1
|
|
Zhang Q, Xie Z, Li Y, Zhu Q, Shi H, Zhao R, Yang X, Tian J, Ma L. The potential of Lycium barbarum miR166a in kidney cancer treatment. Exp Cell Res 2023; 423:113455. [PMID: 36584744 DOI: 10.1016/j.yexcr.2022.113455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Predator species of animal can absorb plant microRNA that can regulate target gene expression and physiological function across species. The herb Lycium barbarum, a traditional Chinese medicine, has a wide range of antitumor effects. However, there are no reports on the effects of microRNA derived from it on the cross-border regulation of renal cell carcinoma (RCC). We performed in vitro and in vivo experiments to explore the role and mechanism of the L. barbarum-derived microRNA miR166a (Lb-miR166a) in cross-border regulation of RCC. Our mRNA sequencing analysis showed that Lb-miR166a regulates the expression of various genes in tumor cells, including 1232 upregulated genes and 581 downregulated genes, which were enriched to 1094 Gene Ontology entries and 43 Kyoto Encyclopedia of Genes and Genomes pathways. In vitro cell experiments confirmed that Lb-miR166a can inhibit the proliferation of RCC cells, promote the apoptosis of tumor cells, and inhibit the invasion and metastasis of tumor cells by regulating the expression of related genes. Furthermore, our in vivo tumor-bearing experiment showed that subcutaneous tumor formation volume decreased in Lb-miR166a mice, along with the number of liver metastases. This study elucidates the role and mechanism of Lb-miR166a in RCC treatment (Fig. 1). Our results further mechanistically confirm the antitumor properties of L. barbarum. Our study may contribute to the clinical development of a targeted drug for RCC treatment.
Collapse
|
2
|
|
Mioc M, Mioc A, Racoviceanu R, Ghiulai R, Prodea A, Milan A, Barbu Tudoran L, Oprean C, Ivan V, Șoica C. The Antimelanoma Biological Assessment of Triterpenic Acid Functionalized Gold Nanoparticles. Molecules 2023; 28. [PMID: 36615613 DOI: 10.3390/molecules28010421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
One of several promising strategies for increasing the bioavailability and therapeutic potential of high-lipophilic biologically active compounds is gold nanoparticle formulation. The current study describes the synthesis and biological antimelanoma evaluation of three triterpen-functionalized gold nanoparticles, obtained using our previously reported antimelanoma benzotriazole-triterpenic acid esters. Functionalized gold nanoparticle (GNP) formation was validated through UV-VIS and FTIR spectroscopy. The conjugate's cytotoxic effects were investigated using HaCaT healthy keratinocytes and A375 human melanoma cells. On A375 cells, all three conjugates demonstrated dose-dependent cytotoxic activity, but no significant cytotoxic effects were observed on normal HaCaT keratinocytes. GNP-conjugates were found to be more cytotoxic than their parent compounds. After treatment with all three GNP-conjugates, 4,6'-diamidino-2-phenylindole (DAPI) staining revealed morphological changes consistent with apoptosis in A375 melanoma cells. Quantitative real-time polymerase chain reaction (RT-qPCR) analysis revealed that the triterpene-GNP conjugate treated A375 melanoma cells had a fold change increase in Bcl-2-associated X protein (BAX) expression and a fold change decrease in B-cell lymphoma 2 (Bcl-2) expression. In A735 melanoma cells, high-resolution respirometry studies revealed that all three GNP-conjugates act as selective inhibitors of mitochondrial function. Furthermore, by examining the effect on each mitochondrial respiratory rate, the results indicate that all three conjugates are capable of increasing the production of reactive oxygen species (ROS), an apoptosis trigger in cancer cells.
Collapse
Affiliation(s)
- Marius Mioc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Alexandra Mioc
- Research Centre for Pharmaco-Toxicological Evaluation, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Department of Anatomy, Physiology, Pathophysiology, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Roxana Racoviceanu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Roxana Ghiulai
- Research Centre for Pharmaco-Toxicological Evaluation, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Department of Pharmacology-Pharmacotherapy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Alexandra Prodea
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Andreea Milan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Lucian Barbu Tudoran
- Electron Microscopy Laboratory "Prof. C. Craciun", Faculty of Biology & Geology, "Babes-Bolyai" University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania
- Electron Microscopy Integrated Laboratory, National Institute for R & D of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania
| | - Camelia Oprean
- Department of Chemistry and Toxicology, OncoGen Centre, County Hospital 'Pius Branzeu', Blvd. Liviu Rebreanu 156, 300736 Timisoara, Romania
- Department of Drug Analysis, Food and Environmental Chemistry, Legislation, Management and Pharmaceutical Marketing, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Viviana Ivan
- Department of Internal Medicine II, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Codruța Șoica
- Research Centre for Pharmaco-Toxicological Evaluation, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Department of Pharmacology-Pharmacotherapy, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| |
Collapse
|
3
|
|
Surducan DA, Racea RC, Cabuta M, Olariu I, Macasoi I, Rusu LC, Chiriac SD, Chioran D, Dinu S, Pricop MO. Eugenol Induces Apoptosis in Tongue Squamous Carcinoma Cells by Mediating the Expression of Bcl-2 Family. Life (Basel) 2022; 13. [PMID: 36675971 DOI: 10.3390/life13010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Head and neck squamous cell carcinoma is highly aggressive type of cancer for which the available treatment often causes patients severe side effects. Eugenol (Eug) is the major active constituent of clove essential oil and is known to possess antitumor properties. The present study aimed to assess the in vitro cytotoxicity of eugenol in SCC-4, tongue squamous carcinoma cells, and also in HGF, human gingival fibroblasts. Both cell lines were treated with five concentrations of Eug (0.1-1 mM) for 72 h. Cellular viability was assessed, followed by cellular morphological evaluation and by staining of the nuclei and cytoskeleton. RT-PCR was conducted in order to find the effect eugenol had on the expression on Bad, Bax, and Bcl-2 genes. Eugenol induced a dose-dependent decrease in viability in both cell lines, with the SCC-4 cells being significantly more affected. HGF cells detached from the plate at the highest concentrations used, while SCC-4 cells changed their morphology in a dose-dependent manner, with rounding, floating cells, and confluency loss being observed. Apoptotic-like signs such as chromatin and actin filaments condensation were clearly seen in SCC-4 cells, while RT-PCR revealed a significantly increased expression of pro-apoptotic genes Bax and Bad. Therefore, eugenol exerts its cytotoxic effect in tongue squamous cell carcinoma through inducing apoptosis.
Collapse
|
4
|
|
Lee J, Keam B, Kim S, Heo JN, Joung E, Kim M, Kim TM, Kim DW, Heo DS. The antitumor activity of a novel GCN2 inhibitor in head and neck squamous cell carcinoma cell lines. Transl Oncol 2022; 27:101592. [PMID: 36436443 DOI: 10.1016/j.tranon.2022.101592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND General control nonderepressible 2 (GCN2) senses amino acid deprivation and activates activating transcription factor 4 (ATF4), which regulates many adaptive genes. We evaluated the impact of AST-0513, a novel GCN2 inhibitor, on the GCN2-ATF4 pathway. Additionally, we evaluated the antitumor effects of AST-0513 in amino acid deprivation in head and neck squamous cell carcinoma (HNSCC) cell lines. METHODS GCN2 expression in HNSCC patient tissues was measured by immunohistochemistry. Five HNSCC cell lines (SNU-1041, SNU-1066, SNU-1076, Detroit-562, FaDu) grown under amino acid deprivation conditions, were treated with AST-0513. After AST-0513 treatment, cell proliferation was measured by CCK-8 assay. Flow cytometry was used to evaluate apoptosis and cell cycle phase. In addition, immunoblotting was performed to evaluate the effect of AST-0513 on the GCN2-ATF4 pathway, cell cycle arrest, and apoptosis. RESULTS We demonstrated that GCN2 was highly expressed in HNSCC patient tissues. AST-0513 inhibited the GCN2-ATF4 pathway in all five HNSCC cell lines. Inhibiting the GCN2-ATF4 pathway during amino acid deprivation reduced HNSCC cell proliferation and prevented adaptation to nutrient stress. Moreover, AST-0513 treatment led to p21 and Cyclin B1 accumulation and G2/M phase cycle arrest. Also, apoptosis was increased, consistent with increased bax expression, increased bcl-xL phosphorylation, and decreased bcl-2 expression. CONCLUSION A novel GCN2 inhibitor, AST-0513, inhibited the GCN2-ATF4 pathway and has antitumor activity that inhibits proliferation and promotes cell cycle arrest and apoptosis. Considering the high expression of GCN2 in HNSCC patients, these results suggest the potential role of GCN2 inhibitor for the treatment of HNSCC.
Collapse
|
5
|
|
Qin R, You F, Zhao Q, Xie X, Peng C, Zhan G, Han B. Naturally derived indole alkaloids targeting regulated cell death (RCD) for cancer therapy: from molecular mechanisms to potential therapeutic targets. J Hematol Oncol 2022; 15. [PMID: 36104717 DOI: 10.1186/s13045-022-01350-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Regulated cell death (RCD) is a critical and active process that is controlled by specific signal transduction pathways and can be regulated by genetic signals or drug interventions. Meanwhile, RCD is closely related to the occurrence and therapy of multiple human cancers. Generally, RCD subroutines are the key signals of tumorigenesis, which are contributed to our better understanding of cancer pathogenesis and therapeutics. Indole alkaloids derived from natural sources are well defined for their outstanding biological and pharmacological properties, like vincristine, vinblastine, staurosporine, indirubin, and 3,3′-diindolylmethane, which are currently used in the clinic or under clinical assessment. Moreover, such compounds play a significant role in discovering novel anticancer agents. Thus, here we systemically summarized recent advances in indole alkaloids as anticancer agents by targeting different RCD subroutines, including the classical apoptosis and autophagic cell death signaling pathways as well as the crucial signaling pathways of other RCD subroutines, such as ferroptosis, mitotic catastrophe, necroptosis, and anoikis, in cancer. Moreover, we further discussed the cross talk between different RCD subroutines mediated by indole alkaloids and the combined strategies of multiple agents (e.g., 3,10-dibromofascaplysin combined with olaparib) to exhibit therapeutic potential against various cancers by regulating RCD subroutines. In short, the information provided in this review on the regulation of cell death by indole alkaloids against different targets is expected to be beneficial for the design of novel molecules with greater targeting and biological properties, thereby facilitating the development of new strategies for cancer therapy.
Collapse
|
6
|
|
El-Sayed WA, Alminderej FM, Mounier MM, Nossier ES, Saleh SM, F Kassem A. New 1,2,3-Triazole-Coumarin-Glycoside Hybrids and Their 1,2,4-Triazolyl Thioglycoside Analogs Targeting Mitochondria Apoptotic Pathway: Synthesis, Anticancer Activity and Docking Simulation. Molecules 2022; 27:5688. [PMID: 36080455 DOI: 10.3390/molecules27175688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Toxicity and resistance to newly synthesized anticancer drugs represent a challenging phenomenon of intensified concern arising from variation in drug targets and consequently the prevalence of the latter concern requires further research. The current research reports the design, synthesis, and anticancer activity of new 1,2,3-triazole-coumarin-glycosyl hybrids and their 1,2,4-triazole thioglycosides as well as acyclic analogs. The cytotoxic activity of the synthesized products was studied against a panel of human cancer cell lines. Compounds 8, 10, 16 and 21 resulted in higher activities against different human cancer cells. The impact of the hybrid derivative 10 upon different apoptotic protein markers, including cytochrome c, Bcl-2, Bax, and caspase-7 along with its effect on the cell cycle was investigated. It revealed a mitochondria-apoptotic effect on MCF-7 cells and had the ability to upregulate pro-apoptotic Bax protein and downregulate anti-apoptotic Bcl-2 protein and thus implies the apoptotic fate of the cells. Furthermore, the inhibitory activities against EGFR, VEGFR-2 and CDK-2/cyclin A2 kinases for 8, 10 and 21 were studied to detect the mechanism of their high potency. The coumarin-triazole-glycosyl hybrids 8 and 10 illustrated excellent broad inhibitory activity (IC50= 0.22 ± 0.01, 0.93 ± 0.42 and 0.24 ± 0.20 μM, respectively, for compound 8), (IC50 = 0.12 ± 0.50, 0.79 ± 0.14 and 0.15± 0. 60 μM, respectively, for compound 10), in comparison with the reference drugs, erlotinib, sorafenib and roscovitine (IC50 = 0.18 ± 0.05, 1.58 ± 0.11 and 0.46 ± 0.30 μM, respectively). In addition, the docking study was simulated to afford better rationalization and put insight into the binding affinity between the promising derivatives and their targeted enzymes and that might be used as an optimum lead for further modification in the anticancer field.
Collapse
|
7
|
|
Singh D, Yodun T, Kumar G, Ahmad Tali J, Tiwari H, Singh J, Nargotra A, Samykutty A, Singh S, Shankar R. Synthesis of 3-N-/O-/S-methyl-imidazo[1,2-a] pyridine derivatives for caspase-3 mediated apoptosis induced anticancer activity. Bioorg Chem 2022; 125:105882. [DOI: 10.1016/j.bioorg.2022.105882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
|
An W, Zhang Y, Lai H, Zhang Y, Zhang H, Zhao G, Liu M, Li Y, Lin X, Cao S. Alpinia katsumadai Hayata induces growth inhibition and autophagy‑related apoptosis by regulating the AMPK and Akt/mTOR/p70S6K signaling pathways in cancer cells. Oncol Rep 2022; 48:142. [PMID: 35730618 DOI: 10.3892/or.2022.8353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Alpinia katsumadai Hayata (AKH), a widely used traditional Chinese medicine, exerts various biological functions, including anti-inflammatory, antioxidant, anti-microbial and anti-asthmatic effects. However, studies on its anticancer activity and associated mechanisms are limited. The present study investigated the effects of ethanol extract from AKH on the viability of various human cancer and normal liver LX-2 cells using Cell Counting Kit-8 assay. Apoptosis was detected by Hoechst 33342/PI staining and Annexin-V-FITC/PI double staining. Autophagy was examined by Ad-GFP-LC3B transfection. The association between AKH-induced autophagy and apoptosis was investigated by pre-treatment of the cells with the autophagy inhibitors, 3-methyladenine (3MA) and bafilomycin A1 (Baf-A1), followed by treatment with AKH. The expression levels of cleaved poly(ADP-ribose) polymerase (PARP), caspase-8, caspase-3, caspase-9, phosphorylated (p-)AMP-activated protein kinase (AMPK), Akt, mTOR and p70S6K were examined using western blot analysis. The in vivo antitumor activity of AKH was investigated in nude mice bearing A549 lung cancer xenografts. The components of AKH were detected by liquid chromatography mass spectrometry-ion trap-time-of-flight mass spectrometry. The results revealed that AKH significantly inhibited the proliferation of various cancer cells with the half maximal inhibitory concentration (IC50) values of 203–284 µg/ml; however, its inhibitory effect was much less prominent against normal liver LX-2 cells with an IC50 value of 395 µg/ml. AKH markedly induced apoptosis and autophagy, and upregulated the protein expression of cleaved-caspase-3, caspase-8, caspase-9 and cleaved PARP in a concentration-dependent manner. Of note, the autophagy inhibitors (3MA and Baf-A1) significantly attenuated its pro-apoptotic effects on human pancreatic cancer Panc-28 and lung cancer A549 cells. Furthermore, AKH significantly increased the levels of p-AMPK, and decreased those of p-Akt, p-mTOR and p-p70S6K in Panc-28 and A549 cells. AKH markedly inhibited the growth of A549 tumor xenografts in vivo. In addition, a total of nine compounds were detected from AKH. The present study demonstrates that AKH markedly inhibits the growth and induces autophagy-related apoptosis in cancer cells by regulating the AMPK and Akt/mTOR/p70S6K signaling pathways. AKH and/or its active fractions may thus have potential to be developed as novel anticancer agents for clinical use.
Collapse
Affiliation(s)
- Weixiao An
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yuxi Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Honglin Lai
- Department of Pharmacy, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yangyang Zhang
- Department of Pharmacy, Dongying Hospital of Traditional Chinese Medicine, Dongying, Shandong 257055, P.R. China
| | - Hongmei Zhang
- Rizhao Hospital of Traditional Chinese Medicine, Rizhao, Shandong 276801, P.R. China
| | - Ge Zhao
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Minghua Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yang Li
- Department of International Trade, School of International Traded and Economics, University of International Business and Economics, Beijing 100029, P.R. China
| | - Xiukun Lin
- Delisi Group Co. Ltd., Zhucheng, Shandong 262200, P.R. China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
9
|
|
Noser AA, Shehadi IA, Abdelmonsef AH, Salem MM. Newly Synthesized Pyrazolinone Chalcones as Anticancer Agents via Inhibiting the PI3K/Akt/ERK1/2 Signaling Pathway. ACS Omega 2022; 7:25265-77. [PMID: 35910116 DOI: 10.1021/acsomega.2c02181] [Citation(s) in RCA: 8] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A series of novel pyrazolinone chalcones 3-9 have been synthesized through the condensation of azo pyrazolinone derivatives with various aromatic aldehydes. Spectroscopic techniques and elemental analysis have both corroborated this. Furthermore, all compounds were screened in silico for their ability to inhibit cancer proliferation and metastasis by targeting the PI3K/Akt signaling pathway. This inhibitory pathway might be an efficient approach for the death of cancer cells, angiogenesis, and metastasis prevention. Our results indicated that only compound 6b was the top-ranked. It demonstrated the highest binding energies of -11.1 and -10.7 kcal/mol against the target proteins PI3K and Akt, respectively; thus, it was chosen for in vitro studies. Compound 6b exhibited the most effective cytotoxic impact against the Caco cell line with IC50 of 23.34 ± 0.14 μM. Furthermore, it showed significant inhibition of PI3K/Akt proteins and oxidative stress, leading to elevated Bax and p53 expression, reduced Bcl-2 expression, and triggered cell cycle arrest at the sub-G0/G1 phase. Additionally, it showed significant downregulation of the Raf-1 gene, leading to ERK1/2 protein inhibition. These findings demonstrate that compound 6b obeyed Lipinski's rule of five and might be used as a favored scaffold for cancer treatment by inhibiting proliferation and metastasis via inhibition of the PI3K/Akt and Raf-1/ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Ahmed
A. Noser
- Organic
Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Ihsan A. Shehadi
- Department
of Chemistry, Pure and Applied Chemistry Research Group, College of
Sciences, University of Sharjah, Sharjah 27272, UAE
| | | | - Maha M. Salem
- Biochemistry
Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
10
|
|
Azadi S, Mohammadi E, Tafazzoli-Shadpour M, Tabatabaei M. Effects of chemically EGFR targeting on non-targeted physical cell behaviors in 2D and 3D microfluidic cultures of invasive and non-invasive breast cancer cell lines. Biochem Biophys Res Commun 2022; 622:1-7. [PMID: 35841768 DOI: 10.1016/j.bbrc.2022.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cancer development comprehends changes in cell structural and physical states. Cancer cells are softer than normal cells, produce higher contractile forces, and migrate more easily. While chemotherapy, targets proteins involved in biological behaviors, it may affect cell physicomechanical state due to the interconnections among signaling pathways. Here we treated non-invasive and invasive breast cancer cell lines by targeting EGRF which modulates major biological behaviors. We quantified migration potential of cancer cells in a microfluidic device, and evaluated expression of proteins associated with physical behaviors. Results indicated significant alterations in physical behaviors, with a higher impact on invasive cells. The anti-cancer synergy between biological and physical behaviors was shown by decreasing actin, vinculin, and myosin II content and altered distribution, limiting cell invasion in 3D collagen structure, accompanied by decreasing cell viability and vimentin expression as the EMT biomarker. The center point of changes in physical behaviors was in cytoskeletal remodeling by chemical treatment, potentially through lower contractile force generation and less development of focal adhesions and stress fibers. The synergy between physical and chemical pathways can be used in enhancing anti-cancer drug efficacy.
Collapse
Affiliation(s)
- Shohreh Azadi
- Cell Engineering and Biomicrofluidics Systems Lab, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Ehsan Mohammadi
- Cell Engineering and Biomicrofluidics Systems Lab, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Mohammad Tafazzoli-Shadpour
- Cell Engineering and Biomicrofluidics Systems Lab, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Mohammad Tabatabaei
- Cell Engineering and Biomicrofluidics Systems Lab, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
11
|
|
Trang CNM, Chi HNQ, Manh NK, Son HN, Ngo D, Long LT. The Chloroform Extracts of Vietnamese Sophora flavescens Ait. Inhibit the Proliferation of HepG2 Cells through Apoptosis Induction. Applied Sciences 2022; 12:5906. [DOI: 10.3390/app12125906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The present study evaluated the effects of Sophora flavescens Ait. root extract on the proliferation of human hepatoma cell line HepG2. HPLC-UV analysis showed that the highest matrine and oxymatrine contents were obtained in the chloroform extract, compared to ethanol extract and ethyl acetate extract. The morphological analysis revealed that the chloroform extract of Sophora flavescens Ait. (SFA-CHCl3 extract) induced alterations of HepG2 cell morphology, resulting in the shrinkage of cells, the formation of debris, and cell detachment. The proliferation of HepG2 cells was inhibited by SFA-CHCl3 extract treatment. Cell cycle analysis exhibited that the cell proportion of the G0/G1 phase of HepG2 cells with SFA-CHCl3 extract treatment was decreased, while the cell proportion of the G2/M phase was increased. Flow cytometry analysis indicated a dramatic increase in the apoptotic percentage of HepG2 cells over the time of SFA-CHCl3 extract treatment. The SFA-CHCl3 extract also caused morphological changes in HepG2 nuclear, including chromatin condensation and DNA fragmentation. SFA-CHCl3 extract treatment induced the bax up-regulation and the bcl-2 down-regulation in HepG2 cells. These results revealed that SFA-CHCl3 extract could be a potential apoptosis inducer in HepG2 cells.
Collapse
|
12
|
|
Gonçalves Correa ND, Silva FD, Vieira DP, Soares CRJ, de Queiroz AAA. In vitro cytotoxic data on Se-methylselenocysteine conjugated to dendritic poly(glycerol) against human squamous carcinoma cells. J Biomater Sci Polym Ed 2022; 33:651-67. [PMID: 34809530 DOI: 10.1080/09205063.2021.2008788] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Polymeric nanoparticles acting as sources of selenium (Se) are currently an interesting topic in cancer chemotherapy. In this study, polyglycerol dendrimer (DPGLy) was functionalized with seleno-methyl-selenocysteine (SeMeCys) by means of Steglich esterification with 4-dimethylaminopyridine/(l-ethyl-3-(3-dimethylaminopropyl)carbodiimide) (EDC/DMAP) and cerium chloride as cocatalyst in acetonitrile at quantitative yields of 98 ± 1%. The SeMeCys coupling DPGLy efficiency vs. time were determined by Fourier Transform infrared spectroscopy (FTIR) and ultraviolet-visible (UV-Vis) spectroscopy. The cytotoxic effects of SeMeCys-DPGLy on the Chinese Hamster ovary cell line (CHO-K1) and head and neck squamous cell carcinoma (HNSCC) cells line were assessed by MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay. No signs of general toxicity of SeMeCys-DPGLy against CHO-K1 cells were detectable at which cell viability was greater than 98%. MTS assays revealed that SeMeCys-DPGLy reduced HNSCC cell viability and proliferation at higher doses and long incubation times.
Collapse
|
13
|
|
Jayathilake AG, Kadife E, Kuol N, Luwor RB, Nurgali K, Su XQ. Krill oil supplementation reduces the growth of CT-26 orthotopic tumours in Balb/c mice. BMC Complement Med Ther 2022; 22:34. [PMID: 35120511 DOI: 10.1186/s12906-022-03521-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Background We have previously reported that the free fatty acid extract (FFAE) of krill oil (KO) significantly inhibits the proliferation and migration, and induces apoptosis of colorectal cancer (CRC) cells. This study aimed to investigate the in vivo efficacy of various doses of KO supplementation on the inhibition of CRC tumour growth, molecular markers of proliferation, angiogenesis, apoptosis, the epidermal growth factor receptor (EGFR) and its downstream molecular signalling. Methods Male Balb/c mice were randomly divided into four groups with five in each group. The control (untreated) group received standard chow diet; and other three groups received KO supplementation at 5%, 10%, and 15% of their daily dietary intake respectively for three weeks before and after the orthotopic implantation of CT-26 CRC cells in their caecum. The expression of cell proliferation marker Ki-67 and angiogenesis marker CD-31 were assessed by immunohistochemistry. The expression of EGFR, phosphorylated EGFR (pEGFR), protein kinase B (AKT), pAKT, extracellular signal-regulated kinase (ERK1/2), pERK1/2, cleaved caspase-7, cleaved poly (ADP-ribose) polymerase (PARP), and DNA/RNA damage were determined by western blot. Results KO supplementation reduced the CRC tumour growth in a dose-dependent manner; with 15% of KO being the most effective in reduction of tumour weight and volume (68.5% and 68.3% respectively, P < 0.001), inhibition of cell proliferation by 69.9% (P < 0.001) and microvessel density by 72.7% (P < 0.001). The suppressive effects of KO on EGFR and its downstream signalling, ERK1/2 and AKT, were consistent with our previous in vitro observations. Furthermore, KO exhibited pro-apoptotic effects on tumour cells as indicated by an increase in the expression of cleaved PARP by 3.9-fold and caspase-7 by 8.9-fold. Conclusions This study has demonstrated that KO supplementation reduces CRC tumour growth by inhibiting cancer cell proliferation and blood vessel formation and inducing apoptosis of tumour cells. These anti-cancer effects are associated with the downregulation of the EGFR signalling pathway and activation of caspase-7, PARP cleavage, and DNA/RNA damage. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03521-4.
Collapse
Affiliation(s)
| | - Elif Kadife
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia
| | - Nyanbol Kuol
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia
| | - Rodney Brain Luwor
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia.,Department of Medicine, Western Health, The University of Melbourne, Melbourne, Australia.,Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Sciences (AIMSS), Melbourne, Australia
| | - Xiao Qun Su
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia.
| |
Collapse
|
14
|
|
Wang L, Cheng L, Ma L, Ahmad Farooqi A, Qiao G, Zhang Y, Ye H, Liu M, Huang J, Yang X, Lin X, Cao S. Alnustone inhibits the growth of hepatocellular carcinoma via ROS- mediated PI3K/Akt/mTOR/p70S6K axis. Phytother Res 2021. [PMID: 34847624 DOI: 10.1002/ptr.7337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alnustone, a diarylheptane compound, exhibits potent growth inhibition against hepatocellular carcinoma (HCC) BEL-7402 cells. However, the underlying mechanisms associated with its anticancer activity remain unknown. In the present study, we evaluated the anticancer effect of alnustone against several human cancers focused on HCC and the possible associated mechanisms. The results showed that alnustone significantly inhibited the growth of several cancer cells by CCK-8 assay. Alnustone markedly induced apoptosis and decreased mitochondrial membrane potential in BEL-7402 and HepG2 cells. Alnustone inhibited the expression of proteins related to apoptosis and PI3K/Akt/mTOR/p70S6K pathways and generated ROS production in BEL-7402 and HepG2 cells. Moreover, N-acetyl-L-cysteine (NAC, a ROS inhibitor) could significantly reverse the effects of alnustone on the growth inhibition of BEL-7402 and HepG2 cells and the expression of proteins related to apoptosis and PI3K/Akt/mTOR signaling pathway in HepG2 cells. Furthermore, alnustone significantly inhibited tumor growth of HepG2 xenografts, obviously induced apoptosis in the tumor tissues and improved the pathological condition of liver tissues of mice in vivo. The study provides evidence that alnustone is effective against HCC via ROS-mediated PI3K/Akt/mTOR/p70S6K pathway and the compound has the potential to be developed as a novel anticancer agent for the treatment of HCC clinically.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Li Cheng
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Li Ma
- Rizhao Hospital of Traditional Chinese Medicine, Rizhao, Shandong, China
| | - Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Gan Qiao
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuxi Zhang
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Hanlin Ye
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Minghua Liu
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianlin Huang
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaohui Yang
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiukun Lin
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China.,Delisi Group Co. Ltd., Zhucheng, Shandong, China
| | - Shousong Cao
- Department of Pharmacology, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
15
|
|
Gunathilaka TL, Dilrangi KH, Ranasinghe P, Samarakoon KW, Peiris LDC. Mechanistic Insight into Apoptotic Induction in Human Rhabdomyosarcoma and Breast Adenocarcinoma Cells by Chnoospora minima: A Sri Lankan Brown Seaweed. Pharmaceuticals (Basel) 2021; 14:1154. [PMID: 34832937 DOI: 10.3390/ph14111154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The current study determined the cytotoxic and apoptotic potential of the polyphenol-rich methanol extract of Chnoospora minima (C. minima) and its fractions against human breast adenocarcinoma (MCF-7) and rhabdomyosarcoma (RMS) cells. MTT and neutral red assays were used to determine cytotoxicity. The clonogenic assay evaluated the antineoplastic activity, while the apoptotic activity was determined by cellular morphological changes, caspase 3/7 activity, and DNA fragmentation. Morphological alterations in apoptosis were observed by an inverted phase-contrast microscope and Hoechst 33342 staining methods. The total phenolic, flavonoids, alkaloids, and antioxidant activity in the hexane and chloroform fractions were determined, based on their cytotoxic activity. The hexane fraction of C. minima effectively reduced the cell growth that is concentration-dependent in human RMS and MCF-7 cell lines. It also exhibited low cytotoxicity on Vero cells. The characteristic cellular and nuclear apoptotic morphological features were observed. A noticeable caspase 3/7 activation and the fragmented DNA were detected only in the hexane fraction treated RMS cells, whereas MCF-7 cells showed low caspase 3/7 activation due to a lack of caspase 3 and no evidence of having a typical ladder pattern of apoptosis. Further analysis revealed that the hexane fraction-treated RMS cells upregulated the p53 gene twofold (2.72) compared to the p21 (0.77) gene, whereas in the MCF-7 cells, a 2.21-fold upregulation of p53 was observed compared to the p21 (0.64) gene. The hexane fraction exhibited moderate total phenolics, flavonoids, alkaloids content, and antioxidant activity. According to the different antioxidant mechanisms, hexane and chloroform fractions showed the highest antioxidant activities by FRAP and ORAC assays, respectively. GC-MS analysis of hexane fraction revealed the presence of methyl tetradecanoate (38.314%) as the most abundant compound. The study’s findings highlighted that the non-polar compounds present in the hexane fraction of C. minima suppressed cell proliferation and induced apoptosis-mediated cell death in RMS and MCF-7 cells, mainly via the activation of the p53 gene. Hence, the isolation of compounds is warranted. However, more studies are required to understand the mechanistic insights of these observations.
Collapse
|
16
|
|
Abstract
Many anti-cancer drugs, including paclitaxel and etoposide, have originated and been developed from natural products, and traditional herbal medicines have fewer adverse effects and lesser toxicity than anti-tumor reagents. Therefore, we developed a novel complex herbal medicine, JI017, which mediates endoplasmic reticulum (ER) stress and apoptosis through the Nox4–PERK–CHOP signaling pathway in ovarian cancer cells. JI017 treatment increases the expression of GRP78, ATF4, and CHOP and the phosphorylation of PERK and eIF2α via the upregulation of Nox4. Furthermore, it increases the release of intracellular reactive oxygen species (ROS), the production of intracellular Ca2+, and the activation of exosomal GRP78 and cell lysate GRP78. Combination treatment using the sarco/endoplasmic reticulum Ca2+-ATPase inhibitor thapsigargin (TG) and JI017 reportedly induces increased ER stress and cell death in comparison to the control; however, knockdown experiments of PERK and CHOP indicated suppressed apoptosis and ER stress in JI017-treated ovarian cancer cells. Furthermore, targeting Nox4 using specific siRNA and pharmacological ROS inhibitors, including N-acetylcystein and diphenylene iodonium, blocked apoptosis and ER stress in JI017-treated ovarian cancer cells. In the radioresistant ovarian cancer model, when compared to JI017 alone, JI017 co-treatment with radiation induced greater cell death and resulted in overcoming radioresistance by inhibiting epithelial–mesenchymal-transition-related phenomena such as the reduction of E-cadherin and the increase of N-cadherin, vimentin, Slug, and Snail. These findings suggest that JI017 is a powerful anti-cancer drug for ovarian cancer treatment and that its combination treatment with radiation may be a novel therapeutic strategy for radioresistant ovarian cancer.
Collapse
Affiliation(s)
| | - Seong-Gyu Ko
- Correspondence: ; Tel.: +82-2-961-0329; Fax: +82-2-961-1165
| |
Collapse
|
17
|
|
Luo YH, Wang C, Xu WT, Zhang Y, Zhang T, Xue H, Li YN, Fu ZR, Wang Y, Jin CH. 18β-Glycyrrhetinic Acid Has Anti-Cancer Effects via Inducing Apoptosis and G2/M Cell Cycle Arrest, and Inhibiting Migration of A549 Lung Cancer Cells. Onco Targets Ther 2021; 14:5131-44. [PMID: 34712051 DOI: 10.2147/OTT.S322852] [Citation(s) in RCA: 10] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background 18β-glycyrrhetinic acid (18β-Gly), which is extracted from licorice root, has various pharmacological properties; however, its anti-cancer effects on lung cancer cells have not been fully established. Purpose In this study, we investigated the underlying molecular mechanisms of 18β-Gly. Results Our results showed that 18β-Gly had significant cytotoxic effects and no apparent side effects. 18β-Gly induced mitochondria-dependent apoptosis of A549 lung cancer cells. In addition, after treatment with 18β-Gly, intracellular reactive oxygen species (ROS) levels were significantly increased, and G2/M cell cycle arrest and inhibition of cell migration were induced via the mitogen-activated protein kinase (MAPK)/signal transducer and activator of transcription 3 (STAT3)/nuclear factor kappa (NF-κB) signaling pathways. After pretreatment with the ROS scavenger N-acetyl-L-cysteine or MAPK inhibitors, the expression levels of phosphorylated p38 (p-p38), phosphorylated c-Jun N-terminal kinase, inhibitor of nuclear factor kappa B, cleaved caspase-3 (cle-cas-3), cleaved poly (ADP ribose) polymerase (cle-PARP), p-p53, p27, p21, and E-cadherin were decreased; and levels of phosphorylated extracellular signal-regulated kinase, p-STAT3, NF-κB, Bcl-2, cyclin B1, cyclase-dependent kinase 1/2 (CDK1/2), N-cadherin, vimentin, and snail homolog 1 (SNAI 1) were increased. In addition, the percentage of cells in the G2/M phase was decreased, and inhibition of migration was reduced. Conclusion In summary, 18β-Gly induced apoptosis and G2/M cell cycle arrest and inhibited migration via the ROS/MAPK/STAT3/NF-κB signaling pathways in A549 lung cancer cells. Therefore, 18β-Gly is a novel promising candidate for the treatment of lung cancer.
Collapse
Affiliation(s)
- Ying-Hua Luo
- Department of Grass Science, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Cheng Wang
- Pharmacy Department, Daqing Oilfield General Hospital, Daqing, 163001, People's Republic of China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Hui Xue
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Yan-Nan Li
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Zhong-Ren Fu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Ying Wang
- College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China.,College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China.,National Coarse Cereals Engineering Research Center, Daqing, 163319, People's Republic of China
| |
Collapse
|
18
|
|
Yu W, Zhang X, Zhang W, Xiong M, Lin Y, Chang M, Xu L, Lu Y, Liu Y, Zhang J. 19-Hydroxybufalin inhibits non-small cell lung cancer cell proliferation and promotes cell apoptosis via the Wnt/β-catenin pathway. Exp Hematol Oncol 2021; 10:48. [PMID: 34696818 DOI: 10.1186/s40164-021-00243-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Bufadienolides derived from the skin of toads are often regarded as the main active components with antitumor effects. 19-Hydroxybufalin (19-HB) is a monomer of bufadienolides; however, its effects and underlying molecular mechanisms on tumor growth remain to be ascertained. In this report, we focused on the antitumor effects of 19-HB on non-small cell lung cancer to provide a scientific basis for its further development and utilization. Methods The antitumor effects of 19-HB on the human NSCLC cell lines NCI-H1299 and NCI-H838 were examined in vitro. The cells were treated with different concentrations of 19-HB, and the inhibition of cell growth was measured by CCK-8 and colony formation assays. Furthermore, cell apoptosis was analyzed by flow cytometry, TUNEL staining, JC-1 staining, and western blotting. The effects on migration and invasion were evaluated by wound-healing assay, transwell assay, and western blotting. Finally, the antitumor effects of 19-HB were evaluated in vivo using a xenograft mouse model. Results 19-HB-treated NSCLC cells showed inhibited cell viability and increased apoptosis. The expression levels of cleaved caspase-3, cleaved-PARP, and Bax/Bcl-2 were upregulated, while the mitochondrial membrane potential decreased. In contrast, migration, invasion, as well as the expression of MMP2, MMP7, MMP9, the epithelial–mesenchymal transition-related proteins N-cadherin and Vimentin, and the transcription factors Snail and Slug were inhibited. Furthermore, the expression levels of the key molecules in the Wnt/β-catenin signaling pathway (CyclinD1, c-Myc, and β-catenin) were decreased. In vivo, the growth of xenograft tumors in nude mice was also significantly inhibited by 19-HB, and there were no significant changes in biochemical indicators of hepatic and renal function. Conclusions 19-HB inhibited the proliferation, migration, and invasion, and promoted the apoptosis of NSCLC cells via the Wnt/β-catenin pathway. In addition, 19-HB inhibited the growth of xenograft tumors in nude mice with little toxicity to the liver and kidney. Thus, 19-HB may be a potential antitumor agent for treating NSCLC. Supplementary Information The online version contains supplementary material available at 10.1186/s40164-021-00243-0.
Collapse
Affiliation(s)
- Wei Yu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment With Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China.,School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Xiao Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Wei Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Minggang Xiong
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Yuhan Lin
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Ming Chang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Lin Xu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment With Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China. .,Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, 518055, Guangdong, China.
| | - Yun Liu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment With Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China.
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China. .,Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
19
|
|
Tsai TH, Lieu AS, Huang TY, Kwan AL, Lin CL, Hsu YC. RTA404, an Activator of Nrf2, Activates the Checkpoint Kinases and Induces Apoptosis through Intrinsic Apoptotic Pathway in Malignant Glioma. J Clin Med 2021; 10:4805. [PMID: 34768325 DOI: 10.3390/jcm10214805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background: Malignant glioma (MG) is an aggressive malignant brain tumor. Despite advances in multidisciplinary treatment, overall survival rates remain low. A trifluoroethyl amide derivative of 2-cyano-3-,12-dioxoolean-1,9-dien-28-oic acid (CDDO), CDDO–trifluoroethyl amide (CDDO–TFEA) is a nuclear erythroid 2-related factor 2/antioxidant response element pathway activator. RTA404 is used to inhibit proliferation and induce apoptosis in cancer cells. However, its effect on tumorigenesis in glioma is unclear. Methods: This in vitro study evaluated the effects of RTA404 on MG cells. We treated U87MG cell lines with RTA404 and performed assessments of apoptosis and cell cycle distributions. DNA content and apoptosis induction were subjected to flow cytometry analysis. The mitotic index was assessed based on MPM-2 expression. Protein expression was analyzed through Western blotting. Results: RTA404 significantly inhibited the cell viability and induced cell apoptosis on the U87MG cell line. The Annexin-FITC/PI assay revealed significant changes in the percentage of apoptotic cells. Treatment with RTA404 led to a significant reduction in the U87MG cells’ mitochondrial membrane potential. A significant rise in the percentage of caspase-3 activity was detected in the treated cells. In addition, these results suggest that cells pass the G2 checkpoint without cell cycle arrest by RTA404 treatment in the MPM-2 staining. An analysis of CHK1, CHK2, and p-CHK2 expression suggested that the DNA damage checkpoint system seems also to be activated by RTA404 treatment in established U87MG cells. Therefore, RTA404 may not only activate the DNA damage checkpoint system, it may also exert apoptosis in established U87MG cells. Conclusions: RTA404 inhibits the cell viability of gliomas and induces cancer cell apoptosis through intrinsic apoptotic pathway in Malignant glioma. In addition, the DNA damage checkpoint system seems also to be activated by RTA404. Taken together, RTA404 activated the DNA damage checkpoint system and induced apoptosis through intrinsic apoptotic pathways in established U87MG cells.
Collapse
|
20
|
|
Almalki Z, Algregri M, Alhosin M, Alkhaled M, Damiati S, Zamzami MA. In vitro cytotoxicity of curcuminoids against head and neck cancer HNO97 cell line. BRAZ J BIOL 2021; 83:e248708. [PMID: 34468533 DOI: 10.1590/1519-6984.248708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a malignant tumour of Head and Neck Cancer (HNC). The recent therapeutic approaches used to treat cancer have adverse side effects. The natural agents exhibiting anticancer activities are generally considered to have a robust therapeutic potential. Curcuminoids, one of the major active compounds of the turmeric herb, are used as a therapeutic agent for several diseases including cancer. In this study, the cytotoxicity of curcuminoids was investigated against OSCC cell line HNO97. Our data showed that curcuminoids significantly inhibits the proliferation of HNO97 in a time and dose-dependent manner (IC50=35 μM). Cell cycle analysis demonstrated that curcuminoids increased the percentage of G2/M phase cell populations in the treated groups. Treating HNO97 cells with curcuminoids led to cell shrinking and increased detached cells, which are the typical appearance of apoptotic cells. Moreover, flow cytometry analysis revealed that curcuminoids significantly induced apoptosis in a time-dependent manner. Furthermore, as a response to curcuminoids treatment, comet tails were formed in cell nuclei due to the induction of DNA damage. Curcuminoids treatment reduced the colony formation capacity of HNO97 cells and induced morphological changes. Overall, these findings demonstrate that curcuminoids can in vitro inhibit HNC proliferation and metastasis and induce apoptosis.
Collapse
Affiliation(s)
- Z Almalki
- King Abdulaziz University, Faculty of Science, Department of Biochemistry, Jeddah, Saudi Arabia
| | - M Algregri
- King Abdulaziz University, King Fahad Medical Research Canter, Jeddah, Saudi Arabia
| | - M Alhosin
- King Abdulaziz University, Faculty of Science, Department of Biochemistry, Jeddah, Saudi Arabia
| | - M Alkhaled
- University of Jeddah, Faculty of Science, Department of Biological Sciences, Jeddah, Saudi Arabia
| | - S Damiati
- King Abdulaziz University, Faculty of Science, Department of Biochemistry, Jeddah, Saudi Arabia
| | - M A Zamzami
- King Abdulaziz University, Faculty of Science, Department of Biochemistry, Jeddah, Saudi Arabia
| |
Collapse
|
21
|
|
Abd Wahab NA, Abas F, Othman I, Naidu R. Diarylpentanoid (1,5-bis(4-hydroxy-3-methoxyphenyl)-1,4-pentadiene-3-one) (MS13) Exhibits Anti-proliferative, Apoptosis Induction and Anti-migration Properties on Androgen-independent Human Prostate Cancer by Targeting Cell Cycle-Apoptosis and PI3K Signalling Pathways. Front Pharmacol 2021; 12:707335. [PMID: 34366863 DOI: 10.3389/fphar.2021.707335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Diarylpentanoids exhibit a high degree of anti-cancer activity and stability in vitro over curcumin in prostate cancer cells. Hence, this study aims to investigate the effects of a diarylpentanoid, 1,5-bis(4-hydroxy-3-methoxyphenyl)-1,4-pentadiene-3-one (MS13) on cytotoxicity, anti-proliferative, apoptosis-inducing, anti-migration properties, and the underlying molecular mechanisms on treated androgen-independent prostate cancer cells, DU 145 and PC-3. A cell viability assay has shown greater cytotoxicity effects of MS13-treated DU 145 cells (EC50 7.57 ± 0.2 µM) and PC-3 cells (EC50 7.80 ± 0.7 µM) compared to curcumin (EC50: DU 145; 34.25 ± 2.7 µM and PC-3; 27.77 ± 6.4 µM). In addition, MS13 exhibited significant anti-proliferative activity against AIPC cells compared to curcumin in a dose- and time-dependent manner. Morphological observation, increased caspase-3 activity, and reduced Bcl-2 protein levels in these cells indicated that MS13 induces apoptosis in a time- and dose-dependent. Moreover, MS13 effectively inhibited the migration of DU 145 and PC-3 cells. Our results suggest that cell cycle-apoptosis and PI3K pathways were the topmost significant pathways impacted by MS13 activity. Our findings suggest that MS13 may demonstrate the anti-cancer activity by modulating DEGs associated with the cell cycle-apoptosis and PI3K pathways, thus inhibiting cell proliferation and cell migration as well as inducing apoptosis in AIPC cells.
Collapse
Affiliation(s)
- Nurul Azwa Abd Wahab
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, Serdang, Malaysia.,Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
22
|
|
Zhang D, Xiao X, Song D, Chen S, Zhang Z, Cao S, Liu M. Atractylenolide III induces apoptosis by regulating the Bax/Bcl-2 signaling pathway in human colorectal cancer HCT-116 Cells in vitro and in vivo. Anticancer Drugs 2021. [PMID: 34261915 DOI: 10.1097/CAD.0000000000001136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Atractylodes is the dry root of atractylodes macrocephala koidz and has been commonly used as a traditional Chinese medicine (TCM). Atractylenolide III, a main component of atractylodes, has displayed significant effects on anti-inflammation and anticancer. However, the effects of atractylenolide III on growth inhibition and apoptosis induction in colon cancer remain unclear. The results showed that atractylenolide III significantly inhibited the cell growth and induce cellular apoptosis in HCT-116 cells in a concentration dependence manner in vitro. Mechanistic studies further showed that atractylenolide III could regulate the Bax/Bcl-2 apoptotic signaling pathway through promoting the expression of proapoptotic related gene/proteins Bax, caspase-9 and caspase-3 but inhibiting the expression of antiapoptotic related gene/protein Bcl-2 in HCT-116 cells. Furthermore, atractylenolide III also significantly inhibited the tumor growth of HCT-116 tumor xenografts bearing in nude mice through inducing apoptosis by upregulation of the expressions of Bax, cleaved caspase-3 and p53 but downregulation of the expressions of Bcl-2 in HCT-116 tumor tissues in vivo. The studies may provide the scientific rationale for the understanding of the anticancer effect of atractylenolide III. Therefore, atractylenolide III may have the potential to be developed as a promising novel anticancer agent for the treatment of colorectal cancer clinically.
Collapse
|
23
|
|
Abstract
Simple Summary Breast cancer remains the most commonly diagnosed cancer and the leading cause of cancer death among females worldwide. It is a highly heterogeneous disease, classified according to hormone and growth factor receptor expression. Patients with triple negative breast cancer (TNBC) (estrogen receptor-negative/progesterone receptor-negative/human epidermal growth factor receptor (HER2)-negative) and hormone-independent HER2 overexpressing subtypes still represent highly aggressive behavior, metastasis, poor prognosis, and drug resistance. Thus, new alternative anticancer agents based on the use of natural products have been receiving enormous attention. In this regard, curcumin is a promising lead in cancer drug discovery due its ability to modulate a diverse range of molecular targets and signaling pathways. The current review has emphasized the underlying mechanism of curcumin anticancer action mediated through the modulation of PI3K/Akt/mTOR, JAK/STAT, MAPK, NF-ĸB, p53, Wnt/β-catenin, apoptosis, and cell cycle pathways in hormone-independent breast cancer, providing frameworks for future studies and insights to improve its efficiency in clinical practice. Abstract Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer death among women worldwide. Despite the overall successes in breast cancer therapy, hormone-independent HER2 negative breast cancer, also known as triple negative breast cancer (TNBC), lacking estrogens and progesterone receptors and with an excessive expression of human epidermal growth factor receptor 2 (HER2), along with the hormone-independent HER2 positive subtype, still remain major challenges in breast cancer treatment. Due to their poor prognoses, aggressive phenotype, and highly metastasis features, new alternative therapies have become an urgent clinical need. One of the most noteworthy phytochemicals, curcumin, has attracted enormous attention as a promising drug candidate in breast cancer prevention and treatment due to its multi-targeting effect. Curcumin interrupts major stages of tumorigenesis including cell proliferation, survival, angiogenesis, and metastasis in hormone-independent breast cancer through the modulation of multiple signaling pathways. The current review has highlighted the anticancer activity of curcumin in hormone-independent breast cancer via focusing on its impact on key signaling pathways including the PI3K/Akt/mTOR pathway, JAK/STAT pathway, MAPK pathway, NF-ĸB pathway, p53 pathway, and Wnt/β-catenin, as well as apoptotic and cell cycle pathways. Besides, its therapeutic implications in clinical trials are here presented.
Collapse
|
24
|
|
Li MH, Liao X, Li C, Wang TT, Sun YS, Yang K, Jiang PW, Shi ST, Zhang WX, Zhang K, Li C, Yang P. Lycorine hydrochloride induces reactive oxygen species-mediated apoptosis via the mitochondrial apoptotic pathway and the JNK signaling pathway in the oral squamous cell carcinoma HSC-3 cell line. Oncol Lett 2021; 21:236. [PMID: 33613725 DOI: 10.3892/ol.2021.12497] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Poor drug efficacy is a prominent cause of oral squamous cell carcinoma (OSCC) treatment failure. Although increased efforts in developing OSCC therapeutic strategies have been achieved in recent decades, the 5-year survival rate of patients with OSCC remains poor and effective drugs to treat OSCC are lacking. The aim of the present study was to investigate the apoptotic effect caused by lycorine hydrochloride (LH) and to identify its mechanism in the OSCC HSC-3 cell line. The findings demonstrated that LH effectively induced HSC-3 cell apoptosis and cell cycle arrest at the G0/G1 phase, resulting in the inhibition of cell proliferation. Furthermore, it was found that LH increased reactive oxygen species (ROS) production, triggered mitochondrial membrane potential (MMP) disorder, enhanced the protein expression levels of Bax, Bim, cleaved caspase-9, caspase-3 and poly(ADP-ribose) polymerase 1 and decreased Mcl-1 expression. The protein expression levels of important members of the JNK signaling pathway, including phosphorylated (p)-JNK, p-mitogen-activated protein kinase kinase 4 and p-c-Jun, were significantly increased in LH-treated cells, accompanied by an increase in ROS. However, N-acetyl cysteine (NAC), a potent antioxidant, reversed the upregulated mRNA expression of c-Jun, as well as the enhanced ROS production, the disorder of MMP and the apoptosis of HSC-3 cells induced by LH. These results suggested that LH may induce HSC-3 cell apoptosis via the ROS-mediated mitochondrial apoptotic pathway and the JNK signaling pathway, which indicated that LH may be a potential drug candidate for anti-OSCC therapy.
Collapse
Affiliation(s)
- Min-Hui Li
- Center of Science and Research, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China.,School of Basic Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xin Liao
- Center of Science and Research, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China.,School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Chen Li
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Tian-Tian Wang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Yi-Song Sun
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Kang Yang
- School of Basic Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Pei-Wen Jiang
- School of Basic Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Song-Ting Shi
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Wen-Xin Zhang
- School of Basic Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Kun Zhang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Chao Li
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan 610041, P.R. China
| | - Ping Yang
- School of Basic Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
25
|
|
Patel P, Umapathy D, Manivannan S, Nadar VM, Venkatesan R, Joseph Arokiyam VA, Pappu S, Ponnuchamy K. A doxorubicin-platinum conjugate system: impacts on PI3K/AKT actuation and apoptosis in breast cancer cells. RSC Adv 2021; 11:4818-28. [PMID: 35424411 DOI: 10.1039/d0ra06708c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In recent years, the development of a nano-conjugate system for drug delivery applications has gained attention among researchers. Keeping this in mind, in this study, we developed a doxorubicin-platinum conjugate system that targeted breast cancer cell lines. To achieve this, we developed platinum nanoparticles using polyvinylpyrrolidone (PVP). High resolution-transmission electron microscopy (HR-TEM) revealed the occurrence of octopod-shaped platinum nanoparticles. Subsequently, doxorubicin (DOX) was conjugated on the surface of the as-prepared platinum octopods via an in situ stirring method. The physicochemical characterization of the doxorubicin-platinum conjugate system revealed that the PVP of PtNPs interacts with the NH2 group of doxorubicin via electrostatic interaction/hydrogen bonding. Besides, the doxorubicin-platinum conjugate system exhibited a sustained drug release profile within the cancer cells. Furthermore, the evaluation of the in vitro anticancer efficacy of the doxorubicin-platinum conjugate system in breast cancer cells (MCF-7 and MDA-MB-231) unveiled the induction of apoptosis via intracellular ROS and DNA damage, rather than free DOX and PtNPs. Remarkably, we also perceived that the doxorubicin-platinum conjugate system was strong enough to down-regulate the PI3K/AKT signalling pathway. As a result, the tumour suppressor gene PTEN was activated, which led to the stimulation of a mitochondrion-based intrinsic apoptotic pathway and its downstream caspases, triggering cell death. Hence, our findings suggested that a biologically stable doxorubicin-platinum conjugate system could be an imperative therapeutic agent for anticancer therapy in the near future.
Collapse
Affiliation(s)
- Puja Patel
- Food Chemistry and Molecular Cancer Biology Lab, Department of Animal Health and Management, Alagappa University Karaikudi 630 003 India
| | - Devan Umapathy
- Molecular Oncology Lab, Department of Biochemistry, Bharathidasan University Tiruchirappalli 620 024 Tamil Nadu India
| | - Selvambigai Manivannan
- Department of Biomedical Science, Centre for Membrane Interactions and Dynamics (CMIAD), The University of Sheffield Western Bank Sheffield S10 2TN UK
| | - Vinita Manimaran Nadar
- Food Chemistry and Molecular Cancer Biology Lab, Department of Animal Health and Management, Alagappa University Karaikudi 630 003 India
| | - Rajiu Venkatesan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University Hangzhou 310027 China
| | | | - Srinivasan Pappu
- Phage Therapy and Molecular Biology Lab, Department of Animal Health and Management, Alagappa University Karaikudi 630003 Tamil Nadu India
| | - Kumar Ponnuchamy
- Food Chemistry and Molecular Cancer Biology Lab, Department of Animal Health and Management, Alagappa University Karaikudi 630 003 India
| |
Collapse
|
26
|
|
Abstract
Holamine and funtumine, steroidal alkaloids with strong and diverse pharmacological activities are commonly found in the Apocynaceae family of Holarrhena. The selective anti-proliferative and cell cycle arrest effects of holamine and funtumine on cancer cells have been previously reported. The present study evaluated the anti-proliferative mechanism of action of these two steroidal alkaloids on cancer cell lines (HT-29, MCF-7 and HeLa) by exploring the mitochondrial depolarization effects, reactive oxygen species (ROS) induction, apoptosis, F-actin perturbation, and inhibition of topoisomerase-I. The apoptosis-inducing effects of the compounds were studied by flow cytometry using the APOPercentageTM dye and Caspase-3/7 Glo assay kit. The two compounds showed a significantly greater cytotoxicity in cancer cells compared to non-cancer (normal) fibroblasts. The observed antiproliferative effects of the two alkaloids presumably are facilitated through the stimulation of apoptosis. The apoptotic effect was elicited through the modulation of mitochondrial function, elevated ROS production, and caspase-3/7 activation. Both compounds also induced F-actin disorganization and inhibited topoisomerase-I activity. Although holamine and funtumine appear to have translational potential for the development of novel anticancer agents, further mechanistic and molecular studies are recommended to fully understand their anticancer effects.
Collapse
Affiliation(s)
- Jelili A. Badmus
- Department of Medical Biosciences, University of the Western Cape, 7535 Bellville, Western Cape, South Africa; (J.A.B.); (O.E.E.)
| | - Okobi E. Ekpo
- Department of Medical Biosciences, University of the Western Cape, 7535 Bellville, Western Cape, South Africa; (J.A.B.); (O.E.E.)
| | - Jyoti R. Sharma
- DSI/Mintek-Nanotechnology Innovation Centre-BioLabels Node, Department of Biotechnology, University of the Western Cape, 7535 Bellville, Western Cape, South Africa; (J.R.S.); (N.R.S.S.); (M.M.)
| | - Nicole Remaliah S. Sibuyi
- DSI/Mintek-Nanotechnology Innovation Centre-BioLabels Node, Department of Biotechnology, University of the Western Cape, 7535 Bellville, Western Cape, South Africa; (J.R.S.); (N.R.S.S.); (M.M.)
| | - Mervin Meyer
- DSI/Mintek-Nanotechnology Innovation Centre-BioLabels Node, Department of Biotechnology, University of the Western Cape, 7535 Bellville, Western Cape, South Africa; (J.R.S.); (N.R.S.S.); (M.M.)
| | - Ahmed A. Hussein
- Department of Chemistry, Cape Peninsula University of Technology, 7535 Bellville, Western Cape, South Africa;
| | - Donavon C. Hiss
- Department of Medical Biosciences, University of the Western Cape, 7535 Bellville, Western Cape, South Africa; (J.A.B.); (O.E.E.)
- Correspondence:
| |
Collapse
|
27
|
|
Lan H, Fang-liang Z, Pan Z, Xian-wen W, Yi-lan J, Ying-chun H, Duan-fang L, De-liang C. Yi Qi Jie Du Formula and Salinomycin Combination Treatment Mediates Nasopharyngeal Carcinoma Stem Cell Proliferation, Migration and Apoptosis via CD44/Ras Signaling Pathway. Digital Chinese Medicine 2020; 3:297-308. [DOI: 10.1016/j.dcmed.2020.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
28
|
|
Khodadust R, Alpsoy A, Ünsoy G, GÜndÜz U. Poly (I:C)- and doxorubicin-loaded magnetic dendrimeric nanoparticles affect the apoptosis-related gene expressions in MCF-7 cells. ACTA ACUST UNITED AC 2020; 44:133-44. [PMID: 32922121 DOI: 10.3906/biy-1912-71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Use of nanoparticles as drug carrier vectors has great potential to circumvent the limitations associated with chemotherapy, including drug resistance and destructive side effects. For this purpose, magnetic generation 4 dendrimeric nanoparticles were prepared to carry chemotherapeutic agent doxorubicin (G4-DOX) and immune modulator polyinosinic:polycytidylic acid [Poly(I:C)]. As previously reported, DOX and Poly(I:C) was loaded onto G4 nanoparticles (PIC-G4-DOX). Cellular internalization study using confocal microscopy demonstrated high levels of cellular internalization of PIC-G4-DOX nanoparticles by MCF-7 cells. This resulted in higher efficacy of PIC-G4-DOX nanoparticles in killing MCF-7 breast cancer cells. Alteration in the expression levels of selected genes was determined by RT-qPCR analyses. Proapoptotic NOXA, PUMA, and BAX genes were upregulated, and SURVIVIN, APOLLON, and BCL-2 genes were downregulated, indicating the cell-killing effectiveness of PIC-G4-DOX nanoparticles. Gene expression analysis provided some insights into the possible molecular mechanisms on cytotoxicity of DOX and Poly(I:C) delivered through G4 magnetic nanoparticles. The results demonstrated that PIC-G4-DOX can be useful for targeted delivery affecting apoptotic pathways, resulting in an advanced degree of cancer-cell–killing. They are promising for targeting cancer-cells because of their stability, biocompatibility, higher internalization, and toxicity.
Collapse
Affiliation(s)
- Rouhollah Khodadust
- Department of Biotechnology, Middle East Technical University, Ankara Turkey.,Department of Biotechnology, Hamidiye Health Science Institute, University of Health Science-Turkey, İstanbul Turkey
| | - Aktan Alpsoy
- Department of Biological Sciences, Middle East Technical University, Ankara Turkey
| | - Gözde Ünsoy
- Department of Biotechnology, Middle East Technical University, Ankara Turkey
| | - Ufuk GÜndÜz
- Department of Biotechnology, Middle East Technical University, Ankara Turkey.,Department of Biological Sciences, Middle East Technical University, Ankara Turkey
| |
Collapse
|
29
|
|
Zhang H, Wang Y, Mehmood K, Chang YF, Tang Z, Li Y. Treatment of tibial dyschondroplasia with traditional Chinese medicines: "Lesson and future directions". Poult Sci 2020; 99:6422-33. [PMID: 33248557 DOI: 10.1016/j.psj.2020.08.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tibial dyschondroplasia (TD) is a metabolic tibiotarsal bone disease in rapidly growing birds throughout the world, which is characterized by gait disorders, reduced growth, and in an unrecoverable lameness in many cases. The short production cycle in chickens, long metabolism cycle in most of the drugs with the severe drug residue, and high treatment cost severely restrict the enthusiasm for the treatment of TD. Traditional Chinese medicine (TCM) has been used for the prevention, treatment, and cure of avian bone diseases. Previously, a couple of traditional Chinese medicines has been reported being useful in treating TD. This review will discuss the TCM used in TD and the alternative TCM to treat TD. Selecting a TCM approach and its pharmacologic effects on TD chickens mainly focused on the differentiation, proliferation, and apoptosis of chondrocytes, angiogenesis, matrix metabolism, oxidative damage, cytokines, and calcification of cartilage in tibia.
Collapse
|
30
|
|
Zhang Y, Zhu M, Krishna Mohan S, Hao Z. Crocin treatment promotes the oxidative stress and apoptosis in human thyroid cancer cells FTC-133 through the inhibition of STAT/JAK signaling pathway. J Biochem Mol Toxicol 2021; 35:e22608. [PMID: 32886819 DOI: 10.1002/jbt.22608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Thyroid cancer is the most frequent endocrine malignancy, which accounts for nearly 1% of all the cancer worldwide. Crocin has a diverse biological function, such as anti-cancer, anti-inflammatory and antioxidant functions, specifically in the respiratory related diseases. Using in vitro techniques, this work was intended to illuminate the anti-cancer effect of crocin in follicular thyroid carcinoma (FTC) (FT 133 cells), and the potential molecular mechanism convoluted. The outcome of the present work showed that crocin was able to prevent the proliferation and triggering the apoptosis in a dose-dependent mode, of FTC-133 cells by methyl thiazolyldiphenyl-tetrazolium bromide and staining assay (acridine orange/propiduim iodide [PI], 4',6-diamidino-2-phenylindole, and PI dye). Crocin did not show toxicity to the normal thyroid (PCCL3) cells. Crocin-induced reactive oxygen species, mitochondrial membrane potential activity, caspase-8 and -9, lipid peroxidation (thiobarbituric acid reactive substances) activity while suppressing antioxidant activity (superoxide dismutase, catalase, and glutathione) in FTC-133 cells. In addition, crocin was also participated in a halting of the proteins related to cell cycle, cyclin D1, and pro-apoptotic proteins; Bax and caspase-3 expression, together with the elevation of anti-apoptotic factor Bcl-2. Further, crocin have a dual inhibition of two major pathways, nuclear factor-κB, extracellular signal-regulated kinase, and janus kinase-signal transducer and activator of transcription signaling pathways. In conclusion, crocin can inhibit follicular thyroid carcinoma proliferation and promote cell apoptosis.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Head and Neck Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Meng Zhu
- Department of Head and Neck Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Surapaneni Krishna Mohan
- Department of Biochemistry, Panimalar Medical College Hospital & Research Institute, Chennai, Tamil Nadu, India
| | - Zhi Hao
- Department of Head and Neck Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| |
Collapse
|
31
|
|
Saralamma VVG, Vetrivel P, Lee HJ, Kim SM, Ha SE, Murugesan R, Kim EH, Heo JD, Kim GS. Comparative proteomic analysis uncovers potential biomarkers involved in the anticancer effect of Scutellarein in human gastric cancer cells. Oncol Rep 2020; 44:939-58. [PMID: 32705238 DOI: 10.3892/or.2020.7677] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Scutellarein (SCU), a flavone that belongs to the flavonoid family and abundantly present in Scutellaria baicalensis a flowering plant in the family Lamiaceae, has been reported to exhibit anticancer effects in several cancer cell lines including gastric cancer (GC). Although our previous study documented the mechanisms of Scutellarein‑induced cytotoxic effects, the literature shows that the proteomic changes that are associated with the cellular response to SCU have been poorly understood. To avoid adverse side‑effects and significant toxicity of chemotherapy in patients who react poorly, biomarkers anticipating therapeutic responses are imperative. In the present study, we utilized a comparative proteomic analysis to identify proteins associated with Scutellarein (SCU)‑induced cell death in GC cells (AGS and SNU484), by integrating two‑dimensional gel electrophoresis (2‑DE), mass spectrometry (MS), and bioinformatics to analyze the proteins. Proteomic analysis between SCU‑treated and DMSO (control) samples successfully identified 41 (AGS) and 31 (SNU484) proteins by MALDI‑TOF/MS analysis and protein database search. Comparative proteomics analysis between AGS and SNU484 cells treated with SCU revealed a total of 7 protein identities commonly expressed and western blot analysis validated a subset of identified critical proteins, which were consistent with those of the 2‑DE outcome. Molecular docking studies also confirmed the binding affinity of SCU towards these critical proteins. Phosphatidylinositol 4,5‑bisphosphate 3‑kinase catalytic subunit β isoform (PIK3CB) protein expression was accompanied by a distinct group of cellular functions, including cell growth, and proliferation. Cancerous inhibitor of protein phosphatase 2A (CIP2A), is one of the oncogenic molecules that have been shown to promote tumor growth and resistance to apoptosis and senescence‑inducing therapies. In the present study, both PIK3CB and CIP2A proteins were downregulated in SCU‑treated cells, which boosts our previous results of SCU to induce apoptosis and inhibits GC cell growth by regulating these critical proteins. The comparative proteomic analysis has yielded candidate biomarkers of response to SCU treatment in GC cell models and further validation of these biomarkers will help the future clinical development of SCU as a novel therapeutic drug.
Collapse
Affiliation(s)
- Venu Venkatarame Gowda Saralamma
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Preethi Vetrivel
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Ho Jeong Lee
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, Jinju, Gyeongnam 52834, Republic of Korea
| | - Seong Min Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Sang Eun Ha
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Rajeswari Murugesan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641043, India
| | - Eun Hee Kim
- Department of Nursing Science, International University of Korea, Jinju, Gyeongnam 52833, Republic of Korea
| | - Jeong Doo Heo
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, Jinju, Gyeongnam 52834, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| |
Collapse
|
32
|
|
Zhang X, Cheng Y, Zhou Q, Huang H, Dong Y, Yang Y, Zhao M, He Q. The Effect of Chinese Traditional Medicine Huaiqihuang (HQH) on the Protection of Nephropathy. Oxid Med Cell Longev 2020; 2020:2153912. [PMID: 32655761 DOI: 10.1155/2020/2153912] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022]
Abstract
Kidney disease is one of the common diseases with high morbidity and high mortality, which brings a huge burden to the society and the patient's family. The pathogenesis, treatment, and prognosis of kidney diseases are related to oxidative stress, inflammation, mitochondrial damage, and immune dysfunction. However, existing treatments always cause some damage to the kidneys. Kidney disease and immunosuppressant used together often lead to drug toxicity, patients with weakened immunity, organic rupture of the normal structure of the kidney, damage to the physiological function of the kidney, etc. Huaiqihuang is a kind of traditional Chinese medicine with a history of more than one thousand years. According to research, Robinia pseudoacacia can regulate the immune function by regulating oxidative stress, calcium inflow, and mitochondrial ATP. At the same time, it is also involved in regulating the ways of cell death, such as apoptosis, autophagy, ferroptosis, pyroptosis, and clockophagy, to reduce kidney damage, which has important clinical value. This article reviews the exact mechanism and clinical application of Huaiqihuang in different types of nephropathy. The aim is to provide new ideas for the treatment of clinical nephropathy.
Collapse
|
33
|
|
Li J, Han B, Sun G, Zheng Z, Mu Y, Chi J. Chinese patent medicine (Jinlong Capsule) for gastric cancer: Protocol for a systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e20532. [PMID: 32502010 DOI: 10.1097/MD.0000000000020532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND JLC has been widely applied as a promising adjunctive drug for GC. However, the exact effects and safety of JLC have yet to be systematically investigated. We aimed to summarize the efficacy and safety of JLC for the treatment of advanced GC through the meta-analysis, in order to provide scientific reference for the design of future clinical trials. METHODS The protocol followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols. Relevant randomized controlled trials were searched from Cochrane Library, PubMed, Web of Science (WOS), Excerpt Medica Database (Embase), Chinese Biomedical Literature Database (CBM), China National Knowledge Infrastructure (CNKI), China Scientific Journal Database (VIP), and Wanfang Database. Papers in English or Chinese published from their inception to January 2020 will be included without any restrictions.Study selection and data extraction will be performed independently by 2 investigators. The clinical outcomes including overall response rate, complete response rate, overall survival, Disease-free survival, quality of life (QoL), immune function, and adverse events, were systematically evaluated. Review Manager 5.3 and Stata 14.0 were used for data analysis, and the quality of the studies was also evaluated. RESULTS AND CONCLUSION The findings of this research will be published in a peer-reviewed journal, and provide more evidence-based guidance in clinical practice. INTERNATIONAL PLATFORM OF REGISTERED SYSTEMATIC REVIEW AND META-ANALYSIS PROTOCOLS (INPLASY) REGISTRATION NUMBER:: INPLASY202040105. URL: https://inplasy.com/inplasy-2020-4-0105/.
Collapse
Affiliation(s)
| | | | - Guangzong Sun
- Department of Emergency, People's Hospital of Weifang Binhai Economic and Technological Development Zone
| | - Zhong Zheng
- Department of Gastroenterology, Weifang People's Hospital, Weifang
| | - Ying Mu
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng
| | - Jingxia Chi
- Quality Management Office, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong Province, China
| |
Collapse
|
34
|
|
Neergheen VS, Hip Kam A, Pem Y, Ramsaha S, Bahorun T. Regulation of cancer cell signaling pathways as key events for therapeutic relevance of edible and medicinal mushrooms. Semin Cancer Biol 2020. [DOI: 10.1016/j.semcancer.2020.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 12/25/2022]
|
35
|
|
L SW, Lee CH, Lin MS, Chi CW, Chen YJ, Wang GS, Liao KW, Chiu LP, Wu SH, Huang DM, Chen L, Shen YS. ZnO Nanoparticles Induced Caspase-Dependent Apoptosis in Gingival Squamous Cell Carcinoma through Mitochondrial Dysfunction and p70S6K Signaling Pathway. Int J Mol Sci 2020; 21:E1612. [PMID: 32111101 DOI: 10.3390/ijms21051612] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/07/2020] [Accepted: 02/20/2020] [Indexed: 12/17/2022] Open
Abstract
Zinc oxide nanoparticles (ZnO-NPs) are increasingly used in sunscreens, food additives, pigments, rubber manufacture, and electronic materials. Several studies have shown that ZnO-NPs inhibit cell growth and induce apoptosis by the production of oxidative stress in a variety of human cancer cells. However, the anti-cancer property and molecular mechanism of ZnO-NPs in human gingival squamous cell carcinoma (GSCC) are not fully understood. In this study, we found that ZnO-NPs induced growth inhibition of GSCC (Ca9-22 and OECM-1 cells), but no damage in human normal keratinocytes (HaCaT cells) and gingival fibroblasts (HGF-1 cells). ZnO-NPs caused apoptotic cell death of GSCC in a concentration-dependent manner by the quantitative assessment of oligonucleosomal DNA fragmentation. Flow cytometric analysis of cell cycle progression revealed that sub-G1 phase accumulation was dramatically induced by ZnO-NPs. In addition, ZnO-NPs increased the intracellular reactive oxygen species and specifically superoxide levels, and also decreased the mitochondrial membrane potential. ZnO-NPs further activated apoptotic cell death via the caspase cascades. Importantly, anti-oxidant and caspase inhibitor clearly prevented ZnO-NP-induced cell death, indicating the fact that superoxide-induced mitochondrial dysfunction is associated with the ZnO-NP-mediated caspase-dependent apoptosis in human GSCC. Moreover, ZnO-NPs significantly inhibited the phosphorylation of ribosomal protein S6 kinase (p70S6K kinase). In a corollary in vivo study, our results demonstrated that ZnO-NPs possessed an anti-cancer effect in a zebrafish xenograft model. Collectively, these results suggest that ZnO-NPs induce apoptosis through the mitochondrial oxidative damage and p70S6K signaling pathway in human GSCC. The present study may provide an experimental basis for ZnO-NPs to be considered as a promising novel anti-tumor agent for the treatment of gingival cancer.
Collapse
Affiliation(s)
- Shih-Wei Wang
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan;
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Hsing Lee
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ming-Shen Lin
- Department of Water Resources and Environmental Engineering, Tamkang University, New Taipei City 251, Taiwan;
| | - Chih-Wen Chi
- Department of Nursing, MacKay Medical College, New Taipei City 252, Taiwan;
- Department of Medical Research, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Yu-Jen Chen
- MacKay Junior College of Medicine, Nursing, and Management, Taipei 112, Taiwan;
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Guo-Shou Wang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan; (G.-S.W.); (K.-W.L.)
- Department of Orthopaedics, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan; (G.-S.W.); (K.-W.L.)
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Li-Pin Chiu
- General Education Center, University of Taipei, Taipei 100, Taiwan;
- Division of General Surgery, Taipei City Hospital, Taipei 103, Taiwan
| | - Shu-Hui Wu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 350, Taiwan;
| | - Dong-Ming Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 350, Taiwan;
- Correspondence: (D.-M.H.); (L.C.); (Y.-S.S.); Tel.: +886-37-246-166 (ext. 38105) (D.-M.H.); +886-2-2621-5656 (ext. 2682) (L.C.); +886-2-2636-0303 (ext. 1422) (Y.-S.S.)
| | - Luke Chen
- Department of Water Resources and Environmental Engineering, Tamkang University, New Taipei City 251, Taiwan;
- Correspondence: (D.-M.H.); (L.C.); (Y.-S.S.); Tel.: +886-37-246-166 (ext. 38105) (D.-M.H.); +886-2-2621-5656 (ext. 2682) (L.C.); +886-2-2636-0303 (ext. 1422) (Y.-S.S.)
| | - Yung-Shuen Shen
- Institute of Geriatric Welfare Technology and Science, MacKay Medical College, New Taipei City 252, Taiwan
- Correspondence: (D.-M.H.); (L.C.); (Y.-S.S.); Tel.: +886-37-246-166 (ext. 38105) (D.-M.H.); +886-2-2621-5656 (ext. 2682) (L.C.); +886-2-2636-0303 (ext. 1422) (Y.-S.S.)
| |
Collapse
|
36
|
|
Wang TH, Leu YL, Chen CC, Shieh TM, Lian JH, Chen CY. Psorachromene Suppresses Oral Squamous Cell Carcinoma Progression by Inhibiting Long Non-coding RNA GAS5 Mediated Epithelial-Mesenchymal Transition. Front Oncol 2019; 9:1168. [PMID: 31750253 DOI: 10.3389/fonc.2019.01168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/17/2019] [Indexed: 12/14/2022] Open
Abstract
The extract of the seeds of Psoralea corylifolia Linn. (P. corylifolia) have been shown to display anti-tumor activity. However, the prospects of the active compounds from this plant in the treatment of oral squamous cell carcinoma (OSCC) remains unclear. In the present study, the antitumor effects of psorachromene, a flavonoid extracted from the seeds of P. corylifolia, were investigated using cells and animal models of OSCC; the downstream regulatory mechanisms were also elucidated. The results showed that psorachromene significantly repressed cell proliferation, migration, and invasiveness and increased the toxic effects of chemotherapeutic agents against OSCC cells. The repressive effects of psorachromene were attributable to the inhibition of EGFR-Slug signaling, and the induction of G2/M arrest and apoptosis in the OSCC cells. Additionally, we found that psorachromene induced the expression of tumor suppressor long non-coding ribonucleic acid (RNA) growth arrest-specific transcript 5 (GAS5) and the activation of its downstream anticancer mechanisms. Animal experiments also showed noticeable inhibition of tumor growth, without significant physiological toxicity. The findings indicate that psorachromene displays anti-tumor activity in OSCC, and warrants further investigation as a potential agent for clinical application.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan.,Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Tao-Yuan, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan.,Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chin-Chuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Tao-Yuan, Taiwan
| | - Tzong-Ming Shieh
- Department of Dental Hygiene, China Medical University, Taichung, Taiwan
| | - Jang-Hau Lian
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| |
Collapse
|
37
|
|
Niu Y, Zhang J, Tong Y, Li J, Liu B. Physcion 8-O-β-glucopyranoside induced ferroptosis via regulating miR-103a-3p/GLS2 axis in gastric cancer. Life Sci 2019; 237:116893. [DOI: 10.1016/j.lfs.2019.116893] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [What about the content of this article? (0)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/10/2019] [Accepted: 09/19/2019] [Indexed: 01/18/2023]
|