dose-dependent manner (p < 0.01). Our conclusions affirmed that pre-incubation with green tea extract (80 μg/ml) and EGCG (60 μmol/L) significantly reversed the impacts of TGF-β in Hela and SiHa cells by decreasing Vimentin, ZEB, Slug, Snail, and Twist and increasing E-cadherin expression. The molecular mechanism of green tea extract and EGCG for TGF-β-induced EMT inhibition interfered with ROS generation and Smad signaling. Green tea extract and EGCG could significantly decrease ROS levels, the phosphorylation of Smad2/3, the translocation, DNA binding, and activity of Smads in cervical cancer cell lines treated with TGF-β1 (p < 0.01). CONCLUSION EGCG and green tea extract suppressed TGF-β-induced EMT in Hela and SiHa cells, and the underlying molecular mechanism may be related to the ROS generation and Smad signaling pathway.
Collapse
Affiliation(s)
- Mohammad Panji
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahideh Behmard
- Student Research Committee, Department of Midwifery, School of Medical, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Zahra Zare
- Department of Biology, Farhangian University, Tehran, Iran
| | - Monireh Malekpour
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hasan Nejadbiglari
- Department of Nursing, Sirjan Branch, Islamic Azad University, Sirjan, Iran
| | - Saeede Yavari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Tina Nayerpour Dizaj
- Department of Medical Biotechnology, Faculty of Modern Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azadeh Safaeian
- Department of Physiology, Faculty of Medicine, Shahid Sadoughy University of Medical Sciences, Yazd, Iran
| | - Narges Maleki
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Islamic Azad University-Tehran North Branch, Tehran, Iran
| | - Mojtaba Abbasi
- Veterinary Medicine, Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Omid Abazari
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Maryam Shabanzadeh
- Department of Medical Radiation, Faculty of Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Parisa Khanicheragh
- Department of Clinical Biochemistry, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
10
|
Panji M, Behmard V, Zare Z, Malekpour M, Nejadbiglari H, Yavari S, Nayerpour Dizaj T, Safaeian A, Bakhshi A, Abazari O, Abbasi M, Khanicheragh P, Shabanzadeh M. Synergistic effects of green tea extract and paclitaxel in the induction of mitochondrial apoptosis in ovarian cancer cell lines. Gene 2021; 787:145638. [PMID: 33848578 DOI: 10.1016/j.gene.2021.145638] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Green tea is a natural compound with anti-neoplastic properties. Paclitaxel (PTX) is a natural anti-tumor medication used to manage patients with advanced ovarian cancer. This manuscript evaluated the cytotoxic effects of green tea extract combined with PTX drug in two human ovarian cancer cell lines (p53-negative cell line, SKOV-3; and mutant type p53 cell line, OVCAR-3) and underlying mechanisms. METHODS The human ovarian cancer cell lines were treated with green tea extract, PTX, and green tea plus PTX for 24 h, and cell viability was assessed using the MTT method. Flow cytometric analyses were carried out to detect apoptosis. For the apoptotic process, quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting analysis were applied to study pAkt, Bax, Bcl-2, Cytochrome C (Cyt-C), cleaved-caspase-3, and cleaved-caspase-9 levels after drug treatments. RESULTS Our results pointed out that various green tea (25 and 50 µg/ml) concentrations combined with PTX (20 and 40 µg/ml) synergistically inhibited cell viability of cancer cells more than green tea or PTX alone after 24 h of treatment. Also, green tea and PTX combination induced apoptosis in ovarian cancer cells by blocking the phosphorylation of Akt and the expression of Bcl-2 while inducing Bax, Cyt-C, cleaved-caspase 3, and cleaved-caspase 9. CONCLUSION Our results showed that the combination of green tea and PTX could be more potent than the individual drug to induce cytotoxicity and apoptosis in ovarian cancer cells.
Collapse
Affiliation(s)
- Mohammad Panji
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahideh Behmard
- Student Research Committee, Department of Midwifery, School of Medical, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Zahra Zare
- Department of Biology, Farhangian University, Tehran, Iran
| | - Monireh Malekpour
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hasan Nejadbiglari
- Department of Nursing, Sirjan Branch, Islamic Azad University, Sirjan, Iran
| | - Saeede Yavari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Tina Nayerpour Dizaj
- Department of Medical Biotechnology, Faculty of Modern Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azadeh Safaeian
- Department of Physiology, Faculty of Medicine, Shahid Sadoughy University of Medical Sciences, Yazd, Iran
| | - Ali Bakhshi
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Omid Abazari
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| | - Mojtaba Abbasi
- Veterinary Medicine, Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Parisa Khanicheragh
- Department of Clinical Biochemistry, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Maryam Shabanzadeh
- Department of Medical Radiation, Faculty of Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| |
Collapse
|
11
|
The Protein-Binding Behavior of Platinum Anticancer Drugs in Blood Revealed by Mass Spectrometry. Pharmaceuticals (Basel) 2021; 14:ph14020104. [PMID: 33572935 PMCID: PMC7911130 DOI: 10.3390/ph14020104] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin and its analogues are widely used as chemotherapeutic agents in clinical practice. After being intravenously administrated, a substantial amount of platinum will bind with proteins in the blood. This binding is vital for the transport, distribution, and metabolism of drugs; however, toxicity can also occur from the irreversible binding between biologically active proteins and platinum drugs. Therefore, it is very important to study the protein-binding behavior of platinum drugs in blood. This review summarizes mass spectrometry-based strategies to identify and quantitate the proteins binding with platinum anticancer drugs in blood, such as offline high-performance liquid chromatography/inductively coupled plasma mass spectrometry (HPLC–ICP-MS) combined with electrospray ionization mass spectrometry (ESI-MS/MS) and multidimensional LC–ESI-MS/MS. The identification of in vivo targets in blood cannot be accomplished without first studying the protein-binding behavior of platinum drugs in vitro; therefore, relevant studies are also summarized. This knowledge will further our understanding of the pharmacokinetics and toxicity of platinum anticancer drugs, and it will be beneficial for the rational design of metal-based anticancer drugs.
Collapse
|
12
|
Shariati B, Yektadoost E, Behzadi E, Azmoodeh E, Attar F, Sari S, Akhtari K, Falahati M. Interaction of silica nanoparticles with tau proteins and PC12 cells: Colloidal stability, thermodynamic, docking, and cellular studies. Int J Biol Macromol 2018; 118:1963-1973. [PMID: 30009913 DOI: 10.1016/j.ijbiomac.2018.07.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/03/2018] [Accepted: 07/11/2018] [Indexed: 12/12/2022]
Abstract
Study on the side effects of the nanoparticles (NPs) can provide useful information regarding their biological and medical applications. Herein, the colloidal stability of the silicon dioxide NPs (SiO2 NPs) in the absence and presence of tau was investigated by TEM and DLS techniques. Afterwards, the thermodynamic parameters of interaction between SiO2 NPs and tau were determined by fluorescence spectroscopy and docking studies. Finally, the cytotoxic effects of SiO2 NPs on the viability of PC12 cells were investigated by MTT, AO/EB staining and flow cytometry assays. TEM, DLS, and zeta potential investigations revealed that tau can reduce the colloidal stability of SiO2 NPs. Fluorescence spectroscopy study indicated that SiO2 NPs bound to the tau with high affinity through hydrogen bonds and van der Waals interactions. Docking study also determined that Ser, Thr and Tyr residues provide a polar microenvironment for SiO2 NPs/tau interaction. Cellular studies demonstrated that SiO2 NPs can induce cell mortality through both apoptosis and necrosis mechanisms. Therefore, it may be concluded that the biological systems such as nervous system proteins can affect the colloidal stability of NPs and vice versa NPs in the biological systems can bind to proteins and cell membranes non-specifically and may induce toxicity.
Collapse
Affiliation(s)
- Behdad Shariati
- Pharmaceutical Sciences Research Center, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Elham Yektadoost
- Pharmaceutical Sciences Research Center, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Elham Behzadi
- Pharmaceutical Sciences Research Center, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Elnaz Azmoodeh
- Pharmaceutical Sciences Research Center, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Farnoosh Attar
- Department of Biology, Faculty of Food Industry & Agriculture, Standard Research Institute (SRI), Karaj, Iran
| | - Soyar Sari
- Department of Cellular and Molecular Biology, Faculty of Advance Science and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advance Science and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran.
| |
Collapse
|