101
|
Wang M, Li J, Dong S, Cai X, Simaiti A, Yang X, Zhu X, Luo J, Jiang LH, Du B, Yu P, Yang W. Silica nanoparticles induce lung inflammation in mice via ROS/PARP/TRPM2 signaling-mediated lysosome impairment and autophagy dysfunction. Part Fibre Toxicol 2020; 17:23. [PMID: 32513195 PMCID: PMC7281956 DOI: 10.1186/s12989-020-00353-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/26/2020] [Indexed: 01/26/2023] Open
Abstract
Background Wide applications of nanoparticles (NPs) have raised increasing concerns about safety to humans. Oxidative stress and inflammation are extensively investigated as mechanisms for NPs-induced toxicity. Autophagy and lysosomal dysfunction are emerging molecular mechanisms. Inhalation is one of the main pathways of exposing humans to NPs, which has been reported to induce severe pulmonary inflammation. However, the underlying mechanisms and, more specifically, the interplays of above-mentioned mechanisms in NPs-induced pulmonary inflammation are still largely obscure. Considered that NPs exposure in modern society is often unavoidable, it is highly desirable to develop effective strategies that could help to prevent nanomaterials-induced pulmonary inflammation. Results Pulmonary inflammation induced by intratracheal instillation of silica nanoparticles (SiNPs) in C57BL/6 mice was prevented by PJ34, a poly (ADP-ribose) polymerase (PARP) inhibitor. In human lung bronchial epithelial (BEAS-2B) cells, exposure to SiNPs reduced cell viability, and induced ROS generation, impairment in lysosome function and autophagic flux. Inhibition of ROS generation, PARP and TRPM2 channel suppressed SiNPs-induced lysosome impairment and autophagy dysfunction and consequent inflammatory responses. Consistently, SiNPs-induced pulmonary inflammation was prevented in TRPM2 deficient mice. Conclusion The ROS/PARP/TRPM2 signaling is critical in SiNPs-induced pulmonary inflammation, providing novel mechanistic insights into NPs-induced lung injury. Our study identifies TRPM2 channel as a new target for the development of preventive and therapeutic strategies to mitigate nanomaterials-induced lung inflammation. Graphical abstract ![]()
Collapse
Affiliation(s)
- Mingxiang Wang
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Jin Li
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Shunni Dong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, China
| | - Xiaobo Cai
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.,Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Aili Simaiti
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Xin Yang
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Xinqiang Zhu
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.,The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, P. R. China
| | - Jianhong Luo
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, P. R. China.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Binyang Du
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, China.
| | - Peilin Yu
- Department of Toxicology, and Department of Medical Oncology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.
| | - Wei Yang
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China.
| |
Collapse
|
102
|
Cho YL, Tan HWS, Saquib Q, Ren Y, Ahmad J, Wahab R, He W, Bay BH, Shen HM. Dual role of oxidative stress-JNK activation in autophagy and apoptosis induced by nickel oxide nanoparticles in human cancer cells. Free Radic Biol Med 2020; 153:173-186. [PMID: 32353482 DOI: 10.1016/j.freeradbiomed.2020.03.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 02/08/2023]
Abstract
Nickel oxide nanoparticles (NiO-NPs) are an important group of nanoparticles with increasing applications in many aspects of industry. At present, there is evidence demonstrating the cytotoxic characteristics of NiO-NPs, while the involvement of autophagy in the cytotoxicity of NiO-NPs has not been reported. In this study, we aimed to study the role of autophagy in the cytotoxicity of NiO-NPs and the underlying regulatory mechanisms. First, we provided evidence that NiO-NPs induce autophagy in human cancer cells. Second, we found that the enhanced autophagic flux by NiO-NPs via the generation of intracellular reactive oxygen species (ROS) from mitochondria and the subsequent activation of the JNK pathway. Third, we demonstrated that the activation of JNK is a main force in mediating NiO-NPs-induced apoptosis. Finally, we demonstrated that the autophagic response plays an important protective role against the cytotoxic effect of NiO-NPs. Therefore, this study identifies the dual role of oxidative stress-JNK activation in the biological effects of NiO-NPs via promoting autophagy and mediating apoptosis. Understanding the protective role of autophagy and the underlying mechanism is important for the potential application of NiO-NPs in the biomedical industry.
Collapse
Affiliation(s)
- Yik-Lam Cho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Hayden Weng Siong Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.
| | - Quaiser Saquib
- Zoology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Yi Ren
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Javed Ahmad
- Zoology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Rizwan Wahab
- Zoology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, China.
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; Faculty of Health Sciences, University of Macau, Macau.
| |
Collapse
|
103
|
Wu Y, Jin Y, Sun T, Zhu P, Li J, Zhang Q, Wang X, Jiang J, Chen G, Zhao X. p62/SQSTM1 accumulation due to degradation inhibition and transcriptional activation plays a critical role in silica nanoparticle-induced airway inflammation via NF-κB activation. J Nanobiotechnology 2020; 18:77. [PMID: 32429946 PMCID: PMC7236097 DOI: 10.1186/s12951-020-00634-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Most nanoparticles (NPs) reportedly block autophagic flux, thereby upregulating p62/SQSTM1 through degradation inhibition. p62 also acts as a multifunctional scaffold protein with multiple domains, and is involved in various cellular processes. However, the autophagy substrate-independent role of p62 and its regulation at the transcriptional level upon NPs exposure remain unclear. RESULTS In this work, we exposed BEAS-2b cells and mice to silica nanoparticles (SiNPs), and found that SiNPs increased p62 protein levels in vivo and vitro. Then, we further explored the role and mechanism of SiNPs-stimulated p62 in vitro, and found that p62 degradation was inhibited due to autophagic flux blockade. Mechanistically, SiNPs blocked autophagic flux through impairment of lysosomal capacity rather than defective autophagosome fusion with lysosomes. Moreover, SiNPs stimulated translocation of NF-E2-related factor 2 (Nrf2) to the nucleus from the cytoplasm, which upregulated p62 transcriptional activation through direct binding of Nrf2 to the p62 promoter. Nrf2 siRNA dramatically reduced both the mRNA and protein levels of p62. These two mechanisms led to p62 protein accumulation, thus increasing interleukin (IL)-1 and IL-6 expression. SiNPs activated nuclear factor kappa B (NF-κB), and this effect could be alleviated by p62 knockdown. CONCLUSION SiNPs caused accumulation of p62 through both pre- and post-translational mechanisms, resulting in airway inflammation. These findings improve our understanding of SiNP-induced pulmonary damage and the molecular targets available to mitigate it.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Yang Jin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Tianyu Sun
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Piaoyu Zhu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Jinlong Li
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Qinglin Zhang
- Departments of Gastroenterology, Affiliated to Wuxi People's Hospital, Nanjing Medical University, Wuxi, 214023, China
| | - Xiaoke Wang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China.
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China.
| |
Collapse
|
104
|
Dubovoy V, Nawrocki S, Verma G, Wojtas L, Desai P, Al-Tameemi H, Brinzari TV, Stranick M, Chen D, Xu S, Ma S, Boyd JM, Asefa T, Pan L. Synthesis, Characterization, and Investigation of the Antimicrobial Activity of Cetylpyridinium Tetrachlorozincate. ACS OMEGA 2020; 5:10359-10365. [PMID: 32426592 PMCID: PMC7226859 DOI: 10.1021/acsomega.0c00131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/09/2020] [Indexed: 06/11/2023]
Abstract
Cetylpyridinium tetrachlorozincate (referred to herein as (CP)2ZnCl4) was synthesized and its solid-state structure was elucidated via single-crystal X-ray diffraction (SC-XRD), revealing a stoichiometry of C42H76Cl4N2Zn with two cetylpyridinium (CP) cations per [ZnCl4]2- tetrahedra. Crystal structures at 100 and 298 K exhibited a zig-zag pattern with alternating alkyl chains and zinc units. The material showed potential for application as a broad-spectrum antimicrobial agent, to reduce volatile sulfur compounds (VSCs) generated by bacteria, and in the fabrication of advanced functional materials. Minimum inhibitory concentration (MIC) of (CP)2ZnCl4 was 60, 6, and 6 μg mL-1 for Salmonella enterica, Staphylococcus aureus, and Streptococcus mutans, respectively. The MIC values of (CP)2ZnCl4 were comparable to that of pure cetylpyridinium chloride (CPC), despite the fact that approximately 16% of the bactericidal CPC is replaced with bacteriostatic ZnCl2 in the structure. A modified layer-by-layer deposition technique was implemented to synthesize mesoporous silica (i.e., SBA-15) loaded with approximately 9.0 wt % CPC and 8.9 wt % Zn.
Collapse
Affiliation(s)
- Viktor Dubovoy
- Department
of Chemistry and Chemical Biology, Rutgers,
The State University of New Jersey, 610 Taylor Road, Piscataway, New Jersey 08854, United States
| | - Shiri Nawrocki
- Department
of Medicine, Rutgers Robert Wood Johnson
Medical School, 675 Hoes
Lane West, Piscataway, New
Jersey 08854, United
States
| | - Gaurav Verma
- Department
of Chemistry, University of South Florida, 4202 East Fowler Avenue, Tampa, Florida33620, United States
| | - Lukasz Wojtas
- Department
of Chemistry, University of South Florida, 4202 East Fowler Avenue, Tampa, Florida33620, United States
| | - Primit Desai
- Department
of Biochemistry and Microbiology, Rutgers,
The State University of New Jersey, 71 Lipman Drive, New Brunswick, New Jersey 08854, United States
| | - Hassan Al-Tameemi
- Department
of Biochemistry and Microbiology, Rutgers,
The State University of New Jersey, 71 Lipman Drive, New Brunswick, New Jersey 08854, United States
| | - Tatiana V. Brinzari
- Colgate-Palmolive
Company, 909 River Road, Piscataway, New Jersey 08854, United States
| | - Michael Stranick
- Colgate-Palmolive
Company, 909 River Road, Piscataway, New Jersey 08854, United States
| | - Dailin Chen
- Colgate-Palmolive
Company, 338 Qingnian
Road, Economic Development Zone, Guangzhou 510620, China
| | - Shaopeng Xu
- Colgate-Palmolive
Company, 338 Qingnian
Road, Economic Development Zone, Guangzhou 510620, China
| | - Shengqian Ma
- Department
of Chemistry, University of South Florida, 4202 East Fowler Avenue, Tampa, Florida33620, United States
| | - Jeffrey M. Boyd
- Department
of Biochemistry and Microbiology, Rutgers,
The State University of New Jersey, 71 Lipman Drive, New Brunswick, New Jersey 08854, United States
| | - Tewodros Asefa
- Department
of Chemistry and Chemical Biology, Rutgers,
The State University of New Jersey, 610 Taylor Road, Piscataway, New Jersey 08854, United States
- Department
of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, New Jersey 08854, United States
| | - Long Pan
- Colgate-Palmolive
Company, 909 River Road, Piscataway, New Jersey 08854, United States
| |
Collapse
|
105
|
He H, Zou Z, Wang B, Xu G, Chen C, Qin X, Yu C, Zhang J. Copper Oxide Nanoparticles Induce Oxidative DNA Damage and Cell Death via Copper Ion-Mediated P38 MAPK Activation in Vascular Endothelial Cells. Int J Nanomedicine 2020; 15:3291-3302. [PMID: 32494130 PMCID: PMC7229313 DOI: 10.2147/ijn.s241157] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
Background Inhaled nanoparticles can cross pulmonary air–blood barrier into circulation and cause vascular endothelial injury and progression of cardiovascular disease. However, the molecular mechanism underlying the vascular toxicity of copper oxide nanoparticles (CuONPs) remains unclear. We have recently demonstrated that the release of copper ions and the accumulation of superoxide anions contributed to CuONPs-induced cell death in human umbilical vein endothelial cells (HUVECs). Herein, we further demonstrate the mechanism underlying copper ions-induced cell death in HUVECs. Methods and Results CuONPs were suspended in culture medium and vigorously vortexed for several seconds before exposure. After treatment with CuONPs, HUVECs were collected, and cell function assays were conducted to elucidate cellular processes including cell viability, oxidative stress, DNA damage and cell signaling pathways. We demonstrated that CuONPs uptake induced DNA damage in HUVECs as evidenced by γH2AX foci formation and increased phosphorylation levels of ATR, ATM, p53 and H2AX. Meanwhile, we showed that CuONPs exposure induced oxidative stress, indicated by the increase of cellular levels of superoxide anions, the upregulation of protein levels of heme oxygenase-1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), the elevation of the levels of malondialdehyde (MDA), but the reduction of glutathione to glutathione disulfide ratio. We also found that antioxidant N-acetyl-L-cysteine (NAC) could ameliorate CuONPs-induced oxidative stress and cell death. Interestingly, we demonstrated that p38 mitogen-activated protein kinase (MAPK) signaling pathway was activated in CuONPs-treated HUVECs, while p38α MAPK knockdown by siRNA significantly rescued HUVECs from CuONPs-induced DNA damage and cell death. Importantly, we showed that copper ions chelator tetrathiomolybdate (TTM) could alleviate CuONPs-induced oxidative stress, DNA damage, p38 MAPK pathway activation and cell death in HUVECs. Conclusion We demonstrated that CuONPs induced oxidative DNA damage and cell death via copper ions-mediated p38 MAPK activation in HUVECs, suggesting that the release of copper ions was the upstream activator for CuONPs-induced vascular endothelial toxicity, and the copper ions chelator TTM can alleviate CuONPs-associated cardiovascular disease.
Collapse
Affiliation(s)
- Hui He
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, People's Republic of China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, People's Republic of China
| | - Chengzhi Chen
- School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xia Qin
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
106
|
Ali A, Ovais M, Cui X, Rui Y, Chen C. Safety Assessment of Nanomaterials for Antimicrobial Applications. Chem Res Toxicol 2020; 33:1082-1109. [DOI: 10.1021/acs.chemrestox.9b00519] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Arbab Ali
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, P.R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Muhammad Ovais
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - YuKui Rui
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510700, China
| |
Collapse
|
107
|
Sydor MJ, Anderson DS, Steele HBB, Ross JBA, Holian A. Effects of titanium dioxide and zinc oxide nano-materials on lipid order in model membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183313. [PMID: 32304756 DOI: 10.1016/j.bbamem.2020.183313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
Engineered nano-materials (ENM) have been reported to affect lipid membrane permeability in cell models, but a mechanistic understanding of how these materials interact with biological membranes has not been described. To assess mechanisms of permeability, liposomes composed of DOPC, DOPS, or POPC, with or without cholesterol, were used as model membranes for measuring ENM-induced changes to lipid order to improve our understanding of ENM effects on membrane permeability. Liposomes were treated with either titanium dioxide (TiO2) or zinc oxide (ZnO) ENM, and changes to lipid order were measured by time-resolved fluorescence anisotropy of a lipophilic probe, Di-4-ANEPPDHQ. Both ENM increased lipid order in two lipid models differing in headgroup charge. TiO2 increased lipid order of POPC liposomes (neutral charge), while ZnO acted primarily on DOPS liposomes (negative charge). Addition of cholesterol to these models significantly increased lipid order while in some cases attenuated ENM-induced changes to lipid order. To assess the ability of ENM to induce membrane permeability, liposomes composed of the above lipids were assayed for membrane permeability by calcein leakage in response to ENM. Both ENM caused a dose-dependent increase in permeability in all liposome models tested, and the addition of cholesterol to the liposome models neither blocked nor reduced calcein leakage. Together, these experiments show that ENM increased permeability of small molecules (calcein) from model liposomes, and that the magnitude of the effect of ENM on lipid order depended on ENM surface charge, lipid head group charge and the presence of cholesterol in the membrane.
Collapse
Affiliation(s)
- Matthew J Sydor
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, United States of America.
| | - Donald S Anderson
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, United States of America.
| | - Harmen B B Steele
- Department of Chemistry and Biochemistry, University of Montana, Missoula, MT 59812, United States of America; Center for Biomolecular and Structure & Dynamics, University of Montana, Missoula, MT 59812, United States of America.
| | - J B Alexander Ross
- Department of Chemistry and Biochemistry, University of Montana, Missoula, MT 59812, United States of America; Center for Biomolecular and Structure & Dynamics, University of Montana, Missoula, MT 59812, United States of America.
| | - Andrij Holian
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, United States of America.
| |
Collapse
|
108
|
Vysloužil J, Kulich P, Zeman T, Vaculovič T, Tvrdoňová M, Mikuška P, Večeřa Z, Stráská J, Moravec P, Balcar VJ, Šerý O. Subchronic continuous inhalation exposure to zinc oxide nanoparticles induces pulmonary cell response in mice. J Trace Elem Med Biol 2020; 61:126511. [PMID: 32294608 DOI: 10.1016/j.jtemb.2020.126511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/21/2020] [Accepted: 03/18/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVES We used mice as an animal model to investigate the entry of ZnO nanoparticles from the ambient air into the lungs and other organs, subsequent changes in Zn levels and the impact on the transcription of Zn homeostasis-related genes in the lungs. METHODS The mice were exposed to two concentrations of ZnO nanoparticles; lower (6.46 × 104 particles/cm3) and higher (1.93 × 106 particles/cm3), allowed to breathe the nanoparticles in the air for 12 weeks and subjected to necropsy. Characterization of the ZnO nanoparticles was done using transmission electron microscopy (TEM). Energy-dispersive X-ray (EDX) spectroscopy was used to quantify ZnO nanoparticles in the lungs, brain, liver and kidney. The total zinc content in the lungs, brain, liver, kidney, red blood cells and plasma was estimated by inductively coupled plasma mass spectroscopy (ICP-MS). Transcription rate of the genes was evaluated by RealTime PCR. RESULTS The two concentration of ZnO nanoparticles in the ambient air produced two different outcomes. The lower concentration resulted in significant increases in Zn content of the liver while the higher concentration significantly increased Zn in the lungs (p < 0.05). Additionally, at the lower concentration, Zn content was found to be lower in brain tissue (p < 0.05). Using TEM/EDX we detected ZnO nanoparticles inside the cells in the lungs, kidney and liver. Inhaling ZnO NP at the higher concentration increased the levels of mRNA of the following genes in the lungs: Mt2 (2.56 fold), Slc30a1 (1.52 fold) and Slc30a5 (2.34 fold). At the lower ZnO nanoparticle concentration, only Slc30a7 mRNA levels in the lungs were up (1.74 fold). Thus the two air concentrations of ZnO nanoparticles produced distinct effects on the expression of the Zn-homeostasis related genes. CONCLUSION Until adverse health effects of ZnO nanoparticles deposited in organs such as lungs are further investigated and/or ruled out, the exposure to ZnO nanoparticles in aerosols should be avoided or minimised.
Collapse
Affiliation(s)
- Jan Vysloužil
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 967/97, 602 00, Brno, Czech Republic; Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Pavel Kulich
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 967/97, 602 00, Brno, Czech Republic
| | - Tomáš Zeman
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 967/97, 602 00, Brno, Czech Republic
| | - Tomáš Vaculovič
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Michaela Tvrdoňová
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Pavel Mikuška
- Institute of Analytical Chemistry, Academy of Sciences of the Czech Republic, Veveří 967/97, 602 00, Brno, Czech Republic
| | - Zbyněk Večeřa
- Institute of Analytical Chemistry, Academy of Sciences of the Czech Republic, Veveří 967/97, 602 00, Brno, Czech Republic
| | - Jana Stráská
- Regional Centre of Advanced Technologies and Materials, Palacký University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic
| | - Pavel Moravec
- Laboratory of Aerosol Chemistry and Physics, Institute of Chemical Process Fundamentals of the Czech Academy of Sciences, Rozvojová 2/135, 165 02 Prague, Czech Republic
| | - Vladimir J Balcar
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 967/97, 602 00, Brno, Czech Republic; Bosch Institute and Discipline of Anatomy and Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Anderson Stuart Building F13, Sydney, NSW, 2006, Australia
| | - Omar Šerý
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 967/97, 602 00, Brno, Czech Republic; Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| |
Collapse
|
109
|
Areecheewakul S, Adamcakova-Dodd A, Givens BE, Steines BR, Wang Y, Meyerholz DK, Parizek NJ, Altmaier R, Haque E, O’Shaughnessy PT, Salem AK, Thorne PS. Toxicity assessment of metal oxide nanomaterials using in vitro screening and murine acute inhalation studies. NANOIMPACT 2020; 18:100214. [PMID: 32968700 PMCID: PMC7504913 DOI: 10.1016/j.impact.2020.100214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Characterizations and in vitro toxicity screening were performed on metal oxide engineered nanomaterials (ENMs) independently comprising ZnO, CuO, CeO2, Fe2O3, WO3, V2O5, TiO2, Al2O3 and MgO. Nanomaterials that exhibited the highest toxicity responses in the in vitro screening assays (ZnO, CuO, and V2O5) and the lesser explored material WO3 were tested for acute pulmonary toxicity in vivo. Female and male mice (C57Bl/6J) were exposed to aerosolized metal oxide ENMs in a nose-only exposure system and toxicity outcomes (biomarkers of cytotoxicity, immunotoxicity, inflammation, and lung histopathology) at 4 and 24 h after the start of exposure were assessed. The studies were performed as part of the NIEHS Nanomaterials Health Implications Research consortium with the purpose of investigating the effects of ENMs on various biological systems. ENMs were supplied by the Engineered Nanomaterials Resource and Coordination Core. Among the ENMs studied, the highest toxicity was observed for CuO and ZnO NPs in both in vitro and in vivo acute models. Compared to sham-exposed controls, there was a significant increase in bronchoalveolar lavage neutrophils and proinflammatory cytokines and a loss of macrophage viability at both 4 h and 24 h for ZnO and CuO but not seen for V2O5 or WO3. These effects were observed in both female and male mice. The cell viability performed after in vitro exposure to ENMs and assessment of lung inflammation after acute inhalation exposure in vivo were shown to be sensitive endpoints to predict ENM acute toxicity.
Collapse
Affiliation(s)
- Sudartip Areecheewakul
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52246, USA
| | - Andrea Adamcakova-Dodd
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52246, USA
| | - Brittany E. Givens
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52246, USA
| | - Benjamin R. Steines
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52246, USA
| | - Yifang Wang
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA 52246, USA
| | | | - Nathanial J. Parizek
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA 52246, USA
| | - Ralph Altmaier
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52246, USA
| | - Ezazul Haque
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA 52246, USA
| | - Patrick T. O’Shaughnessy
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52246, USA
| | - Aliasger K. Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52246, USA
- Corresponding author
| | - Peter S. Thorne
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52246, USA
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA 52246, USA
- Correspondence to: P.S. Thorne, Department of Occupational and Environmental Health, The University of Iowa, College of Public Health, 145 N. Riverside Dr., S341A CPHB, Iowa City, IA 52242, USA. (A.K. Salem), (P.S. Thorne)
| |
Collapse
|
110
|
Kong T, Zhang SH, Zhang C, Zhang JL, Yang F, Wang GY, Yang ZJ, Bai DY, Shi YY, Liu TQ, Li HL. The Effects of 50 nm Unmodified Nano-ZnO on Lipid Metabolism and Semen Quality in Male Mice. Biol Trace Elem Res 2020; 194:432-442. [PMID: 31264129 DOI: 10.1007/s12011-019-01792-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/19/2019] [Indexed: 11/30/2022]
Abstract
Fifty male mice were exposed to 50 nm unmodified nano-ZnO through intragastric administration for 90 days to detect the long-term effects of unmodified nano-ZnO in mice. Results showed that the blood glucose, serum follicle stimulating hormone, luteinizing hormone, testosterone, and estradiol were significantly decreased (p < 0.05). The serum triglyceride, total cholesterol, and low-density lipoprotein were significantly increased (p < 0.05). The semen quality of the 160 mg/kg·bw group were significantly lowered (p < 0.05). The liver and testis catalase and CuZn-SOD activities were significantly elevated (p < 0.05). The abilities of •OH inhibition in the livers and testes of the 160 mg/kg·bw group were significantly lowered (p < 0.05). The liver and testis MDA levels of the 160 mg/kg·bw group were significantly elevated (p < 0.05). Results indicate that exposure of nano-ZnO could induce lipid metabolism disorder, hyperlipidemia, and reproductive toxicity to male mice through oxidative injury.
Collapse
Affiliation(s)
- Tao Kong
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China.
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China.
| | - Shu-Hui Zhang
- Library of Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Cai Zhang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Ji-Liang Zhang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Fan Yang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Guo-Yong Wang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Zi-Jun Yang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Dong-Ying Bai
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Yun-Yun Shi
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Tian-Qi Liu
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| | - Hai-Long Li
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, 471023, Henan, People's Republic of China
| |
Collapse
|
111
|
He H, Xiao S, Xu G, Wang B, Zou Z, Qin X, Yu C, Zhang J. The NADPH oxidase 4 protects vascular endothelial cells from copper oxide nanoparticles-induced oxidative stress and cell death. Life Sci 2020; 252:117571. [PMID: 32201278 DOI: 10.1016/j.lfs.2020.117571] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/09/2020] [Accepted: 03/17/2020] [Indexed: 01/13/2023]
Abstract
AIMS Nanoparticles (NPs) exposure is associated with increased risk of cardiovascular diseases, but the underlying mechanism is still obscure. In this study, we investigated the role of NADPH oxidase 4 (NOX4) in copper oxide nanoparticles (CuONPs)-induced cytotoxicity in human umbilical vein endothelial cells (HUVECs). MATERIALS AND METHODS Morphology changes were examined under the microscope. Cell viability was determined by MTS assay and Calcein AM assay. Apoptosis and the levels of superoxide anion (O2-) and hydrogen peroxide (H2O2) were measured by fluorescence activated cell sorting (FACS). Oxidative stress was detected by assaying the levels of glutathione/glutathione disulfide (GSH/GSSG) and malondialdehyde (MDA). Protein expression levels were determined by western blotting. KEY FINDINGS We revealed that O2- rather than H2O2 was the major component of reactive oxygen species (ROS) in CuONPs-treated HUVECs. Meanwhile, CuONPs downregulated expression of O2--eliminating enzyme NOX4 both at mRNA and protein levels, but did not affect the expression of SOD2 and catalase. NOX4 knockdown caused more accumulation of O2-, and a further decrease of H2O2 in CuONPs-treated HUVECs, suggesting that NOX4 regulates the conversion of O2- to H2O2 in CuONPs-treated HUVECs. Furthermore, we revealed that NOX4 knockdown aggravated CuONPs-induced oxidative stress, characterized by a decrease of GSH/GSSG ratio, an increase of MDA level, and upregulation of HSPA5 and γH2AX. Finally, we showed that NOX4 knockdown exacerbated CuONPs-induced apoptotic cell death in HUVECs, indicating that NOX4 could protect ECs from CuONPs-induced cell death. SIGNIFICANCE Our study provides the evidence that NOX4 protects vascular endothelial cells from CuONPs-induced oxidative stress and cell death.
Collapse
Affiliation(s)
- Hui He
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Shiquan Xiao
- Reproductive Medicine Center, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xia Qin
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
112
|
Guo L, He N, Zhao Y, Liu T, Deng Y. Autophagy Modulated by Inorganic Nanomaterials. Theranostics 2020; 10:3206-3222. [PMID: 32194863 PMCID: PMC7053187 DOI: 10.7150/thno.40414] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
With the rapid development of nanotechnology, inorganic nanomaterials (NMs) have been widely applied in modern society. As human exposure to inorganic NMs is inevitable, comprehensive assessment of the safety of inorganic NMs is required. It is well known that autophagy plays dual roles in cell survival and cell death. Moreover, inorganic NMs have been proven to induce autophagy perturbation in cells. Therefore, an in-depth understanding of inorganic NMs-modulated autophagy is required for the safety assessment of inorganic NMs. This review presents an overview of a set of inorganic NMs, consisting of iron oxide NMs, silver NMs, gold NMs, carbon-based NMs, silica NMs, quantum dots, rare earth oxide NMs, zinc oxide NMs, alumina NMs, and titanium dioxide NMs, as well as how each modulates autophagy. This review emphasizes the potential mechanisms underlying NMs-induced autophagy perturbation, as well as the role of autophagy perturbation in cell fate determination. Furthermore, we also briefly review the potential roles of inorganic NMs-modulated autophagy in diagnosis and treatment of disease.
Collapse
|
113
|
|
114
|
Keerthana S, Kumar A. Potential risks and benefits of zinc oxide nanoparticles: a systematic review. Crit Rev Toxicol 2020; 50:47-71. [PMID: 32186437 DOI: 10.1080/10408444.2020.1726282] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Zinc oxide nanoparticles are well-known metal oxide nanoparticles having numbers of applications in the field of cosmetology, medicine, and chemistry. However, the number of reports has indicated its toxicity also such as hepatotoxicity, pulmonary toxicity, neurotoxicity, and immunotoxicity. Thus, in this article, we have analyzed the potential risks and benefits of zinc oxide nanoparticles. The data related to risks and benefits of zinc oxide nanoparticles have been extracted from PubMed (from January 2007 to August 2019). A total of 3,892 studies have been published during this period regarding zinc oxide nanoparticles. On the basis of inclusion and exclusion criteria, 277 studies have been included for the analysis of risks and benefits. Emerging reports have indicated both risks and benefits of zinc oxide nanoparticles in concentration- and time-dependent manner under in vitro and in vivo conditions through different mechanism of action. In conclusion, zinc oxide nanoparticles could play a beneficial role in the treatment of various diseases but safety of these particles at particular effective concentration should be thoroughly evaluated.
Collapse
Affiliation(s)
- S Keerthana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| | - A Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| |
Collapse
|
115
|
Liu X, Liao X, Rao X, Wang B, Zhang J, Xu G, Jiang X, Qin X, Chen C, Zou Z. The lysosomal membrane protein LAMP‐2 is dispensable for PINK1/Parkin‐mediated mitophagy. FEBS Lett 2019; 594:823-840. [DOI: 10.1002/1873-3468.13663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/16/2019] [Accepted: 10/27/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Xuemei Liu
- Department of Occupational and Environmental Health Chongqing Medical University China
| | - Xinyi Liao
- Institute of Life Sciences Chongqing Medical University China
| | - Xiyun Rao
- Institute of Life Sciences Chongqing Medical University China
| | - Bin Wang
- Institute of Life Sciences Chongqing Medical University China
| | - Jun Zhang
- Institute of Life Sciences Chongqing Medical University China
| | - Ge Xu
- Institute of Life Sciences Chongqing Medical University China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center Chongqing Medical University China
| | - Xia Qin
- Department of Pharmacy The First Affiliated Hospital of Chongqing Medical University China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health Chongqing Medical University China
- Dongsheng Lung‐Brain Disease Joint Lab Chongqing Medical University China
| | - Zhen Zou
- Institute of Life Sciences Chongqing Medical University China
- Dongsheng Lung‐Brain Disease Joint Lab Chongqing Medical University China
| |
Collapse
|
116
|
Ahmed F, Husain Q, Ansari MO, Shadab GGHA. Antidiabetic and oxidative stress assessment of bio-enzymatically synthesized zinc oxide nanoformulation on streptozotocin-induced hyperglycemic mice. APPLIED NANOSCIENCE 2019. [DOI: 10.1007/s13204-019-01169-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
117
|
Cui D, Ma J, Liang T, Sun L, Meng L, Liang T, Li Q. Selenium nanoparticles fabricated in laminarin polysaccharides solutions exert their cytotoxicities in HepG2 cells by inhibiting autophagy and promoting apoptosis. Int J Biol Macromol 2019; 137:829-835. [DOI: 10.1016/j.ijbiomac.2019.07.031] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/11/2019] [Accepted: 07/04/2019] [Indexed: 01/14/2023]
|
118
|
Liu X, Tu B, Jiang X, Xu G, Bai L, Zhang L, Meng P, Qin X, Chen C, Zou Z. Lysosomal dysfunction is associated with persistent lung injury in dams caused by pregnancy exposure to carbon black nanoparticles. Life Sci 2019; 233:116741. [PMID: 31398419 DOI: 10.1016/j.lfs.2019.116741] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
AIMS Carbon black nanoparticles (CBNPs) are widely used in industrial field. Sensitive stages such as pregnancy are assumed to be more susceptible to stimulus, however whether pregnancy exposure to CBNPs (PrE-to-CBNPs) would cause long-term toxic effects in dams and the underlying mechanisms remain poorly addressed. The present study is aimed to determine the long-term toxic effects of PrE-to-CBNPs in dams. MATERIALS AND METHODS The pregnant mice were randomly divided into control group, low (21 μg/animal), medium (103 μg/animal) and high (515 μg/animal) CBNPs-treated groups. From gestational day (GD) 9 to GD18, the pregnant mice were intranasal exposed. At 49 days after parturition, lung tissues and bronchoalveolar lavage fluid (BALF) were obtained. Weight change, lung histopathology, lung ultrastructural pathology, cell count in BALF, oxidative stress/inflammatory maker and autophagy/lysosome-related protein expression were determined. KEY FINDINGS PrE-to-CBNPs caused a dose-dependent persistent lung injury in mice even 49 days after parturition, including the deteriorative lung histopathological changes, elevation of oxidative stress marker Nrf-2, HO-1 and CHOP, infiltration of macrophage and increased mRNA expression of inflammatory cytokines in the lung tissues and elevation of cells in BALF. However, PrE-to-CBNPs did not induce significant neutrophil infiltration and fibrosis. Moreover, we found that CBNPs could deposit in the lysosomes and decrease cathepsin D (an important hydrolase in lysosome), which might be associated with the dysfunction of lysosome and autophagy. SIGNIFICANCE Our study demonstrated that PrE-to-CBNPs could result in long-term lung injury in dams, and lysosomal dysfunction was probably linked to this process.
Collapse
Affiliation(s)
- Xuemei Liu
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Baijie Tu
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Lulu Bai
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Longbin Zhang
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Pan Meng
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China; Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, PR China.
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, PR China; Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
119
|
Zinc oxide nanoparticles induce necroptosis and inhibit autophagy in MCF-7 human breast cancer cells. Biologia (Bratisl) 2019. [DOI: 10.2478/s11756-019-00325-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
120
|
Keshavan S, Calligari P, Stella L, Fusco L, Delogu LG, Fadeel B. Nano-bio interactions: a neutrophil-centric view. Cell Death Dis 2019; 10:569. [PMID: 31358731 PMCID: PMC6662811 DOI: 10.1038/s41419-019-1806-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/04/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022]
Abstract
Neutrophils are key components of the innate arm of the immune system and represent the frontline of host defense against intruding pathogens. However, neutrophils can also cause damage to the host. Nanomaterials are being developed for a multitude of different purposes and these minute materials may find their way into the body through deliberate or inadvertent exposure; understanding nanomaterial interactions with the immune system is therefore of critical importance. However, whereas numerous studies have focused on macrophages, less attention is devoted to nanomaterial interactions with neutrophils, the most abundant leukocytes in the blood. We discuss the impact of engineered nanomaterials on neutrophils and how neutrophils, in turn, may digest certain carbon-based materials such as carbon nanotubes and graphene oxide. We also discuss the role of the corona of proteins adsorbed onto the surface of nanomaterials and whether nanomaterials are sensed as pathogens by cells of the immune system.
Collapse
Affiliation(s)
- Sandeep Keshavan
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Calligari
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Lorenzo Stella
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Laura Fusco
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Lucia Gemma Delogu
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Bengt Fadeel
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
121
|
Yin X, Li Q, Wei H, Chen N, Wu S, Yuan Y, Liu B, Chen C, Bi H, Guo D. Zinc oxide nanoparticles ameliorate collagen lattice contraction in human tenon fibroblasts. Arch Biochem Biophys 2019; 669:1-10. [DOI: 10.1016/j.abb.2019.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/14/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023]
|
122
|
Kong T, Zhang SH, Zhang C, Zhang JL, Yang F, Wang GY, Yang ZJ, Bai DY, Zhang MY, Wang J, Zhang BH. Long-Term Effects of Unmodified 50 nm ZnO in Mice. Biol Trace Elem Res 2019; 189:478-489. [PMID: 30109551 DOI: 10.1007/s12011-018-1477-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/08/2018] [Indexed: 12/22/2022]
Abstract
Nanometer zinc oxide (nano-ZnO) is widely used in many kinds of fields. However, information about the toxicity and toxic mechanism of nano-ZnO is limited. The aims of this study were to investigate the long-term toxic effects of unmodified 50 nm ZnO administered by gavage in mice. After 90 days, hematological parameters, hepatic and renal functions, and oxidative and anti-oxidative status were measured. Pathological damages in livers, kidneys, and other tissues were also examined by hematoxylin and eosin (H&E) staining. The results showed that oral nano-ZnO exposure induced anemia and damages to liver and kidney, influenced the antioxidant system, and impacted functions of liver and kidney in mice after a 90-day exposure. The main cause for oxidative stress in vivo induced by nano-ZnO might be hydroxyl free radical. The lowest observed adverse effect level (LOAEL) was 40 mg/kg·bw, and the livers, kidneys, lungs, pancreas, and gastrointestinal tracts are the target organs.
Collapse
Affiliation(s)
- Tao Kong
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China.
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China.
| | - Shu-Hui Zhang
- Library of Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Cai Zhang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Ji-Liang Zhang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Fan Yang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Guo-Yong Wang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Zi-Jun Yang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
- Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Dong-Ying Bai
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Meng-Yu Zhang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Jie Wang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| | - Bai-Hao Zhang
- College of Animal Science and Veterinary Medicine, Henan University of Science and Technology, No. 263 Kaiyuan Road, Luoyang, Henan, 471023, People's Republic of China
| |
Collapse
|
123
|
Wan X, Zhang X, Pan W, Liu B, Yu L, Wang H, Li N, Tang B. Ratiometric Fluorescent Quantification of the Size-Dependent Cellular Toxicity of Silica Nanoparticles. Anal Chem 2019; 91:6088-6096. [DOI: 10.1021/acs.analchem.9b00633] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Xiuyan Wan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Xinhao Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Bo Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Longhai Yu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Honghong Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, People’s Republic of China
| |
Collapse
|
124
|
Augustine R, Prasad P, Khalaf IMN. Therapeutic angiogenesis: From conventional approaches to recent nanotechnology-based interventions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:994-1008. [DOI: 10.1016/j.msec.2019.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/06/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
|
125
|
Sharifi M, Hosseinali SH, Saboury AA, Szegezdi E, Falahati M. Involvement of planned cell death of necroptosis in cancer treatment by nanomaterials: Recent advances and future perspectives. J Control Release 2019; 299:121-137. [DOI: 10.1016/j.jconrel.2019.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/31/2022]
|
126
|
Cationic gold nanoparticles elicit mitochondrial dysfunction: a multi-omics study. Sci Rep 2019; 9:4366. [PMID: 30867451 PMCID: PMC6416392 DOI: 10.1038/s41598-019-40579-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/19/2019] [Indexed: 01/05/2023] Open
Abstract
Systems biology is increasingly being applied in nanosafety research for observing and predicting the biological perturbations inflicted by exposure to nanoparticles (NPs). In the present study, we used a combined transcriptomics and proteomics approach to assess the responses of human monocytic cells to Au-NPs of two different sizes with three different surface functional groups, i.e., alkyl ammonium bromide, alkyl sodium carboxylate, or poly(ethylene glycol) (PEG)-terminated Au-NPs. Cytotoxicity screening using THP-1 cells revealed a pronounced cytotoxicity for the ammonium-terminated Au-NPs, while no cell death was seen after exposure to the carboxylated or PEG-modified Au-NPs. Moreover, Au-NR3+ NPs, but not the Au-COOH NPs, were found to trigger dose-dependent lethality in vivo in the model organism, Caenorhabditis elegans. RNA sequencing combined with mass spectrometry-based proteomics predicted that the ammonium-modified Au-NPs elicited mitochondrial dysfunction. The latter results were validated by using an array of assays to monitor mitochondrial function. Au-NR3+ NPs were localized in mitochondria of THP-1 cells. Moreover, the cationic Au-NPs triggered autophagy in macrophage-like RFP-GFP-LC3 reporter cells, and cell death was aggravated upon inhibition of autophagy. Taken together, these studies have disclosed mitochondria-dependent effects of cationic Au-NPs resulting in the rapid demise of the cells.
Collapse
|
127
|
Mohammadinejad R, Moosavi MA, Tavakol S, Vardar DÖ, Hosseini A, Rahmati M, Dini L, Hussain S, Mandegary A, Klionsky DJ. Necrotic, apoptotic and autophagic cell fates triggered by nanoparticles. Autophagy 2019; 15:4-33. [PMID: 30160607 PMCID: PMC6287681 DOI: 10.1080/15548627.2018.1509171] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 07/19/2018] [Accepted: 08/03/2018] [Indexed: 12/15/2022] Open
Abstract
Nanomaterials have gained a rapid increase in use in a variety of applications that pertain to many aspects of human life. The majority of these innovations are centered on medical applications and a range of industrial and environmental uses ranging from electronics to environmental remediation. Despite the advantages of NPs, the knowledge of their toxicological behavior and their interactions with the cellular machinery that determines cell fate is extremely limited. This review is an attempt to summarize and increase our understanding of the mechanistic basis of nanomaterial interactions with the cellular machinery that governs cell fate and activity. We review the mechanisms of NP-induced necrosis, apoptosis and autophagy and potential implications of these pathways in nanomaterial-induced outcomes. Abbreviations: Ag, silver; CdTe, cadmium telluride; CNTs, carbon nanotubes; EC, endothelial cell; GFP, green fluorescent protein; GO, graphene oxide; GSH, glutathione; HUVECs, human umbilical vein endothelial cells; NP, nanoparticle; PEI, polyethylenimine; PVP, polyvinylpyrrolidone; QD, quantum dot; ROS, reactive oxygen species; SiO2, silicon dioxide; SPIONs, superparamagnetic iron oxide nanoparticles; SWCNT, single-walled carbon nanotubes; TiO2, titanium dioxide; USPION, ultra-small super paramagnetic iron oxide; ZnO, zinc oxide.
Collapse
Affiliation(s)
- Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Deniz Özkan Vardar
- Sungurlu Vocational High School, Health Programs, Hitit University, Corum, Turkey
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Salik Hussain
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, School of Medicine, Morgantown, WV, USA
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
128
|
Umar H, Kavaz D, Rizaner N. Biosynthesis of zinc oxide nanoparticles using Albizia lebbeck stem bark, and evaluation of its antimicrobial, antioxidant, and cytotoxic activities on human breast cancer cell lines. Int J Nanomedicine 2018; 14:87-100. [PMID: 30587987 PMCID: PMC6304255 DOI: 10.2147/ijn.s186888] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Biocompatibility and stability of zinc oxide nanoparticles (ZnO NPs) synthesized using plants is an interesting research area of study in nanotechnology, due to its wide applications in biomedical, industrial, cell imaging, and biosensor fields. The present study reports the novel green synthesis of stable ZnO NPs using various concentrations of zinc nitrate (0.01M, 0.05M, 0.1M) and Albizia lebbeck stem bark extracts as an efficient chelating agent. Antimicrobial, antioxidant, cytotoxic, and antiproliferative activities of the synthesized NPs on human breast cancer cell lines were evaluated using different assays. Methods Characterization of the synthesized ZnO NPs were carried out using various spectroscopic and microscopic techniques. Antimicrobial activity evaluation using disc diffusion method, antioxidant activity using hydrogen peroxide (H2O2) free radical scavenging assay and cytotoxic activity on MDA-MB 231 and MCF-7 using tryphan blue dye exclusion and MTT assay. Results The UV–vis spectroscopy result revealed an absorption peak in the range of 370 nm. The involvements of A. lebbeck bioactive compounds in the stabilization of the ZnO NPs were confirmed by X-ray diffraction and Fourier transform infrared analysis. Zeta sizer studies showed an average size of 66.25 nm with a polydisparity index of 0.262. Scanning electron microscopy (SEM) and energy-dispersive X-ray spectroscopy (EDX) analyses results revealed irregular spherical morphology and the presence of primarily Zn, C, O, Na, P, and K, respectively. The biosynthesized ZnO NPs revealed strong antimicrobial potentials against various gram-negative and gram-positive bacterial pathogens. Antioxidant activities carried out using H2O2 free radical scavenging assay revealed higher IC50 values of 48.5, 48.7, and 60.2 µg/mL for 0.1M, 0.05M, and 0.01M ZnO NPs, respectively. Moreover, the biosynthesized ZnO NPs showed significant cytotoxic effects on MDA-MB 231 and MCF-7 breast cancer cell lines (P< 0.001, n≥3) in a concentration-dependent manner. Conclusion Overall, various concentrations of ZnO NPs were synthesized through a stable, simple, and eco-friendly green route via the use of A. lebbeck stem bark extract. The biosynthesized ZnO NPs showed strong antimicrobial, antioxidant and cytotoxic activity against strongly and weakly metastatic breast cancer cell lines.
Collapse
Affiliation(s)
- Huzaifa Umar
- Department of Bioengineering, Institute of Graduate Studies and Research, Cyprus International University, Mersin, Turkey, .,Biotechnology Research Center, Cyprus International University, Mersin, Turkey, .,Bioengineering Department, Cyprus International University, Mersin, Turkey,
| | - Doga Kavaz
- Department of Bioengineering, Institute of Graduate Studies and Research, Cyprus International University, Mersin, Turkey, .,Biotechnology Research Center, Cyprus International University, Mersin, Turkey, .,Bioengineering Department, Cyprus International University, Mersin, Turkey,
| | - Nahit Rizaner
- Department of Bioengineering, Institute of Graduate Studies and Research, Cyprus International University, Mersin, Turkey, .,Biotechnology Research Center, Cyprus International University, Mersin, Turkey, .,Bioengineering Department, Cyprus International University, Mersin, Turkey,
| |
Collapse
|
129
|
Zhang J, Wang B, Wang H, He H, Wu Q, Qin X, Yang X, Chen L, Xu G, Yuan Z, Yi Q, Zou Z, Yu C. Disruption of the superoxide anions-mitophagy regulation axis mediates copper oxide nanoparticles-induced vascular endothelial cell death. Free Radic Biol Med 2018; 129:268-278. [PMID: 30248444 DOI: 10.1016/j.freeradbiomed.2018.09.032] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/20/2018] [Accepted: 09/20/2018] [Indexed: 12/18/2022]
Abstract
Copper oxide nanoparticles (CuONPs) have been widely used in the industrial and pharmaceutical fields; however, their toxicity profile is deeply concerning. Currently, nanomaterials-induced toxicity in the cardiovascular system is receiving increased attention. Our previous toxicological study found that lysosomal deposition of CuONPs triggered vascular endothelial cell death, indicating that the involvement of autophagic dysfunction was crucial for CuONPs-induced toxicity in human umbilical vein endothelial cells (HUVECs). In the current study, we investigated the detailed mechanism underlying the autophagic dysfunction induced by CuONPs. We demonstrated that CuONPs exposure caused accumulation of superoxide anions, which likely resulted from mitochondrial dysfunctions. MnTBAP, a superoxide anions scavenger, alleviated CuONPs-induced HUVECs death, indicating that excessive superoxide anions were directly related to the CuONPs cytotoxicity in HUVECs. Interestingly, we found that mitophagy (a protective mechanism for clearance of damaged mitochondria and excessive superoxide anions) was initiated but failed to be cleared in CuONPs-treated cells, resulting in the accumulation of damaged mitochondria. Inhibition of mitophagy through Atg5 knockout or blocking of mitochondria fission with Mdivi-1 significantly aggravated CuONPs-induced superoxide anions accumulation and cell death, suggesting that mitophagy is a protective mechanism against CuONPs cytotoxicity in HUVECs. In summary, we demonstrate that superoxide anions (originating from damaged mitochondria) are involved in CuONPs-associated toxicity and that impaired mitophagic flux aggravates the accumulation of excessive superoxide anions, which leads to HUVECs death. Our findings indicate that there are crucial roles for superoxide anions and mitophagy in CuONPs-induced toxicity in vascular endothelial cells.
Collapse
Affiliation(s)
- Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Hong Wang
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Hui He
- College of Pharmacy, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Qiong Wu
- College of Pharmacy, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Xia Qin
- College of Pharmacy, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Xi Yang
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Linmu Chen
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Qiying Yi
- Laboratory Animal Center, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China.
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
130
|
Zhang M, Xu C, Jiang L, Qin J. A 3D human lung-on-a-chip model for nanotoxicity testing. Toxicol Res (Camb) 2018; 7:1048-1060. [PMID: 30510678 PMCID: PMC6220735 DOI: 10.1039/c8tx00156a] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022] Open
Abstract
The prevalent application of nanoparticles (NPs) has drawn intense concerns about their impact on the environment and human health. Inhalation of NPs is the major route of NP exposure and has led to adverse effects on the lung. It is of great concern to evaluate the potential hazards of nanoparticles for human health during pulmonary exposure. Here, we proposed a novel 3D human lung-on-a-chip model to recreate the organ-level structure and functions of the human lung that allow to us evaluate the pulmonary toxicity of nanoparticles. The lung-on-a-chip consists of three parallel channels for the co-culture of human vascular endothelial cells and human alveolar epithelial cells sandwiching a layer of Matrigel membrane, which recapitulate the key features of the alveolar capillary barrier in the human lung. Cell-cell interaction, cell-matrix interaction and vascular mechanical cues work synergistically to promote the barrier function of the lung-on-a-chip model. TiO2 nanoparticles and ZnO nanoparticles were applied on the lung-on-a-chip to assay their nanotoxicity on both epithelial cells and endothelial cells. Junction protein expression, increased permeability to macromolecules, dose dependent cytotoxicity, ROS production and apoptosis were assayed and compared on the chip. This lung-on-a-chip model indicated its versatile application in human pulmonary health and safety assessment for nanoparticles, environment, food and drugs.
Collapse
Affiliation(s)
- Min Zhang
- Division of Biotechnology , CAS Key Laboratory of Separation Sciences for Analytical Chemistry , Dalian Institute of Chemical Physics , Chinese Academy of Sciences , Dalian , China .
| | - Cong Xu
- Division of Biotechnology , CAS Key Laboratory of Separation Sciences for Analytical Chemistry , Dalian Institute of Chemical Physics , Chinese Academy of Sciences , Dalian , China .
- University of Chinese Academy of Sciences , Beijing , China
| | - Lei Jiang
- Division of Biotechnology , CAS Key Laboratory of Separation Sciences for Analytical Chemistry , Dalian Institute of Chemical Physics , Chinese Academy of Sciences , Dalian , China .
| | - Jianhua Qin
- Division of Biotechnology , CAS Key Laboratory of Separation Sciences for Analytical Chemistry , Dalian Institute of Chemical Physics , Chinese Academy of Sciences , Dalian , China .
- University of Chinese Academy of Sciences , Beijing , China
- CAS Centre for Excellence in Brain Science and Intelligence Technology , Chinese Academy of Sciences , Shanghai , China
- Institute for Stem Cell and Regeneration , Chinese Academy of Sciences , Beijing , China
| |
Collapse
|
131
|
Feng X, Chen L, Guo W, Zhang Y, Lai X, Shao L, Li Y. Graphene oxide induces p62/SQSTM-dependent apoptosis through the impairment of autophagic flux and lysosomal dysfunction in PC12 cells. Acta Biomater 2018; 81:278-292. [PMID: 30273743 DOI: 10.1016/j.actbio.2018.09.057] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/19/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022]
Abstract
Graphene oxide (GO), as a two-dimensional carbon nanosheet, has been extensively studied for potential biomedical applications due to its notable properties. Although a growing number of studies have investigated the adverse effects of GO nanosheets, the available toxicity data concerning GO's effect on the neuronal cells remain highly limited. In this work, we systematically investigated the toxic responses of commercially available GO on a rat pheochromocytoma-derived PC12 cell line, which was an ideal in vitro model to study the neurotoxicity of GO. GO exerted a significant toxic effect on PC12 cells in a dose- and time-dependent manner. GO treatments under doses of 40, 50, and 60 μg/mL triggered an autophagic response and the blockade of autophagic flux via disrupting lysosome degradation capability. Caspase 9-mediated apoptosis was also observed in GO-treated cells. Moreover, GO-induced apoptosis was relevant to the aberrant accumulation of autophagy substrate p62/SQSTM. Inhibitionofthe accumulation of autophagic substrate alleviated GO-caused apoptotic cell death. Our findings raise a concern for the putative biomedical applications of GO in the form of diagnostic and therapeutic tools, where its systematic biocompatibility should be thoroughly explored. STATEMENT OF SIGNIFICANCE: Graphene oxide (GO) has attracted considerable interests in biomedical fields, which also resulted in numerous safety risks to human bodies. It is urgently required to establish a paradigm for accurately evaluating their adverse effects in biological systems. This study thoroughly explored the neurotoxicity of GO in PC12 cells. We found GO triggered an increased autophagic response and the impairment of autophagic flux, which was functionally involved in cell apoptosis. Inhibitionofexcessive accumulation of autophagic cargo attenuated apoptotic cell death. Our findings highlight deep considerations on the regulation mechanism of autophagy-lysosomes-apotosis-axis, which will contribute to a better understanding of the neurotoxicity of graphene-family nanomaterials, and provide a new insight in the treatment of cancer cells at nanoscale levels.
Collapse
|
132
|
Jiang X, Tang Q, Zhang J, Wang H, Bai L, Meng P, Qin X, Xu G, Bose DD, Wang B, Chen C, Zou Z. Autophagy-dependent release of zinc ions is critical for acute lung injury triggered by zinc oxide nanoparticles. Nanotoxicology 2018; 12:1068-1091. [PMID: 30317896 DOI: 10.1080/17435390.2018.1513094] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Pulmonary exposure to zinc oxide nanoparticles (ZnONPs) could cause acute lung injury (ALI), but the underlying molecular mechanism remains unclear. Herein, we established a ZnONPs-induced ALI mouse model, characterized by the histopathological changes (edema and infiltration of inflammatory cells in lung tissues), and the elevation of total protein and cytokine interleukin-6 in bronchoalveolar lavage fluid in time- and dose-dependent manners. This model also exhibited features like the disturbance of redox-state (reduced of glutathione to glutathione disulfide ratio, elevation of heme oxygenase-1 and superoxide dismutase 2), the decrease of adenosine triphosphate synthesis and the release of zinc ions in the lung tissues. Interestingly, we found that ZnONPs exposure caused the accumulation of autophagic vacuoles and the elevation of microtubule-associated proteins 1A/1B light chain (LC)3B-II and p62, indicating the impairment of autophagic flux. Our data indicated that the above process might be regulated by the activation of AMP-activated protein kinase but not the mammalian target of rapamycin pathway. The association between ZnONPs-induced ALI and autophagy was further verified by a classical autophagy inhibitor, 3-methyladenine (3-MA). 3-MA administration reduced the accumulation of autophagic vacuoles, the expression of LC3B-II and p62, followed by a significant attenuation of histopathological changes, inflammation, and oxidative stress. More importantly, 3-MA could directly decrease the release of zinc ions in lung tissues. Taken together, our study provides the evidence that ZnONPs-induced pulmonary toxicity is autophagy-dependent, suggests that limiting the release of zinc ions by inhibiting autophagy could be a feasible strategy for the prevention of ZnONPs-associated pulmonary toxicity.
Collapse
Affiliation(s)
- Xuejun Jiang
- a Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center , Chongqing Medical University , Chongqing , People's Republic of China.,b Laboratory of Tissue and Cell Biology, Experimental Teaching and Management Center , Chongqing Medical University , Chongqing , People's Republic of China
| | - Qianghu Tang
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Jun Zhang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Hong Wang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Lulu Bai
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Pan Meng
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Xia Qin
- e Department of Pharmacy , The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Ge Xu
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Diptiman D Bose
- f Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences , Western New England University , Springfield , MA , USA
| | - Bin Wang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Chengzhi Chen
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China.,g Post-doctoral Research Stations of Nursing Science, School of Nursing , Chongqing Medical University , Chongqing , People's Republic of China
| | - Zhen Zou
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| |
Collapse
|
133
|
He G, Ma Y, Zhu Y, Yong L, Liu X, Wang P, Liang C, Yang C, Zhao Z, Hai B, Pan X, Liu Z, Liu X, Mao C. Cross Talk Between Autophagy and Apoptosis Contributes to ZnO Nanoparticle-Induced Human Osteosarcoma Cell Death. Adv Healthc Mater 2018; 7:e1800332. [PMID: 29900694 PMCID: PMC6310009 DOI: 10.1002/adhm.201800332] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/18/2018] [Indexed: 12/13/2022]
Abstract
Killing osteosarcoma cells by zinc oxide nanoparticles (NPs) and its underlying subcellular mechanism are never studied. Here, it is found that the NPs induce cross talk between apoptosis and autophagy, which leads to osteosarcoma cell death. Specifically, the NP uptake promotes autophagy by inducing accumulation of autophagosomes along with impairment of lysosomal functions. The autophagy further causes the uptaken NPs to release zinc ions by promoting their dissolution. These intracellular zinc ions, together with those that are originally released from the extracellular NPs and flowed into the cells, collectively target and damage mitochondria to produce reactive oxygen species (ROS). Then the ROS inhibit cell proliferation by arresting S phase and trigger apoptosis by extrinsic and intrinsic pathways, ultimately leading to cell death. More importantly, suppression of the early stage autophagy restores cell viability by abolishing apoptosis whereas blockade of the late stage autophagy inversely enhances apoptosis. In contrast, inhibition of apoptosis shows a limited ability to restore cell viability but obviously enhance autophagy. Notably, cell viability is strongly ameliorated by the combination of inhibitors for both the late stage autophagy and the apoptosis. These findings provide a mechanistic understanding of the NP-directed autophagy and apoptosis in osteosarcoma cells.
Collapse
Affiliation(s)
- Guanping He
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yunlong Ma
- The Center for Pain Medicine, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Ye Zhu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Lei Yong
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiao Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Peng Wang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chen Liang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chenlong Yang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhigang Zhao
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Bao Hai
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiaoyu Pan
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhongjun Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiaoguang Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
134
|
Jiang J, Pi J, Cai J. The Advancing of Zinc Oxide Nanoparticles for Biomedical Applications. Bioinorg Chem Appl 2018; 2018:1062562. [PMID: 30073019 PMCID: PMC6057429 DOI: 10.1155/2018/1062562] [Citation(s) in RCA: 478] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 05/13/2018] [Accepted: 05/21/2018] [Indexed: 12/15/2022] Open
Abstract
Zinc oxide nanoparticles (ZnO NPs) are used in an increasing number of industrial products such as rubber, paint, coating, and cosmetics. In the past two decades, ZnO NPs have become one of the most popular metal oxide nanoparticles in biological applications due to their excellent biocompatibility, economic, and low toxicity. ZnO NPs have emerged a promising potential in biomedicine, especially in the fields of anticancer and antibacterial fields, which are involved with their potent ability to trigger excess reactive oxygen species (ROS) production, release zinc ions, and induce cell apoptosis. In addition, zinc is well known to keep the structural integrity of insulin. So, ZnO NPs also have been effectively developed for antidiabetic treatment. Moreover, ZnO NPs show excellent luminescent properties and have turned them into one of the main candidates for bioimaging. Here, we summarize the synthesis and recent advances of ZnO NPs in the biomedical fields, which will be helpful for facilitating their future research progress and focusing on biomedical fields.
Collapse
Affiliation(s)
- Jinhuan Jiang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jiang Pi
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jiye Cai
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
- Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|
135
|
Sipos A, Kim KJ, Chow RH, Flodby P, Borok Z, Crandall ED. Alveolar epithelial cell processing of nanoparticles activates autophagy and lysosomal exocytosis. Am J Physiol Lung Cell Mol Physiol 2018; 315:L286-L300. [PMID: 29722567 DOI: 10.1152/ajplung.00108.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Using confocal microscopy, we quantitatively assessed uptake, processing, and egress of near-infrared (NIR)-labeled carboxylated polystyrene nanoparticles (PNP) in live alveolar epithelial cells (AEC) during interactions with primary rat AEC monolayers (RAECM). PNP fluorescence intensity (content) and colocalization with intracellular vesicles in a cell were determined over the entire cell volume via z stacking. Isotropic cuvette-based microfluorimetry was used to determine PNP concentration ([PNP]) from anisotropic measurements of PNP content assessed by confocal microscopy. Results showed that PNP uptake kinetics and steady-state intracellular content decreased as diameter increased from 20 to 200 nm. For 20-nm PNP, uptake rate and steady-state intracellular content increased with increased apical [PNP] but were unaffected by inhibition of endocytic pathways. Intracellular PNP increasingly colocalized with autophagosomes and/or lysosomes over time. PNP egress exhibited fast Ca2+ concentration-dependent release and a slower diffusion-like process. Inhibition of microtubule polymerization curtailed rapid PNP egress, resulting in elevated vesicular and intracellular PNP content. Interference with autophagosome formation led to slower PNP uptake and markedly decreased steady-state intracellular content. At steady state, cytosolic [PNP] was higher than apical [PNP], and vesicular [PNP] (~80% of intracellular PNP content) exceeded both cytosolic and intracellular [PNP]. These data are consistent with the following hypotheses: 1) autophagic processing of nanoparticles is essential for maintenance of AEC integrity; 2) altered autophagy and/or lysosomal exocytosis may lead to AEC injury; and 3) intracellular [PNP] in AEC can be regulated, suggesting strategies for enhancement of nanoparticle-driven AEC gene/drug delivery and/or amelioration of AEC nanoparticle-related cellular toxicity.
Collapse
Affiliation(s)
- Arnold Sipos
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Kwang-Jin Kim
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California , Los Angeles, California.,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California , Los Angeles, California
| | - Robert H Chow
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California , Los Angeles, California.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Per Flodby
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zea Borok
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Edward D Crandall
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Pathology, Keck School of Medicine, University of Southern California , Los Angeles, California.,Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California , Los Angeles, California
| |
Collapse
|
136
|
Iron(III)-Tannic Molecular Nanoparticles Enhance Autophagy effect and T 1 MRI Contrast in Liver Cell Lines. Sci Rep 2018; 8:6647. [PMID: 29703912 PMCID: PMC5923259 DOI: 10.1038/s41598-018-25108-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023] Open
Abstract
Herein, a new molecular nanoparticle based on iron(III)-tannic complexes (Fe–TA NPs) is presented. The Fe–TA NPs were simply obtained by mixing the precursors in a buffered solution at room temperature, and they exhibited good physicochemical properties with capability of inducing autophagy in both hepatocellular carcinoma cells (HepG2.2.15) and normal rat hepatocytes (AML12). The Fe–TA NPs were found to induce HepG2.2.15 cell death via autophagic cell death but have no effect on cell viability in AML12 cells. This is possibly due to the much higher uptake of the Fe–TA NPs by the HepG2.2.15 cells via the receptor-mediated endocytosis pathway. As a consequence, enhancement of the T1 MRI contrast was clearly observed in the HepG2.2.15 cells. The results demonstrate that the Fe–TA NPs could provide a new strategy combining diagnostic and therapeutic functions for hepatocellular carcinoma. Additionally, because of their autophagy-inducing properties, they can be applied as autophagy enhancers for prevention and treatment of other diseases.
Collapse
|
137
|
Zhang J, Zou Z, Wang B, Xu G, Wu Q, Zhang Y, Yuan Z, Yang X, Yu C. Lysosomal deposition of copper oxide nanoparticles triggers HUVEC cells death. Biomaterials 2018; 161:228-239. [DOI: 10.1016/j.biomaterials.2018.01.048] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/20/2018] [Accepted: 01/27/2018] [Indexed: 12/16/2022]
|
138
|
Wang L, Chen C, Guo L, Li Q, Ding H, Bi H, Guo D. Zinc oxide nanoparticles induce murine photoreceptor cell death via mitochondria-related signaling pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:1102-1113. [PMID: 29488395 DOI: 10.1080/21691401.2018.1446018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Zinc oxide (ZnO) nanoparticles can exhibit toxic effect on cells and tissues, which may be involved in the excessive generation of reactive oxygen species (ROS) and the consequent mitochondria-mediated apoptotic pathway. Nevertheless, the detailed mechanism remains unclear. In this study, we explored the effects of ZnO nanoparticles on the expressions of cytochrome c, ATP level, mitochondrial membrane potential, ROS, apoptosis, total antioxidant enzyme activities and apoptotic-related protein levels in murine photoreceptor cells as well as the changes of proteomic profiling. Moreover, we also performed the bioinformatics analysis for the differentially expressed proteins. Our results show that ZnO nanoparticles induce the release of cytochorme c, decrease the intracellular ATP level, collapse the mitochondrial membrane potential, elevate the ROS level, inhibit total antioxidant enzyme activities and increase the Bax and Caspase 3 levels whereas it decrease the Bcl-2 expression, leading to cell death. Proteomic analysis reveals the differentially expressed proteins are involved in cytochrome c oxidase activity and oxidative phosphorylation. Protein-protein interaction analysis confirms the differentially expressed proteins are closely associated with the clusters related to apoptotic signaling pathway and oxidative phosphorylation-associated proteins. Our results indicate that mitochondria play a central role in ZnO nanoparticle-induced murine photoreceptor cell death.
Collapse
Affiliation(s)
- Ling Wang
- a Clinical Medical College, Jining Medical University , Jining , Shandong Province , China
| | - Chao Chen
- b Department of Ophthalmology , The First People's Hospital of Jining , Jining , Shandong Province , China
| | - Lijie Guo
- c College of Life Sciences , Qingdao Agricultural University , Qingdao , Shandong Province , China
| | - Qin Li
- d Department of Integration of Chinese and Western Medicine , The Affiliated Yantai Yuhuangding Hospital of Qingdao University , Yantai , Shandong Province , China
| | - Hongyan Ding
- e Jiangsu Provincial Key Laboratory for Interventional Medical Devices , Huaiyin Institute of Technology , Huaian , Jiangsu Province , China
| | - Hongsheng Bi
- f Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases , Eye Institute of Shandong University of Traditional Chinese Medicine , Jinan , Shandong Province , China
| | - Dadong Guo
- f Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases , Eye Institute of Shandong University of Traditional Chinese Medicine , Jinan , Shandong Province , China
| |
Collapse
|
139
|
Sharma A, Gorey B, Casey A. In vitro comparative cytotoxicity study of aminated polystyrene, zinc oxide and silver nanoparticles on a cervical cancer cell line. Drug Chem Toxicol 2018; 42:9-23. [PMID: 29359584 DOI: 10.1080/01480545.2018.1424181] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nanoparticles use in nano-biotechnology applications have increased significantly with Aminated polystyrene amine (AmPs NP), Zinc oxide (ZnO NP), and Silver (Ag NP) nanoparticles utilized in wide variety of consumer products. This has presented a number of concerns due to their increased exposure risks and associated toxicity on living systems. Changes in the structural and physicochemical properties of nanoparticles can lead to changes in biological activities. This study investigates, compares, and contrasts the potential toxicity of AmPs, ZnO and Ag NPs on an in vitro model (HeLa cells) and assesses the associated mechanism for their corresponding cytotoxicity relative to the surface material. It was noted that NPs exposure attributed to the reduction in cell viability and high-level induction of oxidative stress. All three test particles were noted to induce ROS to varying degrees which is irrespective of the attached surface group. Cell cycle analysis indicated a G2/M phase cell arrest, with the corresponding reduction in G0/G1 and S phase cells resulting in caspase-mediated apoptotic cell death. These findings suggest that all three NPs resulted in the decrease in cell viability, increase intracellular ROS production, induce cell cycle arrest at the G2/M phase and finally result in cell death by caspase-mediated apoptosis, which is irrespective of their differences in physiochemical properties and attached surface groups.
Collapse
Affiliation(s)
- Akash Sharma
- a NANOLAB Research Centre , Focas Institute, Dublin Institute of Technology , Dublin 8 , Ireland.,b School of Physics, Clinical and Optometric Sciences , Dublin Institute of Technology , Dublin , Ireland
| | - Brian Gorey
- a NANOLAB Research Centre , Focas Institute, Dublin Institute of Technology , Dublin 8 , Ireland
| | - Alan Casey
- a NANOLAB Research Centre , Focas Institute, Dublin Institute of Technology , Dublin 8 , Ireland.,b School of Physics, Clinical and Optometric Sciences , Dublin Institute of Technology , Dublin , Ireland
| |
Collapse
|
140
|
Wang B, Zhang J, Chen C, Xu G, Qin X, Hong Y, Bose DD, Qiu F, Zou Z. The size of zinc oxide nanoparticles controls its toxicity through impairing autophagic flux in A549 lung epithelial cells. Toxicol Lett 2017; 285:51-59. [PMID: 29289694 DOI: 10.1016/j.toxlet.2017.12.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
Zinc oxide nanoparticles (ZnONPs) widely used in various products, have been concerned with its impact on human health, in particular, on the risk of pulmonary toxicity. Our previous study indicated that ZnONPs could harness autophagy and impair the autophagic flux, which was positively linked to ZnONPs-induced toxicity. The objective of this study was to investigate whether ZnONPs-induced impairment of autophagic flux and cell death in lung epithelial cells is related to the size of ZnONPs. We demonstrate that ZnONPs with the average size of 50 nm could induce toxic effects in A549 lung epithelial cells, including accumulation of autophagosomes (the elevation of LC3B-II/LC3B-I ratio), impaired autophagic flux (the increase of p62 expression), the release of intracellular zinc ions (the increase of FluoZin-3 signal and ZnT1 mRNA expression), mitochondrial damage (the decrease of TMRE signal), lysosomal dysfunction (the aberrant expression of LAMP-2), oxidative stress (the increase of DCFH-DA signal and HO-1 expression) and cell death. Interestingly, ZnONPs with the average size of 200 nm failed to induce autophagy-mediated toxicity. Taken together, our results indicate that the size of ZnONPs is closely correlated with its toxicity, which is probably mediated by induction of impaired autophagic flux. This finding provides an insight into better understating of ZnONPs-associated toxicity, and mitigating the risk to humans and allowing the safer application.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing 400016, China; Post-doctoral Research Stations of Nursing Science, School of Nursing, Chongqing Medical University, Chongqing 400016, China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yueling Hong
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Diptiman D Bose
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
141
|
Zhang B, Hou R, Zou Z, Luo T, Zhang Y, Wang L, Wang B. Mechanically induced autophagy is associated with ATP metabolism and cellular viability in osteocytes in vitro. Redox Biol 2017; 14:492-498. [PMID: 29096322 PMCID: PMC5680519 DOI: 10.1016/j.redox.2017.10.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/17/2017] [Accepted: 10/25/2017] [Indexed: 12/16/2022] Open
Abstract
Both mechanical loading and intracellular autophagy play important roles in bone homeostasis; however, their relationship remains largely unexplored. The objectives of this study were to determine whether osteocytes undergo autophagy upon fluid shear stress (FSS) loading and to determine the correlation between mechanically induced autophagy and ATP metabolism. Autophagic vacuoles were observed by transmission electron microscopy (TEM) in osteocyte-like MLO-Y4 cells subjected to FSS. Increased autophagic flux was further confirmed by the increased amount of the LC3-II isoform and the degradation of p62. Fluorescent puncta distributed in the cytoplasm were observed in the GFP-LC3 transformed cells subjected to FSS. Furthermore, FSS-induced ATP release and synthesis in osteocytes were attenuated by inhibiting autophagy with 3-MA. After FSS exposure, a high ratio of cell death was observed in cultures pretreated with 3-MA, an autophagy inhibitor, with no significantly different Caspase 3/7 activity. Our results indicated that FSS induces protective autophagy in osteocytes and that mechanically induced autophagy is associated with ATP metabolism and osteocyte survival. From the clinical perspective, it may be possible to enhance skeletal cell survival with drugs that modulate the autophagic state, and the autophagy-related pathway could be a potential target for the prevention of ageing-related bone disorders. Fluid flow shear stress (FSS) induces activation of autophagic flux in MLO-Y4 osteocytes. FSS-induced autophagy promoted ATP metabolism in MLO-Y4 osteocytes. Inhibited autophagy decreased FSS-induced ATP release. FSS-induced autophagy was beneficial to the osteocyte survival after FSS exposure.
Collapse
Affiliation(s)
- Bingbing Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Rutao Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zhen Zou
- Department of Medical Laboratory Technology, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Tiantian Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yang Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Bin Wang
- Department of Medical Laboratory Technology, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
142
|
Qin X, Qiu F, Zou Z. TRIM25 is associated with cisplatin resistance in non-small-cell lung carcinoma A549 cell line via downregulation of 14-3-3σ. Biochem Biophys Res Commun 2017; 493:568-572. [PMID: 28867193 DOI: 10.1016/j.bbrc.2017.08.151] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/30/2017] [Indexed: 12/30/2022]
Abstract
Lung cancer, in particular, non-small cell lung cancer (NSCLC), is the leading cause of cancer-related mortality. Cis-Diamminedichloroplatinum (cisplatin, CDDP) as first-line chemotherapy for NSCLC, but resistance occurs frequently. We previously reported that Tripartite motif protein 25 (TRIM25) was highly expressed in cisplatin-resistant human lung adenocarcinoma A549 cells (A549/CDDP) in comparison with its parental A549 cells. Herein, we take a further step to demonstrate the association of TRIM25 and cisplatin resistance and also the underlying mechanisms. Knockdown of TRIM25 by RNA interference in A549/CDDP cells decreased half maximal inhibitory concentration (IC50) values and promoted apoptosis in response to cisplatin, whereas overexpression of TRIM25 had opposite effects. More importantly, we found that concomitant knockdown of 14-3-3σ and TRIM25 absolutely reversed the decreased MDM2, increased p53, increased cleaved-Capsese3 and decreased IC50 value induced by knockdown of TRIM25 individually, suggesting that TRIM25 mediated cisplatin resistance primarily through downregulation of 14-3-3σ. Our results indicate that TRIM25 is associated with cisplatin resistance and 14-3-3σ-MDM2-p53 signaling pathway is involved in this process, suggesting targeting TRIM25 may be a potential strategy for the reversal of cisplatin resistance.
Collapse
Affiliation(s)
- Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China.
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, 400016 Chongqing, China.
| |
Collapse
|