1
|
Gao X, Jin Y, Zhu W, Wu X, Wang J, Guo C. Regulation of Eukaryotic Translation Initiation Factor 4E as a Potential Anticancer Strategy. J Med Chem 2023; 66:12678-12696. [PMID: 37725577 DOI: 10.1021/acs.jmedchem.3c00636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Eukaryotic translation initiation factors (eIFs) are highly expressed in cancer cells, especially eIF4E, the central regulatory node driving cancer cell growth and a potential target for anticancer drugs. eIF4E-targeting strategies primarily focus on inhibiting eIF4E synthesis, interfering with eIF4E/eIF4G interactions, and targeting eIF4E phosphorylation and peptide inhibitors. Although some small-molecule inhibitors are in clinical trials, no eIF4E inhibitors are available for clinical use. We provide an overview of the regulatory mechanisms of eIF4E and summarize the progress in developing and discovering eIF4E inhibitor strategies. We propose that interference with eIF4E/eIF4G interactions will provide a new perspective for the design of eIF4E inhibitors and may be a preferred strategy.
Collapse
Affiliation(s)
- Xintao Gao
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yonglong Jin
- The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Wenyong Zhu
- Department of Thoracic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Xiaochen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jing Wang
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou 014030, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
2
|
Du L, Wang D, Nagle PW, Groen AAH, Zhang H, Muijs CT, Plukker JTM, Coppes RP. Role of mTOR through Autophagy in Esophageal Cancer Stemness. Cancers (Basel) 2022; 14:cancers14071806. [PMID: 35406578 PMCID: PMC9040713 DOI: 10.3390/cancers14071806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 02/05/2023] Open
Abstract
Esophageal cancer (EC) is a highly aggressive disease with a poor prognosis. Therapy resistance and early recurrences are major obstacles in reaching a better outcome. Esophageal cancer stem-like cells (CSCs) seem tightly related with chemoradiation resistance, initiating new tumors and metastases. Several oncogenic pathways seem to be involved in the regulation of esophageal CSCs and might harbor novel therapeutic targets to eliminate CSCs. Previously, we identified a subpopulation of EC cells that express high levels of CD44 and low levels of CD24 (CD44+/CD24-), show CSC characteristics and reside in hypoxic niches. Here, we aim to clarify the role of the hypoxia-responding mammalian target of the rapamycin (mTOR) pathway in esophageal CSCs. We showed that under a low-oxygen culture condition and nutrient deprivation, the CD44+/CD24- population is enriched. Since both low oxygen and nutrient deprivation may inhibit the mTOR pathway, we next chemically inhibited the mTOR pathway using Torin-1. Torin-1 upregulated SOX2 resulted in an enrichment of the CD44+/CD24- population and increased sphere formation potential. In contrast, stimulation of the mTOR pathway using MHY1485 induced the opposite effects. In addition, Torin-1 increased autophagic activity, while MHY1485 suppressed autophagy. Torin-1-mediated CSCs upregulation was significantly reduced in cells treated with autophagy inhibitor, hydroxychloroquine (HCQ). Finally, a clearly defined CD44+/CD24- CSC population was detected in EC patients-derived organoids (ec-PDOs) and here, MHY1485 also reduced this population. These data suggest that autophagy may play a crucial role in mTOR-mediated CSCs repression. Stimulation of the mTOR pathway might aid in the elimination of putative esophageal CSCs.
Collapse
Affiliation(s)
- Liang Du
- Section Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (L.D.); (D.W.); (P.W.N.); (A.A.H.G.)
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
- Graduate School, Shantou University Medical College, Shantou 515041, China
| | - Da Wang
- Section Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (L.D.); (D.W.); (P.W.N.); (A.A.H.G.)
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Peter W. Nagle
- Section Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (L.D.); (D.W.); (P.W.N.); (A.A.H.G.)
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
- Medical Research Council (MRC) Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Andries A. H. Groen
- Section Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (L.D.); (D.W.); (P.W.N.); (A.A.H.G.)
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Hao Zhang
- Department of Pathology, Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China;
- Department of General Surgery, First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Christina T. Muijs
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - John Th. M. Plukker
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Robert P. Coppes
- Section Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (L.D.); (D.W.); (P.W.N.); (A.A.H.G.)
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
- Correspondence:
| |
Collapse
|
3
|
Pang Y, Liu L, Mu H, Priya Veeraraghavan V. Nobiletin promotes osteogenic differentiation of human osteoblastic cell line (MG-63) through activating the BMP-2/RUNX-2 signaling pathway. Saudi J Biol Sci 2021; 28:4916-4920. [PMID: 34466066 PMCID: PMC8381068 DOI: 10.1016/j.sjbs.2021.06.070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/06/2021] [Accepted: 06/24/2021] [Indexed: 11/01/2022] Open
Abstract
Nobiletin (NOB) is polymethoxy flavonoids, which plentifully there in Citrus depressa and they demonstrate numerous pharmacological effects. NOB has an anti-proliferative effect, attenuates ovalbumin-treated eosinophilic airway inflammation and Type II collagen treated arthritis. NOB noticeably inhibits bone resorption and renovates bone loss in mice model, but role of NOB in bone metabolism is unclear. Human bone is a important organ that sustains its homeostasis among bone resorpting osteoclasts and bone developing osteoblasts. The balances of among these two kind of cell outcomes are implicated in bone remodeling. The current study designed to explore possessions of NOB on differentiation and proliferation of MG-63 cells and contribution of morphogenetic protein signaling. Cell proliferation was analyzed by MTT, mineralization analysis by alizarin red staining and morphogenetic signaling protein by RT-PCR. No stimulus outcome of NOB on cell proliferation was found at days of 1, 3 and 7. Accumulation of calcium was augmented after that treatment of NOB. The mRNA expression of BMP-2, COL-I, ALP, OCN, RUNX2 and COL1A1 augmented markedly with NOB supplement. Hence, NOB can stimulate osteogenic differentiation of MG-63, almost certainly by promoting RUNX2 and BMP-2 signaling and this result might provide to its action on stimulation of osteoblast development, differentiation and augments of bone mass.
Collapse
Affiliation(s)
- Ying Pang
- Dental Clinic, Cangzhou Central Hospital, Cangzhou City, Hebei Province 061000, China
| | - Lili Liu
- Dental Clinic, Cangzhou Central Hospital, Cangzhou City, Hebei Province 061000, China
| | - Hong Mu
- Dental Clinic, Cangzhou Central Hospital, Cangzhou City, Hebei Province 061000, China
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
| |
Collapse
|
4
|
Mardanshahi A, Gharibkandi NA, Vaseghi S, Abedi SM, Molavipordanjani S. The PI3K/AKT/mTOR signaling pathway inhibitors enhance radiosensitivity in cancer cell lines. Mol Biol Rep 2021; 48:1-14. [PMID: 34357550 DOI: 10.1007/s11033-021-06607-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/29/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Radiotherapy is one of the most common types of cancer treatment modalities. Radiation can affect both cancer and normal tissues, which limits the whole delivered dose. It is well documented that radiation activates phosphatidylinositol 3-kinase (PI3K) and AKT signaling pathway; hence, the inhibition of this pathway enhances the radiosensitivity of tumor cells. The mammalian target of rapamycin (mTOR) is a regulator that is involved in autophagy, cell growth, proliferation, and survival. CONCLUSION The inhibition of mTOR as a downstream mediator of the PI3K/AKT signaling pathway represents a vital option for more effective cancer treatments. The combination of PI3K/AKT/mTOR inhibitors with radiation can increase the radiosensitivity of malignant cells to radiation by autophagy activation. Therefore, this review aims to discuss the impact of such inhibitors on the cell response to radiation.
Collapse
Affiliation(s)
- Alireza Mardanshahi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasrin Abbasi Gharibkandi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Samaneh Vaseghi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sajjad Molavipordanjani
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
5
|
Lim JJ, Li X, Lehmler HJ, Wang D, Gu H, Cui JY. Gut Microbiome Critically Impacts PCB-induced Changes in Metabolic Fingerprints and the Hepatic Transcriptome in Mice. Toxicol Sci 2021; 177:168-187. [PMID: 32544245 DOI: 10.1093/toxsci/kfaa090] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Polychlorinated biphenyls (PCBs) are ubiquitously detected and have been linked to metabolic diseases. Gut microbiome is recognized as a critical regulator of disease susceptibility; however, little is known how PCBs and gut microbiome interact to modulate hepatic xenobiotic and intermediary metabolism. We hypothesized the gut microbiome regulates PCB-mediated changes in the metabolic fingerprints and hepatic transcriptome. Ninety-day-old female conventional and germ-free mice were orally exposed to the Fox River Mixture (synthetic PCB mixture, 6 or 30 mg/kg) or corn oil (vehicle control, 10 ml/kg), once daily for 3 consecutive days. RNA-seq was conducted in liver, and endogenous metabolites were measured in liver and serum by LC-MS. Prototypical target genes of aryl hydrocarbon receptor, pregnane X receptor, and constitutive androstane receptor were more readily upregulated by PCBs in conventional conditions, indicating PCBs, to the hepatic transcriptome, act partly through the gut microbiome. In a gut microbiome-dependent manner, xenobiotic, and steroid metabolism pathways were upregulated, whereas response to misfolded proteins-related pathways was downregulated by PCBs. At the high PCB dose, NADP, and arginine appear to interact with drug-metabolizing enzymes (ie, Cyp1-3 family), which are highly correlated with Ruminiclostridium and Roseburia, providing a novel explanation of gut-liver interaction from PCB-exposure. Utilizing the Library of Integrated Network-based Cellular Signatures L1000 database, therapeutics targeting anti-inflammatory and endoplasmic reticulum stress pathways are predicted to be remedies that can mitigate PCB toxicity. Our findings demonstrate that habitation of the gut microbiota drives PCB-mediated hepatic responses. Our study adds knowledge of physiological response differences from PCB exposure and considerations for further investigations for gut microbiome-dependent therapeutics.
Collapse
Affiliation(s)
- Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195
| | - Xueshu Li
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa 52242; and
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, Iowa 52242; and
| | - Dongfang Wang
- Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, School of Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195
| |
Collapse
|
6
|
Rhaponticin suppresses osteosarcoma through the inhibition of PI3K-Akt-mTOR pathway. Saudi J Biol Sci 2021; 28:3641-3649. [PMID: 34220214 PMCID: PMC8241634 DOI: 10.1016/j.sjbs.2021.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 02/04/2023] Open
Abstract
Osteosarcoma is the frequent pediatric bone cancer where pediatric osteosarcoma incidences are more than 10% within the population. Most of the patients with osteosarcoma fall within the age of 15-30 years. Therefore, in this research, we examined the anticancer effect of Rhaponticin against the human osteosarcoma (MG-63) cells. The cytotoxicity of Rhaponticin on the MC3T3-E1 and MG-63 cells was examined through the MTT assay. The intracellular ROS accumulation, cell nuclear morphological alterations, apoptotic cell death and nuclear damages, and MMP status of Rhaponticin administered MG-63 cells were inspected by fluorescent staining techniques. The cell migration was assessed through scratch assay. The mRNA expressions of PI3K-Akt-mTOR signaling proteins were studied by RT-PCR analysis. Rhaponticin showed potent cytotoxicity, substantially inhibited the MG-63 cell growth, and displayed morphological alterations. However, rhaponticin did not affect the MC3T3-E1 cell viability. Rhaponticin administered MG-63 cells demonstrated augmented intracellular ROS accretion, weakened MMP, increased nuclear damages, and increased apoptosis. Rhaponticin effectively down-regulated the PI3K-Akt-mTOR signaling cascade in the MG-63 cells. These outcomes proved that the Rhaponticin can be a hopeful chemotherapeutic agent in the future to treat human osteosarcoma.
Collapse
|
7
|
Huang H, Zhou M, Ruan L, Wang D, Lu H, Zhang J, Chen J, Hu Y, Chai Z. AMPK mediates the neurotoxicity of iron oxide nanoparticles retained in mitochondria or lysosomes. Metallomics 2020; 11:1200-1206. [PMID: 31241124 DOI: 10.1039/c9mt00103d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Environmental factors may play a critical role in the etiology and pathogenesis of Parkinson's disease (PD). However, the association of PD with specific chemical species remains largely unknown. Here we prepared three kinds of iron oxide nanoparticles and examined their cytotoxicity in a cellular model of PD. We found that lysosome-targeted nanoparticles showed significant cytotoxicity in SH-SY5Y cells. Inhibition of AMPK could aggravate the neurotoxicity of lysosome-targeted nanoparticles as well as mitochondrion-targeted nanoparticles. Alteration of mitochondrial membrane potentials was found to be in agreement with the neurotoxicity of iron nanoparticles. These results suggested an important role of AMPK in regulating iron nanoparticle-associated neurotoxicity.
Collapse
Affiliation(s)
- Hui Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengxue Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lifo Ruan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dongqing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China.
| | - Huiru Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China.
| | - Jiayu Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China.
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifang Chai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multidisciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
8
|
Kao CH, Ryu SW, Kim MJ, Wen X, Wimalarathne O, Paull TT. Growth-Regulated Hsp70 Phosphorylation Regulates Stress Responses and Prion Maintenance. Mol Cell Biol 2020; 40:e00628-19. [PMID: 32205407 PMCID: PMC7261718 DOI: 10.1128/mcb.00628-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/05/2020] [Accepted: 03/18/2020] [Indexed: 11/20/2022] Open
Abstract
Maintenance of protein homeostasis in eukaryotes under normal growth and stress conditions requires the functions of Hsp70 chaperones and associated cochaperones. Here, we investigate an evolutionarily conserved serine phosphorylation that occurs at the site of communication between the nucleotide-binding and substrate-binding domains of Hsp70. Ser151 phosphorylation in yeast Hsp70 (Ssa1) is promoted by cyclin-dependent kinase (Cdk1) during normal growth. Phosphomimetic substitutions at this site (S151D) dramatically downregulate heat shock responses, a result conserved with HSC70 S153 in human cells. Phosphomimetic forms of Ssa1 also fail to relocalize in response to starvation conditions, do not associate in vivo with Hsp40 cochaperones Ydj1 and Sis1, and do not catalyze refolding of denatured proteins in vitro in cooperation with Ydj1 and Hsp104. Despite these negative effects on HSC70/HSP70 function, the S151D phosphomimetic allele promotes survival of heavy metal exposure and suppresses the Sup35-dependent [PSI+ ] prion phenotype, consistent with proposed roles for Ssa1 and Hsp104 in generating self-nucleating seeds of misfolded proteins. Taken together, these results suggest that Cdk1 can downregulate Hsp70 function through phosphorylation of this site, with potential costs to overall chaperone efficiency but also advantages with respect to reduction of metal-induced and prion-dependent protein aggregate production.
Collapse
Affiliation(s)
- Chung-Hsuan Kao
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Seung W Ryu
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Min J Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Xuemei Wen
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Oshadi Wimalarathne
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Tanya T Paull
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
9
|
Cheng J, Wang X, Qiu L, Li Y, Marraiki N, Elgorban AM, Xue L. Green synthesized zinc oxide nanoparticles regulates the apoptotic expression in bone cancer cells MG-63 cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 202:111644. [PMID: 31770706 DOI: 10.1016/j.jphotobiol.2019.111644] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 12/26/2022]
Abstract
Management of degenerative spine pathologies frequently leads to the need for bone growth. Rehmanniae Radix (RR), a Chinese herbal formulation was found to exhibit numerous therapeutic properties including its potent effect against cancer cell lines. However, the underlying mechanism through which the Zinc oxide nanoparticles (ZnONPs) synthesized from Rehmanniae Radix exerts its anti-cancer activity against osteosarcoma cell line MG-63 needs to be explored. Therefore, the study was performed to evaluate the anticancer, cytotoxicity and apoptotic effectiveness of ZnONPs from RR against MG-63 cells. Characterization studies such UV-vis spectroscopy, FTIR, TEM and XRD analysis were performed. Cytotoxicity assay, mitochondrial membrane potential (MMP), morphological examination of cells and formation of reactive oxygen species (ROS), and apoptosis inducing ability of RR were evaluated by various procedures. Western blot analysis of apoptotic markers such as Bax, caspase-3 and caspase-9 were also performed. RR was found to inhibit growth of MG-63 cells at increasing dose. AO/EB staining confirmed the apoptotic efficacy of ZnONPs induced by RR in MG-63 cells. ZnONPs was also found to initiate increased generation of ROS and decreased MMP. Decreased MMP has resulted in increased levels of apoptotic proteins Bax, caspase-3 and caspase-9 and induction of apoptosis was substantiated by western blot analysis. The outcomes of the work propose that ZnONPs from RR exhibits strong anticancer action and inducing apoptosis on MG-63 cells via stimulating increased generation of ROS. Thus, ZnONPs from RR might be used as a hopeful drug target against several types of cancer cell lines.
Collapse
Affiliation(s)
- Jun Cheng
- Department of Orthopedics, Chongqing Three Gorges Central Hospital, Chongqing Province 404000, China
| | - Xiaofeng Wang
- Department of Neurosurgery, Weinan Central Hospital, Weinan, Shaanxi Province 714000, China
| | - Lei Qiu
- Department of Oncology, Zhucheng Hospital of Traditional Chinese Medicine, Zhucheng, Shandong Province 262200, China
| | - Yunkai Li
- Emergency Surgery the No.4 Hospital Jinan, Shangdong Province 250031, China
| | - Najat Marraiki
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdallah M Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Li Xue
- Department of Orthopaedics, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiao Tong University Medical School, Chengdu, Sichuan Province 610031, China.
| |
Collapse
|
10
|
Ruan L, Wang M, Zhou M, Lu H, Zhang J, Gao J, Chen J, Hu Y. Doxorubicin–Metal Coordinated Micellar Nanoparticles for Intracellular Codelivery and Chemo/Chemodynamic Therapy in Vitro. ACS APPLIED BIO MATERIALS 2019; 2:4703-4707. [DOI: 10.1021/acsabm.9b00879] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Lifo Ruan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mengxue Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huiru Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jiayu Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
11
|
Wang D, Cui L, Chang X, Guan D. Biosynthesis and characterization of zinc oxide nanoparticles from Artemisia annua and investigate their effect on proliferation, osteogenic differentiation and mineralization in human osteoblast-like MG-63 Cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 202:111652. [PMID: 31760374 DOI: 10.1016/j.jphotobiol.2019.111652] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 01/08/2023]
Abstract
The Biocompatibility and stability of nanoparticles using plants have been widely investigated due to its applications in the biomedical industry. Currently, there is a growing interest in nanoparticles in bone remodelling. Artemisia annua is an herbal plant commonly used in the treatment of various ailments. This study investigated the zinc oxide nanoparticles (ZnO NPs) using the green synthesis technique from A. annua and the effects of A. annua ZnO-NPs on osteoblast differentiation and inhibition of osteoclast formation. The formulated ZnO-NPs from A. annua were characterized by using various spectroscopic and microscopic methods Fourier transform-infrared spectroscopy (FT-IR), transmission electron microscopy (TEM), X-ray diffraction (XRD), and UV-Visible spectroscopy. The disc diffusion method was adopted to test the antimicrobial efficacy of ZnO-NPs. The viability of MG-63 cells were assayed by MTT test and Osteogenic-related assays like Real-time PCR and Mineralization assay were adopted to determine the effects of A. annua ZnO-NPs on the multiplication and differentiation of human osteoblast-like MG-63 cells. The characterization of A. annua ZnO-NPs revealed the crystalline nature with high zinc content and the presence of bioactive compounds from A. annua extract. The synthesized A. annua ZnO-NPs indicate significant antimicrobial potential. Besides, A. annua ZnO-NPs enhanced the proliferation, differentiation, and mineralization without causing significant cytotoxic impact on MG-63 cells. These effects indicate that A. annua ZnO-NPs can both stimulate bone formation via the differentiation of MG-63 cells. Hence, it was concluded that A. annua ZnO-NPs can be a promising agent for the treatment of bone deformities and bone-related diseases, however further research also required to explore the clear mechanism of A. annua ZnO-NPs in the formation and differentiation of MG-63 cells.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Lihuang Cui
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Xin Chang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Dehong Guan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China.
| |
Collapse
|
12
|
The dietary flavonoid isoliquiritigenin is a potent cytotoxin for human neuroblastoma cells. Neuronal Signal 2019; 3:NS20180201. [PMID: 32269833 PMCID: PMC7104307 DOI: 10.1042/ns20180201] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor of early childhood; it accounts for approximately 8–10% of all childhood cancers and is the most common cancer in children in the first year of life. Patients in the high-risk group have a poor prognosis, with relapses being common and often refractory to drug treatment in those that survive. Moreover, the drug treatment itself can lead to a range of long-term sequelae. Therefore, there is a critical need to identify new therapeutics for NB. Isoliquiritigenin (ISLQ) is a naturally-occurring, dietary chalcone-type flavonoid with a range of biological effects that depend on the cell type and context. ISLQ has potential as an anticancer agent. Here we show that ISLQ has potent cytotoxic effects on SK-N-BE(2) and IMR-32 human NB cells, which carry amplification of the MYCN gene, the main prognostic marker of poor survival in NB. ISLQ was found to increase cellular reactive oxygen species (ROS). The cytotoxic effect of ISLQ was blocked by small molecule inhibitors of oxidative stress-induced cell death, and by the antioxidant N-acetyl-l-cysteine (NAC). Combined treatment of either SK-N-B-E(2) or IMR-32 cells with ISLQ and the anticancer agent cisplatin resulted in loss of cell viability that was greater than that induced by cisplatin alone. This study provides proof-of-principle that ISLQ is a potent cytotoxin for MYCN-amplified human NB cells. This is an important first step in rationalizing the further study of ISLQ as a potential adjunct therapy for high-risk NB.
Collapse
|
13
|
Kumar P, Awasthi A, Nain V, Issac B, Puria R. Novel insights into TOR signalling in Saccharomyces cerevisiae through Torin2. Gene 2018; 669:15-27. [DOI: 10.1016/j.gene.2018.05.081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/06/2018] [Accepted: 05/21/2018] [Indexed: 12/18/2022]
|