51
|
Lin YR, Chan CH, Lee HT, Cheng SJ, Yang JW, Chang SJ, Lin SF, Chen GY. Remote Magnetic Control of Autophagy in Mouse B-Lymphoma Cells with Iron Oxide Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E551. [PMID: 30987307 PMCID: PMC6524120 DOI: 10.3390/nano9040551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022]
Abstract
Autophagy is the spontaneous degradation of intracellular proteins and organelles in response to nutrient deprivation. The phagocytosis of iron oxide nanoparticles (IONPs) results in intracellular degradation that can be exploited for use in cancer treatment. Non-invasive magnetic control has emerged as an important technology, with breakthroughs achieved in areas such as magneto-thermal therapy and drug delivery. This study aimed to regulate autophagy in mouse B-lymphoma cells (A20) through the incorporation of IONPs-quantum dots (QDs). We hypothesized that with the application of an external magnetic field after phagocytosis of IONPs-QDs, autophagy of intracellular IONPs-QDs could be regulated in a non-invasive manner and subsequently modulate the regulation of inflammatory responses. The potential of this approach as a cancer treatment method was explored. The application of IONPs and an external magnetic force enabled the non-invasive regulation of cell autophagy and modulation of the self-regulatory function of cells. The combination of non-invasive magnetic fields and nanotechnology could provide a new approach to cancer treatment.
Collapse
Affiliation(s)
- You-Rong Lin
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
| | - Chia-Hao Chan
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Gynecologic Oncology Section Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu 300, Taiwan.
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan.
| | - Hui-Ting Lee
- Division of Allergy, Immunology and Rheumatology, MacKay Memorial Hospital, Taipei 10491, Taiwan.
| | - Sheng-Jen Cheng
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
| | - Jia-Wei Yang
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
| | - Shing-Jyh Chang
- Gynecologic Oncology Section Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu 300, Taiwan.
| | - Shien-Fong Lin
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
| | - Guan-Yu Chen
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan.
| |
Collapse
|
52
|
Qi S, Qian J, Chen F, Zhou P, Yue J, Tang F, Zhang Y, Gong S, Shang G, Cui C, Xu Y. Expression of autophagy‑associated proteins in rat dental irreversible pulpitis. Mol Med Rep 2019; 19:2749-2757. [PMID: 30816453 PMCID: PMC6423575 DOI: 10.3892/mmr.2019.9944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 01/25/2019] [Indexed: 11/23/2022] Open
Abstract
Autophagy serves an important role in numerous diseases, as well as in infection and inflammation. Irreversible pulpitis (IP) is one of the most common inflammatory endodontic diseases, and autophagy has been reported to regulate IP in vitro. However, the level of autophagy in the IP pathogenic process in vivo remains unknown. The aim of the current study was, thus, to investigate the levels of autophagy-associated proteins in rats with IP in vivo. A rat dental IP model was successfully constructed, and five different time points (0, 1, 3, 5 and 7 days) were investigated. The levels of the autophagy-related 5 (ATG5), ATG7, light chain 3 (LC3) and Beclin-1 proteins exhibited a time-dependent increase in rats with IP, whereas the levels of mammalian target of rapamycin and p62/sequestosome 1 were decreased. In addition, the levels of ATG proteins were specifically increased in odontoblasts and microvascular endothelial cells in pulpitis tissue. Based on these findings, autophagy may serve an important role in IP, and the present study data provide a new insight into the IP pathogenesis and treatment.
Collapse
Affiliation(s)
- Shengcai Qi
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jun Qian
- Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Fubo Chen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Peng Zhou
- Jiangsu Key Laboratory of Oral Diseases, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jing Yue
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Fengqin Tang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yiming Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Shiqiang Gong
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Guangwei Shang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Chun Cui
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
53
|
Lee K, Lee J, Kwak M, Cho YL, Hwang B, Cho MJ, Lee NG, Park J, Lee SH, Park JG, Kim YG, Kim JS, Han TS, Cho HS, Park YJ, Lee SJ, Lee HG, Kim WK, Jeung IC, Song NW, Bae KH, Min JK. Two distinct cellular pathways leading to endothelial cell cytotoxicity by silica nanoparticle size. J Nanobiotechnology 2019; 17:24. [PMID: 30722792 PMCID: PMC6362579 DOI: 10.1186/s12951-019-0456-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/11/2019] [Indexed: 12/18/2022] Open
Abstract
Background Silica nanoparticles (SiNPs) are widely used for biosensing and diagnostics, and for the targeted delivery of therapeutic agents. Safety concerns about the biomedical and clinical applications of SiNPs have been raised, necessitating analysis of the effects of their intrinsic properties, such as sizes, shapes, and surface physicochemical characteristics, on human health to minimize risk in biomedical applications. In particular, SiNP size-associated toxicological effects, and the underlying molecular mechanisms in the vascular endothelium remain unclear. This study aimed to elucidate the detailed mechanisms underlying the cellular response to exposure to trace amounts of SiNPs and to determine applicable size criteria for biomedical application. Methods To clarify whether these SiNP-mediated cytotoxicity due to induction of apoptosis or necrosis, human ECs were treated with SiNPs of four different non-overlapping sizes under low serum-containing condition, stained with annexin V and propidium iodide (PI), and subjected to flow cytometric analysis (FACS). Two types of cell death mechanisms were assessed in terms of production of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress induction, and autophagy activity. Results Spherical SiNPs had a diameter of 21.8 nm; this was further increased to 31.4, 42.9, and 56.7 nm. Hence, we investigated these effects in human endothelial cells (ECs) treated with these nanoparticles under overlap- or agglomerate-free conditions. The 20-nm SiNPs, but not SiNPs of other sizes, significantly induced apoptosis and necrosis. Surprisingly, the two types of cell death occurred independently and through different mechanisms. Apoptotic cell death resulted from ROS-mediated ER stress. Furthermore, autophagy-mediated necrotic cell death was induced through the PI3K/AKT/eNOS signaling axis. Together, the present results indicate that SiNPs within a diameter of < 20-nm pose greater risks to cells in terms of cytotoxic effects. Conclusion These data provide novel insights into the size-dependence of the cytotoxic effects of silica nanoparticles and the underlying molecular mechanisms. The findings are expected to inform the applicable size range of SiNPs to ensure their safety in biomedical and clinical applications. Electronic supplementary material The online version of this article (10.1186/s12951-019-0456-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kyungmin Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Minjeong Kwak
- Center for Nano-Bio Measurement, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Young-Lai Cho
- Research Center for Metabolic Regulation, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Byungtae Hwang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Min Ji Cho
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Na Geum Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Jongjin Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Sang-Hyun Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jang-Seong Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Tae-Su Han
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Young-Jun Park
- Research Center for Metabolic Regulation, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seon-Jin Lee
- Immunotherapy Convergence Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Convergence Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Won Kon Kim
- Research Center for Metabolic Regulation, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - In Cheul Jeung
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero Seocho-gu, Seoul, 06591, Republic of Korea
| | - Nam Woong Song
- Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| | - Kwang-Hee Bae
- Research Center for Metabolic Regulation, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea. .,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
54
|
Gulla S, Lomada D, Srikanth VV, Shankar MV, Reddy KR, Soni S, Reddy MC. Recent advances in nanoparticles-based strategies for cancer therapeutics and antibacterial applications. J Microbiol Methods 2019. [DOI: 10.1016/bs.mim.2019.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
55
|
Du Z, Chen S, Cui G, Yang Y, Zhang E, Wang Q, Lavin MF, Yeo AJ, Bo C, Zhang Y, Li C, Liu X, Yang X, Peng C, Shao H. Silica nanoparticles induce cardiomyocyte apoptosis via the mitochondrial pathway in rats following intratracheal instillation. Int J Mol Med 2018; 43:1229-1240. [PMID: 30628656 PMCID: PMC6365031 DOI: 10.3892/ijmm.2018.4045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022] Open
Abstract
Diseases of the cardiac system caused by silicon dioxide exposure have captured wide public attention. Upon entering the blood circulation, ultrafine particles have the potential to influence cardiomyocytes, leading to myocardial ischemia or even cardiac failure, and the molecular mechanisms remain to be completely elucidated. In this study, the toxicity of ultrafine particles on cardiomyocytes from rats exposed to silica nanoparticles was observed. Rats were randomly divided into a normal saline control group and three exposure groups (2, 5 and 10 mg/kg·body weight) that were intratracheally treated with 60-nm silica nanoparticles. Alterations in body weight, routine blood factors and myocardial enzymes, histopathological and microstructural alterations, apoptosis and the expression of apoptosis-associated proteins were assessed at the end of the exposure period. The silicon levels in the heart and serum, and myocardial enzymes in exposed rats were significantly increased in a dose-dependent manner. In addition, exposure to the silica nanoparticles caused notable histological and ultrastructural alterations in the hearts of these animals. Furthermore, a significant apoptotic effect was observed in the exposure groups. The present data suggest that silica nanoparticles may enter the circulatory system through the lungs, and are distributed to the heart causing cardiovascular injury. Silica nanoparticle-induced apoptosis via the mitochondrial pathway may serve an important role in observed cardiac damage.
Collapse
Affiliation(s)
- Zhongjun Du
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Shangya Chen
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Guanqun Cui
- Department of Respiratory Medicine, Qilu Children's Hospital of Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Ye Yang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Enguo Zhang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Qiang Wang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Martin F Lavin
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Abrey J Yeo
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Cunxiang Bo
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Yu Zhang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Chao Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Xiaoshan Liu
- Department of Radiology, Shandong Tumor Hospital, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250117, P.R. China
| | - Xu Yang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Cheng Peng
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| | - Hua Shao
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong 250062, P.R. China
| |
Collapse
|
56
|
Breznan D, Das DD, MacKinnon-Roy C, Bernatchez S, Sayari A, Hill M, Vincent R, Kumarathasan P. Physicochemical Properties Can Be Key Determinants of Mesoporous Silica Nanoparticle Potency in Vitro. ACS NANO 2018; 12:12062-12079. [PMID: 30475590 DOI: 10.1021/acsnano.8b04910] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nanoforms of mesoporous silica (mSiNPs) are increasingly applied in medicine, imaging, energy storage, catalysis, biosensors, and bioremediation. The impact of their physicochemical properties on health and the environment remain to be elucidated. In this work, newly synthesized mesoporous silica (sizes: 25, 70, 100, 170, and 600 nm; surface functionalization: pristine, C3-, and C11-COOH moieties) were assessed for cytotoxicity and induction of inflammatory responses in vitro (A549, THP-1, J774A.1 cells). All toxicity end points were integrated to obtain simple descriptors of biological potencies of these mSiNPs. The findings indicate that mSiNPs are less bioactive than the nonporous reference SiNP used in this study. The C3-COOH-modified mSiNPs were generally less cytotoxic than their pristine and C11-modified counterparts in the nanorange (≤100 nm). Carboxyl-modified mSiNPs affected inflammatory marker release across all sizes with cell-type specificity, suggesting a potential for immunomodulatory effects. Surface area, size, extent of agglomeration, ζ-potential, and surface modification appeared to be important determinants of cytotoxicity of mSiNPs based on association tests. Pathway analysis identified particle and cell-type-specific alteration of cellular pathways and functions by mSiNPs. The integration of exposure-related biological responses of multiple cell lines to mSiNPs allowed for a comprehensive evaluation of the impact of physicochemical factors on their toxicity characteristics. The integrated multilevel toxicity assessment approach can be valuable as a hazard screening tool for safety evaluations of emerging nanomaterials for regulatory purpose.
Collapse
Affiliation(s)
| | | | | | | | - Abdelhamid Sayari
- Department of Chemistry and Biomolecular Sciences , University of Ottawa , Ottawa , Ontario K1N 6N5 , Canada
| | | | | | | |
Collapse
|
57
|
Asweto CO, Hu H, Liang S, Wang L, Liu M, Yang H, Duan J, Sun Z. Gene profiles to characterize the combined toxicity induced by low level co-exposure of silica nanoparticles and benzo[a]pyrene using whole genome microarrays in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 163:47-55. [PMID: 30036756 DOI: 10.1016/j.ecoenv.2018.07.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/12/2018] [Accepted: 07/15/2018] [Indexed: 06/08/2023]
Abstract
Several studies have suggested that air pollutants combine exposure have greater adverse effects. However, limited studies were available on the combined toxicity of silica nanoparticles (SiNPs) and benzo[a]pyrene (B[a]P). The study was to evaluate the toxic effect and mechanisms of low-dose exposure of SiNPs, B[a]P and co-exposure in zebrafish embryos. In this study, zebrafish embryos received intravenous microinjection of SiNPs and B[a]P, and then was used to select differentially expressed genes by microarray analysis. Multiple bioinformatics analyses and STC analysis were done to identify key genes, pathways and biological processes and the expression trend of genes in each group. 1) 3065 differentially expressed genes were identified in zebrafish embryos. 2) These differentially expressed genes were involved in multiple biological processes and cellular processes such as immunity, response to stimuli, cell proliferation, adhesion, signaling transduction, and embryonic development. 3) Dynamic Gene Network analysis was used to identify a subgroup of 26 core genes that involved in multiple biological processes and cellular processes. 4) Pathway analysis and Signal-net analysis indicated that the MAPK signaling pathway, calcium signaling pathway, p53 signaling pathway, PI3k/Akt signaling pathway, and several pathways associated with immune response were the most prominent significant pathways induced by co-exposure of SiNPs and B[a]P in zebrafish embryos. Our study demonstrated that the molecular actions of co-treated with SiNPs and B[a]P on the immune system, inflammatory process and cardiovascular development had more severe toxicity than single exposure.
Collapse
Affiliation(s)
- Collins Otieno Asweto
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Hejing Hu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Shuang Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Lijing Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Mengdi Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Han Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| |
Collapse
|
58
|
Guo C, Ma R, Liu X, Xia Y, Niu P, Ma J, Zhou X, Li Y, Sun Z. Silica nanoparticles induced endothelial apoptosis via endoplasmic reticulum stress-mitochondrial apoptotic signaling pathway. CHEMOSPHERE 2018; 210:183-192. [PMID: 29990757 DOI: 10.1016/j.chemosphere.2018.06.170] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/20/2018] [Accepted: 06/27/2018] [Indexed: 06/08/2023]
Abstract
Along with their extensively application, human exposure to amorphous silica nanoparticles (SiNPs) has highly increased. Accumulative toxicological researches have provided the scientific correlation between SiNPs exposure and cardiovascular diseases. Endothelial apoptosis is vital in the initiation and progression of atherosclerosis. However, molecular details between SiNPs and endothelial apoptosis remain unidentified. Here, we investigated the uptake and toxic mechanism of SiNPs using HUVECs (Human umbilical vein endothelial cells). Consequently, at 24-h exposure, SiNPs were located freely or within membrane-bound agglomerates in the cytosol, especially in mitochondrial and endoplasmic reticulum (ER) regions with swelled mitochondria, cristae rupture or aggregated ER. Further, we demonstrated that SiNPs induced endothelial apoptosis as evidenced by the Annexin V/PI staining and flow cytometry determination. In line with the ultrastructure alterations, SiNPs triggered mitochondrial ROS generation, ΔΨm collapse, cytosolic Ca2+ overload, as well as ER stress confirmed by enhanced ER staining, up-regulated GRP78/BiP and XBP1 splicing. More notably, in line with the induction of apoptosis, SiNPs-induced ER stress-associated activation of CHOP, caspase-12, and IRE1α/JNK pathways, which may regulate the BCL2 family member as evidenced by a increased proapoptotic BAX while a decline of anti-apoptotic Bcl-2, ultimately facilitate the mitochondria-mediated apoptotic caspase cascade as confirmed by the upregulated expressions of cytochrome c, Caspase-9 and -3. Altogether, our results indicated the activation of ER stress-mitochondria cascade-mediated apoptotic pathways may be a key mechanism among the SiNPs-induced endothelial apoptosis.
Collapse
Affiliation(s)
- Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Ru Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xiaoying Liu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Yinye Xia
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Piye Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Junxiang Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xianqing Zhou
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Zhiwei Sun
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
59
|
Yang X, Feng L, Zhang Y, Hu H, Shi Y, Liang S, Zhao T, Fu Y, Duan J, Sun Z. Cytotoxicity induced by fine particulate matter (PM 2.5) via mitochondria-mediated apoptosis pathway in human cardiomyocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 161:198-207. [PMID: 29885615 DOI: 10.1016/j.ecoenv.2018.05.092] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/20/2018] [Accepted: 05/29/2018] [Indexed: 06/08/2023]
Abstract
Although the strongly causal associations were between fine particulate matter (PM2.5) and cardiovascular disease, the toxic effect and potential mechanism of PM2.5 on heart was poorly understood. Thus, the aim of this study was to evaluate the cardiac toxicity of PM2.5 exposure on human cardiomyocytes (AC16). The cell viability was decreased while the LDH release was increased in a dose-dependent way after AC16 exposed to PM2.5. The reactive oxygen species (ROS) generation and production of malondialdehyde (MDA) were increased followed by the decreasing in superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). The damage of mitochondria was observed by ultra-structural analysis and MMP measurement. The apoptotic rate of AC16 were markedly elevated which was triggered by PM2.5. In addition, the proteins involved in mitochondria- mediated apoptosis pathway were measured. The protein levels of Caspase-3, Caspase-9 and Bax were up-regulated while the anti-apoptotic protein, Bcl-2 was down-regulated after AC16 exposed to PM2.5. In summary, our results demonstrated that mitochondria-mediated apoptosis pathway played a critical role in PM2.5-induced myocardial cytotoxicity in AC16, which suggested that PM2.5 may contribute to cardiac dysfunction.
Collapse
Affiliation(s)
- Xiaozhe Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Lin Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yannan Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Hejing Hu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yanfeng Shi
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Shuang Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Tong Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yang Fu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
60
|
Shen N, Zhang R, Zhang HR, Luo HY, Shen W, Gao X, Guo DZ, Shen J. Inhibition of retinal angiogenesis by gold nanoparticles via inducing autophagy. Int J Ophthalmol 2018; 11:1269-1276. [PMID: 30140628 DOI: 10.18240/ijo.2018.08.04] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/26/2018] [Indexed: 01/13/2023] Open
Abstract
AIM To investigate the effect of gold nanoparticles on retinal angiogenesis in vitro and in vivo, and to reveal the possible mechanism. METHODS Seed growth method was used to synthesize gold nanoparticles (GNPs). The size, zeta potential, absorption spectrum and morphology of GNPs were identified using Malvern Nano-ZS, multimode reader (BioTek synergy2) and transmission electron microscope. Cell viability was analyzed using cell counting kit-8 method and cell growth was assessed with EdU kit. Transwell chamber was used to investigate cell migration. Tube formation method was used to assess the angiogenic property in vitro. Oxygen induced retinopathy (OIR) model was used to investigate the effect of GNPs on retinal angiogenesis. Confocal microscope and Western blot were used to study the possible mechanism of GNPs inhibited angiogenesis. RESULTS The GNPs synthesized were uniform and well dispersed. GNPs of 10 µg/mL and 20 µg/mL were able to inhibit human umbilical vein endothelial cells proliferation (50% and 72% separately, P<0.001), migration (54% and 83% separately, P<0.001) and tube formation (52% and 90% separately, P<0.001). Further data showed that GNPs were able to improve the retinopathy in an OIR model. The possible mechanism might be that GNPs were able to induce autophagy significantly (P<0.05). CONCLUSION The present study suggests that GNPs are able to inhibit retinal neovascularization in vitro and in vivo. GNPs might be a potential nanomedicine for the treatment of retinal angiogenesis.
Collapse
Affiliation(s)
- Ni Shen
- Department of Ophthalmology, Shanghai Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.,Department of Ophthalmology, Changhai Hospital, the Second Military Medical Univerisity, Shanghai 200433, China
| | - Rui Zhang
- Department of Ophthalmology, Changhai Hospital, the Second Military Medical Univerisity, Shanghai 200433, China
| | - Hao-Rui Zhang
- Department of Ophthalmology, Changhai Hospital, the Second Military Medical Univerisity, Shanghai 200433, China.,Company 6 of Basic Medical School, the Second Military Medical University, Shanghai 200433, China
| | - Hao-Yang Luo
- School of Life Science, Fudan University, Shanghai 200082, China
| | - Wei Shen
- Department of Ophthalmology, Changhai Hospital, the Second Military Medical Univerisity, Shanghai 200433, China
| | - Xin Gao
- Department of Ophthalmology, Changhai Hospital, the Second Military Medical Univerisity, Shanghai 200433, China
| | - Da-Zhi Guo
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing 100037, China
| | - Jie Shen
- Department of Ophthalmology, Shanghai Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
61
|
Chen L, Liu J, Zhang Y, Zhang G, Kang Y, Chen A, Feng X, Shao L. The toxicity of silica nanoparticles to the immune system. Nanomedicine (Lond) 2018; 13:1939-1962. [PMID: 30152253 DOI: 10.2217/nnm-2018-0076] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Silicon-based materials and their oxides are widely used in drug delivery, dietary supplements, implants and dental fillers. Silica nanoparticles (SiNPs) interact with immunocompetent cells and induce immunotoxicity. However, the toxic effects of SiNPs on the immune system have been inadequately reviewed. The toxicity of SiNPs to the immune system depends on their physicochemical properties and the cell type. Assessments of immunotoxicity include determining cell dysfunctions, cytotoxicity and genotoxicity. This review focuses on the immunotoxicity of SiNPs and investigates the underlying mechanisms. The main mechanisms were proinflammatory responses, oxidative stress and autophagy. Considering the toxicity of SiNPs, surface and shape modifications may mitigate the toxic effects of SiNPs, providing a new way to produce these nanomaterials with less toxic impaction.
Collapse
Affiliation(s)
- Liangjiao Chen
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 510140, PR China
| | - Jia Liu
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Yanli Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Guilan Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Aijie Chen
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xiaoli Feng
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| |
Collapse
|
62
|
Wang J, Li Y, Duan J, Yang M, Yu Y, Feng L, Yang X, Zhou X, Zhao Z, Sun Z. Silica nanoparticles induce autophagosome accumulation via activation of the EIF2AK3 and ATF6 UPR pathways in hepatocytes. Autophagy 2018; 14:1185-1200. [PMID: 29940794 DOI: 10.1080/15548627.2018.1458174] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Autophagy dysfunction is a potential toxic effect of nanoparticles. Previous studies have indicated that silica nanoparticles (SiNPs) induce macroautophagy/autophagy dysfunction, while the precise mechanisms remain uncertain. Hence, the present study investigated the molecular mechanisms by which SiNPs enhanced autophagosome synthesis, which then contributed to autophagy dysfunction. First, the effects of SiNPs on autophagy and autophagic flux were verified using transmission electron microscopy, laser scanning confocal microscopy, and western blot assays. Then, the activation of endoplasmic reticular (ER) stress was validated to be through the EIF2AK3 and ATF6 UPR pathways but not the ERN1-XBP1 pathway, along with the upregulation of downstream ATF4 and DDIT3. Thereafter, the ER stress inhibitor 4-phenylbutyrate (4-PBA) was used to verify that SiNP-induced autophagy could be influenced by ER stress. Furthermore, specialized lentiviral shRNA were employed to determine that autophagy was induced via specific activation of the EIF2AK3 and ATF6 UPR pathways. Finally, the 2 autophagic genes LC3B and ATG12 were found to be transcriptionally upregulated by downstream ATF4 and DDIT3 in ER stress, which contributed to the SiNP-enhanced autophagosome synthesis. Taken together, these data suggest that SiNPs induced autophagosome accumulation via the activation of the EIF2AK3 and ATF6 UPR pathways in hepatocytes, which offers a new insight into detailed molecular mechanisms underlying SiNP-induced autophagy dysfunction, and specifically how UPR pathways regulate key autophagic genes. This work provides novel evidence for the study of toxic effects and risk assessment of SiNPs.
Collapse
Affiliation(s)
- Ji Wang
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Li
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Junchao Duan
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Man Yang
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Yu
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Lin Feng
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Xiaozhe Yang
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Xianqing Zhou
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Zhendong Zhao
- c MOH Key Laboratory of Systems Biology of Pathogens , Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing , China
| | - Zhiwei Sun
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| |
Collapse
|
63
|
Guo C, Wang J, Jing L, Ma R, Liu X, Gao L, Cao L, Duan J, Zhou X, Li Y, Sun Z. Mitochondrial dysfunction, perturbations of mitochondrial dynamics and biogenesis involved in endothelial injury induced by silica nanoparticles. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 236:926-936. [PMID: 29074197 DOI: 10.1016/j.envpol.2017.10.060] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 10/09/2017] [Accepted: 10/14/2017] [Indexed: 05/15/2023]
Abstract
As silica nanoparticles (SiNPs) pervade the global economy, however, the followed emissions during the manufacturing, use, and disposal stages inevitably bring an environmental release, potentially result in harmful impacts. Endothelial dysfunction precedes cardiovascular disease, and is often accompanied by mitochondrial impairment and dysfunction. We had reported endothelial dysfunction induced by SiNPs, however, the related mechanisms by which SiNPs interact with mitochondria are not well understood. In the present study, we examined SiNPs-induced mitochondrial dysfunction, and further demonstrated their adverse effects on mitochondrial dynamics and biogenesis in endothelial cells (HUVECs). Consequently, SiNPs entered mitochondria, caused mitochondrial swelling, cristae disruption and even disappearance. Further analyses revealed SiNPs increased the intracellular level of mitochondrial reactive oxygen species, eventually resulting in the collapse of mitochondrial membrane potential, impairments in ATP synthesis, cellular respiration and the activities of three ATP-dependent enzymes (including Na+/K+-ATPase, Ca2+-ATPase and Ca2+/Mg2+-ATPase), as well as an elevated intracellular calcium level. Furthermore, mitochondria in SiNPs-treated HUVECs displayed a fission phenotype. Accordingly, dysregulation of the key gene expressions (FIS1, DRP1, OPA1, Mfn1 and Mfn2) involved in fission/fusion event further certified the SiNPs-induced perturbation of mitochondrial dynamics. Meanwhile, SiNPs-treated HUVECs displayed declined levels of mitochondrial DNA copy number, PGC-1α, NRF1 and also TFAM, indicating an inhibition of mitochondrial biogenesis triggered by SiNPs via PGC-1α-NRF1-TFAM signaling. Overall, SiNPs triggered endothelial toxicity through mitochondria as target, including the induction of mitochondrial dysfunction, as well as the perturbations of their dynamics and biogenesis.
Collapse
Affiliation(s)
- Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Ji Wang
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Li Jing
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Ru Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xiaoying Liu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Lifang Gao
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Lige Cao
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Junchao Duan
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xianqing Zhou
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Zhiwei Sun
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| |
Collapse
|
64
|
Duan J, Liang S, Yu Y, Li Y, Wang L, Wu Z, Chen Y, Miller MR, Sun Z. Inflammation-coagulation response and thrombotic effects induced by silica nanoparticles in zebrafish embryos. Nanotoxicology 2018; 12:470-484. [PMID: 29658397 PMCID: PMC6157531 DOI: 10.1080/17435390.2018.1461267] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Nowadays, nanotechnology environmental health and safety (nanoEHS) is gaining attention. We previously found that silica nanoparticles (SiNPs) could induce vascular endothelial damage. However, the subsequent toxicologic response to SiNPs-induced endothelial damage was still largely unknown. In this study, we explored the inflammation–coagulation response and thrombotic effects of SiNPs in endothelial cells and zebrafish embryos. For in vitro study, swollen mitochondria and autophagosome were observed in ultrastructural analysis. The cytoskeleton organization was disrupted by SiNPs in vascular endothelial cells. The release of proinflammatory and procoagulant cytokines including IL-6, IL-8, MCP-1, PECAM-1, TF and vWF, were markedly elevated in a dose-dependent manner. For in vivo study, based on the NOAEL for dosimetry selection, and using two transgenic zebrafish, Tg(mpo:GFP) and Tg(fli-1:EGFP), SiNPs-induced neutrophil-mediated inflammation and impaired vascular endothelial cells. With the dosage higher than NOAEL, SiNPs significantly decreased blood flow and velocity, exhibiting a blood hypercoagulable state in zebrafish embryos. The thrombotic effect was assessed by o-dianisidine staining, showed that an increasing of erythrocyte aggregation occurred in SiNPs-treated zebrafish. Microarray analysis was used to screen the possible genes for inflammation–coagulation response to SiNPs in zebrafish, and the JAK1/TF signaling pathway was further verified by qRT-PCR and Western blot assays. For in-deepth study, il6st was knocked down with specific morpholinos. The whole-mount in situ hybridization and qRT-PCR analysis showed that the expression jak1 and f3b were attenuated in il6st knockdown groups. In summary, our data demonstrated that SiNPs could induce inflammation–coagulation response and thrombotic effects via JAK1/TF signaling pathway.
Collapse
Affiliation(s)
- Junchao Duan
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Shuang Liang
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Yu
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Li
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Lijing Wang
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Zehao Wu
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yueyue Chen
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Mark R Miller
- c BHF Centre for Cardiovascular Science, Queens Medical Research Institute, The University of Edinburgh , Edinburgh , UK
| | - Zhiwei Sun
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| |
Collapse
|
65
|
Wan H, Chen J, Zhu X, Liu L, Wang J, Zhu X. Titania-Coated Gold Nano-Bipyramids for Blocking Autophagy Flux and Sensitizing Cancer Cells to Proteasome Inhibitor-Induced Death. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700585. [PMID: 29593960 PMCID: PMC5867123 DOI: 10.1002/advs.201700585] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/16/2017] [Indexed: 05/18/2023]
Abstract
Targeting protein degradation is recognized as a valid approach to cancer therapy. The ubiquitin-proteasome system (UPS) and the autophagy-lysosome pathway are two major pathways for intracellular protein degradation. Proteasome inhibitors such as bortezomib are clinically approved for treating malignancies, but to date, they are still unsatisfactory for cancer therapy. This study identifies titania-coated gold nano-bipyramid (NBP/TiO2) nanostructures as an autophagic flux inhibitor, as the smallest NBP/TiO2 nanostructures induce significant autophagosome accumulation in human glioblastoma U-87 MG cells via blocking the autophagosome-lysosome fusion process and inhibiting lysosomal degradation. Further study indicates that NBP/TiO2 nanostructures reduce the intracellular level of mature cathepsin B and directly inhibit the proteolytic activity of cathepsin B, thereby further inhibiting trypsin-like proteolytic activity, which is a potential cotarget for UPS inhibition. NBP/TiO2 nanostructures interact synergistically with bortezomib to suppress the viability of U-87 MG cells, as the combined treatment synergistically induces the intracellular accumulation of ubiquitinated protein and endoplasmic reticulum stress. In addition, photothermal therapy further synergistically reduces the cell viability. In summary, this study suggests that NBP/TiO2 nanostructures function as a promising anticancer agent in combination with proteasome inhibitors.
Collapse
Affiliation(s)
- Hong‐Ye Wan
- State Key Laboratory of Quality Research in Chinese MedicineMacau Institute for Applied Research in Medicine and HealthMacau University of Science and TechnologyAvenida Wai LongTaipaMacau SARChina
| | - Jian‐Li Chen
- State Key Laboratory of Quality Research in Chinese MedicineMacau Institute for Applied Research in Medicine and HealthMacau University of Science and TechnologyAvenida Wai LongTaipaMacau SARChina
| | - Xingzhong Zhu
- Department of PhysicsThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese MedicineMacau Institute for Applied Research in Medicine and HealthMacau University of Science and TechnologyAvenida Wai LongTaipaMacau SARChina
| | - Jianfang Wang
- Department of PhysicsThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Xiao‐Ming Zhu
- State Key Laboratory of Quality Research in Chinese MedicineMacau Institute for Applied Research in Medicine and HealthMacau University of Science and TechnologyAvenida Wai LongTaipaMacau SARChina
| |
Collapse
|
66
|
Fan C, Graff P, Vihlborg P, Bryngelsson IL, Andersson L. Silica exposure increases the risk of stroke but not myocardial infarction-A retrospective cohort study. PLoS One 2018; 13:e0192840. [PMID: 29481578 PMCID: PMC5826533 DOI: 10.1371/journal.pone.0192840] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 01/31/2018] [Indexed: 02/02/2023] Open
Abstract
Introduction Work-related exposure to silica is a global health hazard that causes diseases such as silicosis. Some studies have also reported that silica exposure is linked to elevated cardiovascular disease mortality. However, these diagnoses have not been investigated in detail and there have been few studies on morbidity. The aim of this study is to examine morbidity and mortality from different cardiovascular diseases among silica-exposed Swedish foundry workers. Methods Historical and contemporary measurements (1968–2006) of respiratory silica exposure were matched to job categories, individual foundries, and 4 time periods (1968–1979, 1980–1989, 1990–1999, 2000–2006) using a mixed model. Morbidity and mortality data for the studied cohorts were matched against the General Population Registry. Statistical analyses were performed with SPSS and STATA, and the data were stratified by age, gender, and year. Results Mortality from cardiovascular disease (SMR 1.3; 95% CI 1.2–1.4) and stroke (SMR 1.6, 95% CI 1.2–2.1) was significantly elevated among the studied population. The cohort also exhibited significantly elevated morbidity from stroke (SIR 1.34; 95% CI 1.2–1.5) but not myocardial infarction. The mean age at the time of first morbidity from stroke was 64 years, with 36% of the cases occurring before the age of 60. Conclusions Swedish foundry workers exposed to respirable silica exhibit elevated morbidity and mortality from stroke, but not from myocardial infarction. Our results also suggest a relationship between silica exposure and morbidity from stroke at a younger age than the general population.
Collapse
Affiliation(s)
- Chenjing Fan
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Pål Graff
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.,National Institute of Occupational Health (STAMI), Oslo, Norway
| | - Per Vihlborg
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ing-Liss Bryngelsson
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Lena Andersson
- Department of Occupational and Environmental Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
67
|
Wang W, Zeng C, Feng Y, Zhou F, Liao F, Liu Y, Feng S, Wang X. The size-dependent effects of silica nanoparticles on endothelial cell apoptosis through activating the p53-caspase pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 233:218-225. [PMID: 29096294 DOI: 10.1016/j.envpol.2017.10.053] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 08/24/2017] [Accepted: 10/12/2017] [Indexed: 06/07/2023]
Abstract
With the growing production and applications of silica nanoparticles (SiNPs), human exposure to these nanoparticles continues to increase. However, the possible hazards that SiNP exposure may pose to human cardiovascular system and the underlying mechanisms remain unclear. In the present study, the flow cytometry was employed to investigate the potential of four sizes (10, 25, 50, 100 nm) of SiNPs to induce the apoptosis of human umbilical vein endothelial cells (HUVECs) in culture. The apoptotic pathway was also explored through the determination of the protein expression and/or activation of p53, Bcl-2, Bax, caspases-9, -7, -3, and PARP by western blot. The results showed that all the four sizes of SiNPs could significantly elicit apoptosis in HUVECs at the tested concentrations (1, 5, 25 μg/mL), compared with the negative control (p < 0.05, p < 0.01). Moreover, the apoptotic rates were increased with the elevating levels and decreasing sizes of administrative SiNPs, showing both dose- and size-dependent effect relationships. Interestingly, the enhancing phosphorylation of p53 protein (Ser15), decreasing ratio of Bcl-2/Bax protein, and elevating activation of the downstream proteins, caspase-9, -7, -3 and PARP, were also observed with the decreasing sizes of tested SiNPs, indicating that the p53-caspase pathway is the main way of the SiNP-mediated apoptosis in HUVECs and that the size is an important parameter that determines the SiNPs' potential to induce cellular response.
Collapse
Affiliation(s)
- Wuxiang Wang
- The School of Public Health, University of South China, Hengyang 421001, China
| | - Can Zeng
- The School of Public Health, University of South China, Hengyang 421001, China
| | - Yuqin Feng
- The College of Materials Science and Engineering, Jilin University, Changchun 130022, China
| | - Furong Zhou
- The School of Public Health, University of South China, Hengyang 421001, China
| | - Fen Liao
- The School of Public Health, University of South China, Hengyang 421001, China
| | - Yuanfeng Liu
- The School of Public Health, University of South China, Hengyang 421001, China
| | - Shaolong Feng
- The School of Public Health, University of South China, Hengyang 421001, China.
| | - Xinming Wang
- The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China.
| |
Collapse
|
68
|
Hu H, Shi Y, Zhang Y, Wu J, Asweto CO, Feng L, Yang X, Duan J, Sun Z. Comprehensive gene and microRNA expression profiling on cardiovascular system in zebrafish co-exposured of SiNPs and MeHg. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 607-608:795-805. [PMID: 28711009 DOI: 10.1016/j.scitotenv.2017.07.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 06/07/2023]
Abstract
Air pollution has been shown to increase cardiovascular diseases. However, little attention has been paid to the combined effects of PM and air pollutants on the cardiovascular system. To explore this, a high-throughput sequencing technology was used to determine combined effects of silica nanoparticles (SiNPs) and MeHg in zebrafish. Our study demonstrated that SiNPs and MeHg co-exposure could cause significant changes in mRNA and miRNA expression patterns in zebrafish. The differentially expressed (DE) genes in profiles 17 and 26 of STC analysis suggest that SiNPs and MeHg co-exposure had more proinflammatory and cardiovascular toxicity in zebrafish than single exposure. Major gene functions associated with cardiovascular system in the co-exposed zebrafish were discerned from the dynamic-gene-network, including stxbp1a, celf4, ahr1b and bai2. In addition, the prominently expressed pathway of cardiac muscle contraction was targeted by 3 DE miRNAs identified by the miRNA-pathway-network (dre-miR-7147, dre-miR-26a and dre-miR-375), which included 23 DE genes. This study presents a global view of the combined SiNPs and MeHg toxicity on the dynamic expression of both mRNAs and miRNAs in zebrafish, and could serve as fundamental research clues for future studies, especially on cardiovascular system toxicity.
Collapse
Affiliation(s)
- Hejing Hu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yanfeng Shi
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yannan Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Jing Wu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Collins Otieno Asweto
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Lin Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Xiaozhe Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
69
|
Anozie UC, Dalhaimer P. Molecular links among non-biodegradable nanoparticles, reactive oxygen species, and autophagy. Adv Drug Deliv Rev 2017; 122:65-73. [PMID: 28065863 DOI: 10.1016/j.addr.2017.01.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 12/15/2022]
Abstract
For nanoparticles to be successful in combating diseases in the clinic in the 21st century and beyond, they must localize to target areas of the body and avoid damaging non-target, healthy tissues. Both soft and stiff, bio-degradable and non-biodegradable nanoparticles are anticipated to be used to this end. It has been shown that stiff, non-biodegradable nanoparticles cause reactive oxygen species (ROS) generation and autophagy in a variety of cell lines in vitro. Both responses can lead to significant remodeling of the cytosol and even apoptosis. Thus these are crucial cellular functions to understand. Improved assays have uncovered crucial roles of the Akt/mTOR signaling pathway in both ROS generation and autophagy initiation after cells have internalized stiff, non-biodegradable nanoparticles over varying geometries in culture. Of particular - yet unresolved - interest is how these nanoparticles cause the activation of these pathways. This article reviews the most recent advances in nanoparticle generation of ROS and autophagy initiation with a focus on stiff, non-biodegradable technologies. We provide experimental guidelines to the reader for fleshing out the effects of their nanoparticles on the above pathways with the goal of tuning nanoparticle design.
Collapse
|
70
|
Hu H, Zhang Y, Shi Y, Feng L, Duan J, Sun Z. Microarray-based bioinformatics analysis of the combined effects of SiNPs and PbAc on cardiovascular system in zebrafish. CHEMOSPHERE 2017; 184:1298-1309. [PMID: 28679150 DOI: 10.1016/j.chemosphere.2017.06.112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 06/20/2017] [Accepted: 06/27/2017] [Indexed: 06/07/2023]
Abstract
With rapid development of nanotechnology and growing environmental pollution, the combined toxic effects of SiNPs and pollutants of heavy metals like lead have received global attentions. The aim of this study was to explore the cardiovascular effects of the co-exposure of SiNPs and lead acetate (PbAc) in zebrafish using microarray and bioinformatics analysis. Although there was no other obvious cardiovascular malformation except bleeding phenotype, bradycardia, angiogenesis inhibition and declined cardiac output in zebrafish co-exposed of SiNPs and PbAc at NOAEL level, significant changes were observed in mRNA and microRNA (miRNA) expression patterns. STC-GO analysis indicated that the co-exposure might have more toxic effects on cardiovascular system than that exposure alone. Key differentially expressed genes were discerned out based on the Dynamic-gene-network, including stxbp1a, ndfip2, celf4 and gsk3b. Furthermore, several miRNAs obtained from the miRNA-Gene-Network might play crucial roles in cardiovascular disease, such as dre-miR-93, dre-miR-34a, dre-miR-181c, dre-miR-7145, dre-miR-730, dre-miR-129-5p, dre-miR-19d, dre-miR-218b, dre-miR-221. Besides, the analysis of miRNA-pathway-network indicated that the zebrafish were stimulated by the co-exposure of SiNPs and PbAc, which might cause the disturbance of calcium homeostasis and endoplasmic reticulum stress. As a result, cardiac muscle contraction might be deteriorated. In general, our data provide abundant fundamental research clues to the combined toxicity of environmental pollutants and further in-depth verifications are needed.
Collapse
Affiliation(s)
- Hejing Hu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yannan Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yanfeng Shi
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Lin Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| |
Collapse
|
71
|
Murugadoss S, Lison D, Godderis L, Van Den Brule S, Mast J, Brassinne F, Sebaihi N, Hoet PH. Toxicology of silica nanoparticles: an update. Arch Toxicol 2017; 91:2967-3010. [PMID: 28573455 PMCID: PMC5562771 DOI: 10.1007/s00204-017-1993-y] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 05/18/2017] [Indexed: 12/18/2022]
Abstract
Large-scale production and use of amorphous silica nanoparticles (SiNPs) have increased the risk of human exposure to SiNPs, while their health effects remain unclear. In this review, scientific papers from 2010 to 2016 were systematically selected and sorted based on in vitro and in vivo studies: to provide an update on SiNPs toxicity and to address the knowledge gaps indicated in the review of Napierska (Part Fibre Toxicol 7:39, 2010). Toxicity of SiNPs in vitro is size, dose, and cell type dependent. SiNPs synthesized by wet route exhibited noticeably different biological effects compared to thermal route-based SiNPs. Amorphous SiNPs (particularly colloidal and stöber) induced toxicity via mechanisms similar to crystalline silica. In vivo, route of administration and physico-chemical properties of SiNPs influences the toxicokinetics. Adverse effects were mainly observed in acutely exposed animals, while no significant signs of toxicity were noted in chronically dosed animals. The correlation between in vitro and in vivo toxicity remains less well established mainly due to improper-unrealistic-dosing both in vitro and in vivo. In conclusion, notwithstanding the multiple studies published in recent years, unambiguous linking of physico-chemical properties of SiNPs types to toxicity, bioavailability, or human health effects is not yet possible.
Collapse
Affiliation(s)
- Sivakumar Murugadoss
- Unit for Lung Toxicology, Katholieke Universiteit Leuven, Herestraat 49, O&N1, Room: 07.702, box 706, 3000 Louvain, Belgium
| | - Dominique Lison
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Université Catholique de Louvain, Avenue E. Mounier 52/B1.52.12, 1200 Brussels, Belgium
| | - Lode Godderis
- Department of Occupational, Environmental and Insurance Medicine, Katholieke Universiteit Leuven, Kapucijnenvoer 35 block d, box 7001, 3000 Louvain, Belgium
| | - Sybille Van Den Brule
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Université Catholique de Louvain, Avenue E. Mounier 52/B1.52.12, 1200 Brussels, Belgium
| | - Jan Mast
- EM-unit, Center for Veterinary and Agrochemical Studies and Research (CODA-CERVA), Groeselenberg 99, Uccle, 1180 Brussels, Belgium
| | - Frederic Brassinne
- EM-unit, Center for Veterinary and Agrochemical Studies and Research (CODA-CERVA), Groeselenberg 99, Uccle, 1180 Brussels, Belgium
| | - Noham Sebaihi
- General Quality and Safety, Metrology Department, National Standards, North Gate-Office 2A29, Bd du Roi Albert II, 16, 1000 Brussels, Belgium
| | - Peter H. Hoet
- Unit for Lung Toxicology, Katholieke Universiteit Leuven, Herestraat 49, O&N1, Room: 07.702, box 706, 3000 Louvain, Belgium
| |
Collapse
|
72
|
Duan J, Hu H, Feng L, Yang X, Sun Z. Silica nanoparticles inhibit macrophage activity and angiogenesis via VEGFR2-mediated MAPK signaling pathway in zebrafish embryos. CHEMOSPHERE 2017; 183:483-490. [PMID: 28570891 DOI: 10.1016/j.chemosphere.2017.05.138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/16/2017] [Accepted: 05/23/2017] [Indexed: 06/07/2023]
Abstract
The safety evaluation of silica nanoparticles (SiNPs) are getting great attention due to its widely-used in food sciences, chemical industry and biomedicine. However, the adverse effect and underlying mechanisms of SiNPs on cardiovascular system, especially on angiogenesis is still unclear. This study was aimed to illuminate the possible mechanisms of SiNPs on angiogenesis in zebrafish transgenic lines, Tg(fli-1:EGFP) and Albino. SiNPs caused the cardiovascular malformations in a dose-dependent manner via intravenous microinjection. The incidences of cardiovascular malformations were observed as: Pericardial edema > Bradycardia > Blood deficiency. The area of subintestinal vessels (SIVs) was significant reduced in SiNPs-treated groups, accompanied with the weaken expression of vascular endothelial cells in zebrafish embryos. Using neutral red staining, the quantitative number of macrophage was declined; whereas macrophage inhibition rate was elevated in a dose-dependent way. Furthermore, SiNPs significantly decreased the mRNA expression of macrophage activity related gene, macrophage migration inhibitory factor (MIF) and the angiogenesis related gene, vascular endothelial growth factor receptor 2 (VEGFR2). The protein levels of p-Erk1/2 and p-p38 MAPK were markedly decreased in zebrafish exposed to SiNPs. Our results implicate that SiNPs inhibited the macrophage activity and angiogenesis via the downregulation of MAPK singaling pathway.
Collapse
Affiliation(s)
- Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China.
| | - Hejing Hu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Lin Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Xiaozhe Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| |
Collapse
|
73
|
Yu Y, Duan J, Li Y, Li Y, Jing L, Yang M, Wang J, Sun Z. Silica nanoparticles induce liver fibrosis via TGF-β 1/Smad3 pathway in ICR mice. Int J Nanomedicine 2017; 12:6045-6057. [PMID: 28860765 PMCID: PMC5573053 DOI: 10.2147/ijn.s132304] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The liver is one of the target organs of silica nanoparticles (SiO2 NPs) but the toxic mechanism on the liver still remains unclear. This study aimed to explore the hepatic toxicity and its mechanism through repeated intravenous exposure to SiO2 NPs in ICR mice. Results indicated that SiO2 NPs could be distributed in hepatocytes, Kupffer cells, and hepatic stellate cells, and induce hepatic dysfunction as well as granuloma formation in the liver. The increase of lipid peroxide level and decrease of antioxidant enzyme activities in the liver indicated that SiO2 NPs could induce hepatic oxidative damage. SiO2 NPs induced hepatocytes’ apoptosis shown by morphological examination and TUNEL assay. The results of Masson’s trichrome staining and hydroxyproline assay showed hyperplasia of collagen fibers in the liver, suggesting SiO2 NPs caused liver fibrosis, and it was promoted by oxidative damage and hepatocytes’ apoptosis. The results of Western blot analysis and immunohistochemical staining indicated that the activation of TGF-β1/Smad3 signaling pathway played an important role in this pathophysiological process. The results suggested that oxidative damage and hepatocyte apoptosis activated TGF-β1/Smad3 signaling pathway, and thus promoted the process of liver fibrosis induced by intravenous injection of SiO2 NPs in mice. This study, for the first time, investigated liver fibrosis and its related mechanism induced by repeated intravenous exposure of amorphous SiO2 NPs, and provides important experimental evidence for safety evaluation of SiO2 NPs, especially in biomedical application.
Collapse
Affiliation(s)
- Yang Yu
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Junchao Duan
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Yang Li
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Yanbo Li
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Li Jing
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Man Yang
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Ji Wang
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Zhiwei Sun
- School of Public Health, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
74
|
Dornhof R, Maschowski C, Osipova A, Gieré R, Seidl M, Merfort I, Humar M. Stress fibers, autophagy and necrosis by persistent exposure to PM2.5 from biomass combustion. PLoS One 2017; 12:e0180291. [PMID: 28671960 PMCID: PMC5495337 DOI: 10.1371/journal.pone.0180291] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/13/2017] [Indexed: 02/06/2023] Open
Abstract
Fine particulate matter (PM2.5) can adversely affect human health. Emissions from residential energy sources have the largest impact on premature mortality globally, but their pathological and molecular implications on cellular physiology are still elusive. In the present study potential molecular consequences were investigated during long-term exposure of human bronchial epithelial BEAS-2B cells to PM2.5, collected from a biomass power plant. Initially, we observed that PM2.5 did not affect cellular survival or proliferation. However, it triggered an activation of the stress response p38 MAPK which, along with RhoA GTPase and HSP27, mediated morphological changes in BEAS-2B cells, including actin cytoskeletal rearrangements and paracellular gap formation. The p38 inhibitor SB203580 prevented phosphorylation of HSP27 and ameliorated morphological changes. During an intermediate phase of long-term exposure, PM2.5 triggered proliferative regression and activation of an adaptive stress response necessary to maintain energy homeostasis, including AMPK, repression of translational elongation, and autophagy. Finally, accumulation of intracellular PM2.5 promoted lysosomal destabilization and cell death, which was dependent on lysosomal hydrolases and p38 MAPK, but not on the inflammasome and pyroptosis. TEM images revealed formation of protrusions and cellular internalization of PM2.5, induction of autophagosomes, amphisomes, autophagosome-lysosomal fusion, multiple compartmental fusion, lysosomal burst, swollen mitochondria and finally necrosis. In consequence, persistent exposure to PM2.5 may impair epithelial barriers and reduce regenerative capacity. Hence, our results contribute to a better understanding of PM-associated lung and systemic diseases on the basis of molecular events.
Collapse
Affiliation(s)
- Regina Dornhof
- Institute of Pharmaceutical Sciences, Pharmaceutical Biology and Biotechnology, Albert-Ludwigs University Freiburg, Freiburg, Germany
| | - Christoph Maschowski
- Institute of Earth and Environmental Sciences, Albert-Ludwigs University Freiburg, Freiburg, Germany
| | - Anastasiya Osipova
- Institute of Pharmaceutical Sciences, Pharmaceutical Biology and Biotechnology, Albert-Ludwigs University Freiburg, Freiburg, Germany
| | - Reto Gieré
- Department of Earth and Environmental Science and Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maximilian Seidl
- Institute for Surgical Pathology, Faculty of Medicine, Albert-Ludwigs University Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, Albert-Ludwigs University Freiburg, Freiburg, Germany
| | - Irmgard Merfort
- Institute of Pharmaceutical Sciences, Pharmaceutical Biology and Biotechnology, Albert-Ludwigs University Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs University Freiburg, Freiburg, Germany
- * E-mail: (IM); (MH)
| | - Matjaz Humar
- Institute of Pharmaceutical Sciences, Pharmaceutical Biology and Biotechnology, Albert-Ludwigs University Freiburg, Freiburg, Germany
- * E-mail: (IM); (MH)
| |
Collapse
|
75
|
Yan Z, Wang W, Wu Y, Wang W, Li B, Liang N, Wu W. Zinc oxide nanoparticle-induced atherosclerotic alterations in vitro and in vivo. Int J Nanomedicine 2017; 12:4433-4442. [PMID: 28652743 PMCID: PMC5476650 DOI: 10.2147/ijn.s134897] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Engineered zinc oxide nanoparticles (ZnO-NPs) are currently being produced in high tonnage. Exposure to ZnO-NPs presents potential risks to cardiovascular system. Thus far, the toxicological effects of ZnO-NPs on cardiovascular system have not been well characterized. In this study, human coronary artery endothelial cells (HCAECs) were exposed to ZnO-NPs directly or indirectly using a transwell coculture system with human alveolar epithelial cell line A549 to mimic the lung/circulation interaction. It was shown that levels of proinflammatory mediators (interleukin-8 [IL-8] and tumor necrosis factor-α [TNF-α]) and biomarkers of atherosclerogenesis (heme oxygenase-1 [HO-1] and platelet endothelial cell adhesion molecules-1 [PECAM-1]) in the supernatants of culture media were significantly increased. Pretreatment of A549 cells on the apical side of the coculture system with the phagocytosis inhibitor cytochalasin B (CB) blocked ZnO-NP-induced HO-1 and PECAM-1 expression in HCAEC, indicating that endocytosis of ZnO-NPs by alveolar epithelial cells was involved in ZnO-NP-induced HO-1 or PECAM-1 expression in endothelial cells. Moreover, Wistar rats were intratracheally instilled with ZnO-NP suspension and high fat diet (positive control). ZnO-NP treatment induced lung and systemic inflammation, dyslipidemia, increased levels of serum HO-1 and PECAM-1, and aortic pathological damage. Taken together, exposure to ZnO-NPs could induce atherosclerotic alterations, which might involve phagocytosis of nanoparticles and inflammation in the lung.
Collapse
Affiliation(s)
- Zhen Yan
- College of Public Health, Zhengzhou University, Zhengzhou
| | - Wenjun Wang
- School of Public Health, Jining Medical University, Jining
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou
| | - Wei Wang
- School of Public Health, Jining Medical University, Jining
| | - Bing Li
- College of Public Health, Zhengzhou University, Zhengzhou
| | - Ning Liang
- College of Public Health, Zhengzhou University, Zhengzhou
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, People's Republic of China
| |
Collapse
|
76
|
Wu Q, Jin R, Feng T, Liu L, Yang L, Tao Y, Anderson JM, Ai H, Li H. Iron oxide nanoparticles and induced autophagy in human monocytes. Int J Nanomedicine 2017; 12:3993-4005. [PMID: 28603414 PMCID: PMC5457122 DOI: 10.2147/ijn.s135189] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Superparamagnetic iron oxide nanoparticles have been widely used in biomedical applications, but understanding of their interactions with the biological immune system is relatively limited. This work is focused on dextran-coated iron oxide nanoparticles and their induced autophagy in human monocytes. We found that these nanoparticles can be taken up by human monocytes, followed by localization within vesicles or free in cytoplasm. Autophagosome formation was observed with increased expression of LC3II protein, the specific marker of autophagy. The autophagy substrate p62 was degraded in a dose-dependent manner, and autophagy was blocked by autophagy (or lysosome) inhibitors alone or along with iron oxide nanoparticles, indicating that autophagosome accumulation was mainly due to autophagy induction, rather than blockade of autophagy flux. Interestingly, iron oxide nanoparticles increased the viability of human monocytes, but the mechanism was not clear. We further found that inhibition of autophagy mostly attenuated the survival of cells, with acceleration of the inflammation induced by these nanoparticles. Taken together, autophagic activation in human monocytes may play a protective role against the cytotoxicity of iron oxide nanoparticles.
Collapse
Affiliation(s)
- QiHong Wu
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital
| | - RongRong Jin
- National Engineering Research Center for Biomaterials
| | - Ting Feng
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital
| | - Li Liu
- National Engineering Research Center for Biomaterials
| | - Li Yang
- National Engineering Research Center for Biomaterials
| | - YuHong Tao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - James M Anderson
- Department of Biomedical Engineering.,Department of Pathology, Case Western Reserve University, Cleveland, OH, US
| | - Hua Ai
- National Engineering Research Center for Biomaterials.,Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital.,Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
77
|
Orlando A, Cazzaniga E, Tringali M, Gullo F, Becchetti A, Minniti S, Taraballi F, Tasciotti E, Re F. Mesoporous silica nanoparticles trigger mitophagy in endothelial cells and perturb neuronal network activity in a size- and time-dependent manner. Int J Nanomedicine 2017; 12:3547-3559. [PMID: 28507435 PMCID: PMC5428814 DOI: 10.2147/ijn.s127663] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Purpose Mesoporous silica nanoparticles (MSNPs) are excellent candidates for biomedical applications and drug delivery to different human body areas, the brain included. Although toxicity at cellular level has been investigated, we are still far from using MSNPs in the clinic, because the mechanisms involved in the cellular responses activated by MSNPs have not yet been elucidated. Materials and methods This study used an in vitro multiparametric approach to clarify relationships among size, dose, and time of exposure of MSNPs (0.05–1 mg/mL dose range), and cellular responses by analyzing the morphology, viability, and functionality of human vascular endothelial cells and neurons. Results The results showed that 24 hours of exposure of endothelial cells to 250 nm MSNPs exerted higher toxicity in terms of mitochondrial activity and membrane integrity than 30 nm MSN at the same dose. This was due to induced cell autophagy (in particular mitophagy), probably consequent to MSNP cellular uptake (>20%). Interestingly, after 24 hours of treatment with 30 nm MSNPs, very low MSNP uptake (<1%) and an increase in nitric oxide production (30%, P<0.01) were measured. This suggests that MSNPs were able to affect endothelial functionality from outside the cells. These differences could be attributed to the different protein-corona composition of the MSNPs used, as suggested by sodium dodecyl sulfate polyacrylamide-gel electrophoresis analysis of the plasma proteins covering the MSNP surface. Moreover, doses of MSNPs up to 0.25 mg/mL perturbed network activity by increasing excitability, as detected by multielectrode-array technology, without affecting neuronal cell viability. Conclusion These results suggest that MSNPs may be low-risk if prepared with a diameter <30 nm and if they reach human tissues at doses <0.25 mg/mL. These important advances could help the rational design of NPs intended for biomedical uses, demonstrating that careful toxicity evaluation is necessary before using MSNPs in patients.
Collapse
Affiliation(s)
- Antonina Orlando
- Nanomedicine Center, School of Medicine and Surgery, University of Milano-Bicocca, Monza
| | - Emanuela Cazzaniga
- Nanomedicine Center, School of Medicine and Surgery, University of Milano-Bicocca, Monza
| | | | - Francesca Gullo
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Andrea Becchetti
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Stefania Minniti
- Nanomedicine Center, School of Medicine and Surgery, University of Milano-Bicocca, Monza
| | - Francesca Taraballi
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI).,Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI).,Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Francesca Re
- Nanomedicine Center, School of Medicine and Surgery, University of Milano-Bicocca, Monza
| |
Collapse
|
78
|
Blood-brain barrier dysfunction induced by silica NPs in vitro and in vivo : Involvement of oxidative stress and Rho-kinase/JNK signaling pathways. Biomaterials 2017; 121:64-82. [DOI: 10.1016/j.biomaterials.2017.01.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 01/03/2017] [Accepted: 01/03/2017] [Indexed: 01/03/2023]
|
79
|
Use of Zebrafish Larvae as a Multi-Endpoint Platform to Characterize the Toxicity Profile of Silica Nanoparticles. Sci Rep 2016; 6:37145. [PMID: 27872490 PMCID: PMC5131651 DOI: 10.1038/srep37145] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/25/2016] [Indexed: 12/22/2022] Open
Abstract
Nanomaterials are being extensively produced and applied in society. Human and environmental exposures are, therefore, inevitable and so increased attention is being given to nanotoxicity. While silica nanoparticles (NP) are one of the top five nanomaterials found in consumer and biomedical products, their toxicity profile is poorly characterized. In this study, we investigated the toxicity of silica nanoparticles with diameters 20, 50 and 80 nm using an in vivo zebrafish platform that analyzes multiple endpoints related to developmental, cardio-, hepato-, and neurotoxicity. Results show that except for an acceleration in hatching time and alterations in the behavior of zebrafish embryos/larvae, silica NPs did not elicit any developmental defects, nor any cardio- and hepatotoxicity. The behavioral alterations were consistent for both embryonic photomotor and larval locomotor response and were dependent on the concentration and the size of silica NPs. As embryos and larvae exhibited a normal touch response and early hatching did not affect larval locomotor response, the behavior changes observed are most likely the consequence of modified neuroactivity. Overall, our results suggest that silica NPs do not cause any developmental, cardio- or hepatotoxicity, but they pose a potential risk for the neurobehavioral system.
Collapse
|
80
|
Wu J, Shi Y, Asweto CO, Feng L, Yang X, Zhang Y, Hu H, Duan J, Sun Z. Co-exposure to amorphous silica nanoparticles and benzo[a]pyrene at low level in human bronchial epithelial BEAS-2B cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:23134-23144. [PMID: 27591886 DOI: 10.1007/s11356-016-7559-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 08/29/2016] [Indexed: 06/06/2023]
Abstract
Both ultrafine particles (UFP) and polycyclic aromatic hydrocarbons (PAHs) are widely present in the environment, thus increasing their chances of exposure to human in the daily life. However, the study on the combined toxicity of UFP and PAHs on respiratory system is still limited. In this study, we examined the potential interactive effects of silica nanoparticles (SiNPs) and benzo[a]pyrene (B[a]P) in bronchial epithelial cells (BEAS-2B). Cells were exposed to SiNPs and B[a]P alone or in combination for 24 h. Co-exposure to SiNPs and B[a]P enhanced the malondialdehyde (MDA) contents and reduced superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities significantly, while the reactive oxygen species (ROS) generation had a slight increase in the exposed groups compared to the control but not statistically significant. Cell cycle arrest induced by the co-exposure showed a significant percentage increase in G2/M phase cells and a decrease in G0/G1 phase cells. In addition, there was a significant increase in BEAS-2B cells multinucleation as well as DNA damage. Cellular apoptosis was markedly increased even at the low-level co-exposure. Our results suggest that co-exposure to SiNPs and B[a]P exerts synergistic and additive cytotoxic and genotoxic effects.
Collapse
Affiliation(s)
- Jing Wu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yanfeng Shi
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Collins Otieno Asweto
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Lin Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Xiaozhe Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yannan Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hejing Hu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
81
|
Guo C, Yang M, Jing L, Wang J, Yu Y, Li Y, Duan J, Zhou X, Li Y, Sun Z. Amorphous silica nanoparticles trigger vascular endothelial cell injury through apoptosis and autophagy via reactive oxygen species-mediated MAPK/Bcl-2 and PI3K/Akt/mTOR signaling. Int J Nanomedicine 2016; 11:5257-5276. [PMID: 27785026 PMCID: PMC5066858 DOI: 10.2147/ijn.s112030] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Environmental exposure to silica nanoparticles (SiNPs) is inevitable due to their widespread application in industrial, commercial, and biomedical fields. In recent years, most investigators focus on the evaluation of cardiovascular effects of SiNPs in vivo and in vitro. Endothelial injury and dysfunction is now hypothesized to be a dominant mechanism in the development of cardiovascular diseases. This study aimed to explore interaction of SiNPs with endothelial cells, and extensively investigate the exact effects of reactive oxygen species (ROS) on the signaling molecules and cytotoxicity involved in SiNPs-induced endothelial injury. Significant induction of cytotoxicity as well as oxidative stress, apoptosis, and autophagy was observed in human umbilical vein endothelial cells following the SiNPs exposure (P<0.05). The oxidative stress was induced by ROS generation, leading to redox imbalance and lipid peroxidation. SiNPs induced mitochondrial dysfunction, characterized by membrane potential collapse, and elevated Bax and declined bcl-2 expression, ultimately leading to apoptosis, and also increased number of autophagosomes and autophagy marker proteins, such as LC3 and p62. Phosphorylated ERK, PI3K, Akt, and mTOR were significantly decreased, but phosphorylated JNK and p38 MAPK were increased in SiNPs-exposed endothelial cells. In contrast, all of these stimulation phenomena were effectively inhibited by N-acetylcysteine. The N-acetylcysteine supplement attenuated SiNPs-induced endothelial toxicity through inhibition of apoptosis and autophagy via MAPK/Bcl-2 and PI3K/Akt/mTOR signaling, as well as suppression of intracellular ROS property via activating antioxidant enzyme and Nrf2 signaling. In summary, the results demonstrated that SiNPs triggered autophagy and apoptosis via ROS-mediated MAPK/Bcl-2 and PI3K/Akt/mTOR signaling in endothelial cells, and subsequently disturbed the endothelial homeostasis and impaired endothelium. Our findings may provide experimental evidence and explanation for cardiovascular diseases triggered by SiNPs. Furthermore, results hint that the application of antioxidant may provide a novel way for safer use of nanomaterials.
Collapse
Affiliation(s)
- Caixia Guo
- Department of Occupational and Environmental Health, School of Public Health
- Beijing Key Laboratory of Environmental Toxicology
| | - Man Yang
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Li Jing
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Ji Wang
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Yang Yu
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Yang Li
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Junchao Duan
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Xianqing Zhou
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Zhiwei Sun
- Beijing Key Laboratory of Environmental Toxicology
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
82
|
Xing R, Li KL, Zhou YF, Su YY, Yan SQ, Zhang KL, Wu SC, Sima YH, Zhang KQ, He Y, Xu SQ. Impact of fluorescent silicon nanoparticles on circulating hemolymph and hematopoiesis in an invertebrate model organism. CHEMOSPHERE 2016; 159:628-637. [PMID: 27348562 DOI: 10.1016/j.chemosphere.2016.06.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/03/2016] [Accepted: 06/13/2016] [Indexed: 05/07/2023]
Abstract
Silicon nanoparticles (SiNPs) have attractive potential applications in biological and medical fields, and yet their impact on animals is still controversial, and there have been no reports of their effects on hematopoiesis. In this study, the effects of SiNPs on hemocytes and hematopoiesis were investigated by administering SiNPs via a vascular injection into an invertebrate model, the silkworm. Our results show that the ability of SiNPs to enter different types of circulating hemocytes and their impact on those hemocytes differed significantly. Rapid accumulation of SiNPs was observed in granulocytes, oenocytoids, and spherulocytes, which have immune functions in the circulating hemolymph, whereas SiNPs did not easily enter prohemocytes, which can differentiate into granulocytes, oenocytoids, and spherulocytes and replenish them. The SiNPs that entered the hemocytes initiated autophagy and apoptosis via the lysosomal/mitochondrial pathway. High-dose SiNPs weakly stimulated lysosomal activity in hematopoietic organs, but did not lead to a significant increase in reactive oxygen species or severe autophagy or apoptosis in the organ tissues. We suggest that the damage caused by high-dose SiNPs to hematopoiesis is self-healing, because few SiNPs entered the hematopoietic stem cells in the circulating hemolymph, so the damage to the hematopoietic tissues was limited.
Collapse
Affiliation(s)
- Rui Xing
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China; National Engineering Laboratory for Modern Silk (NESER), Soochow University, Suzhou, 215123, China
| | - Kai-Le Li
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China; National Engineering Laboratory for Modern Silk (NESER), Soochow University, Suzhou, 215123, China
| | - Yan-Feng Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, China
| | - Yuan-Yuan Su
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, China
| | - Si-Qi Yan
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China; National Engineering Laboratory for Modern Silk (NESER), Soochow University, Suzhou, 215123, China
| | - Kai-Long Zhang
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China; National Engineering Laboratory for Modern Silk (NESER), Soochow University, Suzhou, 215123, China
| | - Si-Cong Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, China
| | - Yang-Hu Sima
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China; National Engineering Laboratory for Modern Silk (NESER), Soochow University, Suzhou, 215123, China
| | - Ke-Qin Zhang
- National Engineering Laboratory for Modern Silk (NESER), Soochow University, Suzhou, 215123, China; Research Center of Cooperative Innovation for Functional Organic/Polymer Material Micro/Nanofabrication, Soochow University, Suzhou, 215123, China.
| | - Yao He
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, China.
| | - Shi-Qing Xu
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China; National Engineering Laboratory for Modern Silk (NESER), Soochow University, Suzhou, 215123, China.
| |
Collapse
|
83
|
Shukur A, Whitehead D, Seifalian A, Azzawi M. The influence of silica nanoparticles on small mesenteric arterial function. Nanomedicine (Lond) 2016; 11:2131-46. [DOI: 10.2217/nnm-2016-0124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To determine the influence of silica nanoparticles (SiNPs) on small arterial function; both ex vivo and in vivo. Methods: Mono-dispersed dye-encapsulated SiNPs (97.85 ± 2.26 nm) were fabricated and vasoconstrictor and vasodilator responses of mesenteric arteries assessed. Results: We show that while exposure to SiNPs under static conditions, attenuated endothelial dependent dilator responses ex vivo, attenuation was only evident at lower agonist concentrations, when exposed under flow conditions or after intravenous administration in vivo. Pharmacological inhibition studies suggest that SiNPs may interfere with the endothelial dependent hyperpolarizing factor vasodilator pathway. Conclusion: The dosage dependent influence of SiNPs on arterial function will help identify strategies for their safe clinical administration.
Collapse
Affiliation(s)
- Ali Shukur
- School of Healthcare Science, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Debra Whitehead
- School of Science & the Environment, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Alexander Seifalian
- Centre for Nanotechnology & Regenerative Medicine, UCL Division of Surgery & Interventional Science, University College London, London, UK
| | - May Azzawi
- School of Healthcare Science, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
84
|
Mukherjee S, Patra CR. Therapeutic application of anti-angiogenic nanomaterials in cancers. NANOSCALE 2016; 8:12444-12470. [PMID: 27067119 DOI: 10.1039/c5nr07887c] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vasculature, plays a vital role in physiological and pathological processes (embryonic development, wound healing, tumor growth and metastasis). The overall balance of angiogenesis inside the human body is maintained by pro- and anti-angiogenic signals. The processes by which drugs inhibit angiogenesis as well as tumor growth are called the anti-angiogenesis technique, a most promising cancer treatment strategy. Over the last couple of decades, scientists have been developing angiogenesis inhibitors for the treatment of cancers. However, conventional anti-angiogenic therapy has several limitations including drug resistance that can create problems for a successful therapeutic strategy. Therefore, a new comprehensive treatment strategy using antiangiogenic agents for the treatment of cancer is urgently needed. Recently researchers have been developing and designing several nanoparticles that show anti-angiogenic properties. These nanomedicines could be useful as an alternative strategy for the treatment of various cancers using anti-angiogenic therapy. In this review article, we critically focus on the potential application of anti-angiogenic nanomaterial and nanoparticle based drug/siRNA/peptide delivery systems in cancer therapeutics. We also discuss the basic and clinical perspectives of anti-angiogenesis therapy, highlighting its importance in tumor angiogenesis, current status and future prospects and challenges.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad - 500007, Telangana, India.
| | | |
Collapse
|
85
|
DNA Hypermethylation of CREB3L1 and Bcl-2 Associated with the Mitochondrial-Mediated Apoptosis via PI3K/Akt Pathway in Human BEAS-2B Cells Exposure to Silica Nanoparticles. PLoS One 2016; 11:e0158475. [PMID: 27362941 PMCID: PMC4928798 DOI: 10.1371/journal.pone.0158475] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/16/2016] [Indexed: 01/12/2023] Open
Abstract
The toxic effects of silica nanoparticles (SiNPs) are raising concerns due to its widely applications in biomedicine. However, current information about the epigenetic toxicity of SiNPs is insufficient. In this study, the epigenetic regulation of low-dose exposure to SiNPs was evaluated in human bronchial epithelial BEAS-2B cells over 30 passages. Cell viability was decreased in a dose- and passage-dependent manner. The apoptotic rate, the expression of caspase-9 and caspase-3, were significantly increased induced by SiNPs. HumanMethylation450 BeadChip analysis identified that the PI3K/Akt as the primary apoptosis-related pathway among the 25 significant altered processes. The differentially methylated sites of PI3K/Akt pathway involved 32 differential genes promoters, in which the CREB3L1 and Bcl-2 were significant hypermethylated. The methyltransferase inhibitor, 5-aza, further verified that the DNA hypermethylation status of CREB3L1 and Bcl-2 were associated with downregulation of their mRNA levels. In addition, mitochondrial-mediated apoptosis was triggered by SiNPs via the downregulation of PI3K/Akt/CREB/Bcl-2 signaling pathway. Our findings suggest that long-term low-dose exposure to SiNPs could lead to epigenetic alterations.
Collapse
|
86
|
Yu X, Hong F, Zhang YQ. Bio-effect of nanoparticles in the cardiovascular system. J Biomed Mater Res A 2016; 104:2881-97. [PMID: 27301683 DOI: 10.1002/jbm.a.35804] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/07/2016] [Indexed: 12/21/2022]
Abstract
Nanoparticles (NPs; < 100 nm) are increasingly being applied in various fields due to their unique physicochemical properties. The increase in human exposure to NPs has raised concerns regarding their health and safety profiles. The potential correlation between NP exposure and several cardiovascular (CV) events has been demonstrated. The aim of this review is to provide a comprehensive evaluation of the current knowledge regarding the bio-toxic impacts of titanium oxide, silver, silica, carbon black, carbon nanotube, and zinc oxide NPs exposure on the CV system in terms of in vivo and in vitro experiments, which is not fully understood presently. Moreover, the potential toxic mechanisms of NPs in the CV system that are still being questioned are elaborately discussed, and the underlying capacity of NPs used in medicine for CV events are summarized. It will be an important instrument to extrapolate relevant data for human CV risk evaluation and management. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2881-2897, 2016.
Collapse
Affiliation(s)
- Xiaohong Yu
- Department of Applied Biology, School of Basic Medical and Biological Sciences, Soochow University, RM702-2303, Renai Road No. 199, Dushuhu Higher Edu. Town, Suzhou, 215123, People's Republic of China
| | - Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, 223300, China. .,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China.
| | - Yu-Qing Zhang
- Department of Applied Biology, School of Basic Medical and Biological Sciences, Soochow University, RM702-2303, Renai Road No. 199, Dushuhu Higher Edu. Town, Suzhou, 215123, People's Republic of China
| |
Collapse
|
87
|
Lorscheidt S, Lamprecht A. Safety assessment of nanoparticles for drug delivery by means of classic in vitro assays and beyond. Expert Opin Drug Deliv 2016; 13:1545-1558. [DOI: 10.1080/17425247.2016.1198773] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Stefan Lorscheidt
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
- FDE (EA4267), University of Burgundy/Franche-Comté, Besançon, France
| |
Collapse
|
88
|
Giannakou C, Park MV, de Jong WH, van Loveren H, Vandebriel RJ, Geertsma RE. A comparison of immunotoxic effects of nanomedicinal products with regulatory immunotoxicity testing requirements. Int J Nanomedicine 2016; 11:2935-52. [PMID: 27382281 PMCID: PMC4922791 DOI: 10.2147/ijn.s102385] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nanomaterials (NMs) are attractive for biomedical and pharmaceutical applications because of their unique physicochemical and biological properties. A major application area of NMs is drug delivery. Many nanomedicinal products (NMPs) currently on the market or in clinical trials are most often based on liposomal products or polymer conjugates. NMPs can be designed to target specific tissues, eg, tumors. In virtually all cases, NMPs will eventually reach the immune system. It has been shown that most NMs end up in organs of the mononuclear phagocytic system, notably liver and spleen. Adverse immune effects, including allergy, hypersensitivity, and immunosuppression, have been reported after NMP administration. Interactions of NMPs with the immune system may therefore constitute important side effects. Currently, no regulatory documents are specifically dedicated to evaluate the immunotoxicity of NMs or NMPs. Their immunotoxicity assessment is performed based on existing guidelines for conventional substances or medicinal products. Due to the unique properties of NMPs when compared with conventional medicinal products, it is uncertain whether the currently prescribed set of tests provides sufficient information for an adequate evaluation of potential immunotoxicity of NMPs. The aim of this study was therefore, to compare the current regulatory immunotoxicity testing requirements with the accumulating knowledge on immunotoxic effects of NMPs in order to identify potential gaps in the safety assessment. This comparison showed that immunotoxic effects, such as complement activation-related pseudoallergy, myelosuppression, inflammasome activation, and hypersensitivity, are not readily detected by using current testing guidelines. Immunotoxicity of NMPs would be more accurately evaluated by an expanded testing strategy that is equipped to stratify applicable testing for the various types of NMPs.
Collapse
Affiliation(s)
- Christina Giannakou
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven; Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
| | - Margriet Vdz Park
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven
| | - Wim H de Jong
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven
| | - Henk van Loveren
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven; Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
| | - Rob J Vandebriel
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven
| | - Robert E Geertsma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven
| |
Collapse
|
89
|
Duan J, Hu H, Li Q, Jiang L, Zou Y, Wang Y, Sun Z. Combined toxicity of silica nanoparticles and methylmercury on cardiovascular system in zebrafish (Danio rerio) embryos. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 44:120-7. [PMID: 27163730 DOI: 10.1016/j.etap.2016.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 04/29/2016] [Accepted: 05/01/2016] [Indexed: 05/12/2023]
Abstract
This study was to investigate the combined toxicity of silica nanoparticles (SiNPs) and methylmercury (MeHg) on cardiovascular system in zebrafish (Danio rerio) embryos. Ultraviolet absorption analysis showed that the co-exposure system had high absorption and stability. The dosages used in this study were based on the NOAEL level. Zebrafish embryos exposed to the co-exposure of SiNPs and MeHg did not show any cardiovascular malformation or atrioventricular block, but had an inhibition effect on bradycardia. Using o-Dianisidine for erythrocyte staining, the cardiac output of zebrafish embryos was decreased gradually in SiNPs, MeHg, co-exposure groups, respectively. Co-exposure of SiNPs and MeHg enhanced the vascular endothelial damage in Tg(fli-1:EGFP) transgenic zebrafish line. Moreover, the co-exposure significantly activated the oxidative stress and inflammatory response in neutrophils-specific Tg(mpo:GFP) transgenic zebrafish line. This study suggested that the combined toxic effects of SiNPs and MeHg on cardiovascular system had more severe toxicity than the single exposure alone.
Collapse
Affiliation(s)
- Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Hejing Hu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Qiuling Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Lizhen Jiang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yang Zou
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yapei Wang
- Department of Chemistry, Renmin University of China, Beijing 100872, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
90
|
Duan J, Yu Y, Li Y, Wang Y, Sun Z. Inflammatory response and blood hypercoagulable state induced by low level co-exposure with silica nanoparticles and benzo[a]pyrene in zebrafish (Danio rerio) embryos. CHEMOSPHERE 2016; 151:152-62. [PMID: 26943738 DOI: 10.1016/j.chemosphere.2016.02.079] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 05/21/2023]
Abstract
Given the severe situation of world-wide particulate matter air pollution, it is urgent to explore the combined effects of particulate matter components on cardiovascular system. Using zebrafish model, this study was aimed to determine whether the low level co-exposure to silica nanoparticles (SiNPs) and benzo[a]pyrene (B[a]P) had a pronounced cardiovascular toxicity than the single exposure to either SiNPs or B[a]P alone. The FTIR and TGA analysis showed that the co-exposure system possessed of high absorption and thermal stability. Embryos exposed to SiNPs or B[a]P alone did not show cardiac toxicity phenotype at the NOAEL level. However, embryos co-exposed to SiNPs and B[a]P exhibited pericardial edema and bradycardia. While ROS generation remained unaffected, the co-exposure induced significant neutrophil-mediated inflammation and caused erythrocyte aggregation in caudal vein of embryos. Microarray analysis and STC analysis were performed to screen the cardiovascular-related differential expression genes and the expression trend of genes in each group. The co-exposure of SiNPs and B[a]P significantly enhanced the expression of proinflammatory and procoagulant genes. Moreover, the co-exposure markedly increased the phosphorylated AP-1/c-Jun and induced TF expression, but not NF-κB p65. This study for the first time demonstrated the inflammatory response and blood hypercoagulable state were triggered by the combination of SiNPs and B[a]P at low level exposure.
Collapse
Affiliation(s)
- Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China.
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China
| | - Yapei Wang
- Department of Chemistry, Renmin University of China, Beijing, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, PR China.
| |
Collapse
|
91
|
Nakahara R, Makino J, Kamiya T, Hara H, Adachi T. Caffeic acid phenethyl ester suppresses monocyte adhesion to the endothelium by inhibiting NF-κB/NOX2-derived ROS signaling. J Clin Biochem Nutr 2016; 58:174-9. [PMID: 27257341 PMCID: PMC4865596 DOI: 10.3164/jcbn.15-94] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/23/2015] [Indexed: 01/17/2023] Open
Abstract
Caffeic acid phenethyl ester (CAPE), one of the major polyphenols, exhibits anti-oxidative, anti-bacterial, and anti-cancer properties. Atherosclerosis is a chronic inflammatory disease, the progression of which is closely related to the accumulated adhesion of inflammatory monocytes/macrophages to the endothelium. We herein determined whether CAPE and its derivatives suppressed THP-1 cell adhesion to human umbilical vein endothelial cells (HUVEC). Of the four polyphenols tested, CAPE significantly suppressed the 12-O-tetradecanoylphorbol 13-acetate (TPA)-elicited expression of cluster for differentiation (CD) 11b, 14, and 36, and this was accompanied by the inhibition of THP-1 cell adhesion to HUVEC. CAPE also suppressed the activation of TPA-elicited nuclear factor-κB (NF-κB) and accumulation of NADPH oxidase 2 (NOX2)-derived reactive oxygen species (ROS), but did not affect extracellular signal-regulated kinase (ERK) phosphorylation. Taken together, these results demonstrated that CAPE suppressed THP-1 cell adhesion to HUVEC through, at least in part, the NF-κB, NOX2, and ROS-derived signaling axis.
Collapse
Affiliation(s)
- Risa Nakahara
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Junya Makino
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Tetsuro Kamiya
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Hirokazu Hara
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Tetsuo Adachi
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| |
Collapse
|
92
|
Hu H, Li Q, Jiang L, Zou Y, Duan J, Sun Z. Genome-wide transcriptional analysis of silica nanoparticle-induced toxicity in zebrafish embryos. Toxicol Res (Camb) 2016; 5:609-620. [PMID: 30090375 PMCID: PMC6062350 DOI: 10.1039/c5tx00383k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/14/2016] [Indexed: 12/28/2022] Open
Abstract
Although silica nanoparticles (SiNPs) have a promising application in biomedical fields, there is still a lack of comprehensive understanding of genome-wide transcriptional analysis. This study aims to clarify the toxic effect and molecular mechanisms of SiNPs in zebrafish embryos based on microarray analysis and bioinformatics analysis. Microarray data analysis demonstrated that SiNP-induced toxicity in zebrafish embryos affected expression of 2515 genes, including 1107 genes that were up-regulated and 1408 genes that were down-regulated. These differentially expressed genes were subjected to bioinformatics analysis for exploring the biological processes triggered by SiNPs in zebrafish embryos. Gene ontology analysis showed that SiNPs caused significant changes in gene expression patterns related to many important functions, including response to stimuli, immune response, cellular processes, and embryonic development. In addition, pathway analysis and Signal-net analysis indicated that the gap junction, vascular smooth muscle contraction, and metabolic pathways, apoptosis, the MAPK signaling pathway, the calcium signaling pathway and the JAK-STAT signaling pathway were the most prominent significant pathways in SiNP-induced toxicity in zebrafish embryos. In addition, the results from qRT-PCR and western blot analysis showed that the IL-6 dependent JAK1/STAT3 signaling pathway was activated by SiNPs in zebrafish embryos. In summary, our data will provide compelling clues for further exploration of SiNP-induced toxicity in zebrafish embryos.
Collapse
Affiliation(s)
- Hejing Hu
- Department of Toxicology and Sanitary Chemistry , School of Public Health , Capital Medical University , Beijing 100069 , P.R. China
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , P.R. China . ; ; ; Tel: +86 010 83911868, +86 010 83911507
| | - Qiuling Li
- Department of Toxicology and Sanitary Chemistry , School of Public Health , Capital Medical University , Beijing 100069 , P.R. China
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , P.R. China . ; ; ; Tel: +86 010 83911868, +86 010 83911507
| | - Lizhen Jiang
- Department of Toxicology and Sanitary Chemistry , School of Public Health , Capital Medical University , Beijing 100069 , P.R. China
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , P.R. China . ; ; ; Tel: +86 010 83911868, +86 010 83911507
| | - Yang Zou
- Department of Toxicology and Sanitary Chemistry , School of Public Health , Capital Medical University , Beijing 100069 , P.R. China
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , P.R. China . ; ; ; Tel: +86 010 83911868, +86 010 83911507
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry , School of Public Health , Capital Medical University , Beijing 100069 , P.R. China
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , P.R. China . ; ; ; Tel: +86 010 83911868, +86 010 83911507
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry , School of Public Health , Capital Medical University , Beijing 100069 , P.R. China
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , P.R. China . ; ; ; Tel: +86 010 83911868, +86 010 83911507
| |
Collapse
|
93
|
Autophagy upregulation promotes macrophages to escape mesoporous silica nanoparticle (MSN)-induced NF-κB-dependent inflammation. Inflamm Res 2016; 65:325-41. [PMID: 26860538 DOI: 10.1007/s00011-016-0919-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Our previous studies (Int J Nanomed 10:22, 2015) have indicated that a single large dose of mesoporous silica nanoparticles (MSNs) can induce severe and selective nephrotoxicity, which is closely related to inflammation mediated by the NF-κB pathway. However, the effect of MSNs on other organs and the interactions of nanomaterials with biological systems remain rudimentary. OBJECTIVE This study aimed to clarify the biological behaviour and influence of MSNs on macrophages. METHODS The mice received a single intraperitoneal injection of a suspension of 150, 300 of 600 mg/kg MSNs, and RAW 264.7 cells were treated with MSNs at various concentrations and times. Cell viability was determined by MTT assay and LDH release assay. The NF-κB pathway and the target proinflammatory cytokines IL-1β and TNF-α were determined by western blotting or ELISA. Autophagy is considered as an emerging mechanism of nanomaterials. So the autophagic ultrastructural analysis, the determination of Beclin-1 and LC3 expression, and the calculation of LC3II dots were employed to verify autophagy activation. In addition, RNA interference, autophagy agonist and inhibitor were used to explore the role of autophagy in inflammation. RESULTS The results indicated that MSNs are internalized into macrophages and induce cytotoxicity in a dose- and time-dependent manner. The NF-κB pathway, IL-1β and TNF-α were induced and released by MSNs. The levels of Beclin-1 and LC3II dots were obviously up-regulated by MSNs, which indicated that autophagy was induced in the MSN-treated cells. Moreover, the enhanced autophagy can attenuate the inflammation mediated by the NF-κB pathway, whereas the inhibition of autophagy can contribute to inflammation. CONCLUSIONS In summary, our results suggest that autophagy may be a possible protective factor in inflammation induced by MSNs in macrophages.
Collapse
|
94
|
Jiang Y, Yang N, Zhang H, Sun B, Hou C, Ji C, Zheng J, Liu Y, Zuo P. Enhanced in vivo antitumor efficacy of dual-functional peptide-modified docetaxel nanoparticles through tumor targeting and Hsp90 inhibition. J Control Release 2016; 221:26-36. [DOI: 10.1016/j.jconrel.2015.11.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/27/2015] [Indexed: 01/19/2023]
|
95
|
Wang Y, Kaur G, Chen Y, Santos A, Losic D, Evdokiou A. Bioinert Anodic Alumina Nanotubes for Targeting of Endoplasmic Reticulum Stress and Autophagic Signaling: A Combinatorial Nanotube-Based Drug Delivery System for Enhancing Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2015; 7:27140-27151. [PMID: 26556288 DOI: 10.1021/acsami.5b07557] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Although nanoparticle-based targeted delivery systems have gained promising achievements for cancer therapy, the development of sophisticated strategies with effective combinatorial therapies remains an enduring challenge. Herein, we report the fabrication of a novel nanomaterial, so-called anodic alumina nanotubes (AANTs) for proof-of-concept cancer therapy by targeting cell signaling networks. This strategy is to target autophagic and endoplasmic reticulum (ER) stress signaling by using thapsigargin (TG)-loaded AANTs cotreated with an autophagy inhibitor 3-methyladenine (3-MA). We first show that AANTs are nontoxic and can activate autophagy in different cell types including human fibroblast cells (HFF), human monocyte cells (THP-1), and human breast cancer cells (MDA-MB 231-TXSA). Treatment with 3-MA at a nontoxic dose reduced the level of autophagy induced by AANTs, and consequently sensitized breast cancer cells to AANTs-induced cellular stresses. To target autophagic and ER stress signaling networking, breast cancer cells were treated with 3-MA together with AANTs loaded with the prototype ER stress inducer TG. We demonstrated that 3-MA enhanced the cancer cell killing effect of AANTs loaded with TG. This effect was associated with enhanced ER stress signaling due to the combination effect of TG and 3-MA. These findings not only demonstrate the excellent biocompatibility of AANTs as novel biomaterials but also provide new opportunities for developing ER- and autophagy-targeted delivery systems for future clinical cancer therapy.
Collapse
Affiliation(s)
- Ye Wang
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Gagandeep Kaur
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Yuting Chen
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Abel Santos
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Dusan Losic
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Andreas Evdokiou
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| |
Collapse
|
96
|
Duan J, Yu Y, Li Y, Li Y, Liu H, Jing L, Yang M, Wang J, Li C, Sun Z. Low-dose exposure of silica nanoparticles induces cardiac dysfunction via neutrophil-mediated inflammation and cardiac contraction in zebrafish embryos. Nanotoxicology 2015; 10:575-85. [PMID: 26551753 DOI: 10.3109/17435390.2015.1102981] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The toxicity mechanism of nanoparticles on vertebrate cardiovascular system is still unclear, especially on the low-level exposure. This study was to explore the toxic effect and mechanisms of low-dose exposure of silica nanoparticles (SiNPs) on cardiac function in zebrafish embryos via the intravenous microinjection. The dosage of SiNPs was based on the no observed adverse effect level (NOAEL) of malformation assessment in zebrafish embryos. The mainly cardiac toxicity phenotypes induced by SiNPs were pericardial edema and bradycardia but had no effect on atrioventricular block. Using o-Dianisidine for erythrocyte staining, the cardiac output of zebrafish embryos was decreased in a dose-dependent manner. Microarray analysis and bioinformatics analysis were performed to screen the differential expression genes and possible pathway involved in cardiac function. SiNPs induced whole-embryo oxidative stress and neutrophil-mediated cardiac inflammation in Tg(mpo:GFP) zebrafish. Inflammatory cells were observed in atrium of SiNPs-treated zebrafish heart by histopathological examination. In addition, the expression of TNNT2 protein, a cardiac contraction marker in heart tissue had been down-regulated compared to control group using immunohistochemistry. Confirmed by qRT-PCR and western blot assays, results showed that SiNPs inhibited the calcium signaling pathway and cardiac muscle contraction via the down-regulated of related genes, such as ATPase-related genes (atp2a1l, atp1b2b, atp1a3b), calcium channel-related genes (cacna1ab, cacna1da) and the regulatory gene tnnc1a for cardiac troponin C. Moreover, the protein level of TNNT2 was decreased in a dose-dependent manner. For the first time, our results demonstrated that SiNPs induced cardiac dysfunction via the neutrophil-mediated cardiac inflammation and cardiac contraction in zebrafish embryos.
Collapse
Affiliation(s)
- Junchao Duan
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| | - Yang Yu
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| | - Yang Li
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| | - Yanbo Li
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| | - Hongcui Liu
- c Hunter Biotechnology Inc. , Hangzhou, Zhejiang Province , P.R. China
| | - Li Jing
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| | - Man Yang
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| | - Ji Wang
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| | - Chunqi Li
- c Hunter Biotechnology Inc. , Hangzhou, Zhejiang Province , P.R. China
| | - Zhiwei Sun
- a School of Public Health, Capital Medical University , Beijing , P.R. China .,b Beijing Key Laboratory of Environmental Toxicology, Capital Medical University , Beijing , P.R. China , and
| |
Collapse
|
97
|
Gao J, Zhang X, Yu M, Ren G, Yang Z. Cognitive deficits induced by multi-walled carbon nanotubes via the autophagic pathway. Toxicology 2015; 337:21-9. [DOI: 10.1016/j.tox.2015.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/26/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
|
98
|
Huang D, Zhou H, Gao J. Nanoparticles modulate autophagic effect in a dispersity-dependent manner. Sci Rep 2015; 5:14361. [PMID: 26394839 PMCID: PMC4585824 DOI: 10.1038/srep14361] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/26/2015] [Indexed: 12/19/2022] Open
Abstract
Autophagy plays a key role in human health and disease, especially in cancer and neurodegeneration. Many autophagy regulators are developed for therapy. Diverse nanomaterials have been reported to induce autophagy. However, the underlying mechanisms and universal rules remain unclear. Here, for the first time, we show a reliable and general mechanism by which nanoparticles induce autophagy and then successfully modulate autophagy via tuning their dispersity. Various well-designed univariate experiments demonstrate that nanomaterials induce autophagy in a dispersity-dependent manner. Aggregated nanoparticles induce significant autophagic effect in comparison with well-dispersed nanoparticles. As the highly stable nanoparticles may block autophagic degradation in autolysosomes, endocytosis and intracellular accumulation of nanoparticles can be responsible for this interesting phenomenon. Our results suggest dispersity-dependent autophagic effect as a common cellular response to nanoparticles, reveal the relationship between properties of nanoparticles and autophagy, and offer a new alternative way to modulate autophagy.
Collapse
Affiliation(s)
- Dengtong Huang
- The Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hualu Zhou
- The Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jinhao Gao
- The Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
99
|
Grützner V, Unger RE, Baier G, Choritz L, Freese C, Böse T, Landfester K, Kirkpatrick CJ. Enzyme-responsive nanocomposites for wound infection prophylaxis in burn management: in vitro evaluation of their compatibility with healing processes. Int J Nanomedicine 2015; 10:4111-24. [PMID: 26150717 PMCID: PMC4484651 DOI: 10.2147/ijn.s81263] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Responsive, theranostic nanosystems, capable of both signaling and treating wound infections, is a sophisticated approach to reduce the most common and potentially traumatizing side effects of burn wound treatment: slowed wound healing due to prophylactic anti-infective drug exposure as well as frequent painful dressing changes. Antimicrobials as well as dye molecules have been incorporated into biodegradable nanosystems that release their content only in the presence of pathogens. Following nanocarrier degradation by bacterial enzymes, any infection will thus emit a visible signal and be effectively treated at its source. In this study, we investigated the effect of fluorescent-labeled hyaluronan nanocapsules containing polyhexanide biguanide and poly-L-lactic acid nanoparticles loaded with octenidine on primary human dermal microvascular endothelial cells, which play a major role in cutaneous wound healing. Microscopic and flow cytometric analysis indicated a time-dependent uptake of both the nanocapsules and the nanoparticles. However, enzyme immunoassays showed no significant influence on the expression of pro-inflammatory cell adhesion molecules and cytokines by the endothelial cells. Under angiogenic-stimulating conditions, the potential to form capillary-like structures in co-culture with dermal fibroblasts was not inhibited. Furthermore, cytotoxicity studies (the MTS and crystal violet assay) after short- and long-term exposure to the materials demonstrated that both systems exhibited less toxicity than solutions of the antiseptic agents alone in comparable concentrations. The results indicate that responsive antimicrobial nanocomposites could be used as an advanced drug delivery system and a promising addition to current best practice wound infection prophylaxis with few side effects.
Collapse
Affiliation(s)
- Verena Grützner
- REPAIR-Lab, Institute of Pathology, University Medical Center, Mainz, Germany
| | - Ronald E Unger
- REPAIR-Lab, Institute of Pathology, University Medical Center, Mainz, Germany
| | - Grit Baier
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Lars Choritz
- Department of Ophthalmology, University Clinic, Magdeburg, Germany
| | - Christian Freese
- REPAIR-Lab, Institute of Pathology, University Medical Center, Mainz, Germany
| | - Thomas Böse
- REPAIR-Lab, Institute of Pathology, University Medical Center, Mainz, Germany
| | | | - C James Kirkpatrick
- REPAIR-Lab, Institute of Pathology, University Medical Center, Mainz, Germany
| |
Collapse
|