1
|
Al-Hawary SIS, Abdalkareem Jasim S, Altalbawy FMA, Kumar A, Kaur H, Pramanik A, Jawad MA, Alsaad SB, Mohmmed KH, Zwamel AH. miRNAs in radiotherapy resistance of cancer; a comprehensive review. Cell Biochem Biophys 2024; 82:1665-1679. [PMID: 38805114 DOI: 10.1007/s12013-024-01329-2] [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] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
While intensity-modulated radiation therapy-based comprehensive therapy increases outcomes, cancer patients still have a low five-year survival rate and a high recurrence rate. The primary factor contributing to cancer patients' poor prognoses is radiation resistance. A class of endogenous non-coding RNAs, known as microRNAs (miRNAs), controls various biological processes in eukaryotes. These miRNAs influence tumor cell growth, death, migration, invasion, and metastasis, which controls how human carcinoma develops and spreads. The correlation between the unbalanced expression of miRNAs and the prognosis and sensitivity to radiation therapy is well-established. MiRNAs have a significant impact on the regulation of DNA repair, the epithelial-to-mesenchymal transition (EMT), and stemness in the tumor radiation response. But because radio resistance is a complicated phenomena, further research is required to fully comprehend these mechanisms. Radiation response rates vary depending on the modality used, which includes the method of delivery, radiation dosage, tumor stage and grade, confounding medical co-morbidities, and intrinsic tumor microenvironment. Here, we summarize the possible mechanisms through which miRNAs contribute to human tumors' resistance to radiation.
Collapse
Affiliation(s)
| | | | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka, 560069, India
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Divison of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | | | - Salim Basim Alsaad
- Department of Pharmaceutics, Al-Hadi University College, Baghdad, 10011, Iraq
| | | | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
2
|
Wu L, Bai L, Dai W, Wu Y, Xi P, Zhang J, Zheng L. Ginsenoside Rg3: A Review of its Anticancer Mechanisms and Potential Therapeutic Applications. Curr Top Med Chem 2024; 24:869-884. [PMID: 38441023 DOI: 10.2174/0115680266283661240226052054] [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: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Traditional Chinese Medicine (TCM) has a long history of treating various diseases and is increasingly being recognized as a complementary therapy for cancer. A promising natural compound extracted from the Chinese herb ginseng is ginsenoside Rg3, which has demonstrated significant anticancer effects. It has been tested in a variety of cancers and tumors and has proven to be effective in suppressing cancer. OBJECTIVES This work covers various aspects of the role of ginsenoside Rg3 in cancer treatment, including its biological functions, key pathways, epigenetics, and potential for combination therapies, all of which have been extensively researched and elucidated. The study aims to provide a reference for future research on ginsenoside Rg3 as an anticancer agent and a support for the potential application of ginsenoside Rg3 in cancer treatment.
Collapse
Affiliation(s)
- Lei Wu
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Bai
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenshu Dai
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontier Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yaping Wu
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pengjun Xi
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jie Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lily Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Fang X, Li J, Zhang M, Yang L, Wang Y, Liu X, Zhang J. Pharmacokinetic investigation on the mechanism of interaction of anti-breast cancer calycosin with albumin: In vitro. ARAB J CHEM 2023; 16:105175. [DOI: 10.1016/j.arabjc.2023.105175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
4
|
Ilhan H. Nanoarchitectonics of the Effects of Curcumin Carbon Dot-Decorated Chitosan Nanoparticles on Proliferation and Apoptosis-Related Gene Expressions in HepG2 Hepatocellular Carcinoma Cells. ACS OMEGA 2023; 8:33554-33563. [PMID: 37744806 PMCID: PMC10515349 DOI: 10.1021/acsomega.3c03405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023]
Abstract
This study examines the potential anticancer properties of curcumin carbon nanodot-decorated chitosan nanoparticles (CCM@CD/CS-NP) in HepG2 hepatocellular carcinoma cells. CCM@CD/CS-NPs were synthesized, and their size, morphology, and elemental analysis were characterized. The combination of curcumin carbon dots and chitosan in the form of a nanoparticle has a number of benefits, including improved solubility and bioavailability of curcumin, enhanced stability and biocompatibility of carbon dots, and sustained release of the drug due to the mucoadhesive properties of chitosan. The purpose of this research was to examine the efficacy of curcumin carbon dot-decorated chitosan nanoparticles as an anticancer agent in the treatment of HepG2 cell lines. The cell proliferation and apoptosis-related gene expressions in HepG2 cells were assessed to investigate the potential use of nanoparticles in vitro. The IC50 value for the inhibitory effect of CCM@CD/CS-NPs on cell growth and proliferation was determined to be 323.61 μg/mL at 24 h and 267.73 μg/mL at 48 h. Increased caspase-3 and -9 activation shows that CCM@CD/CS-NPs promoted apoptosis in HepG2 cells. It was also shown that the overexpression of Bax and the downregulation of Bcl-2 were responsible for the apoptotic impact of CCM@CD/CS-NPs. The nanoparticles have been shown to have minimal toxicity to normal liver cells, indicating their potential as a safe and effective treatment for HepG2. These novel nanomaterials effectively suppressed tumor development and boosted the rate of apoptotic cell death.
Collapse
Affiliation(s)
- Hasan Ilhan
- Department of Chemistry,
Faculty of Science, Ordu University, Ordu 52200, Turkey
| |
Collapse
|
5
|
Ghasemitarei M, Ghorbi T, Yusupov M, Zhang Y, Zhao T, Shali P, Bogaerts A. Effects of Nitro-Oxidative Stress on Biomolecules: Part 1-Non-Reactive Molecular Dynamics Simulations. Biomolecules 2023; 13:1371. [PMID: 37759771 PMCID: PMC10527456 DOI: 10.3390/biom13091371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Plasma medicine, or the biomedical application of cold atmospheric plasma (CAP), is an expanding field within plasma research. CAP has demonstrated remarkable versatility in diverse biological applications, including cancer treatment, wound healing, microorganism inactivation, and skin disease therapy. However, the precise mechanisms underlying the effects of CAP remain incompletely understood. The therapeutic effects of CAP are largely attributed to the generation of reactive oxygen and nitrogen species (RONS), which play a crucial role in the biological responses induced by CAP. Specifically, RONS produced during CAP treatment have the ability to chemically modify cell membranes and membrane proteins, causing nitro-oxidative stress, thereby leading to changes in membrane permeability and disruption of cellular processes. To gain atomic-level insights into these interactions, non-reactive molecular dynamics (MD) simulations have emerged as a valuable tool. These simulations facilitate the examination of larger-scale system dynamics, including protein-protein and protein-membrane interactions. In this comprehensive review, we focus on the applications of non-reactive MD simulations in studying the effects of CAP on cellular components and interactions at the atomic level, providing a detailed overview of the potential of CAP in medicine. We also review the results of other MD studies that are not related to plasma medicine but explore the effects of nitro-oxidative stress on cellular components and are therefore important for a broader understanding of the underlying processes.
Collapse
Affiliation(s)
- Maryam Ghasemitarei
- Department of Physics, Sharif University of Technology, Tehran 14588-89694, Iran
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
| | - Tayebeh Ghorbi
- Department of Physics, Sharif University of Technology, Tehran 14588-89694, Iran
| | - Maksudbek Yusupov
- School of Engineering, New Uzbekistan University, Tashkent 100007, Uzbekistan
- School of Engineering, Central Asian University, Tashkent 111221, Uzbekistan
- Laboratory of Thermal Physics of Multiphase Systems, Arifov Institute of Ion-Plasma and Laser Technologies, Academy of Sciences of Uzbekistan, Tashkent 100125, Uzbekistan
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
| | - Yuantao Zhang
- School of Electrical Engineering, Shandong University, Jinan 250061, China
| | - Tong Zhao
- School of Electrical Engineering, Shandong University, Jinan 250061, China
| | - Parisa Shali
- Research Unit Plasma Technology, Department of Applied Physics, Faculty of Engineering and Agriculture, Ghent University, 9000 Ghent, Belgium
| | - Annemie Bogaerts
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
6
|
Yang S, Song Y, Hu Y, Chen H, Yang D, Song X. Multifaceted Roles of Copper Ions in Anticancer Nanomedicine. Adv Healthc Mater 2023; 12:e2300410. [PMID: 37027332 DOI: 10.1002/adhm.202300410] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/27/2023] [Indexed: 04/08/2023]
Abstract
The significantly increased copper level in tumor tissues and serum indicates the close association of copper ions with tumor development, making copper ions attractive targets in the development of novel tumor treatment methods. The advanced nanotechnology developed in the past decades provides great potential for tumor therapy, among which Cu-based nanotherapeutic systems have received greater attention. Herein, the multifaceted roles of copper ions in cancer progression are summarized and the recent advances in the copper-based nanostructures or nanomedicines for different kinds of tumor therapies including copper depletion therapy, copper-based cytotoxins, copper-ion-based chemodynamic therapy and its combination with other treatments, and copper-ion-induced ferroptosis and cuproptosis activation are discussed. Furthermore, the perspectives for the further development of copper-ion-based nanomedicines for tumor therapy and clinic translation are presented by the authors.
Collapse
Affiliation(s)
- Siyuan Yang
- Department of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, P. R. China
| | - Yingnan Song
- Department of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, P. R. China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, 550025, P. R. China
| | - Yanling Hu
- Nanjing Polytechnic Institute, 210048, Nanjing, China
| | - HongJin Chen
- Department of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, P. R. China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, 550025, P. R. China
| | - Dongliang Yang
- School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), 211816, 30 South Puzhu Road, Nanjing, China
| | - Xuejiao Song
- School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), 211816, 30 South Puzhu Road, Nanjing, China
| |
Collapse
|
7
|
Erukainure OL, Oyenihi OR, Amaku JF, Chukwuma CI, Nde AL, Salau VF, Matsabisa MG. Cannabis sativa L. modulates altered metabolic pathways involved in key metabolisms in human breast cancer (MCF-7) cells: A metabolomics study. Heliyon 2023; 9:e16156. [PMID: 37215911 PMCID: PMC10196869 DOI: 10.1016/j.heliyon.2023.e16156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
The present study investigated the ability of Cannabis sativa leaves infusion (CSI) to modulate major metabolisms implicated in cancer cells survival, as well as to induce cell death in human breast cancer (MCF-7) cells. MCF-7 cell lines were treated with CSI for 48 h, doxorubicin served as the standard anticancer drug, while untreated MCF-7 cells served as the control. CSI caused 21.2% inhibition of cell growth at the highest dose. Liquid chromatography-mass spectroscopy (LC-MS) profiling of the control cells revealed the presence of carbohydrate, vitamins, oxidative, lipids, nucleotides, and amino acids metabolites. Treatment with CSI caused a 91% depletion of these metabolites, while concomitantly generating selenomethionine, l-cystine, deoxyadenosine triphosphate, cyclic AMP, selenocystathionine, inosine triphosphate, adenosine phosphosulfate, 5'-methylthioadenosine, uric acid, malonic semialdehyde, 2-methylguanosine, ganglioside GD2 and malonic acid. Metabolomics analysis via pathway enrichment of the metabolites revealed the activation of key metabolic pathways relevant to glucose, lipid, amino acid, vitamin, and nucleotide metabolisms. CSI caused a total inactivation of glucose, vitamin, and nucleotide metabolisms, while inactivating key lipid and amino acid metabolic pathways linked to cancer cell survival. Flow cytometry analysis revealed an induction of apoptosis and necrosis in MCF-7 cells treated with CSI. High-performance liquid chromatography (HPLC) analysis of CSI revealed the presence of cannabidiol, rutin, cinnamic acid, and ferulic. These results portray the antiproliferative potentials of CSI as an alternative therapy for the treatment and management of breast cancer as depicted by its modulation of glucose, lipid, amino acid, vitamin, and nucleotide metabolisms, while concomitantly inducing cell death in MCF-7 cells.
Collapse
Affiliation(s)
- Ochuko L. Erukainure
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Omolola R. Oyenihi
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - James F. Amaku
- Department of Chemistry, Michael Okpara University of Agriculture, Umudike, Abia State, Nigeria
| | - Chika I. Chukwuma
- Center for Quality of Health and Living, Faculty of Health Sciences, Central University of Technology, Bloemfontein 9301, South Africa
| | - Adeline Lum Nde
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Veronica F. Salau
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Motlalepula G. Matsabisa
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
8
|
Hanif A, Ibrahim AH, Ismail S, Al-Rawi SS, Ahmad JN, Hameed M, Mustufa G, Tanwir S. Cytotoxicity against A549 Human Lung Cancer Cell Line via the Mitochondrial Membrane Potential and Nuclear Condensation Effects of Nepeta paulsenii Briq., a Perennial Herb. Molecules 2023; 28:molecules28062812. [PMID: 36985784 PMCID: PMC10054104 DOI: 10.3390/molecules28062812] [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: 12/04/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
The genus Nepeta belongs to the largest Lamiaceae family, with 300 species, which are distributed throughout the various regions of Africa, Asia, India, and America. Along with other plant families distinguished by their medicinal and therapeutic values, the Nepeta genus of Lameaceae remains relatively valuable. Hence, the phytochemicals of N. paulsenii Briq. were extracted using different plant parts, i.e., leaves, stem, roots, flowers, and the whole plant by using various solvents (ethanol, water, and ethyl acetate), obtaining 15 fractions. Each extract of dried plant material was analyzed by FT-IR and GC-MS to identify the chemical constituents. The cytotoxicity of each fraction was analyzed by MTT assay and mitochondrial membrane potential and nuclear condensation assays against lung cancer cells. Among the ethyl acetate and ethanolic extracts, the flowers showed the best results, with IC50 values of 51.57 μg/mL and 50.58 μg/mL, respectively. In contrast, among the water extracts of the various plant segments, the stem showed the best results, with an IC50 value of 123.80 μg/mL. 5-flourouracil was used as the standard drug, providing an IC50 value of 83.62 μg/mL. The Hoechst 33342 stain results indicated apoptotic features, i.e., chromatin dissolution and broken down, fragmented, and crescent-shaped nuclei. The ethanolic extracts of the flowers showed more pronounced apoptotic effects on the cells. The mitochondrial membrane potential indicated that rhodamine 123 fluorescence signals suppressed mitochondrial potential due to the treatment with the extracts. Again, the apoptotic index of the ethanolic extract of the flowers remained the highest. Hence it can be concluded that the flower part of N. paulsenii Briq. was found to be the most active against the A459 human lung cancer cell line.
Collapse
Affiliation(s)
- Aqsa Hanif
- Department of Botany, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Ahmad H Ibrahim
- Pharmacy Department, Faculty of Pharmacy, Tishk International University, 100mt. St., Near Baz Interaction, Erbil 44001, KRG, Iraq
| | - Sidra Ismail
- Incharge Health Officer, BHU 418 GB, Faisalabad 37150, Pakistan
| | - Sawsan S Al-Rawi
- Biology Education Department, Faculty of Education, Tishk International University, 100mt. St., Near Baz Interaction, Erbil 44001, KRG, Iraq
| | - Jam Nazeer Ahmad
- Department of Entomology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Mansoor Hameed
- Department of Botany, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Ghulam Mustufa
- Centre of Agricultural Biochemistry and Biotechnology (CABB), University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Samina Tanwir
- Department of Botany, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| |
Collapse
|
9
|
Studies on 1,4-Quinone Derivatives Exhibiting Anti-Leukemic Activity along with Anti-Colorectal and Anti-Breast Cancer Effects. Molecules 2022; 28:molecules28010077. [PMID: 36615273 PMCID: PMC9822417 DOI: 10.3390/molecules28010077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC), breast cancer, and chronic myeloid leukemia (CML) are life-threatening malignancies worldwide. Although potent therapeutic and screening strategies have been developed so far, these cancer types are still major public health problems. Therefore, the exploration of more potent and selective new agents is urgently required for the treatment of these cancers. Quinones represent one of the most important structures in anticancer drug discovery. We have previously identified a series of quinone-based compounds (ABQ-1-17) as anti-CML agents. In the current work, ABQ-3 was taken to the National Cancer Institute (NCI) for screening to determine its in vitro antiproliferative effects against a large panel of human tumor cell lines at five doses. ABQ-3 revealed significant growth inhibition against HCT-116 CRC and MCF-7 breast cancer cells with 2.00 µM and 2.35 µM GI50 values, respectively. The MTT test also showed that ABQ-3 possessed anticancer effects towards HCT-116 and MCF-7 cells with IC50 values of 5.22 ± 2.41 μM and 7.46 ± 2.76 μM, respectively. Further experiments indicated that ABQ-3 induced apoptosis in both cell lines, and molecular docking studies explicitly suggested that ABQ-3 exhibited DNA binding in a similar fashion to previously reported compounds. Based on in silico pharmacokinetic prediction, ABQ-3 might display drug-like features enabling this compound to become a lead molecule for future studies.
Collapse
|
10
|
Real-Time Monitoring of the Cytotoxic and Antimetastatic Properties of Cannabidiol in Human Oral Squamous Cell Carcinoma Cells Using Electric Cell-Substrate Impedance Sensing. Int J Mol Sci 2022; 23:ijms232415842. [PMID: 36555480 PMCID: PMC9785110 DOI: 10.3390/ijms232415842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/19/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Cannabidiol (CBD) is an active natural compound that is extracted from Cannabis sativa. Previous studies show that CBD is a nonpsychotropic compound with significant anticancer effects. This study determines its cytotoxic effect on oral cancer cells and OEC-M1 cells and compares the outcomes with a chemotherapeutic drug, cisplatin. This study has investigated the effect of CBD on the viability, apoptosis, morphology, and migration of OEC-M1 cells. Electric cell-substrate impedance sensing (ECIS) is used to measure the change in cell impedance for cells that are treated with a series concentration of CBD for 24 h. AlamarBlue and annexin V/7-AAD staining assays show that CBD has a cytotoxic effect on cell viability and induces cell apoptosis. ECIS analysis shows that CBD decreases the overall resistance and morphological parameters at 4 kHz in a concentration-dependent manner. There is a significant reduction in the wound-healing recovery rate for cells that are treated with 30 μM CBD. This study demonstrates that ECIS can be used for in vitro screening of new chemotherapy and is more sensitive, functional, and comprehensive than traditional biochemical assays. CBD also increases cytotoxicity on cell survival and the migration of oral cancer cells, so it may be a therapeutic drug for oral cancer.
Collapse
|
11
|
Wang Y, Li S, Weng L, Du H, Wang J, Xu X. LASS2 overexpression enhances early apoptosis of lung cancer cells through the caspase‑dependent pathway. Oncol Rep 2022; 48:220. [PMID: 36300249 DOI: 10.3892/or.2022.8435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 06/27/2022] [Indexed: 11/06/2022] Open
Abstract
In a previous study by the authors, the longevity assurance homolog 2 (LASS2) gene was determined to inhibit activity of vacuolar H+‑ATPase (V‑ATPase) by combining with the C subunit (ATP6L) of V‑ATPase. However, the influence of LASS2 overexpression and silencing on apoptosis of human lung cancer cells 95D or 95C remains unclear. Thus, the effect of LASS2 on apoptosis and its potential mechanisms were investigated in 95D and 95C cells. Using the lentiviral transfection method, lentiviral vectors of LASS2 overexpression and silencing were transfected into 95D and 95C cells, respectively. The apoptotic ability of tumor cells was observed by flow cytometry. The expression levels of LASS2, Bcl‑2, Bax, cytochrome c, caspase‑9, and caspase‑3 were detected by western blotting. CCK‑8 assay was used to detect the growth ability of tumor cells in vitro. Flow cytometric analysis revealed that LASS2 overexpression could promote the early apoptosis of lung cancer cells 95D. CCK‑8 assay demonstrated that LASS2 overexpression inhibited the proliferation of 95D cells. Additionally, LASS2 overexpression decreased the expression of Bcl‑2, induced the release of cytochrome c from mitochondria, and promoted the activation of caspase‑9 and caspase‑3. There was a significant difference in the expression of Bcl‑2, cytochrome c, caspase‑9 and caspase‑3 in the LASS2‑overexpression group compared with the normal and negative control groups. Alternatively, the aforementioned experiments in lung cancer cells 95C following LASS2 silencing produced the opposite effects. LASS2 may induce early apoptosis of lung cancer cells by influencing the caspase‑dependent mitochondrial pathway.
Collapse
Affiliation(s)
- Yamei Wang
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Shirong Li
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Lixin Weng
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Hua Du
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Jingyuan Wang
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Xiaoyan Xu
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| |
Collapse
|
12
|
Bae C, Kim H, Kook YM, Lee C, Kim C, Yang C, Park MH, Piao Y, Koh WG, Lee K. Induction of ferroptosis using functionalized iron-based nanoparticles for anti-cancer therapy. Mater Today Bio 2022; 17:100457. [PMID: 36388450 PMCID: PMC9641001 DOI: 10.1016/j.mtbio.2022.100457] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
Ferroptosis, a cell death pathway that is induced in response to iron, has recently attracted remarkable attention given its emerging therapeutic potential in cancer cells. The need for a promising modality to improve chemotherapy's efficacy through this pathway has been urgent in recent years, and this non-apoptotic cell death pathway accumulates reactive oxygen species (ROS) and is subsequently involved in lipid peroxidation. Here, we report cancer-targeting nanoparticles that possess highly efficient cancer-targeting ability and minimal systemic toxicity, thereby leading to ferroptosis. To overcome the limit of actual clinical application, which is the ultimate goal due to safety issues, we designed safe nanoparticles that can be applied clinically. Nanoparticles containing ferroptosis-dependent iron and FDA-approved hyaluronic acid (FHA NPs) are fabricated by controlling physicochemical properties, and the FHA NPs specifically induce ROS production and lipid peroxidation in cancer cells without affecting normal cells. The excellent in vivo anti-tumor therapeutic effect of FHA NPs was confirmed in the A549 tumor-bearing mice model, indicating that the induction of FHA NP-mediated cell death via the ferroptosis pathway could serve as a powerful platform in anticancer therapy. We believe that this newly proposed FHA NP-induced ferroptosis strategy is a promising system that offers the potential for efficient cancer treatment and provides insight into the safe design of nanomedicines for clinical applications.
Collapse
Affiliation(s)
- Chaewon Bae
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyerim Kim
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yun-Min Kook
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Chaedong Lee
- Graduate School of Convergence Science and Technology, Seoul National University, 145 Gwanggyo-ro, Yeongtong-gu, Suwon-si, 16229, Republic of Korea
| | - Changheon Kim
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chungmo Yang
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min Hee Park
- THEDONEE, 1208, 156, Gwanggyo-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16506, Republic of Korea
| | - Yuanzhe Piao
- Graduate School of Convergence Science and Technology, Seoul National University, 145 Gwanggyo-ro, Yeongtong-gu, Suwon-si, 16229, Republic of Korea
- Advanced Institutes of Convergence Technology, Seoul National University, 145, Gwanggyo-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute for Convergence Science, 145, Gwanggyo-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, Republic of Korea
- Corresponding author. Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
13
|
Aghamohammadi M, Zolghadr L, Nezhad NS, Ahmadpour Yazdi H, Esfahani AJ, Gheibi N. Investigating the effects of quercetin fatty acid esters on apoptosis, mechanical properties, and expression of ERK in melanoma cell line (A375). Life Sci 2022; 310:121007. [PMID: 36181863 DOI: 10.1016/j.lfs.2022.121007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/19/2022] [Accepted: 09/23/2022] [Indexed: 11/19/2022]
Abstract
AIMS Malignant melanoma (MM) is the most fatal skin cancer with a critical increase in the number of cases in the last decades. Recent studies have shown the antitumor potential of active biological phytochemical structures of flavonoids for the prevention and treatment of cancerous cells. In this study, two quercetin fatty acid esters (α-linolenic acid (ALA) and linoleic acid (LA)) compounds were evaluated in terms of inducing apoptotic human melanoma cells (A375) death in vitro. MAIN METHODS The MTT assay was utilized for comparing the effects of quercetin, ALA, and LA on A375 cell viability concentrations of 5, 25, 35, 50, and 100μg/mL for 24 and 48 h to obtain IC50. To detect the effects on apoptosis and to determine p-ERK/ERK apoptosis-related signaling pathway proteins level, flow-cytometry and western blot were used. Finally, the nano-mechanical properties of the melanoma A375 membrane structure containing elastic modulus value and cell-cell adhesion forces were investigated using Atomic Force Microscopy (AFM). Statistical data was analyzed in GraphPad v.8.0.0. KEY FINDINGS The most significant A375 cell viability amplified effect of Q-LA was observed with a half-maximal inhibitory concentration (IC50 = 35 μg/ml, 48 h), proportional to dose. Ester compounds, especially Q-LA, showed the highest cell proliferation inhibition with improved elastic modulus, cell-cell adhesion forces (253 ± 11.2), and elevated apoptosis-inducing effect (p < 0.01**). Moreover, Q-LA significantly decreased the mean levels of p-ERK phosphorylation (0.1439) and, subsequently, apoptosis in A375 cells. SIGNIFICANCE The data presented in this study confirmed the antitumor activity of ester compounds against A375 cells, high-lighting the ability of the tested compounds to induce apoptosis.
Collapse
Affiliation(s)
| | - Leila Zolghadr
- Department of Chemistry, Imam Khomeini International University, Qazvin, Iran
| | | | - Hossein Ahmadpour Yazdi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Commuicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Azam Janati Esfahani
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Commuicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Commuicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
14
|
Zeynali-Moghaddam S, Kheradmand F, Gholizadeh-Ghaleh Aziz S, Abroon S. Combination effects of capecitabine, irinotecan and 17-AAG on colorectal cancer cell line (HT-29). Ann Med Surg (Lond) 2022; 78:103850. [PMID: 35734719 PMCID: PMC9207062 DOI: 10.1016/j.amsu.2022.103850] [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: 04/05/2022] [Revised: 05/19/2022] [Accepted: 05/22/2022] [Indexed: 11/21/2022] Open
Abstract
Objevtive Evasion of apoptosis is a major feature of cancer cells, therefore designing treatment strategies to target apoptotic pathways seems effective. In this study, we investigate the effect of 17-AAG (17-allylaminogeldanamycin) alone and in double and triple combination with capecitabine (Cap) and irinotecan (IR) on HT-29 colon cancer cell line apoptosis. Methods Capase-3, 8, 9, p53 and NF-κB genes expression were analyzed by Real-time PCR. DNA laddering assay was performed to confirm Real-time PCR results. Results Our results showed that all single treatment groups elevated expression of caspase-3, 8, and 9 significantly and IR/Cap was the only double combination group that could upregulate caspase-8 and -9. NF-κB was down-regulated in single treatment and IR/Cap double combination group, significantly. 17-AAG mono-treatment and IR/Cap and Cap/17-AAG double combination group significantly upregulated p53 gene expression. Conclusion Our findings showed proapoptotic effects of 17-AAG alone and in combination with Cap and IR. These findings propose 17-AAG in combination with routine chemotherapy, as a new protocol for colorectal cancer combination therapy. Targeted therapy of apoptosis is the main effective way against of cancer cells. 17-AAG alone and in combination with Cap and IR can regulate the pro-apoptotic factors. Combination therapy has more effective than single therapy.
Collapse
|
15
|
Tamtaji OR, Razavi ZS, Razzaghi N, Aschner M, Barati E, Mirzaei H. Quercetin and Glioma: Which signaling pathways are involved? Curr Mol Pharmacol 2022; 15:962-968. [DOI: 10.2174/1874467215666220211094136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/20/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
Gliomas are the most common brain tumors. These tumors commonly exhibit continuous growth without invading surrounding brain tissues. Dominant remedial approaches suffer limited therapy and survival rates. Although some progress has been made in conventional glioma treatments, these breakthroughs have not yet proven sufficient for treating this malignancy. The remedial options are limited given gliomas' aggressive metastasis and drug resistance. Quercetin, a flavonoid, is an anti-oxidative, anti-allergic, antiviral, anti-inflammatory, and anticancer compound. Multiple lines of evidence have shown that Quercetin has anti-tumor effects, documenting this natural compound exerts its pharmacological effects by targeting a variety of cellular and molecular processes, i.e., apoptosis, metastasis, and autophagy. Herein, we summarize various cellular and molecular pathways that are affected by Quercetin in gliomas.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zahra Sadat Razavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Nazanin Razzaghi
- Laboratory Sciences Research Centre, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - Erfaneh Barati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
16
|
In Vitro and In Silico Study of Analogs of Plant Product Plastoquinone to Be Effective in Colorectal Cancer Treatment. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030693. [PMID: 35163957 PMCID: PMC8839215 DOI: 10.3390/molecules27030693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
Plants have paved the way for the attainment of molecules with a wide-range of biological activities. However, plant products occasionally show low biological activities and/or poor pharmacokinetic properties. In that case, development of their derivatives as drugs from the plant world has been actively performed. As plant products, plastoquinones (PQs) have been of high importance in anticancer drug design and discovery; we have previously evaluated and reported the potential cytotoxic effects of a series of PQ analogs. Among these analogs, PQ2, PQ3 and PQ10 were selected for National Cancer Institute (NCI) for in vitro screening of anticancer activity against a wide range of cancer cell lines. The apparent superior anticancer potency of PQ2 on the HCT-116 colorectal cancer cell line than that of PQ3 and PQ10 compared to other tested cell lines has encouraged us to perform further mechanistic studies to enlighten the mode of anti-colorectal cancer action of PQ2. For this purpose, its apoptotic effects on the HCT-116 cell line, DNA binding capacity and several crucial pharmacokinetic properties were investigated. Initially, MTT assay was conducted for PQ2 at different concentrations against HCT-116 cells. Results indicated that PQ2 exhibited significant cytotoxicity in HCT-116 cells with an IC50 value of 4.97 ± 1.93 μM compared to cisplatin (IC50 = 26.65 ± 7.85 μM). Moreover, apoptotic effects of PQ2 on HCT-116 cells were investigated by the annexin V/ethidium homodimer III staining method and PQ2 significantly induced apoptosis in HCT-116 cells compared to cisplatin. Based on the potent DNA cleavage capacity of PQ2, molecular docking studies were conducted in the minor groove of the double helix of DNA and PQ2 presented a key hydrogen bonding through its methoxy moiety. Overall, both in vitro and in silico studies indicated that effective, orally bioavailable drug-like PQ2 attracted attention for colorectal cancer treatment. The most important point to emerge from this study is that appropriate derivatization of a plant product leads to unique biologically active compounds.
Collapse
|
17
|
Münz S, Feger M, Edemir B, Föller M. Up-Regulation of Fibroblast Growth Factor 23 Gene Expression in UMR106 Osteoblast-like Cells with Reduced Viability. Cells 2021; 11:40. [PMID: 35011602 PMCID: PMC8750768 DOI: 10.3390/cells11010040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) controls vitamin D and phosphate homeostasis in the kidney and has additional paracrine effects elsewhere. As a biomarker, its plasma concentration is associated with progression of inflammatory, renal, and cardiovascular diseases. Major stimuli of FGF23 synthesis include active vitamin D and inflammation. Antineoplastic chemotherapy treats cancer by inducing cellular damage ultimately favoring cell death (apoptosis and necrosis) and causing inflammation. Our study explored whether chemotherapeutics and other apoptosis inducers impact on Fgf23 expression. Experiments were performed in osteoblast-like UMR106 cells, Fgf23 gene expression and protein synthesis were determined by qRT-PCR and ELISA, respectively. Viability was assessed by MTT assay and NFκB activity by Western Blotting. Antineoplastic drugs cisplatin and doxorubicin as well as apoptosis inducers procaspase-activating compound 1 (PAC-1), a caspase 3 activator, and serum depletion up-regulated Fgf23 transcripts while reducing cell proliferation and viability. The effect of cisplatin on Fgf23 transcription was paralleled by Il-6 up-regulation and NFκB activation and attenuated by Il-6 and NFκB signaling inhibitors. To conclude, cell viability-decreasing chemotherapeutics as well as apoptosis stimulants PAC-1 and serum depletion up-regulate Fgf23 gene expression. At least in part, Il-6 and NFκB may contribute to this effect.
Collapse
Affiliation(s)
- Sina Münz
- Department of Physiology, University of Hohenheim, 70599 Stuttgart, Germany; (S.M.); (M.F.)
| | - Martina Feger
- Department of Physiology, University of Hohenheim, 70599 Stuttgart, Germany; (S.M.); (M.F.)
| | - Bayram Edemir
- Department of Hematology and Oncology, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany;
| | - Michael Föller
- Department of Physiology, University of Hohenheim, 70599 Stuttgart, Germany; (S.M.); (M.F.)
| |
Collapse
|
18
|
Nam HK, Jeong SR, Pyo MC, Ha SK, Nam MH, Lee KW. Methylglyoxal-Derived Advanced Glycation End Products (AGE4) Promote Cell Proliferation and Survival in Renal Cell Carcinoma Cells through the RAGE/Akt/ERK Signaling Pathways. Biol Pharm Bull 2021; 44:1697-1706. [PMID: 34719646 DOI: 10.1248/bpb.b21-00382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Advanced glycation end products (AGEs) are the products formed through a non-enzymatic reaction of reducing sugars with proteins or lipids. There is a potential for toxicity in the case of AGEs produced through glycation with dicarbonyl compounds including methylglyoxal, glyoxal, and 3-deoxyglucosone. The AGEs bind the receptor for advanced glycation end products (RAGE) and stimulate the mitogen-activated protein (MAP) kinase signaling pathway that can increase the production of matrix metalloproteinases (MMPs). In addition, AGE-induced protein kinase B (Akt) signaling can promote cancer cell proliferation and contribute to many diseases such as kidney cancer. In light of the lack of extensive study of the relationship between methylglyoxal-induced AGEs (AGE4) and renal cancer, we studied the proliferous and anti-apoptotic effects of AGE4 on renal cell carcinoma (RCC) in this study. AGE4 treatment was involved in the proliferation and migration of RCC cells in vitro by upregulating proliferating cell nuclear antigen (PCNA) and MMPs while suppressing apoptotic markers such as Bax and caspase 3. Moreover, Akt and extracellular-signal-regulated kinase (ERK) were phosphorylated in RCC cells with AGE4 treatment. As a result, this study demonstrated that AGE4-RAGE axis can promote the growth ability of RCC by inducing PCNA, MMPs, and inhibiting apoptosis in RCC via the Akt and ERK signaling pathways.
Collapse
Affiliation(s)
- Han-Kyul Nam
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| | - So-Ra Jeong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| | - Min Cheol Pyo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| | - Sang-Keun Ha
- Division of Functional Food Research, Korea Food Research Institute
| | - Mi-Hyun Nam
- Sue Anschutz-Rodgers Eye Center and Department of Ophthalmology, University of Colorado
| | - Kwang-Won Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| |
Collapse
|
19
|
Park S, Nguyen T, Benoit E, Sackett DL, Garmendia-Cedillos M, Pursley R, Boccara C, Gandjbakhche A. Quantitative evaluation of the dynamic activity of HeLa cells in different viability states using dynamic full-field optical coherence microscopy. BIOMEDICAL OPTICS EXPRESS 2021; 12:6431-6441. [PMID: 34745747 PMCID: PMC8548024 DOI: 10.1364/boe.436330] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 05/30/2023]
Abstract
Dynamic full-field optical coherence microscopy (DFFOCM) was used to characterize the intracellular dynamic activities and cytoskeleton of HeLa cells in different viability states. HeLa cell samples were continuously monitored for 24 hours and compared with histological examination to confirm the cell viability states. The averaged mean frequency and magnitude observed in healthy cells were 4.79±0.5 Hz and 2.44±1.06, respectively. In dead cells, the averaged mean frequency was shifted to 8.57±0.71 Hz, whereas the magnitude was significantly decreased to 0.53±0.25. This cell dynamic activity analysis using DFFOCM is expected to replace conventional time-consuming and biopsies-required histological or biochemical methods.
Collapse
Affiliation(s)
- Soongho Park
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49 Convent Dr., Bethesda 20814, USA
| | - Thien Nguyen
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49 Convent Dr., Bethesda 20814, USA
| | - Emilie Benoit
- LLTech SAS-Aquyre Biosciences, 58 Rue du Dessous des Berges, 75013 Paris, France
| | - Dan L. Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49 Convent Dr., Bethesda 20814, USA
| | - Marcial Garmendia-Cedillos
- The Signal Processing and Instrumentation Section, Center for Information Technology, National Institutes of Health, 49 Convent Dr., Bethesda 20814, USA
| | - Randall Pursley
- The Signal Processing and Instrumentation Section, Center for Information Technology, National Institutes of Health, 49 Convent Dr., Bethesda 20814, USA
| | - Claude Boccara
- LLTech SAS-Aquyre Biosciences, 58 Rue du Dessous des Berges, 75013 Paris, France
- Institut Langevin, ESPCI Paris, CNRS, PSL University, 1 rue Jussieu, 75005 Paris, France
| | - Amir Gandjbakhche
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49 Convent Dr., Bethesda 20814, USA
| |
Collapse
|
20
|
Liu H, Liu C, Wang M, Sun D, Zhu P, Zhang P, Tan X, Shi G. Tanshinone IIA affects the malignant growth of Cholangiocarcinoma cells by inhibiting the PI3K-Akt-mTOR pathway. Sci Rep 2021; 11:19268. [PMID: 34588580 PMCID: PMC8481305 DOI: 10.1038/s41598-021-98948-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
In the present study, we aimed to find the target of Tanshinone IIA (Tan-IIA) in Cholangiocarcinoma by network pharmacology-based prediction and investigate the possible mechanism through experimental verification. In this study, we combined Tan-IIA-specific and Cholangiocarcinoma-specific targets with protein–protein interactions (PPI) to construct a Tan-IIA targets-Cholangiocarcinoma network, and network pharmacology approach was applied to identify potential targets and mechanisms of Tan-IIA in the treatment of Cholangiocarcinoma. The anti-cancer effects of Tan-IIA were investigated by using subcutaneous tumorigenic model in nude mice and in the human Cholangiocarcinoma cell lines in vitro. Our results showed that Tan-IIA treatment considerably suppressed the proliferation and migration of Cholangiocarcinoma cells while inducing apoptosis of Cholangiocarcinoma cells. Western blot results demonstrated that the expression of PI3K, p-Akt, p-mTOR, and mTOR were inhibited by Tan-IIA. Meanwhile, After treatment with Tan-IIA, the level of Bcl2 was downregulated and cleaved caspase-3 expression increased. Further studies revealed that the anticancer effects of Tan-IIA were severely mitigated by pretreatment with a PI3K agonist. Our research provides a new anticancer strategy and strengthens support for the use of Tan-IIA as an anticancer drug for the treatment of CCA.
Collapse
Affiliation(s)
- Huayuan Liu
- Department of Medicine, Qingdao University, Qingdao, China
| | - Caiyun Liu
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Mengya Wang
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dongxu Sun
- Graduate School of Dalian Medical University, Dalian, China
| | - Pengcheng Zhu
- Graduate School of Dalian Medical University, Dalian, China
| | - Ping Zhang
- Department of Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Xueying Tan
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Guangjun Shi
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
21
|
Burlacu E, Tanase C. Anticancer Potential of Natural Bark Products-A Review. PLANTS 2021; 10:plants10091895. [PMID: 34579427 PMCID: PMC8467168 DOI: 10.3390/plants10091895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/22/2022]
Abstract
Cell biology, plant-based extracts, structural chemistry, and laboratory in vitro or in vivo experiments are the principal aspects or interfaces that can contribute to discovering new possibilities in cancer therapy and to developing improved chemotherapeutics. Forestry residues can be used for their wealthy resource in polyphenols and other phytoconstituents known for anticancer properties. This review is designed to bring together information on the in vitro or in vivo anticancer potential of woody vascular plants especially the bark extracts (BE) and biosynthesized metallic nanoparticles (BMN) using bark extracts. Type of extracts, main phytoconstituents found in extracts responsible for the anticancer activity, and targeted cancerous cell lines were followed. The literature data were collected via Clarivate Analytics, Science Direct, PubMed, and Google Academic (2011-2021). The search terms were: bark extracts, metallic nanoparticles, silver nanoparticles, gold nanoparticles, anticancer, cytotoxic activity, antiproliferative effect, and antimetastatic potential in vitro and in vivo. All of the search terms listed above were used in different combinations. The literature data highlight the efficaciousness of the BE and BMN as anticancer agents in in vitro experiments and showed the mechanism of action and their advantage of nontoxicity on normal cells. In vitro testing has shown promising results of the BE and BMN effect on different cancer cell lines. In vivo testing is lacking and more data is necessary for drug development on animal models.
Collapse
Affiliation(s)
- Ema Burlacu
- Residency Department, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, 38 Gheorghe Marinescu Street, 540139 Târgu Mureș, Romania;
| | - Corneliu Tanase
- Department of Pharmaceutical Botany, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, 38 Gheorghe Marinescu Street, 540139 Târgu Mureș, Romania
- Correspondence: ; Tel.:+40-744-215-543
| |
Collapse
|
22
|
Kohandel Z, Farkhondeh T, Aschner M, Pourbagher-Shahri AM, Samarghandian S. STAT3 pathway as a molecular target for resveratrol in breast cancer treatment. Cancer Cell Int 2021; 21:468. [PMID: 34488773 PMCID: PMC8422731 DOI: 10.1186/s12935-021-02179-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) induces breast cancer malignancy. Recent clinical and preclinical studies have demonstrated an association between overexpressed and activated STAT3 and breast cancer progression, proliferation, metastasis, and chemoresistance. Resveratrol (RES), a naturally occurring phytoalexin, has demonstrated anti-cancer activity in several disease models. Furthermore, RES has also been shown to regulate the STAT3 signaling cascade via its anti-oxidant and anti-inflammatory effects. In the present review, we describe the STAT3 cascade signaling pathway and address the therapeutic targeting of STAT3 by RES as a tool to mitigate breast cancer.
Collapse
Affiliation(s)
- Zeynab Kohandel
- Department of Biology, Faculty of Sciences, University of Tehran, Tehran, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
23
|
Yabaş E, Şahin-Bölükbaşı S, Şahin-İnan ZD. New water soluble magnesium phthalocyanine as a potential anticancer drug: Cytotoxic and apoptotic effect on different cancer cell lines. J PORPHYR PHTHALOCYA 2021. [DOI: 10.1142/s1088424621500863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although phthalocyanines are usually used a photosensitizers for photodynamic therapy, these works focus on the directly cytotoxic effect of a new water-soluble magnesium phthalocyanine. The new water-soluble magnesium phthalocyanine 2 was synthesized, characterized and investigated for cytotoxic and apoptotic activities. The cytotoxic activities of the compound 2 were determined by using (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cell viability assay on human breast cancer cells (MDA-MB-231, MCF-7), human prostate cancer cells (PC-3), and human healthy lung fibroblast cells (WI-38). The cells were plated and treated 1 to 20 [Formula: see text]M of different concentrations of the compound. MTT assay results indicated that the compound 2 has concentration and time-dependent cytotoxic activities against cancer cells. We also observed that the compound displayed lower toxicity against WI-38 healthy cells than cancer cells at 48 and 72 h. The compound showed a significant cytotoxic activity difference between breast cancer cells and WI-38 healthy cells at 48 and 72 h. Selectivity index of the compound 2 against MCF-7 for 72 h was calculated ¿ 15.62. We also studied the apoptotic and necrotic effect of compound 2using TUNEL and PI staining, respectively. It was found that the synthesized compound 2 increased apoptotic and necrotic cells.
Collapse
Affiliation(s)
- Ebru Yabaş
- Advanced Technology Application and Research Center, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| | - Serap Şahin-Bölükbaşı
- Department of Biochemistry, Faculty of Pharmacy, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| | - Zeynep Deniz Şahin-İnan
- Department of Histology & Embryology, Medicine Faculty, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| |
Collapse
|
24
|
Chae HJ, Kim GJ, Deshar B, Kim HJ, Shin MJ, Kwon H, Youn UJ, Nam JW, Kim SH, Choi H, Suh SS. Anticancer Activity of 2- O-caffeoyl Alphitolic Acid Extracted from the Lichen, Usnea barbata 2017-KL-10. Molecules 2021; 26:molecules26133937. [PMID: 34203232 PMCID: PMC8271524 DOI: 10.3390/molecules26133937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/21/2021] [Accepted: 06/25/2021] [Indexed: 11/16/2022] Open
Abstract
Colorectal cancer is one of the life-threatening ailments causing high mortality and morbidity worldwide. Despite the innovation in medical genetics, the prognosis for metastatic colorectal cancer in patients remains unsatisfactory. Recently, lichens have attracted the attention of researchers in the search for targets to fight against cancer. Lichens are considered mines of thousands of metabolites. Researchers have reported that lichen-derived metabolites demonstrated biological effects, such as anticancer, antiviral, anti-inflammatory, antibacterial, analgesic, antipyretic, antiproliferative, and cytotoxic, on various cell lines. However, the exploration of the biological activities of lichens' metabolites is limited. Thus, the main objective of our study was to evaluate the anticancer effect of secondary metabolites isolated from lichen (Usnea barbata 2017-KL-10) on the human colorectal cancer cell line HCT116. In this study, 2OCAA exhibited concentration-dependent anticancer activities by suppressing antiapoptotic genes, such as MCL-1, and inducing apoptotic genes, such as BAX, TP53, and CDKN1A(p21). Moreover, 2OCAA inhibited the migration and invasion of colorectal cancer cells in a concentration-dependent manner. Taken together, these data suggest that 2OCAA is a better therapeutic candidate for colorectal cancer.
Collapse
Affiliation(s)
- Hae-Jung Chae
- Department of Bioscience, Mokpo National University, Muan-gun, Joennam 58554, Korea; (H.-J.C.); (B.D.); (M.-J.S.)
| | - Geum-Jin Kim
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbukdo 38541, Korea; (G.-J.K.); (H.K.); (J.-W.N.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si, Gyeongsangbukdo 38541, Korea
| | - Barsha Deshar
- Department of Bioscience, Mokpo National University, Muan-gun, Joennam 58554, Korea; (H.-J.C.); (B.D.); (M.-J.S.)
| | - Hyun-Jin Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.-J.K.); (S.-H.K.)
| | - Min-Ji Shin
- Department of Bioscience, Mokpo National University, Muan-gun, Joennam 58554, Korea; (H.-J.C.); (B.D.); (M.-J.S.)
- Department of Biomedicine, Health & Life Convergence Science, BK21 Four, Mokpo National University, Joennam 58554, Korea
| | - Hyukbean Kwon
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbukdo 38541, Korea; (G.-J.K.); (H.K.); (J.-W.N.)
| | - Ui-Joung Youn
- Division of Life Sciences, Korean Polar Research Institute (KOPRI), Incheon 21990, Korea;
- Department of Polar Sciences, University of Science and Technology, Incheon 21990, Korea
| | - Joo-Won Nam
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbukdo 38541, Korea; (G.-J.K.); (H.K.); (J.-W.N.)
| | - Sung-Hak Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.-J.K.); (S.-H.K.)
| | - Hyukjae Choi
- College of Pharmacy, Yeungnam University, Gyeongsan-si, Gyeongsangbukdo 38541, Korea; (G.-J.K.); (H.K.); (J.-W.N.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si, Gyeongsangbukdo 38541, Korea
- Correspondence: (H.C.); (S.-S.S.); Tel.: +82-53-810-2824 (H.C.); +82-61-450-2346 (S.-S.S.)
| | - Sung-Suk Suh
- Department of Bioscience, Mokpo National University, Muan-gun, Joennam 58554, Korea; (H.-J.C.); (B.D.); (M.-J.S.)
- Department of Biomedicine, Health & Life Convergence Science, BK21 Four, Mokpo National University, Joennam 58554, Korea
- Correspondence: (H.C.); (S.-S.S.); Tel.: +82-53-810-2824 (H.C.); +82-61-450-2346 (S.-S.S.)
| |
Collapse
|
25
|
Chen M, Wu W, Liu D, Lv Y, Deng H, Gao S, Gu Y, Huang M, Guo X, Liu B, Zhao B, Pang Q. Evolution and Structure of API5 and Its Roles in Anti-Apoptosis. Protein Pept Lett 2021; 28:612-622. [PMID: 33319655 DOI: 10.2174/0929866527999201211195551] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/30/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
Apoptosis, also named programmed cell death, is a highly conserved physiological mechanism. Apoptosis plays crucial roles in many life processes, such as tissue development, organ formation, homeostasis maintenance, resistance against external aggression, and immune responses. Apoptosis is regulated by many genes, among which Apoptosis Inhibitor-5 (API5) is an effective inhibitor, though the structure of API5 is completely different from the other known Inhibitors of Apoptosis Proteins (IAPs). Due to its high expression in many types of tumors, API5 has received extensive attention, and may be an effective target for cancer treatment. In order to comprehensively and systematically understand the biological roles of API5, we summarized the evolution and structure of API5 and its roles in anti-apoptosis in this review.
Collapse
Affiliation(s)
- Meishan Chen
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Weiwei Wu
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Dongwu Liu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yanhua Lv
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Hongkuan Deng
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Sijia Gao
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yaqi Gu
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Mujie Huang
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Xiao Guo
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Baohua Liu
- Anti-Aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Qiuxiang Pang
- Laboratory of Developmental and Evolutionary Biology, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255049, China
| |
Collapse
|
26
|
Bakır E, Çal T, Aydın Dilsiz S, Canpınar H, Eken A, Ündeğer Bucurgat Ü. Assessment of the cytotoxic, genotoxic, and apoptotic potential of flurbiprofen in HeLa and HepG2 cell lines. J Biochem Mol Toxicol 2021; 35:1-11. [PMID: 33709623 DOI: 10.1002/jbt.22770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/14/2020] [Accepted: 03/02/2021] [Indexed: 11/07/2022]
Abstract
In the literature, the anticancer potential of flurbiprofen isn't fully understood. In this study, the cytotoxic, genotoxic, and apoptotic effects of flurbiprofen were evaluated in human cervical and liver cancer cells. Cytotoxicity was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and it was observed that cytotoxicity increased in a concentration- and time-dependent manner. Genotoxicity was determined using alkaline Comet assay. DNA damage increased in a concentration-dependent manner. Early apoptosis was evaluated using real-time polymerase chain reaction, and it was found that apoptotic gene levels increased while antiapoptotic gene levels decreased. Late apoptosis and cell cycle analyzes were determined using flow cytometry. No evidence of late apoptosis was observed, and no significant arrest was found in the cell cycle. In conclusion, it seems that flurbiprofen has a cytotoxic, genotoxic, and apoptotic effects in both human cancer cell lines. Moreover, the findings indicate that flurbiprofen is effective at the gene level and induces apoptosis with an intracellular pathway.
Collapse
Affiliation(s)
- Elçin Bakır
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Tuğbagül Çal
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sevtap Aydın Dilsiz
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Hande Canpınar
- Department of Basic Oncology, Institute of Cancer, Hacettepe University, Ankara, Turkey
| | - Ayşe Eken
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Ülkü Ündeğer Bucurgat
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
27
|
Sutiningsih D, Ginandjar P, Dian Saraswati L, Mubarika Haryana S. Anticancer Activity of Pasak Bumi Root Extract ( Eurycoma longifolia Jack) on Raji Cells. Pak J Biol Sci 2021; 24:1226-1235. [PMID: 34989200 DOI: 10.3923/pjbs.2021.1226.1235] [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] [Indexed: 06/14/2023]
Abstract
<b>Background and Objective:</b> The use of the roots of the pasak bumi (<i>E. longifolia</i> Jack) to treat cancer has been studied widely, however, the scientific basis of these plants used as an anticancer drug is widely unknown. The purpose of this study was to examine the anticancer activity of ethyl acetate and non-ethyl acetate fractions of pasak bumi roots in Raji cells. <b>Materials and Methods:</b> The cytotoxicity test is using the direct cell count method with trypan blue staining. The growth inhibition is using doubling time analysis of Raji cells. Observation of the apoptotic events of Raji cells used ethidium bromide staining, while observing the expression of p53 protein in Raji cells was done by immunohistochemical staining. <b>Results:</b> The results of the cytotoxicity and doubling time test showed that the activity of the non-ethyl acetate fraction was greater than that of the roots of pasak bumi. The lower concentration of non-ethyl acetate fraction of pasak bumi roots was able to delay the multiplication time of Raji cells which was greater than that of ethyl acetate. The results of the cytotoxicity and doubling time test showed that the activity of the non-ethyl acetate fraction was greater than that of the roots of pasak bumi. <b>Conclusion:</b> It can be concluded that the ethyl acetate and non-ethyl acetate fractions of the roots of pasak bumi have cytotoxic and antiproliferative activity on Raji cells, however they cannot induce apoptosis in Raji cells. The death of Raji cells is through the mechanism of inhibiting Raji cell proliferation as evidenced by an increase in p53 protein expression.
Collapse
|
28
|
Falode JA, Akinmoladun AC, Olaleye MT, Akindahunsi AA. Kigelia africana (Lam.) Benth leaf extract inhibits rat brain and liver mitochondrial membrane permeability transition pore opening. Drug Chem Toxicol 2020; 45:1614-1624. [PMID: 33280443 DOI: 10.1080/01480545.2020.1850757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The effect of Kigelia africana on mitochondrial membrane permeability transition has not been explored. In this study, the effect of a solvent fraction of Kigelia africana leaf extract on mitochondrial membrane permeability transition of rat brain and liver was evaluated. A methanol extract of K. africana leaves was fractionated into different solvents by vacuum liquid chromatography and following preliminary screening, the dichloromethane:ethylacetate (1:1) fraction was selected for further assays. Constituent phytochemicals in the fraction were revealed by thin-layer chromatography and further purification was carried out to characterize the compounds. Brain and liver mitochondria were isolated and used for mitochondrial membrane permeability transition and adenosine triphosphatase assays. Exogenous Ca2+ and Al3+ were used to trigger the mitochondrial membrane permeability transition opening. Physicochemical properties revealed by thin-layer chromatography showed that the isolated compounds were flavonoids. The extract inhibited mitochondrial membrane permeability transition opening in the presence and absence of triggering agents in brain and liver mitochondria. It also inhibited mitochondrial lipid peroxidation and adenosine triphosphatase activity. These results suggest that the extract can limit the rate of apoptosis via inhibition of mitochondrial membrane permeability transition which is pivotal to the mitochondrial apoptotic pathway and is an important therapeutic target in some pathological conditions.
Collapse
Affiliation(s)
- John Adeolu Falode
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, Federal University, Oye-Ekiti, Nigeria.,Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| | - Afolabi Clement Akinmoladun
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| | - Mary Tolulope Olaleye
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| | - Afolabi Akintunde Akindahunsi
- Phytomedicine, Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, School of Sciences, The Federal University of Technology, Akure, Nigeria
| |
Collapse
|
29
|
Zhu XQ, Yang H, Lin MH, Shang HX, Peng J, Chen WJ, Chen XZ, Lin JM. Qingjie Fuzheng Granules regulates cancer cell proliferation, apoptosis and tumor angiogenesis in colorectal cancer xenograft mice via Sonic Hedgehog pathway. J Gastrointest Oncol 2020; 11:1123-1134. [PMID: 33456987 PMCID: PMC7807284 DOI: 10.21037/jgo-20-213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/25/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Sonic Hedgehog (SHh) signaling pathway plays a critical role in cell proliferation, apoptosis, and tumor angiogenesis in various types of malignancies including colorectal cancer (CRC). Qingjie Fuzheng Granules (QFG) is a traditional Chinese medicinal formula, which has been clinically used in various cancer treatments, including CRC. In this study, we explored the potential molecular mechanisms of QFG treatment effects on CRC via the SHh pathway. METHODS A CRC HCT-116 xenograft mouse model was utilized for all experiments. Mice were treated with intra-gastric administration of 1 g/kg of QFG or saline 6 days a week for 28 days (4 weeks). Body weight, length and shortest diameter of the tumor were measured every 3 days. At the end of the treatment, the tumor weight was measured. TUNEL staining assays were used to detect tumor apoptosis. Western blot and immunohistochemistry (IHC) assays were used to detect the expression of relative proteins. RESULTS In our results, QFG inhibited the increase of tumor volume and weight, and exhibited no impact on mouse body weight. Furthermore, QFG significantly decreased the expression of SHh, Smo and Gli proteins, indicating the action of SHh signaling. Consequently, the expression of pro-proliferative survivin, Ki-67, Cyclin-D1 and CDK4 were decreased and expression of anti-proliferative p21 was increased. The pro-apoptotic Bax/Bcl-2 ratio, cle-caspase-3 and TUNEL-positive cell percentage in tumor tissues were increased. Meanwhile, the pro-angiogenic VEGF-A and VEGFR-2 expression was down-regulated. CONCLUSIONS QFG inhibited CRC cell proliferation and promoted CRC cell apoptosis and tumor angiogenesis in vivo through the suppression of SHh pathway, suggesting that QFG could be a potential therapeutic drug for CRC.
Collapse
Affiliation(s)
- Xiao-Qin Zhu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Hong Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Ming-He Lin
- Editorial Department of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hai-Xia Shang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Wu-Jin Chen
- Department of Oncology, Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xu-Zheng Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Jiu-Mao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| |
Collapse
|
30
|
Vo PHT, Nguyen TDT, Tran HT, Nguyen YN, Doan MT, Nguyen PH, Lien GTK, To DC, Tran MH. Cytotoxic components from the leaves of Erythrophleum fordii induce human acute leukemia cell apoptosis through caspase 3 activation and PARP cleavage. Bioorg Med Chem Lett 2020; 31:127673. [PMID: 33161122 DOI: 10.1016/j.bmcl.2020.127673] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/26/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022]
Abstract
Cassaine diterpenoids as erythrofordins A-C (1-3), pseudo-erythrosuamin (4), and erythrofordin U (5) isolated from the leaves of Vietnamese Erythrophleum fordii Oliver were tested cytotoxic activity against human leukemia cancer cells. The results showed that these metabolites exhibited dose-dependent cytotoxicity against human leukemia HL-60 and KG cells with IC50 values ranging from 15.2 ± 1.5 to 42.2 ± 3.6 µM. Treatment with erythrofordin B led to the apoptosis of HL-60 and KG cells due to the activation of caspase 3, caspase 9, and poly (ADP-ribose) polymerase (PARP). Erythrofordin B significantly increased Bak protein expression, but downregulated the anti-apoptotic protein Bcl-2, in HL-60 cells. In silico results demonstrated that erythrofordin B can bind to both the procaspase-3 allosteric site and the PARP-1 active site, with binding energies of -7.36 and -10.76 kcal/mol, respectively. These results indicated that the leaves of Vietnamese E. fordii, which contain cassaine diterpenoids, can induce the apoptosis of human leukemia cancer cells.
Collapse
Affiliation(s)
- Phuong Hien Thi Vo
- University of Science, Vietnam National University Hochiminh City, 227 Nguyen Van Cu, District 5, Hochiminh City 748000, Viet Nam
| | - Thuy Duong Thi Nguyen
- Biomedical Science Department, VNUK Institute for Research & Executive Education, The University of Danang, 158A Le Loi, Hai Chau District, Danang City 551000, Viet Nam
| | - Hoa Thanh Tran
- Biomedical Science Department, VNUK Institute for Research & Executive Education, The University of Danang, 158A Le Loi, Hai Chau District, Danang City 551000, Viet Nam
| | - Yen Nhi Nguyen
- University of Science, Vietnam National University Hochiminh City, 227 Nguyen Van Cu, District 5, Hochiminh City 748000, Viet Nam
| | - Minh Thu Doan
- Biomedical Science Department, VNUK Institute for Research & Executive Education, The University of Danang, 158A Le Loi, Hai Chau District, Danang City 551000, Viet Nam
| | - Phi Hung Nguyen
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay District, Hanoi 122100, Viet Nam
| | - Giang Thi Kim Lien
- The University of Danang, 41 Le Duan, Hai Chau District, Danang City 551000, Viet Nam
| | - Dao Cuong To
- Faculty of Pharmacy, Phenikaa University, Yen Nghia, Ha Dong District, Hanoi 12116, Viet Nam; Phenikaa Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, 167 Hoang Ngan, Cau Giay District, Hanoi 11313, Viet Nam.
| | - Manh Hung Tran
- University of Science, Vietnam National University Hochiminh City, 227 Nguyen Van Cu, District 5, Hochiminh City 748000, Viet Nam.
| |
Collapse
|
31
|
Li ZW, Zhong CY, Wang XR, Li SN, Pan CY, Wang X, Sun XY. Synthesis and Evaluation of the Antitumor Activity of Novel 1-(4-Substituted phenyl)-2-ethyl Imidazole Apoptosis Inducers In Vitro. Molecules 2020; 25:E4293. [PMID: 32962127 PMCID: PMC7570620 DOI: 10.3390/molecules25184293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 01/10/2023] Open
Abstract
Novel imidazole derivatives were designed, prepared, and evaluated in vitro for antitumor activity. The majority of the tested derivatives showed improved antiproliferative activity compared to the positive control drugs 5-FU and MTX. Among them, compound 4f exhibited outstanding antiproliferative activity against three cancer cell lines and was considerably more potent than both 5-FU and MTX. In particular, the selectivity index indicated that the tolerance of normal L-02 cells to 4f was 23-46-fold higher than that of tumor cells. This selectivity was significantly higher than that exhibited by the positive control drugs. Furthermore, compound 4f induced cell apoptosis by increasing the protein expression levels of Bax and decreasing those of Bcl-2 in a time-dependent manner. Therefore, 4f could be a potential candidate for the development of a novel antitumor agent.
Collapse
Affiliation(s)
- Zhen-Wang Li
- College of Animal Science and Technique, Heilongjiang Bayi Agriculture University, Daqing 163319, Heilongjiang, China; (Z.-W.L.); (C.-Y.Z.); (X.-R.W.); (S.-N.L.); (C.-Y.P.); (X.W.)
| | - Chun-Yan Zhong
- College of Animal Science and Technique, Heilongjiang Bayi Agriculture University, Daqing 163319, Heilongjiang, China; (Z.-W.L.); (C.-Y.Z.); (X.-R.W.); (S.-N.L.); (C.-Y.P.); (X.W.)
| | - Xiao-Ran Wang
- College of Animal Science and Technique, Heilongjiang Bayi Agriculture University, Daqing 163319, Heilongjiang, China; (Z.-W.L.); (C.-Y.Z.); (X.-R.W.); (S.-N.L.); (C.-Y.P.); (X.W.)
| | - Shi-Nian Li
- College of Animal Science and Technique, Heilongjiang Bayi Agriculture University, Daqing 163319, Heilongjiang, China; (Z.-W.L.); (C.-Y.Z.); (X.-R.W.); (S.-N.L.); (C.-Y.P.); (X.W.)
| | - Chun-Yuan Pan
- College of Animal Science and Technique, Heilongjiang Bayi Agriculture University, Daqing 163319, Heilongjiang, China; (Z.-W.L.); (C.-Y.Z.); (X.-R.W.); (S.-N.L.); (C.-Y.P.); (X.W.)
| | - Xin Wang
- College of Animal Science and Technique, Heilongjiang Bayi Agriculture University, Daqing 163319, Heilongjiang, China; (Z.-W.L.); (C.-Y.Z.); (X.-R.W.); (S.-N.L.); (C.-Y.P.); (X.W.)
| | - Xian-Yu Sun
- College of Animal Science and Technique, Heilongjiang Bayi Agriculture University, Daqing 163319, Heilongjiang, China; (Z.-W.L.); (C.-Y.Z.); (X.-R.W.); (S.-N.L.); (C.-Y.P.); (X.W.)
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu 215500, Jiangsu, China
| |
Collapse
|
32
|
Retracted
: Syringic acid suppresses oral squamous cell carcinoma SCC131 cell proliferation via modulation of mitochondria‐mediated apoptosis signaling pathways. J Biochem Mol Toxicol 2020; 34:e22586. [DOI: 10.1002/jbt.22586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/28/2020] [Accepted: 07/14/2020] [Indexed: 11/07/2022]
|
33
|
Yang HL, Kuo YT, Vudhya Gowrisankar Y, Lin KY, Hsu LS, Huang PJ, Lin HC, Hseu YC. The Leaf Extracts of Toona sinensis and Fermented Culture Broths of Antrodia camphorata Synergistically Cause Apoptotic Cell Death in Promyelocytic Leukemia Cells. Integr Cancer Ther 2020; 19:1534735420923734. [PMID: 32618215 PMCID: PMC7336824 DOI: 10.1177/1534735420923734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Toona sinensis is a common edible vegetable that is used in
certain Chinese dishes and has importance in folk medicine. The leaf extracts of
T sinensis possess and exhibit anticancer efficacy against
various cancer cell types. In Taiwanese folklore, Antrodia
camphorata, also known as “Niu-Cheng-Zi,” is used in traditional
medicine to treat various illnesses. Its fruit and mycelium possess various
potent antiproliferative properties. Two studies from our group have reported
that T sinensis or A camphorata has the
ability to cause apoptosis in various cancer cells. Conversely, underlying
molecular mechanisms and any beneficial effects remain unknown. This study shows
anticancer efficacy for both T sinensis and A
camphorata co-treatments that target HL-60 cells. The combination
index values indicate that 40 µg/mL of T sinensis and 25 µg/mL
of A camphorata as a combined treatment shows a synergetic
effect, which reduces HL-60 cell proliferation. Alternately, this treatment
exhibited no cytotoxic effects for human umbilical vein endothelial cells.
Western blot data showed that T sinensis and A
camphorata as a combined treatment result in augmented expression
of apoptosis, cytochrome c release, Bcl-2 inhibition, expression of Bax, Fas,
and FasL, as well as the cleavage of Bid in HL-60 cells. Moreover, this combined
treatment overshadowed monotherapy in its ability to inhibit uPAR, MMP-9, MMP-2,
COX-2 expression, and PGE2 secretions. Our study strongly implies
that this combined treatment offers more beneficial effects to suppress and
treat leukemia due to apoptosis-mediated cell inhibition. Further in
vivo studies related to the combined treatment could establish its
future potential.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Ya-Ting Kuo
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | | | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Li-Sung Hsu
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Jane Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Hui-Chang Lin
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, School of Pharmacy, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Research Center of Chinese Herbal Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
34
|
Trametes robiniophila Murr in the treatment of breast cancer. Biomed Pharmacother 2020; 128:110254. [PMID: 32480220 DOI: 10.1016/j.biopha.2020.110254] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/04/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the leading cause of cancer death among women across the world. Trametes robiniophila Murr (Huaier), a traditional herbal medicine, has been used in China to protect human health for about 1600 years. Recent years, Huaier had been proven to be effective for multiple types of malignancies. This systematic review focused on breast cancer treatment, summarizing the curative function of Huaier aqueous extract and polysaccharides in preclinical researches. Huaier could markedly inhibit breast cancer progression with low toxicity, enhance immune response and increase the sensitivity to radiation and chemotherapy. The therapeutic effect of Huaier granule in clinical studies was also included. This review amalgamated the current studies and highlighted the promising role of Huaier and its polysaccharides as complementary alternative medicine in breast cancer treatment.
Collapse
|
35
|
Ma F, Gu X, Liu JQ, Mo LH, Yang G, Geng XR, Liu ZQ, Liu ZG, Yang PC. Inhibition of livin overcomes radioresistance in nasopharyngeal carcinoma cells. PLoS One 2020; 15:e0229272. [PMID: 32119704 PMCID: PMC7051067 DOI: 10.1371/journal.pone.0229272] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 02/03/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND AIMS Radiotherapy is one of the major remedies for the treatment of cancer, including nasopharyngeal carcinoma (NPC). Radioresistance occurs very often in target cells that is a large drawback in cancer treated with radiotherapy. Livin involves the over-growth of cancer cells. This study aims to investigate the role of livin in the radioresistance formation in NPC cells. METHODS NPC cell lines were exposed to small doses of irradiation to establish a cell model of radioresistance, in which the role of livin in the development of radioresistance was evaluated. RESULTS The expression of livin was observed in NPC cells, which was significantly increased after exposing to small doses of irradiation. A negative correlation was detected between livin and Fas expression in NPC cells. Livin formed a complex with heat shock factor-1 (HSF1, the transcription factor of Fas) in NPC cells after irradiation, which sped up ubiquitination of HSF1. Livin was involved in suppressing Fas expression in NPC cells with radioresistance. Exposure to livin inhibitors prevented radioresistance development and overcame the established radioresistance in NPC cells. CONCLUSIONS Livin expression in NPC cells plays a critical role in the development of radioresistance. Depletion of livin increases the sensitiveness of NPC cells to irradiation. Target therapy against livin may have the translational potential for the treatment of NPC.
Collapse
Affiliation(s)
- Fei Ma
- Department of Traditional Chinese Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Xia Gu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Respirology, Affiliated Hospital of Xinan Medical University, Luzhou, China
| | - Jiang-Qi Liu
- Department of Allergy, Longgang ENT Hospital & Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, China
| | - Li-Hua Mo
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Gui Yang
- Department of Otolaryngology, Longgang Central Hospital, Shenzhen, China
| | - Xiao-Rui Geng
- Department of Allergy, Longgang ENT Hospital & Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, China
| | - Zhi-Qiang Liu
- Department of Allergy, Longgang ENT Hospital & Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, China
| | - Zhi-Gang Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Ping-Chang Yang
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
| |
Collapse
|
36
|
Adil Shareef M, Ganapathi T, Khan I, Rani S, Rajanna A, Akbar S, Kumar CG, Babu BN. New Indolyl‐Arylaminopropenone Conjugates: Synthesis, Cytotoxicity and Apoptotic Inducing Studies. ChemistrySelect 2020. [DOI: 10.1002/slct.201904077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Mohd Adil Shareef
- Department of Fluoro-Agrochemicals CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Academy of Scientific and Innovative Research Ghaziabad 201002 India
| | - Thipparapu Ganapathi
- Stem Cell Research Division National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Hyderabad- 500007 Telangana India
| | - Irfan Khan
- Academy of Scientific and Innovative Research Ghaziabad 201002 India
- Organic Synthesis and Process Chemistry Division CSIR-Indian Institute of Chemical Technology Tarnaka, Hyderabad 500007 India
| | - Sunitha Rani
- Organic Synthesis and Process Chemistry Division CSIR-Indian Institute of Chemical Technology Tarnaka, Hyderabad 500007 India
| | - Ajumeera Rajanna
- Stem Cell Research Division National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Hyderabad- 500007 Telangana India
| | - Syed Akbar
- Department of Fluoro-Agrochemicals CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
| | - C. Ganesh Kumar
- Academy of Scientific and Innovative Research Ghaziabad 201002 India
- Organic Synthesis and Process Chemistry Division CSIR-Indian Institute of Chemical Technology Tarnaka, Hyderabad 500007 India
| | - Bathini Nagendra Babu
- Department of Fluoro-Agrochemicals CSIR-Indian Institute of Chemical Technology Hyderabad 500 007 India
- Academy of Scientific and Innovative Research Ghaziabad 201002 India
| |
Collapse
|
37
|
Enabling Combinatorial siRNA Delivery against Apoptosis-Related Proteins with Linoleic Acid and α-Linoleic Acid Substituted Low Molecular Weight Polyethylenimines. Pharm Res 2020; 37:46. [PMID: 32016611 DOI: 10.1007/s11095-020-2770-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Short interfering RNA (siRNA) therapy promises a new era in treatment of breast cancers but effective delivery systems are needed for clinical use. Since silencing complementary targets may offer improved efficacy, this study was undertaken to identify non-viral carriers for combinatorial siRNA delivery for more effective therapy. METHODS A library of lipid-substituted polymers from low molecular weight polyethyleneimine (PEI), linoleic acid (LA) and α-linoleic acid (αLA) with amide or thioester linkages was prepared and investigated for delivering Mcl-1, survivin and STAT5A siRNAs in breast cancer cells. RESULTS The effective polymers formed 80-190 nm particles with similar zeta-potentials, but the serum stability was greater for complexes formed with amide-linked lipid conjugates. The LA and αLA substitutions, with the low molecular weight PEI (1.2 kDa and 2.0 kDa) were able to deliver siRNA effectively to cells and retarded the growth of breast cancer cells. The amide-linked lipid substituents showed higher cellular delivery of siRNA as compared to thioester linkages. Upon combinational delivery of siRNAs, growth of MCF-7 cells was inhibited to a greater extent with 2.0PEI-LA9 mediated delivery of Mcl-1 combined survivin siRNAs as compared to individual siRNAs. The qRT-PCR analysis confirmed the decrease in mRNA levels of target genes with specific siRNAs and 2.0PEI-LA9 was the most effective polymer for delivering siRNAs (either single or in combination). CONCLUSIONS This study yielded effective siRNA carriers for combinational delivery of siRNAs. Careful choice of siRNA combinations will be critical since targeting individual genes might alter the expression of other critical mediators.
Collapse
|
38
|
Rezaieseresht H, Shobeiri SS, Kaskani A. Chenopodium Botrys Essential Oil as A Source of Sesquiterpenes to Induce Apoptosis and G1 Cell Cycle Arrest in Cervical Cancer Cells. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:341-351. [PMID: 33224241 PMCID: PMC7667537 DOI: 10.22037/ijpr.2019.1100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Conducting cell apoptosis pathways is a novel strategy in cancer treatment. This study aimed to explain that C. botrys essential oil could induce apoptosis and arrest the cell cycle in HeLa cells. Cytotoxic and apoptogenic effects of the essential oil of Jerusalem-oak (Chenopodium botrys L.), which was obtained from the aerial parts of the plant, were evaluated in HeLa cells. Cell viability was assessed by MTT and LDH assays, and the mechanism of cell apoptosis was investigated using flow cytometry. Expression of the apoptosis-related genes was assessed using real-time polymerase chain reaction (PCR). GC-MS analysis of the herbal essential oil revealed 37 components. The major components were α-Eudesmol (16.81%), Elemol acetate (13.2%), Elemol (9.0%), and α-Chenopodiol-6-acetate (7.9%). The essential oil inhibited the growth of HeLa cells and increased the expression of p21 and p53. In addition, essential oil treatment increased the sub-G1 DNA content and induced apoptosis due to the increased Bax/Bcl-2 ratio and up-regulation of caspase-3 gene expression. According to the results, C. botrys essential oil exhibited anticancer effects through intrinsic apoptosis pathways and arresting cell proliferation.
Collapse
Affiliation(s)
- Hasan Rezaieseresht
- Traditional and Complementary Medicine Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| | - Saeideh Sadat Shobeiri
- Cellular and Molecular Research center, Sabzevar University of Medical Sciences, Sabzevar, Iran.
- Immunology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Arezou Kaskani
- Department of Chemistry, Sabzevar Branch, Islamic Azad University, Sabzevar, Iran.
| |
Collapse
|
39
|
Wang J, Li X, Li Y, Yao X. Girdin knockdown promotes apoptosis in colorectal cancer via miR-29c-3p/Girdin axis. Transl Cancer Res 2019; 8:2906-2915. [PMID: 35117048 PMCID: PMC8797721 DOI: 10.21037/tcr.2019.12.16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/27/2019] [Indexed: 11/06/2022]
Abstract
Background In recent years, the incidence and mortality of colorectal cancer (CRC) have increased year by year among young people. Increased levels of Girdin expression predict a poor prognosis of CRC, which presents a serious threat to human health globally. Herein, we investigated the role of Girdin in CRC and explored the underlying mechanisms in CRC. Methods The expression of Girdin was detected in human specimens. HCT116 cells with stably expressing or knock-out Girdin protein were successfully constructed to observe the biological function of gene. Protein expression was determined by immunohistochemistry, immunofluorescence and western blot. Results Clinically, overexpression of Girdin was observed in the tumor tissue and poor prognosis of CRC patients. Gain-of-function and loss-of-function assays showed that Girdin promoted CRC cell proliferation in vitro. Mechanistically, Girdin knock-down significantly enhanced apoptosis, the mitochondrial membrane potential dropped, and the reactive oxygen species increased greatly. Last but not least, we analyzed the TargetScan dataset and found that Girdin was a regulated target of hsa-miR-29c-3p in CRC. Luciferase reporter assay was used to verify the interaction between hsa-miR-29c-3p and the 3’UTR of Girdin. Conclusions Our findings suggest that Girdin has a crucial role in CRC progression via miR-29c-3p/Girdin axis, highlighting Girdin as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Junjiang Wang
- Department of General Surgery, The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.,Department of General Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xiaowu Li
- Department of General Surgery, The First Affiliated Hospital & School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Yong Li
- Department of General Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xueqing Yao
- Department of General Surgery, The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.,Department of General Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
40
|
Choi EO, Lee H, HwangBo H, Kwon DH, Kim MY, Ji SY, Hong SH, Kim GY, Park C, Hwang HJ, Moon SK, Yun SJ, Kim WJ, Choi YH. Citrus unshiu peel suppress the metastatic potential of murine melanoma B16F10 cells in vitro and in vivo. Phytother Res 2019; 33:3228-3241. [PMID: 31486124 PMCID: PMC6916627 DOI: 10.1002/ptr.6497] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/30/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022]
Abstract
The peel of Citrus unshiu Marcow. fruits (CU) has long been used as a traditional medicine that has therapeutic effects against pathogenic diseases, including asthma, vomiting, dyspepsia, blood circulation disorders, and various types of cancer. In this study, we investigated the effect of CU peel on metastatic melanoma, a highly aggressive skin cancer, in B16F10 melanoma cells, and in B16F10 cells inoculated‐C57BL/6 mice. Our results show that ethanol extracts of CU (EECU) inhibited cell growth and increased the apoptotic cells in B16F10 cells. EECU also stimulated the induction of mitochondria‐mediated intrinsic pathway, with reduced mitochondrial membrane potential and increased generation of intracellular reactive oxygen species. Furthermore, EECU suppressed the migration, invasion, and colony formation of B16F10 cells. In addition, the oral administration of EECU reduced serum lactate dehydrogenase activity without weight loss, hepatotoxicity, nor nephrotoxicity in B16F10 cell‐inoculated mice. Moreover, EECU markedly suppressed lung hypertrophy, the number and expression of metastatic tumor nodules, and the expression of inflammatory tumor necrosis factor‐alpha in lung tissue. In conclusion, our findings suggest that the inhibitory effect of EECU on the metastasis of melanoma indicates that it may be regarded as a potential therapeutic herbal drug for melanoma.
Collapse
Affiliation(s)
- Eun Ok Choi
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| | - Hyun HwangBo
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| | - Da Hye Kwon
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| | - Su Hyun Hong
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dong-eui University, Busan, Republic of Korea
| | - Hye-Jin Hwang
- Department of Food and Nutrition, College of Nursing, Healthcare Sciences & Human Ecology, Dong-eui University, Busan, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, College of Biotechnology & Natural Resource, Chung-Ang University, Anseong, Republic of Korea
| | - Seok-Joong Yun
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Wun-Jae Kim
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Republic of Korea
| |
Collapse
|
41
|
Nirmala MJ, Durai L, Gopakumar V, Nagarajan R. Anticancer and antibacterial effects of a clove bud essential oil-based nanoscale emulsion system. Int J Nanomedicine 2019; 14:6439-6450. [PMID: 31496696 PMCID: PMC6697666 DOI: 10.2147/ijn.s211047] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 07/09/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE The essential oil derived from clove buds (Syzygium aromaticum) has been used as a chemopreventive agent in Ayurvedic medicine. The antiviral, antibacterial, and anticancer properties of its chemo-skeleton have motivated this study to explore its efficacy in pharmaceutics. METHODS Nanoscale-based emulsions were prepared by employing a spontaneous emulsification technique through self-assembly using varying concentrations of Tween 20 and Tween 80 surfactants. Their physicochemical properties and stability were studied in order to choose an optimum formulation which was clear and stable. The cytotoxicity of the stable oil-based emulsion system was evaluated using MTT assay, colony formation assay, and Annexin V-FITC assay against the thyroid cancer cell line (HTh-7). RESULTS All three methods verified apoptosis and reduction in cancer cell proliferation, making the formulation a promising candidate as an alternative cancer drug. The oil-based emulsion system was also tested for its antibacterial properties against Staphylococcus aureus. Membrane permeability studies proved its efficacy to permeate through cell membrane, thereby increasing the leakage of cytoplasmic contents. CONCLUSION Many current treatments for cancers are aggressive yet ineffective. This study positions the clove bud-based nanoscale emulsion as a suitable candidate for further in vivo studies and trials as a cancer drug.
Collapse
Affiliation(s)
- M Joyce Nirmala
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai600036, Tamil Nadu, India
| | - Latha Durai
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai600036, Tamil Nadu, India
| | - Vineet Gopakumar
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai600036, Tamil Nadu, India
| | - R Nagarajan
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai600036, Tamil Nadu, India
| |
Collapse
|
42
|
Dong Z, Abbas MN, Kausar S, Yang J, Li L, Tan L, Cui H. Biological Functions and Molecular Mechanisms of Antibiotic Tigecycline in the Treatment of Cancers. Int J Mol Sci 2019; 20:ijms20143577. [PMID: 31336613 PMCID: PMC6678986 DOI: 10.3390/ijms20143577] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/14/2022] Open
Abstract
As an FDA-approved drug, glycylcycline tigecycline has been used to treat complicated microbial infections. However, recent studies in multiple hematologic and malignant solid tumors reveal that tigecycline treatment induces cell cycle arrest, apoptosis, autophagy and oxidative stress. In addition, tigecycline also inhibits mitochondrial oxidative phosphorylation, cell proliferation, migration, invasion and angiogenesis. Importantly, combinations of tigecycline with chemotherapeutic or targeted drugs such as venetoclax, doxorubicin, vincristine, paclitaxel, cisplatin, and imatinib, have shown to be promising strategies for cancer treatment. Mechanism of action studies reveal that tigecycline leads to the inhibition of mitochondrial translation possibly through interacting with mitochondrial ribosome. Meanwhile, this drug also interferes with several other cell pathways/targets including MYC, HIFs, PI3K/AKT or AMPK-mediated mTOR, cytoplasmic p21 CIP1/Waf1, and Wnt/β-catenin signaling. These evidences indicate that antibiotic tigecycline is a promising drug for cancer treatment alone or in combination with other anticancer drugs. This review summarizes the biological function of tigecycline in the treatment of tumors and comprehensively discusses its mode of action.
Collapse
Affiliation(s)
- Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Jie Yang
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Lin Li
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Li Tan
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing 400716, China.
- Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing 400716, China.
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing 400716, China.
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing 400716, China.
| |
Collapse
|
43
|
In vitro antimetastatic activity of Agarwood (Aquilaria crassna) essential oils against pancreatic cancer cells. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2015.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
44
|
Ganga Reddy V, Srinivasa Reddy T, Privér SH, Bai Y, Mishra S, Wlodkowic D, Mirzadeh N, Bhargava S. Synthesis of Gold(I) Complexes Containing Cinnamide: In Vitro Evaluation of Anticancer Activity in 2D and 3D Spheroidal Models of Melanoma and In Vivo Angiogenesis. Inorg Chem 2019; 58:5988-5999. [DOI: 10.1021/acs.inorgchem.9b00281] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- V. Ganga Reddy
- Centre for Advanced Materials & Industrial Chemistry, School of Science, RMIT University, G.P.O. Box 2476, Melbourne 3001, Australia
| | - T. Srinivasa Reddy
- Centre for Advanced Materials & Industrial Chemistry, School of Science, RMIT University, G.P.O. Box 2476, Melbourne 3001, Australia
| | - Steven H. Privér
- Centre for Advanced Materials & Industrial Chemistry, School of Science, RMIT University, G.P.O. Box 2476, Melbourne 3001, Australia
| | - Yutao Bai
- Phenomics Laboratory, School of Science, RMIT University, Plenty Road, P.O. Box 71, Bundoora, Victoria 3083, Australia
| | - Shweta Mishra
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Parisar, Indore, Madhya Pradesh 452 001, India
| | - Donald Wlodkowic
- Phenomics Laboratory, School of Science, RMIT University, Plenty Road, P.O. Box 71, Bundoora, Victoria 3083, Australia
| | - Nedaossadat Mirzadeh
- Centre for Advanced Materials & Industrial Chemistry, School of Science, RMIT University, G.P.O. Box 2476, Melbourne 3001, Australia
| | - Suresh Bhargava
- Centre for Advanced Materials & Industrial Chemistry, School of Science, RMIT University, G.P.O. Box 2476, Melbourne 3001, Australia
| |
Collapse
|
45
|
Wang Z, Li C, Xing R, Shao Y, Zhao X, Zhang W, Guo M. β-Integrin mediates LPS-induced coelomocyte apoptosis in sea cucumber Apostichopus japonicus via the integrin/FAK/caspase-3 signaling pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 91:26-36. [PMID: 30339873 DOI: 10.1016/j.dci.2018.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/13/2018] [Accepted: 10/15/2018] [Indexed: 06/08/2023]
Abstract
Lipopolysaccharides (LPS) can induce the apoptosis of coelomocytes in Apostichopus japonicus (A. japonicus), and β-integrin serves as an apoptotic inhibitor during this process. However, the underlying mechanism in invertebrates is largely unknown. Integrin/focal adhesion kinase (FAK) signaling pathway modulates the apoptosis in vertebrates. In this study, a novel FAK was identified from A. japonicus (designated as AjFAK) by β-integrin (designated as AjITGB) -mediated GST-pull down assay. This interaction was further validated in the LPS-exposed coelomocytes through co-immunoprecipitation and immunofluorescence analyses. To investigate the functional role of AjFAK in AjITGB-mediated coelomocyte apoptosis, we cloned the full-length cDNA of AjFAK and characterized its relationship with AjITGB through real-time PCR. The mRNA expression levels of AjFAK exhibited consistent expression patterns with those of AjITGB in our previous work with 0.48- and 0.22-fold decreases at 12 and 96 h in LPS-exposed coelomocytes and in Vibrio splendidus challenged sea cucumber, respectively. Moreover, the expression level of AjFAK decreased to 0.35-fold in AjITGB knockdown treatment by specific small interference RNA (siRNA). We further performed an assay for the apoptotic rate of coelomocytes in AjITGB, AjFAK, and AjITGB/AjFAK silencing conditions and found that their apoptotic percentages increased to 26%, 25%, and 30%, respectively, compared with those of the control. Finally, the expression levels of four caspases from A. japonicus were also investigated to determine the apoptotic effector. After AjITGB or AjFAK was silenced, the mRNA levels of caspase-3 were 6.6-fold and 2.5-fold higher than those of the control, respectively. In addition, the enzymatic activity of caspase-3 was enhanced to 1.82- and 1.79-fold that of the control in the two groups. However, no significant changes were detected in caspase-2/6/8. All our results supported that β-integrin mediated the LPS-induced coelomocyte apoptosis in sea cucumber via the integrin/FAK/caspase-3 signaling pathway.
Collapse
Affiliation(s)
- Zhenhui Wang
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China; College of Life Sciences, Yantai University, Yantai, 264005, PR China.
| | - Ronglian Xing
- College of Life Sciences, Yantai University, Yantai, 264005, PR China
| | - Yina Shao
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Xuelin Zhao
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Ming Guo
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| |
Collapse
|
46
|
Mortezaee K, Salehi E, Mirtavoos-Mahyari H, Motevaseli E, Najafi M, Farhood B, Rosengren RJ, Sahebkar A. Mechanisms of apoptosis modulation by curcumin: Implications for cancer therapy. J Cell Physiol 2019; 234:12537-12550. [PMID: 30623450 DOI: 10.1002/jcp.28122] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Cancer incidences are growing and cause millions of deaths worldwide. Cancer therapy is one of the most important challenges in medicine. Improving therapeutic outcomes from cancer therapy is necessary for increasing patients' survival and quality of life. Adjuvant therapy using various types of antibodies or immunomodulatory agents has suggested modulating tumor response. Resistance to apoptosis is the main reason for radioresistance and chemoresistance of most of the cancers, and also one of the pivotal targets for improving cancer therapy is the modulation of apoptosis signaling pathways. Apoptosis can be induced by intrinsic or extrinsic pathways via stimulation of several targets, such as membrane receptors of tumor necrosis factor-α and transforming growth factor-β, and also mitochondria. Curcumin is a naturally derived agent that induces apoptosis in a variety of different tumor cell lines. Curcumin also activates redox reactions within cells inducing reactive oxygen species (ROS) production that leads to the upregulation of apoptosis receptors on the tumor cell membrane. Curcumin can also upregulate the expression and activity of p53 that inhibits tumor cell proliferation and increases apoptosis. Furthermore, curcumin has a potent inhibitory effect on the activity of NF-κB and COX-2, which are involved in the overexpression of antiapoptosis genes such as Bcl-2. It can also attenuate the regulation of antiapoptosis PI3K signaling and increase the expression of MAPKs to induce endogenous production of ROS. In this paper, we aimed to review the molecular mechanisms of curcumin-induced apoptosis in cancer cells. This action of curcumin could be applicable for use as an adjuvant in combination with other modalities of cancer therapy including radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ensieh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanifeh Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
47
|
Park BS, Choi NE, Lee JH, Kang HM, Yu SB, Kim HJ, Kang HK, Kim IR. Crosstalk between Fisetin-induced Apoptosis and Autophagy in Human Oral Squamous Cell Carcinoma. J Cancer 2019; 10:138-146. [PMID: 30662534 PMCID: PMC6329852 DOI: 10.7150/jca.28500] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/27/2018] [Indexed: 12/19/2022] Open
Abstract
Fisetin (3,3-,4-,7-tetrahydroxyflavone), a naturally occurring flavonoid, has antioxidant, anti-inflammatory, and anticancer effects. Oral squamous cell carcinoma (OSCC) has a 5-year survival rate lower than that of most other carcinomas, and can create functional and aesthetic problems for the patient. New therapies for OSCC are necessary, and treatment using plant-derived natural substances has recently become a trend. It has been suggested that autophagy may play an important role in cancer therapy. Several studies demonstrated that autophagy inhibition enhances apoptotic cell death. Therefore, autophagy inhibition might be a promising therapeutic method against OSCC. Our results showed that fisetin induced apoptotic cell death in human tongue squamous cell line Ca9-22 could be enhanced by inhibition of autophagy. Thus, autophagy process in fisetin treated OSCC might presumed to play a role of pro-survival. The combination of fisetin and an effective autophagy inhibitor could be a potentially adjuvant and useful treatment for oral cancer.
Collapse
Affiliation(s)
- Bong-Soo Park
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,BK21 PLUS Project, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,Institute of Translational Dental Sciences, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Nak-Eun Choi
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Ji Hye Lee
- Department of Oral Pathology, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,BK21 PLUS Project, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,Institute of Translational Dental Sciences, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Hae-Mi Kang
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,BK21 PLUS Project, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Su-Bin Yu
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Hye-Jin Kim
- Department of Dental Hygiene, Dongeui University, Gaya 1-dong, Busanjin-gu, Busan, 47230, South Korea
| | - Hyun-Kyung Kang
- Department of Dental Hygiene, Silla University, 140 Baekyang-daero 700 beon-gil, Busan 46958, South Korea
| | - In-Ryoung Kim
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,Institute of Translational Dental Sciences, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| |
Collapse
|
48
|
Thi Ngo QM, Cao TQ, Woo MH, Min BS. Cytotoxic Lactones from the Pericarps of Litsea japonica. ACTA ACUST UNITED AC 2019. [DOI: 10.20307/nps.2019.25.1.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Quynh-Mai Thi Ngo
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk 38430, Republic of Korea
| | - Thao Quyen Cao
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk 38430, Republic of Korea
| | - Mi Hee Woo
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk 38430, Republic of Korea
| | - Byung Sun Min
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk 38430, Republic of Korea
| |
Collapse
|
49
|
Tran WT, Childs C, Probst H, Farhat G, Czarnota GJ. Imaging Biomarkers for Precision Medicine in Locally Advanced Breast Cancer. J Med Imaging Radiat Sci 2018; 49:342-351. [DOI: 10.1016/j.jmir.2017.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022]
|
50
|
Cerium Oxide Nanoparticles Sensitize Pancreatic Cancer to Radiation Therapy through Oxidative Activation of the JNK Apoptotic Pathway. Cancers (Basel) 2018. [DOI: 10.10.3390/cancers10090303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Side effects of radiation therapy (RT) remain the most challenging issue for pancreatic cancer treatment. Cerium oxide nanoparticles (CONPs) are currently being tested in pre-clinical trials as an adjuvant to sensitize pancreatic cancer cells to RT and protect normal tissues from the harmful side effects. CONPs were not able to significantly affect RT-induced DNA damage in cancer cells, thereby ruling out sensitization through increased mitotic catastrophe. However, activation of c-Jun terminal kinase (JNK), a key driver of RT-induced apoptosis, was significantly enhanced by co-treatment with CONPs and RT in pancreatic cancer cells in vitro and human pancreatic tumors in nude mice in vivo compared to CONPs or RT treatment alone. Further, CONP-driven increase in RT-induced JNK activity was associated with a marked increase in Caspase 3/7 activation, indicative of apoptosis. We have previously shown that CONPs increase reactive oxygen species (ROS) production in cancer cells. ROS has been shown to drive the oxidation of thioredoxin 1 (TRX1) which results in the activation of apoptosis signaling kinase 1 (ASK1). The increase in ASK1 activation following the co-treatment with CONPs followed by RT suggests that the increased JNK activation is the result of increased TRX1 oxidation. The ability of CONPs to sensitize pancreatic cancer cells to RT was mitigated when the TRX1 oxidation was prevented by mutagenesis of a cysteine residue or when the JNK activation was blocked by an inhibitor. Taken together, these data demonstrate an important mechanism for CONPs in specifically killing cancer cells and provide novel insights into the utilization of CONPs as a radiosensitizer and therapeutic agent for pancreatic cancer.
Collapse
|