1
|
Peters K, Staehlke S, Rebl H, Jonitz-Heincke A, Hahn O. Impact of Metal Ions on Cellular Functions: A Focus on Mesenchymal Stem/Stromal Cell Differentiation. Int J Mol Sci 2024; 25:10127. [PMID: 39337612 PMCID: PMC11432215 DOI: 10.3390/ijms251810127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Metals play a crucial role in the human body, especially as ions in metalloproteins. Essential metals, such as calcium, iron, and zinc are crucial for various physiological functions, but their interactions within biological networks are complex and not fully understood. Mesenchymal stem/stromal cells (MSCs) are essential for tissue regeneration due to their ability to differentiate into various cell types. This review article addresses the effects of physiological and unphysiological, but not directly toxic, metal ion concentrations, particularly concerning MSCs. Overloading or unbalancing of metal ion concentrations can significantly impair the function and differentiation capacity of MSCs. In addition, excessive or unbalanced metal ion concentrations can lead to oxidative stress, which can affect viability or inflammation. Data on the effects of metal ions on MSC differentiation are limited and often contradictory. Future research should, therefore, aim to clarify the mechanisms by which metal ions affect MSC differentiation, focusing on aspects such as metal ion interactions, ion concentrations, exposure duration, and other environmental conditions. Understanding these interactions could ultimately improve the design of biomaterials and implants to promote MSC-mediated tissue regeneration. It could also lead to the development of innovative therapeutic strategies in regenerative medicine.
Collapse
Affiliation(s)
- Kirsten Peters
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Susanne Staehlke
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Henrike Rebl
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Anika Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Strasse 142, 18057 Rostock, Germany;
| | - Olga Hahn
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| |
Collapse
|
2
|
Liu G, Lv J, Wang Y, Sun K, Gao H, Li Y, Yao Q, Ma L, Kochshugulova G, Jiang Z. ZnO NPs induce miR-342-5p mediated ferroptosis of spermatocytes through the NF-κB pathway in mice. J Nanobiotechnology 2024; 22:390. [PMID: 38961442 PMCID: PMC11223436 DOI: 10.1186/s12951-024-02672-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Zinc oxide nanoparticle (ZnO NP) is one of the metal nanomaterials with extensive use in many fields such as feed additive and textile, which is an emerging threat to human health due to widely distributed in the environment. Thus, there is an urgent need to understand the toxic effects associated with ZnO NPs. Although previous studies have found accumulation of ZnO NPs in testis, the molecular mechanism of ZnO NPs dominated a decline in male fertility have not been elucidated. RESULTS We reported that ZnO NPs exposure caused testicular dysfunction and identified spermatocytes as the primary damaged site induced by ZnO NPs. ZnO NPs led to the dysfunction of spermatocytes, including impaired cell proliferation and mitochondrial damage. In addition, we found that ZnO NPs induced ferroptosis of spermatocytes through the increase of intracellular chelatable iron content and lipid peroxidation level. Moreover, the transcriptome analysis of testis indicated that ZnO NPs weakened the expression of miR-342-5p, which can target Erc1 to block the NF-κB pathway. Eventually, ferroptosis of spermatocytes was ameliorated by suppressing the expression of Erc1. CONCLUSIONS The present study reveals a novel mechanism in that miR-342-5p targeted Erc1 to activate NF-κB signaling pathway is required for ZnO NPs-induced ferroptosis, and provide potential targets for further research on the prevention and treatment of male reproductive disorders related to ZnO NPs.
Collapse
Affiliation(s)
- Guangyu Liu
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, China
| | - Jing Lv
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, China
| | - Yifan Wang
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, China
| | - Kaikai Sun
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, China
| | - Huimin Gao
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, China
| | - Yuanyou Li
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, China
| | - Qichun Yao
- Animal Husbandry and Veterinary Station of Zhenba County, Hanzhong, 723600, Shaanxi, China
| | - Lizhu Ma
- College of Animal Science and Technology, China Agricultural University, Beijing, 100080, China
| | - Gulzat Kochshugulova
- Department of Food Security, Agrotechnological Faculty, Kozybayev University, 86, Pushkin Street, Petropavlovsk, 150000, Kazakhstan
| | - Zhongliang Jiang
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
3
|
Paslı D, Gürbay A. Assessment of Protective Effects of DTPA, NAC, and Taurine on Possible Cytotoxicity Induced by Individual and Combined Zinc Oxide and Copper Oxide Nanoparticles in SH-SY5Y Cells. Biol Trace Elem Res 2024:10.1007/s12011-024-04161-0. [PMID: 38683268 DOI: 10.1007/s12011-024-04161-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024]
Abstract
The present study investigated the cytotoxic effects of ZnO, CuO, and mixed combinations of them on SH-SY5Y cells. For this purpose, the cells were exposed to various concentrations of these NPs alone for 24-96 h and as a mixture for 24 h. Variations in cell viability were noted. MTT results showed that ZnO and/or CuO NPs decreased cell survival by about 59% at 200 (ZnO, at 24 h) and 800 µg/ml (ZnO and/or CuO, at 72 and 96 h). When the NR assay was used, slight decreases were noted with ZnO NPs at 72 and 96 h. With CuO NPs alone and NPs in a mixture, only the highest concentrations caused 40 and 70% decreases in cell survival, respectively. Especially with NR assays, DTPA, NAC, or taurine provided marked protection. ROS levels were increased with the highest concentration of CuO NPs and with all concentrations of the mixture. The highest concentration of ZnO NPs and the lowest concentration of CuO NPs caused slight decreases in mitochondrial membrane potential levels. Additionally, increases were noted in caspase 3/7 levels with ZnO and CuO NPs alone or with a mixture of them. Intracellular calcium levels were decreased in this system. These findings demonstrated that ZnO and CuO NPs, either separately or in combination, had a modest cytotoxic effect on SH-SY5Y cells. Protection obtained with DTPA, NAC, or taurine against the cytotoxicity of these NPs and the ROS-inducing effect of CuO NPs and the NPs' mixture suggests that oxidative stress might be involved in the cytotoxicity mechanisms of these NPs.
Collapse
Affiliation(s)
- Duygu Paslı
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey
| | - Aylin Gürbay
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
4
|
Esculetin and Fucoidan Attenuate Autophagy and Apoptosis Induced by Zinc Oxide Nanoparticles through Modulating Reactive Astrocyte and Proinflammatory Cytokines in the Rat Brain. TOXICS 2022; 10:toxics10040194. [PMID: 35448455 PMCID: PMC9025201 DOI: 10.3390/toxics10040194] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 12/10/2022]
Abstract
We examined the protective effects of esculetin and fucoidan against the neurotoxicity of ZnO NPs in rats. Ninety rats were divided into nine groups and pre-treated with esculetin or fucoidan 1 h before ZnO NP administration on a daily basis for 2 weeks. Serum and brain homogenates were examined by enzyme-linked immunosorbent assay (ELISA), and neurons, microglia, and astrocytes in the hippocampal region were examined with immunohistochemical analysis. The serum levels of interleukin-1-beta (IL-1β), 3-nitrotyrosine (3-NT), superoxide dismutase (SOD), and 8-hydroxy-2′-deoxyguanosine (8-OHdG) were altered in the ZnO NP treatment groups. Brain IL-1β and TNF-α levels were elevated after ZnO NP administration, and these effects were inhibited by esculetin and fucoidan. SOD, 8-OHdG, and acetylcholinesterase (AChE) levels in the brain were decreased after ZnO NP administration. The brain levels of beclin-1 and caspase-3 were elevated after ZnO NP treatment, and these effects were significantly ameliorated by esculetin and fucoidan. The number of reactive astrocytes measured by counting glial fibrillary acidic protein (GFAP)-positive cells, but not microglia, increased following ZnO NP treatment, and esculetin and fucoidan ameliorated the changes. Esculetin and fucoidan may be beneficial for preventing ZnO NP-mediated autophagy and apoptosis by the modulation of reactive astrocyte and proinflammatory cytokines in the rat brain.
Collapse
|
5
|
Hu B, Cheng Z, Liang S. Advantages and prospects of stem cells in nanotoxicology. CHEMOSPHERE 2022; 291:132861. [PMID: 34774913 DOI: 10.1016/j.chemosphere.2021.132861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
Nanomaterials have been widely used in many fields, especially in biomedical and stem cell therapy. However, the potential risks associated with nanomaterials applications are also gradually increasing. Therefore, effective and robust toxicology models are critical to evaluate the developmental toxicity of nanomaterials. The development of stem cell research provides a new idea of developmental toxicology. Recently, many researchers actively investigated the effects of nanomaterials with different sizes and surface modifications on various stem cells (such as embryonic stem cells (ESCs), adult stem cells, etc.) to study the toxic effects and toxic mechanisms. In this review, we summarized the effects of nanomaterials on the proliferation and differentiation of ESCs, mesenchymal stem cells and neural stem cells. Moreover, we discussed the advantages of stem cells in nanotoxicology compared with other cell lines. Finally, combined with the latest research methods and new molecular mechanisms, we analyzed the application of stem cells in nanotoxicology.
Collapse
Affiliation(s)
- Bowen Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, 830017, China.
| | - Zhanwen Cheng
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Shengxian Liang
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding, 071000, China
| |
Collapse
|
6
|
Li H, Li M, Ran X, Cui J, Wei F, Yi G, Chen W, Luo X, Chen Z. The Role of Zinc in Bone Mesenchymal Stem Cell Differentiation. Cell Reprogram 2022; 24:80-94. [PMID: 35172118 DOI: 10.1089/cell.2021.0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Zinc is an essential trace element for bone growth and bone homeostasis in the human body. Bone mesenchymal stem cells (BMSCs) are multipotent progenitors existing in the bone marrow stroma with the capability of differentiating along multiple lineage pathways. Zinc plays a paramount role in BMSCs, which can be spurred differentiating into osteoblasts, chondrocytes, or adipocytes, and modulates the formation and activity of osteoclasts. The expression of related genes also changed during the differentiation of various cell phenotypes. Based on the important role of zinc in BMSC differentiation, using zinc as a therapeutic approach for bone remodeling will be a promising method. This review explores the role of zinc ion in the differentiation of BMSCs into various cell phenotypes and outlines the existing research on their molecular mechanism.
Collapse
Affiliation(s)
- Huiyun Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Muzhe Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xun Ran
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Juncheng Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Fu Wei
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Guoliang Yi
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Wei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xuling Luo
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhiwei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
7
|
Wang YL, Zheng CM, Lee YH, Cheng YY, Lin YF, Chiu HW. Micro- and Nanosized Substances Cause Different Autophagy-Related Responses. Int J Mol Sci 2021; 22:4787. [PMID: 33946416 PMCID: PMC8124422 DOI: 10.3390/ijms22094787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 02/07/2023] Open
Abstract
With rapid industrialization, humans produce an increasing number of products. The composition of these products is usually decomposed. However, some substances are not easily broken down and gradually become environmental pollutants. In addition, these substances may cause bioaccumulation, since the substances can be fragmented into micro- and nanoparticles. These particles or their interactions with other toxic matter circulate in humans via the food chain or air. Whether these micro- and nanoparticles interfere with extracellular vesicles (EVs) due to their similar sizes is unclear. Micro- and nanoparticles (MSs and NSs) induce several cell responses and are engulfed by cells depending on their size, for example, particulate matter with a diameter ≤2.5 μm (PM2.5). Autophagy is a mechanism by which pathogens are destroyed in cells. Some artificial materials are not easily decomposed in organisms. How do these cells or tissues respond? In addition, autophagy operates through two pathways (increasing cell death or cell survival) in tumorigenesis. Many MSs and NSs have been found that induce autophagy in various cells and tissues. As a result, this review focuses on how these particles interfere with cells and tissues. Here, we review MSs, NSs, and PM2.5, which result in different autophagy-related responses in various tissues or cells.
Collapse
Affiliation(s)
- Yung-Li Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-L.W.); (Y.-F.L.)
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung 406040, Taiwan;
| | - Ya-Yun Cheng
- Department of Environmental Health, Harvard University T.H. Chan School of Public Health, Boston, MA 02115, USA;
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-L.W.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
| | - Hui-Wen Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-L.W.); (Y.-F.L.)
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
8
|
Kunrath MF, Campos MM. Metallic-nanoparticle release systems for biomedical implant surfaces: effectiveness and safety. Nanotoxicology 2021; 15:721-739. [PMID: 33896331 DOI: 10.1080/17435390.2021.1915401] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The current focus of bioengineering for implant devices involves the development of functionalized surfaces, bioactive coatings, and metallic nanoparticles (mNPs) with a controlled release, together with strategies for the application of drugs in situ, aiming at reducing infection rates, with an improvement of clinical outcomes. Controversially, negative aspects, such as cytotoxicity, mNP incorporation, bioaccumulation, acquired autoimmunity, and systemic toxicity have gained attention at the same status of importance, concerning the release of mNPs from these surface systems. The balance between the promising prospects of system releasing mNPs and the undesirable long-term adverse reactions require further investigation. The scarcity of knowledge and the methods of analysis of nanoscale-based systems to control the sequence of migration, interaction, and nanoparticle incorporation with human tissues raise hesitation about their efficacy and safety. Looking ahead, this innovative approach requires additional scientific investigation for permitting an evolution of implants without counterpoints, while updating implant surface technologies to a new level of development. This critical review has explored the promising properties of metals at the nano-scale to promote broad-spectrum bacterial control, allowing for a decrease in using systemic antibiotics. Attempts have also been made to discuss the existing limitations and the future challenges regarding these technologies, besides the negative findings that are explored in the literature.
Collapse
Affiliation(s)
- Marcel F Kunrath
- Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria M Campos
- Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
9
|
Sanità G, Carrese B, Lamberti A. Nanoparticle Surface Functionalization: How to Improve Biocompatibility and Cellular Internalization. Front Mol Biosci 2020; 7:587012. [PMID: 33324678 PMCID: PMC7726445 DOI: 10.3389/fmolb.2020.587012] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
The use of nanoparticles (NP) in diagnosis and treatment of many human diseases, including cancer, is of increasing interest. However, cytotoxic effects of NPs on cells and the uptake efficiency significantly limit their use in clinical practice. The physico-chemical properties of NPs including surface composition, superficial charge, size and shape are considered the key factors that affect the biocompatibility and uptake efficiency of these nanoplatforms. Thanks to the possibility of modifying physico-chemical properties of NPs, it is possible to improve their biocompatibility and uptake efficiency through the functionalization of the NP surface. In this review, we summarize some of the most recent studies in which NP surface modification enhances biocompatibility and uptake. Furthermore, the most used techniques used to assess biocompatibility and uptake are also reported.
Collapse
Affiliation(s)
- Gennaro Sanità
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Annalisa Lamberti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
10
|
Jo HG, Park C, Lee H, Kim GY, Keum YS, Hyun JW, Kwon TK, Choi YH, Hong SH. Inhibition of oxidative stress induced-cytotoxicity by coptisine in V79-4 Chinese hamster lung fibroblasts through the induction of Nrf-2 mediated HO-1 expression. Genes Genomics 2020; 43:17-31. [PMID: 33237503 DOI: 10.1007/s13258-020-01018-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/05/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Coptisine is a natural alkaloid compound and is known to have multiple beneficial effects including antioxidant activity. However, whether it can protect lung fibroblasts from oxidative damage has not been studied yet. OBJECTIVES To investigate the potential inhibitory effect of coptisine against oxidative stress in V79-4 lung fibroblast cells. METHODS V79-4 cells were treated with H2O2 (1 mM) in the presence or absence of coptisine (50 µg/ml), N-acetyl cysteine (NAC, 10 mM) or zinc protoporphyrin IX (ZnPP, 10 µM) for the indicated times. The alleviating effects of coptisine on cytotoxicity, cell cycle arrest, apoptosis, reactive oxygen species (ROS) production, DNA damage, mitochondrial dynamics, and inhibition of ATP production against H2O2 were investigated. Western blot analysis was used to analyze the expression levels of specific proteins. RESULTS Coptisine inhibited H2O2-induced cytotoxicity and DNA damage by blocking abnormal ROS generation. H2O2 treatment caused cell cycle arrest at the G2/M phase accompanied by increased expression of cyclin-dependent kinase (Cdk) inhibitor p21WAF1/CIP1 and decreased expression of cyclin B1 and cyclin A. However, these effects were attenuated in the presence of coptisine or NAC. Coptisine also prevented apoptosis by decreasing the rate of Bax/Bcl-2 expression in H2O2-stimulated cells and suppressing the loss of mitochondrial membrane potential and the cytosolic release of cytochrome c. In addition, the activation of nuclear factor-erythroid-2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) was markedly promoted by coptisine in the presence of H2O2. However, zinc protoporphyrin IX, a potent inhibitor of HO-1, attenuated the ROS scavenging and anti-apoptotic effects of coptisine. CONCLUSIONS Based on current data, we suggest that coptisine can be used as a potential treatment for oxidative stress-related lung disease.
Collapse
Affiliation(s)
- Hyeon-Gyun Jo
- Cheong-Choon Korean Medical Clinic, 47388, Busan, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea
| | - Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-eui University, 47340, Busan, Republic of Korea
| | - Hyesook Lee
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea.,Anti-Aging Research Center, Dong-eui University, 47340, Busan, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, 63243, Jeju, Republic of Korea
| | - Young-Sam Keum
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, 10326, Goyang, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, 63243, Jeju, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, 42601, Daegu, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea. .,Anti-Aging Research Center, Dong-eui University, 47340, Busan, Republic of Korea.
| | - Su Hyun Hong
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea. .,Anti-Aging Research Center, Dong-eui University, 47340, Busan, Republic of Korea.
| |
Collapse
|
11
|
Zhao G, Ge Y, Zhang C, Zhang L, Xu J, Qi L, Li W. Progress of Mesenchymal Stem Cell-Derived Exosomes in Tissue Repair. Curr Pharm Des 2020; 26:2022-2037. [PMID: 32310043 DOI: 10.2174/1381612826666200420144805] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/25/2020] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) are a kind of adult stem cells with self-replication and multidirectional differentiation, which can differentiate into tissue-specific cells under physiological conditions, maintaining tissue self-renewal and physiological functions. They play a role in the pathological condition by lateral differentiation into tissue-specific cells, replacing damaged tissue cells by playing the role of a regenerative medicine , or repairing damaged tissues through angiogenesis, thereby, regulating immune responses, inflammatory responses, and inhibiting apoptosis. It has become an important seed cell for tissue repair and organ reconstruction, and cell therapy based on MSCs has been widely used clinically. The study found that the probability of stem cells migrating to the damaged area after transplantation or differentiating into damaged cells is very low, so the researchers believe the leading role of stem cell transplantation for tissue repair is paracrine secretion, secreting growth factors, cytokines or other components. Exosomes are biologically active small vesicles secreted by MSCs. Recent studies have shown that they can transfer functional proteins, RNA, microRNAs, and lncRNAs between cells, and greatly reduce the immune response. Under the premise of promoting proliferation and inhibition of apoptosis, they play a repair role in tissue damage, which is caused by a variety of diseases. In this paper, the biological characteristics of exosomes (MSCs-exosomes) derived from mesenchymal stem cells, intercellular transport mechanisms, and their research progress in the field of stem cell therapy are reviewed.
Collapse
Affiliation(s)
- Guifang Zhao
- School of Basic Medical Sciences, Jilin Medical University, Jilin 132013, China.,Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan 511518, Guangzhou Province, China
| | - Yiwen Ge
- School of Basic Medical Sciences, Jilin Medical University, Jilin 132013, China
| | - Chenyingnan Zhang
- School of Basic Medical Sciences, Jilin Medical University, Jilin 132013, China
| | - Leyi Zhang
- School of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Junjie Xu
- School of Basic Medical Sciences, Jilin Medical University, Jilin 132013, China
| | - Ling Qi
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan 511518, Guangzhou Province, China.,School of Basic Medical Sciences, Department of Pathophysiology, Jilin Medical University, Jilin 132013, China
| | - Wenliang Li
- School of Pharmacy, Jilin Medical University, Jilin 132013, China
| |
Collapse
|
12
|
Yang D, Zhang M, Gan Y, Yang S, Wang J, Yu M, Wei J, Chen J. Involvement of oxidative stress in ZnO NPs-induced apoptosis and autophagy of mouse GC-1 spg cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 202:110960. [PMID: 32800232 DOI: 10.1016/j.ecoenv.2020.110960] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 05/15/2023]
Abstract
Zinc oxide nanoparticles (ZnO NPs) have been extensively used in various industries and reported to inhibit spermatogenesis, however, ZnO NPs-induced spermatogenesis failure is yet to be fully elucidated. Herein, mouse-derived spermatogonia cell line GC-1 spg cells were treated with ZnO NPs for 24 h in the presence or absence of radical scavenger N-acetyl-L-cysteine (NAC) or autophagy inhibitor 3-methyladenine (3-MA), then cell viability was observed by MTT assay; apoptosis was observed by western blotting analysis and AnnexinV-FITC/PI assay, respectively; autophagy was detected by western blotting analysis and transmission electron microscopy, respectively; and the contents of MDA and GSH and the activities of SOD and GSH-PX were measured by oxidative stress kits. The present study showed that ZnO NPs exposure inhibited viability and induced apoptosis of mouse GC-1 spg cells. Intriguingly, ZnO NPs markedly increased the protein content of LC3-II, the ratio of LC3-II/LC3-I, and the protein levels of ATG 5 and Beclin 1 in the cells. Furthermore, transmission electron microscopy (TEM) showed that autophagic vesicles in the cytoplasm increased significantly in the ZnO NPs-treated cells, indicating that ZnO NPs could induce autophagy of the cells. Oxidative stress could be induced by ZnO NPs; moreover, inhibition of oxidative stress could alleviate the induction of apoptosis and autophagy by ZnO NPs. Inhibition of autophagy by 3-MA could rescue the inhibition of cell viability and induction of apoptosis by ZnO NPs, which indicated that autophagy might have cytotoxic effect on ZnO NPs-induced apoptosis. In summary, oxidative stress was involved in ZnO NPs-induced apoptosis and autophagy of mouse GC-1 spg cells, and autophagy might play a cytotoxic role in ZnO NPs-induced apoptosis.
Collapse
Affiliation(s)
- Dan Yang
- Department of Physiology, Medical College of Nanchang University, Nanchang, 330006, PR China
| | - Meijuan Zhang
- Department of Physiology, Medical College of Nanchang University, Nanchang, 330006, PR China
| | - Yu Gan
- Department of Physiology, Medical College of Nanchang University, Nanchang, 330006, PR China
| | - Si Yang
- Department of Physiology, Medical College of Nanchang University, Nanchang, 330006, PR China
| | - Jinglei Wang
- Department of Physiology, Medical College of Nanchang University, Nanchang, 330006, PR China; Jiangxi Provincial Key Laboratory of Reproductive Physiology and Pathology, Nanchang, 330006, PR China
| | - Mei Yu
- Library, Medical College of Nanchang University, Nanchang, 330006, PR China
| | - Jie Wei
- Department of Physiology, Medical College of Nanchang University, Nanchang, 330006, PR China; Jiangxi Provincial Key Laboratory of Reproductive Physiology and Pathology, Nanchang, 330006, PR China
| | - Jiaxiang Chen
- Department of Physiology, Medical College of Nanchang University, Nanchang, 330006, PR China; Jiangxi Provincial Key Laboratory of Reproductive Physiology and Pathology, Nanchang, 330006, PR China.
| |
Collapse
|
13
|
Li Y, Yang Y, Qing Y, Li R, Tang X, Guo D, Qin Y. Enhancing ZnO-NP Antibacterial and Osteogenesis Properties in Orthopedic Applications: A Review. Int J Nanomedicine 2020; 15:6247-6262. [PMID: 32903812 PMCID: PMC7445529 DOI: 10.2147/ijn.s262876] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022] Open
Abstract
Prosthesis-associated infections and aseptic loosening are major causes of implant failure. There is an urgent need to improve the antibacterial ability and osseointegration of orthopedic implants. Zinc oxide nanoparticles (ZnO-NPs) are a common type of zinc-containing metal oxide nanoparticles that have been widely studied in many fields, such as food packaging, pollution treatment, and biomedicine. The ZnO-NPs have low toxicity and good biological functions, as well as antibacterial, anticancer, and osteogenic capabilities. Furthermore, ZnO-NPs can be easily obtained through various methods. Among them, green preparation methods can improve the bioactivity of ZnO-NPs and strengthen their potential application in the biological field. This review discusses the antibacterial abilities of ZnO-NPs, including mechanisms and influencing factors. The toxicity and shortcomings of anticancer applications are summarized. Furthermore, osteogenic mechanisms and synergy with other materials are introduced. Green preparation methods are also briefly reviewed.
Collapse
Affiliation(s)
- Yuehong Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Yue Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Yun’an Qing
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Ruiyan Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Xiongfeng Tang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Deming Guo
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Yanguo Qin
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
14
|
Park C, Lee H, Han MH, Jeong JW, Kim SO, Jeong SJ, Lee BJ, Kim GY, Park EK, Jeon YJ, Choi YH. Cytoprotective effects of fermented oyster extracts against oxidative stress-induced DNA damage and apoptosis through activation of the Nrf2/HO-1 signaling pathway in MC3T3-E1 osteoblasts. EXCLI JOURNAL 2020; 19:1102-1119. [PMID: 33013267 PMCID: PMC7527492 DOI: 10.17179/excli2020-2376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022]
Abstract
Osteoblast damage by oxidative stress has been recognized as a cause of bone-related disease, including osteoporosis. Recently, we reported that fermented Pacific oyster (Crassostrea gigas) extracts (FO) inhibited osteoclastogenesis and osteoporosis, while promoting osteogenesis. However, since the beneficial potential of FO on osteoblasts is not well known, in the present study, we investigated the cytoprotective effect of FO against oxidative stress in MC3T3-E1 osteoblasts. Our results demonstrated that FO inhibited hydrogen peroxide (H2O2)-induced DNA damage and cytotoxicity through the rescue of mitochondrial function by blocking abnormal ROS accumulation. FO also prevented apoptosis by suppressing loss of mitochondrial membrane potential and cytosolic release of cytochrome c, decreasing the rate of Bax/Bcl-2 expression and reducing the activity of caspase-9 and caspase-3 in H2O2-stimulated MC3T3-E1 osteoblasts, suggesting that FO protected MC3T3-E1 osteoblasts from the induction of caspase dependent- and mitochondria-mediated apoptosis by oxidative stress. In addition, FO markedly promoted the activation of nuclear factor-erythroid-2-related factor 2 (Nrf2), which was associated with the enhanced expression of heme oxygenase-1 (HO-1). However, inhibiting the expression of HO-1 by artificially blocking the expression of Nrf2 using siRNA significantly eliminated the protective effect of FO, indicating that FO activates the Nrf2/HO-1 signaling pathway in MC3T3-E1 osteoblasts to protect against oxidative stress. Based on the present data, FO is thought to be useful as a potential therapeutic agent for the inhibition of oxidative stress in osteoblasts.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong?eui University, Busan, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| | - Min Ho Han
- National Marine Biodiversity Institute of Korea, Seocheon, Republic of Korea
| | - Jin-Woo Jeong
- Freshwater Bioresources Utilization Bureau, Nakdonggang National Institute of Biological Resources, Sangju, Republic of Korea
| | - Sung Ok Kim
- Department of Food Science and Biotechnology, College of Engineering, Kyungsung University, Busan, Republic of Korea
| | - Soon-Jeong Jeong
- Department of Dental Hygiene, College of Health Science, Youngsan University, Yangsan, Republic of Korea
| | - Bae-Jin Lee
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| |
Collapse
|
15
|
Qin X, Tang Q, Jiang X, Zhang J, Wang B, Liu X, Zhang Y, Zou Z, Chen C. Zinc Oxide Nanoparticles Induce Ferroptotic Neuronal Cell Death in vitro and in vivo. Int J Nanomedicine 2020; 15:5299-5315. [PMID: 32884256 PMCID: PMC7436556 DOI: 10.2147/ijn.s250367] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/10/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose Zinc oxide nanoparticles (ZnONPs) are one of the most important nanomaterials that are widely used in the food, cosmetic and medical industries. Humans are often exposed to ZnONPs via inhalation, and they may reach the brain where neurotoxic effects could occur via systemic distribution. However, the mechanisms underlying how ZnONPs produce neurotoxic effects in the brain remain unclear. In this study, we aimed to investigate the novel mechanism involved in ZnONPs-induced neurotoxicity. Methods and Results We demonstrated for the first time that pulmonary exposure to ZnONPs by intratracheal instillation could trigger ferroptosis, a new form of cell death, in the neuronal cells of mouse cerebral cortex. A similar phenomenon was also observed in cultured neuron-like PC-12 cell line. By using a specific inhibitor of ferroptosis ferrostatin-1 (Fer-1), our results showed that inhibition of ferroptosis by Fer-1 could significantly alleviate the ZnONPs-induced neuronal cell death both in vivo and in vitro. Mechanistic investigation revealed that ZnONPs selectively activated the JNK pathway and thus resulted in the ferroptotic phenotypes, JNK inhibitor SP600125 could reverse lipid peroxidation upregulation and ferroptotic cell death induced by ZnONPs in PC-12 cells. Conclusion Taken together, this study not only demonstrates that pulmonary exposure of ZnONPs can induce JNK-involved ferroptotic cell death in mouse cortex and PC-12 cells, but also provides a clue that inhibition of ferroptosis by specific agents or drugs may serve as a feasible approach for reducing the untreatable neurotoxicity induced by ZnONPs.
Collapse
Affiliation(s)
- Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Qianghu Tang
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuemei Liu
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yandan Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China.,Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China.,Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China
| |
Collapse
|
16
|
Ali MM, Bhakta JN. Biosorption of zinc from aqueous solution using leaves of Corchorus olitorius as a low-cost biosorbent. WATER ENVIRONMENT RESEARCH : A RESEARCH PUBLICATION OF THE WATER ENVIRONMENT FEDERATION 2020; 92:821-828. [PMID: 31724269 DOI: 10.1002/wer.1274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/01/2019] [Accepted: 11/06/2019] [Indexed: 05/19/2023]
Abstract
Zinc (Zn) is one of the hazardous metal pollutants commonly found in industrial effluents and poses severe environmental and human health impacts. The present study chosen the leaves of Corchorus olitorius as a potential biosorbent among four different types of leaves employed for removing Zn from aqueous solution. The process parameters-contact time, pH, biosorbent dose, and initial Zn concentration were optimized for maximum removal of Zn using standard protocols. The Fourier-transform infrared spectroscopy study was performed to identify the functional groups involved in Zn biosorption mechanism. The biosorption equilibrium was achieved at 120 min of contact time. The biosorption of Zn was highest at pH 6 and biosorbent dose of 2 g/L. The sorption equilibrium data were well fitted with the Freundlich isotherm model (R2 = 0.995). Highest adsorption capacity of C. olitorius leaves was 11.63 mg/g. It is concluded that the leaves of C. olitorius could be used as a potentially low-cost novel biosorbent to remove Zn from contaminated water. PRACTITIONER POINTS: Optimum Zn sorption process parameters of Corchorus olitorius leaf biosorbent were determined. Zn sorption kinetic data of C. olitorius leaf were well fitted by Freundlich isotherm model. C. olitorius leaf biosorbent showed excellent Zn sorption capacity (11.63 mg/g) from water. Leaves of C. olitorius could be used as a potentially low-cost novel biosorbent.
Collapse
Affiliation(s)
- Md Motakabber Ali
- Department of Ecological Studies & International Center for Ecological Engineering, University of Kalyani, Kalyani, West Bengal, India
| | - Jatindra Nath Bhakta
- Department of Ecological Studies & International Center for Ecological Engineering, University of Kalyani, Kalyani, West Bengal, India
| |
Collapse
|
17
|
Keerthana S, Kumar A. Potential risks and benefits of zinc oxide nanoparticles: a systematic review. Crit Rev Toxicol 2020; 50:47-71. [PMID: 32186437 DOI: 10.1080/10408444.2020.1726282] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- S. Keerthana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| | - A. Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| |
Collapse
|
18
|
Yang Y, Song Z, Wu W, Xu A, Lv S, Ji S. ZnO Quantum Dots Induced Oxidative Stress and Apoptosis in HeLa and HEK-293T Cell Lines. Front Pharmacol 2020; 11:131. [PMID: 32180717 PMCID: PMC7057716 DOI: 10.3389/fphar.2020.00131] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Zinc oxide (ZnO) quantum dot (QD) is a promising inexpensive inorganic nanomaterials, of which potential toxic effects on biological systems and human health should be evaluated before biomedical application. In this study, the cytotoxicity of ZnO QDs was assessed using HeLa cervical cancer cell and HEK-293T human embryonic kidney cell lines. Cell viability was significantly decreased by treatment with 50 µg/ml ZnO QDs after only 6 h, and the cytotoxicity of ZnO QDs was higher in HEK-293T than in HeLa cells. ZnO QDs increased the level of reactive oxygen species and decreased the mitochondria membrane potential in a dose-dependent manner. Several gene expression involved in apoptosis was regulated by ZnO QDs, including bcl-2 gene and caspase. In HeLa cells, ZnO QDs significantly increased early and late apoptosis, but only late apoptosis was affected in HEK-293T cells. These findings will be helpful for future research and application of ZnO QDs in biomedicine.
Collapse
Affiliation(s)
- Yanjie Yang
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Zhenhua Song
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Weixia Wu
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Ao Xu
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuangyu Lv
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shaoping Ji
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
19
|
Park C, Cha HJ, Choi EO, Lee H, Hwang-Bo H, Ji SY, Kim MY, Kim SY, Hong SH, Cheong J, Kim GY, Yun SJ, Hwang HJ, Kim WJ, Choi YH. Isorhamnetin Induces Cell Cycle Arrest and Apoptosis Via Reactive Oxygen Species-Mediated AMP-Activated Protein Kinase Signaling Pathway Activation in Human Bladder Cancer Cells. Cancers (Basel) 2019; 11:cancers11101494. [PMID: 31590241 PMCID: PMC6826535 DOI: 10.3390/cancers11101494] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022] Open
Abstract
Isorhamnetin is an O-methylated flavonol that is predominantly found in the fruits and leaves of various plants, which have been used for traditional herbal remedies. Although several previous studies have reported that this flavonol has diverse health-promoting effects, evidence is still lacking for the underlying molecular mechanism of its anti-cancer efficacy. In this study, we examined the anti-proliferative effect of isorhamnetin on human bladder cancer cells and found that isorhamnetin triggered the gap 2/ mitosis (G2/M) phase cell arrest and apoptosis. Our data showed that isorhamnetin decreased the expression of Wee1 and cyclin B1, but increased the expression of cyclin-dependent kinase (Cdk) inhibitor p21WAF1/CIP1, and increased p21 was bound to Cdk1. In addition, isorhamnetin-induced apoptosis was associated with the increased expression of the Fas/Fas ligand, reduced ratio of B-cell lymphoma 2 (Bcl-2)/Bcl-2 associated X protein (Bax) expression, cytosolic release of cytochrome c, and activation of caspases. Moreover, isorhamnetin inactivated the adenosine 5′-monophosphate-activated protein kinase (AMPK) signaling pathway by diminishing the adenosine triphosphate (ATP) production due to impaired mitochondrial function. Furthermore, isorhamnetin stimulated production of intracellular reactive oxygen species (ROS); however, the interruption of ROS generation using a ROS scavenger led to an escape from isorhamnetin-mediated G2/M arrest and apoptosis. Collectively, this is the first report to show that isorhamnetin inhibited the proliferation of human bladder cancer cells by ROS-dependent arrest of the cell cycle at the G2/M phase and induction of apoptosis. Therefore, our results provide an important basis for the interpretation of the anti-cancer mechanism of isorhamnetin in bladder cancer cells and support the rationale for the need to evaluate more precise molecular mechanisms and in vivo anti-cancer properties.
Collapse
Affiliation(s)
- Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dong-eui University, Busan 47340, Korea;
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea;
| | - Eun Ok Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Hyun Hwang-Bo
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea;
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea;
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - So Young Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Su Hyun Hong
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - JaeHun Cheong
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea;
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea;
| | - Seok Joong Yun
- Department of Urology, College of Medicine, Chungbuk National University, Chungbuk 8644, Korea;
| | - Hye Jin Hwang
- Department of Food and Nutrition, College of Nursing, Healthcare Sciences & Human Ecology, Dong-Eui University, Busan 47340, Korea;
| | - Wun-Jae Kim
- Department of Urology, College of Medicine, Chungbuk National University, Chungbuk 8644, Korea;
- Correspondence: (W.-J.K.); (Y.H.C.); Tel.: +82-43-269-6136 (W.-J.K.); +82-51-850-7413 (Y.H.C.)
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
- Correspondence: (W.-J.K.); (Y.H.C.); Tel.: +82-43-269-6136 (W.-J.K.); +82-51-850-7413 (Y.H.C.)
| |
Collapse
|
20
|
Mousavi M, Hakimian S, Mustafa TA, Aziz FM, Salihi A, Ale-Ebrahim M, Mirpour M, Rasti B, Akhtari K, Shahpasand K, Abou-Zied OK, Falahati M. The interaction of silica nanoparticles with catalase and human mesenchymal stem cells: biophysical, theoretical and cellular studies. Int J Nanomedicine 2019; 14:5355-5368. [PMID: 31409992 PMCID: PMC6643057 DOI: 10.2147/ijn.s210136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/04/2019] [Indexed: 11/23/2022] Open
Abstract
Aim Nanoparticles (NPs) have been receiving potential interests in protein delivery and cell therapy. As a matter of fact, NPs may be used as great candidates in promoting cell therapy by catalase (CAT) delivery into high oxidative stress tissues. However, for using NPs like SiO2 as carriers, the interaction of NPs with proteins and mesenchymal stem cells (MSCs) should be explored in advance. Methods In the present study, the interaction of SiO2 NPs with CAT and human MSCs (hMSCs) was explored by various spectroscopic methods (fluorescence, circular dichroism (CD), UV-visible), molecular docking and dynamics studies, and cellular (MTT, cellular morphology, cellular uptake, lactate dehydrogenase, ROS, caspase-3, flow cytometry) assays. Results Fluorescence study displayed that both dynamic and static quenching mechanisms and hydrophobic interactions are involved in the spontaneous interaction of SiO2 NPs with CAT. CD spectra indicated that native structure of CAT remains stable after interaction with SiO2 NPs. UV-visible study also revealed that the kinetic parameters of CAT such as Km, Vmax, Kcat, and enzyme efficiency were not changed after the addition of SiO2 NPs. Molecular docking and dynamics studies showed that Si and SiO2 clusters interact with hydrophobic residues of CAT and SiO2 cluster causes minor changes in the CAT structure at a total simulation time of 200 ps. Cellular assays depicted that SiO2 NPs induce significant cell mortality, change in cellular morphology, cellular internalization, ROS elevation, and apoptosis in hMSCs at higher concentration than 100 µg/mL (170 µM). Conclusion The current results suggest that low concentrations of SiO2 NPs induce no substantial change or mortality against CAT and hMSCs, and potentially useful carriers in CAT delivery to hMSC.
Collapse
Affiliation(s)
- Mina Mousavi
- Department of Biochemistry and Biophysics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Hakimian
- Department of Biochemistry and Biophysics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Twana Ahmed Mustafa
- Department of Medical Laboratory Technology, Health Technical College, Erbil Polytechnic University, Erbil, Kurdistan Region, Iraq
| | - Falah Mohammad Aziz
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq.,Department of Medical Analysis, Faculty of Science, Tishk International University, Erbil, Iraq
| | - Mahsa Ale-Ebrahim
- Department of Physiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mirsasan Mirpour
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Behnam Rasti
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, Sanandaj, Iran
| | - Koorosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology (RI-SCBT), Tehran, Iran
| | - Osama K Abou-Zied
- Department of Chemistry, Faculty of Science, Sultan Qaboos University, P.O. Box 36, Postal Code 123 Muscat, Oman
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
21
|
Anti-Proliferative and Pro-Apoptotic Effects of Licochalcone A through ROS-Mediated Cell Cycle Arrest and Apoptosis in Human Bladder Cancer Cells. Int J Mol Sci 2019; 20:ijms20153820. [PMID: 31387245 PMCID: PMC6696302 DOI: 10.3390/ijms20153820] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/02/2019] [Accepted: 08/02/2019] [Indexed: 12/20/2022] Open
Abstract
Licochalcone A (LCA) is a chalcone that is predominantly found in the root of Glycyrrhiza species, which is widely used as an herbal medicine. Although previous studies have reported that LCA has a wide range of pharmacological effects, evidence for the underlying molecular mechanism of its anti-cancer efficacy is still lacking. In this study, we investigated the anti-proliferative effect of LCA on human bladder cancer cells, and found that LCA induced cell cycle arrest at G2/M phase and apoptotic cell death. Our data showed that LCA inhibited the expression of cyclin A, cyclin B1, and Wee1, but increased the expression of cyclin-dependent kinase (Cdk) inhibitor p21WAF1/CIP1, and increased p21 was bound to Cdc2 and Cdk2. LCA activated caspase-8 and -9, which are involved in the initiation of extrinsic and intrinsic apoptosis pathways, respectively, and also increased caspase-3 activity, a typical effect caspase, subsequently leading to poly (ADP-ribose) polymerase cleavage. Additionally, LCA increased the Bax/Bcl-2 ratio, and reduced the integrity of mitochondria, which contributed to the discharge of cytochrome c from the mitochondria to the cytoplasm. Moreover, LCA enhanced the intracellular levels of reactive oxygen species (ROS); however, the interruption of ROS generation using ROS scavenger led to escape from LCA-mediated G2/M arrest and apoptosis. Collectively, the present data indicate that LCA can inhibit the proliferation of human bladder cancer cells by inducing ROS-dependent G2/M phase arrest and apoptosis.
Collapse
|
22
|
Eckol Inhibits Particulate Matter 2.5-Induced Skin Keratinocyte Damage via MAPK Signaling Pathway. Mar Drugs 2019; 17:md17080444. [PMID: 31357588 PMCID: PMC6723658 DOI: 10.3390/md17080444] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 01/17/2023] Open
Abstract
Toxicity of particulate matter (PM) towards the epidermis has been well established in many epidemiological studies. It is manifested in cancer, aging, and skin damage. In this study, we aimed to show the mechanism underlying the protective effects of eckol, a phlorotannin isolated from brown seaweed, on human HaCaT keratinocytes against PM2.5-induced cell damage. First, to elucidate the underlying mechanism of toxicity of PM2.5, we checked the reactive oxygen species (ROS) level, which contributed significantly to cell damage. Experimental data indicate that excessive ROS caused damage to lipids, proteins, and DNA and induced mitochondrial dysfunction. Furthermore, eckol (30 μM) decreased ROS generation, ensuring the stability of molecules, and maintaining a steady mitochondrial state. The western blot analysis showed that PM2.5 promoted apoptosis-related protein levels and activated MAPK signaling pathway, whereas eckol protected cells from apoptosis by inhibiting MAPK signaling pathway. This was further reinforced by detailed investigations using MAPK inhibitors. Thus, our results demonstrated that inhibition of PM2.5-induced cell apoptosis by eckol was through MAPK signaling pathway. In conclusion, eckol could protect skin HaCaT cells from PM2.5-induced apoptosis via inhibiting ROS generation.
Collapse
|
23
|
Song WJ, Jeong MS, Choi DM, Kim KN, Wie MB. Zinc Oxide Nanoparticles Induce Autophagy and Apoptosis via Oxidative Injury and Pro-Inflammatory Cytokines in Primary Astrocyte Cultures. NANOMATERIALS 2019; 9:nano9071043. [PMID: 31330912 PMCID: PMC6669602 DOI: 10.3390/nano9071043] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/13/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022]
Abstract
The present study examined the potential toxic concentrations of zinc oxide nanoparticles (ZnO NPs) and associated autophagy and apoptosis-related injuries in primary neocortical astrocyte cultures. Concentrations of ZnO NPs ≥3 μg/mL induced significant toxicity in the astrocytes. At 24 h after exposure to the ZnO NPs, transmission electron microscopy revealed swelling of the endoplasmic reticulum (ER) and increased numbers of autophagolysosomes in the cultured astrocytes, and increased levels of LC3 (microtubule-associated protein 1 light chain 3)-mediated autophagy were identified by flow cytometry. Apoptosis induced by ZnO NP exposure was confirmed by the elevation of caspase-3/7 activity and 4′,6′-diamidino-2-phenylindole (DAPI) staining. Significant (p < 0.05) changes in the levels of glutathione peroxidase, superoxide dismutase, tumor necrosis factor (TNF-α), and interleukin-6 were observed by enzyme-linked immunoassay (ELISA) assay following the exposure of astrocyte cultures to ZnO NPs. Phosphatidylinositol 3-kinase (PI3K)/mitogen-activated protein kinase (MAPK) dual activation was induced by ZnO NPs in a dose-dependent manner. Additionally, the Akt (protein kinase B) inhibitor BML257 and the mTOR (mammalian target of rapamycin) inhibitor rapamycin contributed to the survival of astrocytes. Inhibitors of cyclooxygenase-2 and lipoxygenase attenuated ZnO NP-induced toxicity. Calcium-modulating compounds, antioxidants, and zinc/iron chelators also decreased ZnO NP-induced toxicity. Together, these results suggest that ZnO NP-induced autophagy and apoptosis may be associated with oxidative stress and the inflammatory process in primary astrocyte cultures.
Collapse
Affiliation(s)
- Woo-Ju Song
- Department of Veterinary Toxicology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Myung-Seon Jeong
- Chuncheon Center, Korean Basic Science Institute, Chuncheon 24341, Korea
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, Korea
| | - Dong-Min Choi
- Department of Veterinary Toxicology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Kil-Nam Kim
- Chuncheon Center, Korean Basic Science Institute, Chuncheon 24341, Korea
| | - Myung-Bok Wie
- Department of Veterinary Toxicology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea.
| |
Collapse
|
24
|
Inhibitory effects of CuInS 2 and CdTe nanoparticles on macrophage cytokine production and phagocytosis in vitro. Enzyme Microb Technol 2019; 127:50-57. [PMID: 31088616 DOI: 10.1016/j.enzmictec.2019.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/01/2019] [Accepted: 04/16/2019] [Indexed: 01/24/2023]
Abstract
Macrophages eliminate and destroy invading bacteria and contaminants by engulfing them or secreting cytokines that trigger downstream immune responses. Consequently, impairment of the phagocytic functions of macrophages and/or suppressing their cytokine secretion are dangerous to organisms that rely on immune protection. Accordingly, exposure to environmental nanoparticles (NPs) that display immunomodulatory properties are serious. In this work, two types of NPs, i.e., mild-toxicity CuInS2 NPs and high-toxicity CdTe NPs, were used to evaluate the effects of NP exposure for macrophages. Following incubation for 24 h, THP-1-derived macrophage viability was assessed using an MTT method after exposing the THP-1 cells to different concentrations of CuInS2 or CdTe NPs. Phagocytosis assays demonstrated that both CuInS2 and CdTe NPs impair phagocytic activity toward Staphylococcus aureus (S. aureus). After pretreatment with CuInS2 and CdTe NPs at 4 μmol/L, THP-1 macrophages exhibited decreases in phagocytic ratio from ca. 32.9% to ca. 18.5% and 18.7%, respectively. Since the zeta potentials of intact and weathered CuInS2 NPs were distributed over a wide range from positive to negative, large quantities of intact and weathered CuInS2 NPs bore sufficient positive charge on their surfaces to induce membrane depolarization, thus theoretically providing electrostatic forces between S. aureus and THP-1, which could induce downstream intracellular events that increase phagocytosis. However, real time polymerase chain reaction arrays revealed that transcription of the pro-inflammatory factors IL-1β, IL-6, and TNF-α decreased while that of the anti-inflammatory factor IL-10 increased after treatment with CuInS2 NPs. Furthermore, transcription of TNF-α decreased while IL-10 increased after treatment with CdTe NPs. Thus, both kinds of NPs inhibited phagocytosis of S. aureus by THP-1 to some extent, confirming that immunosuppression can occur when macrophages are exposed to environmental NPs.
Collapse
|
25
|
Kim MK, Kim KB, Lee JY, Kwack SJ, Kwon YC, Kang JS, Kim HS, Lee BM. Risk Assessment of 5-Chloro-2-Methylisothiazol-3(2H)-One/2-Methylisothiazol-3(2H)-One (CMIT/MIT) Used as a Preservative in Cosmetics. Toxicol Res 2019; 35:103-117. [PMID: 31015893 PMCID: PMC6467361 DOI: 10.5487/tr.2019.35.2.103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023] Open
Abstract
The mixture of 5-chloro-2-methylisothiazol-3(2H)-one (CMIT) and 2-methylisothiazol-3(2H)-one (MIT), CMIT/MIT, is a preservative in cosmetics. CMIT/MIT is a highly effective preservative; however, it is also a commonly known skin sensitizer. Therefore, in the present study, a risk assessment for safety management of CMIT/MIT was conducted on products containing 0.0015% of CMIT/MIT, which is the maximum MIT level allowed in current products. The no observed adverse effect level (NOAEL) for CMIT/MIT was 2.8 mg/kg bw/day obtained from a two-generation reproductive toxicity test, and the skin sensitization toxicity standard value for CMIT/MIT, or the no expected sensitization induction level (NESIL), was 1.25 μg/cm2/day in humans. According to a calculation of body exposure to cosmetics use, the systemic exposure dosage (SED) was calculated as 0.00423 mg/kg bw/day when leave-on and rinse-off products were considered. Additionally, the consumer exposure level (CEL) amounted to 0.77512 μg/cm2/day for all representative cosmetics and 0.00584 μg/cm2/day for rinse-off products only. As a result, the non-cancer margin of safety (MOS) was calculated as 633, and CMIT/MIT was determined to be safe when all representative cosmetics were evaluated. In addition, the skin sensitization acceptable exposure level (AEL)/CEL was calculated as 0.00538 for all representative cosmetics and 2.14225 for rinse-off products; thus, CMIT/MIT was considered a skin sensitizer when all representative cosmetics were evaluated. Current regulations indicate that CMIT/MIT can only be used at concentrations 0.0015% or less and is prohibited from use in other cosmetics products. According to the results of this risk assessment, the CMIT/MIT regulatory values currently used in cosmetics are evaluated as appropriate.
Collapse
Affiliation(s)
- Min Kook Kim
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Kyu-Bong Kim
- College of Pharmacy, Dankook University, Cheonan, Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | - Seung Jun Kwack
- College of Natural Science, Changwon National University, Changwon, Korea
| | - Yong Chan Kwon
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Ji Soo Kang
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Hyung Sik Kim
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Byung-Mu Lee
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|