51
|
Zhang R, Li D, Zhao R, Luo D, Hu Y, Wang S, Zhuo X, Iqbal MZ, Zhang H, Han Q, Kong X. Spike structure of gold nanobranches induces hepatotoxicity in mouse hepatocyte organoid models. J Nanobiotechnology 2024; 22:92. [PMID: 38443940 PMCID: PMC10913213 DOI: 10.1186/s12951-024-02363-1] [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: 12/29/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Gold nanoparticles (GNPs) have been extensively recognized as an active candidate for a large variety of biomedical applications. However, the clinical conversion of specific types of GNPs has been hindered due to their potential liver toxicity. The origin of their hepatotoxicity and the underlying key factors are still ambiguous. Because the size, shape, and surfactant of GNPs all affect their properties and cytotoxicity. An effective and sensitive platform that can provide deep insights into the cause of GNPs' hepatotoxicity in vitro is therefore highly desired. METHODS Here, hepatocyte organoid models (Hep-orgs) were constructed to evaluate the shape-dependent hepatotoxicity of GNPs. Two types of GNPs with different nanomorphology, gold nanospheres (GNSs) and spiny gold nanobranches (GNBs), were synthesized as the representative samples. Their shape-dependent effects on mice Hep-orgs' morphology, cellular cytoskeletal structure, mitochondrial structure, oxidative stress, and metabolism were carefully investigated. RESULTS The results showed that GNBs with higher spikiness and tip curvature exhibited more significant cytotoxicity compared to the rounded GNSs. The spike structure of GNBs leads to a mitochondrial damage, oxidative stress, and metabolic disorder in Hep-orgs. Meanwhile, similar trends can be observed in HepG2 cells and mice models, demonstrating the reliability of the Hep-orgs. CONCLUSIONS Hep-orgs can serve as an effective platform for exploring the interactions between GNPs and liver cells in a 3D perspective, filling the gap between 2D cell models and animal models. This work further revealed that organoids can be used as an indispensable tool to rapidly screen and explore the toxic mechanism of nanomaterials before considering their biomedical functionalities.
Collapse
Affiliation(s)
- Rui Zhang
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou, 310000, PR China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Dan Li
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou, 310000, PR China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Ruibo Zhao
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou, 310000, PR China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Dandan Luo
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou, 310000, PR China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Yeting Hu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310030, PR China
| | - Shengyan Wang
- School of Science Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Xiaolu Zhuo
- School of Science Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - M Zubair Iqbal
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou, 310000, PR China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Han Zhang
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou, 310000, PR China.
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China.
| | - Qianqian Han
- National Institutes for Food and Drug Control, Beijing, PR China.
| | - Xiangdong Kong
- Institute for Smart Biomedical Materials, School of Materials Science & Engineering, Zhejiang Sci-Tech University, Hangzhou, 310000, PR China.
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China.
| |
Collapse
|
52
|
Zhang M, Wang W, Zhang D, Zhang Y, Yang Z, Li Y, Fang F, Xue Y, Zhang Y. Copper oxide nanoparticles impairs oocyte meiosis maturation by inducing mitochondrial dysfunction and oxidative stress. Food Chem Toxicol 2024; 185:114441. [PMID: 38218586 DOI: 10.1016/j.fct.2024.114441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/06/2024] [Accepted: 01/07/2024] [Indexed: 01/15/2024]
Abstract
Copper oxides nanoparticles (CuO NPs) are widely used for a variety of industrial and life science applications. In addition to cause neurotoxicity, hepatotoxicity, immunotoxicity, CuO NPs have also been reported to adversely affect the reproductive system in animals; However, little is known about the effects and potential mechanism of CuO NPs exposure on oocyte quality, especially oocyte maturation. In the present study, we reported that CuO NPs exposure impairs the oocyte maturation by disrupting meiotic spindle assembly and chromosome alignment, as well as kinetochore-microtubule attachment. In addition, CuO NPs exposure also affects the acetylation level of α-tubulin in mice oocyte, which hence impairs microtubule dynamics and organization. Besides, CuO NPs exposure would result in the mis-localization of Juno and Ovastacin, which might be one of the critical factors leading to the failure of oocyte maturation. Finally, CuO NPs exposure impairs the mitochondrial distribution and induced high levels of ROS, which led to the accumulation of DNA damage and occurrence of apoptosis. In summary, our results indicated that CuO NPs exposure had potential toxic effects on female fertility and led to the poor oocyte quality in female mice.
Collapse
Affiliation(s)
- Mianqun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China
| | - Wei Wang
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China
| | - Dandan Zhang
- Department of Reproductive Medicine, General Hospital of WanBei Coal Group, Suzhou, 234000, China
| | - Yiwen Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China
| | - Zaishan Yang
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China
| | - Yunsheng Li
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China
| | - Fugui Fang
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China
| | - Yanfeng Xue
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China.
| | - Yunhai Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Key Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding of Anhui Province, Hefei, 230036, China.
| |
Collapse
|
53
|
Wang BJ, Chen YY, Chang HH, Chen RJ, Wang YJ, Lee YH. Zinc oxide nanoparticles exacerbate skin epithelial cell damage by upregulating pro-inflammatory cytokines and exosome secretion in M1 macrophages following UVB irradiation-induced skin injury. Part Fibre Toxicol 2024; 21:9. [PMID: 38419076 PMCID: PMC10900617 DOI: 10.1186/s12989-024-00571-z] [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: 09/06/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Zinc oxide nanoparticles (ZnONPs) are common materials used in skin-related cosmetics and sunscreen products due to their whitening and strong UV light absorption properties. Although the protective effects of ZnONPs against UV light in intact skin have been well demonstrated, the effects of using ZnONPs on damaged or sunburned skin are still unclear. In this study, we aimed to reveal the detailed underlying mechanisms related to keratinocytes and macrophages exposed to UVB and ZnONPs. RESULTS We demonstrated that ZnONPs exacerbated mouse skin damage after UVB exposure, followed by increased transepidermal water loss (TEWL) levels, cell death and epithelial thickness. In addition, ZnONPs could penetrate through the damaged epithelium, gain access to the dermis cells, and lead to severe inflammation by activation of M1 macrophage. Mechanistic studies indicated that co-exposure of keratinocytes to UVB and ZnONPs lysosomal impairment and autophagy dysfunction, which increased cell exosome release. However, these exosomes could be taken up by macrophages, which accelerated M1 macrophage polarization. Furthermore, ZnONPs also induced a lasting inflammatory response in M1 macrophages and affected epithelial cell repair by regulating the autophagy-mediated NLRP3 inflammasome and macrophage exosome secretion. CONCLUSIONS Our findings propose a new concept for ZnONP-induced skin toxicity mechanisms and the safety issue of ZnONPs application on vulnerable skin. The process involved an interplay of lysosomal impairment, autophagy-mediated NLRP3 inflammasome and macrophage exosome secretion. The current finding is valuable for evaluating the effects of ZnONPs for cosmetics applications.
Collapse
Affiliation(s)
- Bour-Jr Wang
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan
- Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan, 70403, Taiwan
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Hui-Hsuan Chang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 406040, Taiwan.
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
54
|
Chen M, Wu T. Nanoparticles and neurodegeneration: Insights on multiple pathways of programmed cell death regulated by nanoparticles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168739. [PMID: 38008311 DOI: 10.1016/j.scitotenv.2023.168739] [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: 07/27/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
Currently, nanoparticles (NPs) are extensively applied in the diagnosis and treatment of neurodegenerative diseases (NDs). With the rapid development and increasing exposure to the public, the potential neurotoxicity associated with NDs caused by NPs has attracted the researchers' attentions but their biosafety assessments are still far behind relevant application studies. Based on recent research, this review aims to conduct a comprehensive and systematic analysis of neurotoxicity induced by NPs. The 191 studies selected according to inclusion and exclusion criteria were imported into the software, and the co-citations and keywords of the included literatures were analyzed to find the breakthrough point of previous studies. According to the available studies, the routes of NPs entering into the normal and injured brain were various, and then to be distributed and accumulated in living bodies. When analyzing the adverse effects induced by NPs, we focused on multiple programmed cell deaths (PCDs), especially ferroptosis triggered by NPs and their tight connection and crosstalk that have been found playing critical roles in the pathogenesis of NDs and their underlying toxic mechanisms. The activation of multiple PCD pathways by NPs provides a scientific basis for the occurrence and development of NDs. Furthermore, the adoption of new methodologies for evaluating the biosafety of NPs would benefit the next generation risk assessment (NGRA) of NPs and their toxic interventions. This would help ensure their safe application and sustainable development in the field of medical neurobiology.
Collapse
Affiliation(s)
- Min Chen
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Tianshu Wu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing 210009, PR China.
| |
Collapse
|
55
|
Vahabirad M, Daei S, Abbasalipourkabir R, Ziamajidi N. Anticancer Action of Silver Nanoparticles in SKBR3 Breast Cancer Cells through Promotion of Oxidative Stress and Apoptosis. BIOMED RESEARCH INTERNATIONAL 2024; 2024:7145339. [PMID: 38410788 PMCID: PMC10896653 DOI: 10.1155/2024/7145339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/04/2023] [Accepted: 01/18/2024] [Indexed: 02/28/2024]
Abstract
Silver nanoparticles (AgNPs) are known as one of the highly utilized NPs owing to their unique characteristics in the field of cancer research. The goal of this research was to explore the oxidative stress, apoptosis, and angiogenesis in SKBR3 breast cancer cells after exposure to AgNPs. The survival rate of SKBR3 cancer cells and MCF-10A normal breast cells was assessed under the effects of different concentrations (0, 32, 64, 128, and 250 μg/ml) by MTT method. The oxidative condition was assessed by measuring reactive oxygen species (ROS) production, total oxidant status (TOS), total antioxidant capacity (TAC), malondialdehyde (MDA), and antioxidant enzyme activity (CAT, GPx, and CAT) using colorimetric-based kits. Flow cytometry and Hoechst 33258 staining were performed to investigate the induction of apoptosis. Furthermore, the expression of Bcl-2-associated X protein (Bax), B-cell lymphoma 2 (Bcl-2), and caspase 3 and 7 activity was measured. The cell migration and vascular endothelial growth factor-A (VEGF-A) gene expression, protein kinase B (AKT), phosphatidylinositol 3-kinase (PI3K) were also studied. The MTT results indicated that AgNPs inhibit the SKBR3 cells' viability in a concentration-dependent way. Besides, AgNPs markedly induced oxidative stress via increasing TOS content, MDA production, reduction of TAC, and regulation of antioxidant enzyme level. Additionally, AgNPs promoted apoptosis as revealed by an enhancement in Bax/Bcl-2 expression ratio. Findings also indicated that AgNPs suppress the expression of genes (VEGF-A, AKT, and PI3K) involved in angiogenesis. Altogether, our data revealed that AgNPs initiate oxidative stress and apoptosis in SKBR3 breast cancer cells, dose dependently.
Collapse
Affiliation(s)
- Mohammad Vahabirad
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajedeh Daei
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
56
|
Wu Y, Li L, Ning Z, Li C, Yin Y, Chen K, Li L, Xu F, Gao J. Autophagy-modulating biomaterials: multifunctional weapons to promote tissue regeneration. Cell Commun Signal 2024; 22:124. [PMID: 38360732 PMCID: PMC10868121 DOI: 10.1186/s12964-023-01346-3] [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: 08/24/2023] [Accepted: 09/29/2023] [Indexed: 02/17/2024] Open
Abstract
Autophagy is a self-renewal mechanism that maintains homeostasis and can promote tissue regeneration by regulating inflammation, reducing oxidative stress and promoting cell differentiation. The interaction between biomaterials and tissue cells significantly affects biomaterial-tissue integration and tissue regeneration. In recent years, it has been found that biomaterials can affect various processes related to tissue regeneration by regulating autophagy. The utilization of biomaterials in a controlled environment has become a prominent approach for enhancing the tissue regeneration capabilities. This involves the regulation of autophagy in diverse cell types implicated in tissue regeneration, encompassing the modulation of inflammatory responses, oxidative stress, cell differentiation, proliferation, migration, apoptosis, and extracellular matrix formation. In addition, biomaterials possess the potential to serve as carriers for drug delivery, enabling the regulation of autophagy by either activating or inhibiting its processes. This review summarizes the relationship between autophagy and tissue regeneration and discusses the role of biomaterial-based autophagy in tissue regeneration. In addition, recent advanced technologies used to design autophagy-modulating biomaterials are summarized, and rational design of biomaterials for providing controlled autophagy regulation via modification of the chemistry and surface of biomaterials and incorporation of cells and molecules is discussed. A better understanding of biomaterial-based autophagy and tissue regeneration, as well as the underlying molecular mechanisms, may lead to new possibilities for promoting tissue regeneration. Video Abstract.
Collapse
Affiliation(s)
- Yan Wu
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Luxin Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Zuojun Ning
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Changrong Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Yongkui Yin
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Kaiyuan Chen
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Lu Li
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Fei Xu
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
57
|
Encinas-Gimenez M, Martin-Duque P, Martín-Pardillos A. Cellular Alterations Due to Direct and Indirect Interaction of Nanomaterials with Nucleic Acids. Int J Mol Sci 2024; 25:1983. [PMID: 38396662 PMCID: PMC10889090 DOI: 10.3390/ijms25041983] [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: 12/26/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Deoxyribonucleic acid (DNA) represents the main reservoir of genetic information in the cells, which is why it is protected in the nucleus. Entry into the nucleus is, in general, difficult, as the nuclear membrane is a selective barrier to molecules longer than 40 kDa. However, in some cases, the size of certain nanoparticles (NPs) allows their internalization into the nucleus, thus causing a direct effect on the DNA structure. NPs can also induce indirect effects on DNA through reactive oxygen species (ROS) generation. In this context, nanomaterials are emerging as a disruptive tool for the development of novel therapies in a broad range of biomedical fields; although their effect on cell viability is commonly studied, further interactions with DNA or indirect alterations triggered by the internalization of these materials are not always clarified, since the small size of these materials makes them perfectly suitable for interaction with subcellular structures, such as the nucleus. In this context, and using as a reference the predicted interactions presented in a computational model, we describe and discuss the observed direct and indirect effects of the implicated nanomaterials on DNA.
Collapse
Affiliation(s)
- Miguel Encinas-Gimenez
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.E.-G.); (A.M.-P.)
- Department of Chemical Engineering and Environmental Technology (IQTMA), University of Zaragoza, 50018 Zaragoza, Spain
- Ciber Bioingeniería y Biomateriales (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pilar Martin-Duque
- Ciber Bioingeniería y Biomateriales (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Desarrollo de Medicamentos de Terapias Avanzadas (DDMTA), Centro de Terapias Avanzadas, Instituto de Salud Carlos lll, 28222 Madrid, Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Ana Martín-Pardillos
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.E.-G.); (A.M.-P.)
- Department of Chemical Engineering and Environmental Technology (IQTMA), University of Zaragoza, 50018 Zaragoza, Spain
- Ciber Bioingeniería y Biomateriales (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
58
|
Monteiro MS, Mesquita MS, Garcia LM, Dos Santos PR, de Marangoni de Viveiros CC, da Fonseca RD, Xavier MA, de Mendonça GW, Rosa SS, Silva SL, Paterno LG, Morais PC, Báo SN. Radiofrequency driving antitumor effect of graphene oxide-based nanocomposites: a Hill model analysis. Nanomedicine (Lond) 2024; 19:397-412. [PMID: 38112257 DOI: 10.2217/nnm-2023-0312] [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] [Indexed: 12/21/2023] Open
Abstract
Aim: This report proposes using the Hill model to assess the benchmark dose, the 50% lethal dose, the cooperativity and the dissociation constant while analyzing cell viability data using nanomaterials to evaluate the antitumor potential while combined with radiofrequency therapy. Materials & methods: A nanocomposite was synthesized (graphene oxide-polyethyleneimine-gold) and the viability was evaluated using two tumor cell lines, namely LLC-WRC-256 and B16-F10. Results: Our findings demonstrated that while the nanocomposite is biocompatible against the LLC-WRC-256 and B16-F10 cancer cell lines in the absence of radiofrequency, the application of radiofrequency enhances the cell toxicity by orders of magnitude. Conclusion: This result points to prospective studies with the tested cell lines using tumor animal models.
Collapse
Affiliation(s)
- Melissa S Monteiro
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| | - Marina S Mesquita
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| | - Leidiane M Garcia
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| | - Paulo R Dos Santos
- Porto Velho Calama Campus, Federal Institute of Rondônia, Porto Velho, Rondônia, 76820-441, Brazil
| | | | - Ronei D da Fonseca
- PRC/DIMAT, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| | - Mary A Xavier
- Faculty of Agronomy & Veterinary, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| | | | - Suélia Srf Rosa
- Faculty of Gama, University of Brasília, Brasília, Distrito Federal, 72444-240, Brazil
| | - Saulo Lp Silva
- Institute of Chemistry, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| | - Leonardo G Paterno
- Institute of Chemistry, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| | - Paulo C Morais
- Institute of Physics, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
- Biotechnology & Genomic Sciences, Catholic University of Brasília, Brasília, Distrito Federal, 70790-160, Brazil
| | - Sônia N Báo
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Distrito Federal, 70910-900, Brazil
| |
Collapse
|
59
|
Li Y, Lu Y, Li J, Li M, Gou H, Sun X, Xu X, Song B, Li Z, Ma Y. Screening of low-toxic zinc oxide nanomaterials and study the apoptosis mechanism of NSC-34 cells. Biotechnol J 2024; 19:e2300443. [PMID: 38403432 DOI: 10.1002/biot.202300443] [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: 08/30/2023] [Revised: 11/10/2023] [Accepted: 01/08/2024] [Indexed: 02/27/2024]
Abstract
With the increasing application of ZnO nanomaterials (ZnO-NMts) in the biomedical field, it is crucial to assess their potential risks to humans and the environment. Therefore, this study aimed to screen for ZnO-NMts with low toxicity and establish safe exposure limits, and investigate their mechanisms of action. The study synthesized 0D ZnO nanoparticles (ZnO NPs) and 3D ZnO nanoflowers (ZnO Nfs) with different morphologies using a hydrothermal approach for comparative research. The ZnO-NMts were characterized using X-ray diffraction (XRD), scanning electron microscopy (SEM), and transmission electron microscopy (TEM). Mouse brain neuronal cells (NSC-34) were incubated with ZnO NMts for 6, 12, and 24 h, and the cell morphology was observed using TEM. The toxic effects of ZnO Nfs on NSC-34 cells were studied using CCK-8 cell viability detection, reactive oxygen species (ROS) measurement, caspase-3 activity detection, Annexin V-FITC/PI apoptosis assay, and mitochondrial membrane potential (Δφm) measurement. The results of the research showed that ZnO-NMts caused cytoplasmic vacuolization and nuclear pyknosis. After incubating cells with 12.5 µg mL-1 ZnO-NMts for 12 h, ZnO NRfs exhibited the least toxicity and ROS levels. Additionally, there was a significant increase in caspase-3 activity, depolarization of mitochondrial membrane potential (Δφm), and the highest rate of early apoptosis.This study successfully identified ZnO NRfs with the lowest toxicity and determined the safe exposure limit to be < 12.5 µg mL-1 (12 h). These findings will contribute to the clinical use of ZnO NRfs with low toxicity and provide a foundation for further research on their potential applications in brain disease treatment.
Collapse
Affiliation(s)
- Yuanyuan Li
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| | - Yan Lu
- Key Laboratory of Atomic and Molecular Physics & Functional Materials of Gansu Province, College of Physics and Electronic Engineering, Northwest Normal University, Lanzhou, China
| | - Jingjing Li
- College of Pharmacy, Gansu University of Traditional Chinese Medicine, Gansu, China
| | - Mei Li
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| | - Huitian Gou
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| | - Xiaolin Sun
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| | - Xiaoli Xu
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| | - Beibei Song
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| | - Zhiyu Li
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| | - Yonghua Ma
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, Gansu, China
| |
Collapse
|
60
|
Choi SH, Hwang HS, Han S, Eom H, Choi JS, Han S, Lee D, Lee SY, Koo H, Kwon HJ, Lim YB. Inhibition of protein-protein interactions using biodegradable depsipeptide nanoassemblies. J Control Release 2024; 366:104-113. [PMID: 38128883 DOI: 10.1016/j.jconrel.2023.12.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Although peptides notoriously have poor intrinsic pharmacokinetic properties, it is well-known that nanostructures with excellent pharmacokinetic properties can be designed. Noticing that peptide inhibitors are generally nonpolar, here, we consolidate the peptide inhibitor targeting intracellular protein-protein interactions (PPIs) as an integral part of biodegradable self-assembled depsipeptide nanostructures (SdPNs). Because the peptide inhibitor has the dual role of PPI inhibition and self-assembly in this design, problems associated with the poor pharmacokinetics of peptides and encapsulation/entrapment processes can be overcome. Optimized SdPNs displayed better tumor targeting and PPI inhibition properties than the comparable small molecule inhibitor in vivo. Kinetics of PPI inhibition for SdPNs were gradual and controllable in contrast to the rapid inhibition kinetics of the small molecule. Because SdPN is modular, any appropriate peptide inhibitor can be incorporated into the platform without concern for the poor pharmacokinetic properties of the peptide.
Collapse
Affiliation(s)
- Se-Hwan Choi
- Department of Materials Science and Engineering, Yonsei University, Seoul, Republic of Korea
| | - Hyun-Seok Hwang
- Department of Materials Science and Engineering, Yonsei University, Seoul, Republic of Korea
| | - Seongryeong Han
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hohyeon Eom
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jun Shik Choi
- Department of Materials Science and Engineering, Yonsei University, Seoul, Republic of Korea; Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Sanghun Han
- Department of Materials Science and Engineering, Yonsei University, Seoul, Republic of Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soo Yeon Lee
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Ho Jeong Kwon
- Chemical Genomics Leader Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Yong-Beom Lim
- Department of Materials Science and Engineering, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
61
|
Alsaleh NB, Aljarbou AM, Assal ME, Assiri MA, Almutairi MM, As Sobeai HM, Alshamrani AA, Almudimeegh S, Hatshan MR, Adil SF. Synthesis, Characterization, and Toxicity Assessment of Zinc Oxide-Doped Manganese Oxide Nanoparticles in a Macrophage Model. Pharmaceuticals (Basel) 2024; 17:168. [PMID: 38399383 PMCID: PMC10892842 DOI: 10.3390/ph17020168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
The doping of engineered nanomaterials (ENMs) is a key tool for manipulating the properties of ENMs (e.g., electromagnetic, optical, etc.) for different therapeutic applications. However, adverse health outcomes and the cellular biointeraction of doped ENMs, compared to undoped counterparts, are not fully understood. Previously, we have shown that doping manganese oxide nanoparticles with ZnO (ZnO-MnO2 NPs) improved their catalytic properties. In this study, we assessed the toxicity of ZnO-MnO2 NPs in Raw 264.7 cells. NPs were prepared via an eco-friendly, co-precipitation method and characterized by several techniques, including transmission and scanning electron microscopy, X-ray diffraction, and Fourier transform infrared. The physicochemical properties of ZnO-MnO2 NPs, including size, morphology, and crystalline structure, were almost identical to MnO2 NPs. However, ZnO-MnO2 NPs showed slightly larger particle aggregates and negative charge in cell culture media. Exposure to ZnO-MnO2 NPs resulted in lower toxicity based on the cell viability and functional assay (phagocytosis) data. Exposure to both NPs resulted in the activation of the cell inflammatory response and the generation of reactive oxygen species (ROS). Despite this, exposure to ZnO-MnO2 NPs was associated with a lower toxicity profile, and it resulted in a higher ROS burst and the activation of the cell antioxidant system, hence indicating that MnO2 NP-induced toxicity is potentially mediated via other ROS-independent pathways. Furthermore, the cellular internalization of ZnO-MnO2 NPs was lower compared to MnO2 NPs, and this could explain the lower extent of toxicity of ZnO-MnO2 NPs and suggests Zn-driven ROS generation. Together, the findings of this report suggest that ZnO (1%) doping impacts cellular biointeraction and the consequent toxicological outcomes of MnO2 NPs in Raw 264.7 cells.
Collapse
Affiliation(s)
- Nasser B. Alsaleh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.M.A.); (M.A.A.); (M.M.A.); (H.M.A.S.); (A.A.A.); (S.A.)
| | - Anas M. Aljarbou
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.M.A.); (M.A.A.); (M.M.A.); (H.M.A.S.); (A.A.A.); (S.A.)
| | - Mohamed E. Assal
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.E.A.); (M.R.H.)
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.M.A.); (M.A.A.); (M.M.A.); (H.M.A.S.); (A.A.A.); (S.A.)
| | - Mohammed M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.M.A.); (M.A.A.); (M.M.A.); (H.M.A.S.); (A.A.A.); (S.A.)
| | - Homood M. As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.M.A.); (M.A.A.); (M.M.A.); (H.M.A.S.); (A.A.A.); (S.A.)
| | - Ali A. Alshamrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.M.A.); (M.A.A.); (M.M.A.); (H.M.A.S.); (A.A.A.); (S.A.)
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.M.A.); (M.A.A.); (M.M.A.); (H.M.A.S.); (A.A.A.); (S.A.)
| | - Mohammad R. Hatshan
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.E.A.); (M.R.H.)
| | - Syed F. Adil
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.E.A.); (M.R.H.)
| |
Collapse
|
62
|
Zhang Z, Miao G, Lu L, Yin H, Wang Y, Wang B, Pan R, Zheng C, Jin X. Crucial physicochemical factors mediating mitochondrial toxicity of nanoparticles at noncytotoxic concentration. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168211. [PMID: 37918742 DOI: 10.1016/j.scitotenv.2023.168211] [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: 08/30/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Nanomaterials have been extensively applied in multiple industries, among which silver nanoparticles (AgNPs), silicon dioxide nanoparticles (SiNPs), and gold nanoparticles (AuNPs) have become representative of widely consumed NPs. Limited knowledge is available regarding the subcellular responses of NPs with different physicochemical properties, i.e. material type and size, under the noncytotoxic concentrations. Macrophages are important sensitive cells exposed to NPs, and mitochondria are sensitive organelles that respond at the subcellular level. Herein, we found that sublethal concentrations of AgNPs and SiNPs, not AuNPs, decreased the mitochondrial membrane potential (MMP) and tubular mitochondria, and further resulted in an increase of ROS level and a decrease of ATP generation. AgNPs and SiNPs can also disturb mitochondrial dynamics manifested as increasing Mfn2 expression and decreasing Drp1 expression. Further assessments for mitochondrial function showed that AgNPs and SiNPs exposure led to a decrease in the gene expressions related to complex I (Ndufa8 and Ndufs2), complex III (Uqcrc2 and Uqcrfs1), complex IV (Cox6b1), and activity of complex I, suggesting their potential roles in impairing cellular respiration. In terms of the effects of NPs with different sizes, stronger toxicity was observed in smaller-sized nanoparticles. Among the above mitochondrial changes, we identified that ROS, ATP, MMP, tubular mitochondria, and expression of Drp1 were relatively sensitive indicators in subcellular response to NPs. With the above sensitive indicators, the comparison of heterogeneity between material type and size of the NPs showed that material type occupied a main influence on subcellular mitochondrial effects. Our finding provided important data on the potential subcellular risks of NPs, and indicated the vital role of material type for a better understanding of the nanomaterial biological safety.
Collapse
Affiliation(s)
- Ze Zhang
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Gan Miao
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Lin Lu
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Hao Yin
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Yingzhu Wang
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Baoqiang Wang
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Ruonan Pan
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Chuer Zheng
- School of Public Health, Qingdao University, Qingdao 266071, PR China
| | - Xiaoting Jin
- School of Public Health, Qingdao University, Qingdao 266071, PR China.
| |
Collapse
|
63
|
Pashootan P, Saadati F, Fahimi H, Rahmati M, Strippoli R, Zarrabi A, Cordani M, Moosavi MA. Metal-based nanoparticles in cancer therapy: Exploring photodynamic therapy and its interplay with regulated cell death pathways. Int J Pharm 2024; 649:123622. [PMID: 37989403 DOI: 10.1016/j.ijpharm.2023.123622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/01/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
Photodynamic therapy (PDT) represents a non-invasive treatment strategy currently utilized in the clinical management of selected cancers and infections. This technique is predicated on the administration of a photosensitizer (PS) and subsequent irradiation with light of specific wavelengths, thereby generating reactive oxygen species (ROS) within targeted cells. The cellular effects of PDT are dependent on both the localization of the PS and the severity of ROS challenge, potentially leading to the stimulation of various cell death modalities. For many years, the concept of regulated cell death (RCD) triggered by photodynamic reactions predominantly encompassed apoptosis, necrosis, and autophagy. However, in recent decades, further explorations have unveiled additional cell death modalities, such as necroptosis, ferroptosis, cuproptosis, pyroptosis, parthanatos, and immunogenic cell death (ICD), which helps to achieve tumor cell elimination. Recently, nanoparticles (NPs) have demonstrated substantial advantages over traditional PSs and become important components of PDT, due to their improved physicochemical properties, such as enhanced solubility and superior specificity for targeted cells. This review aims to summarize recent advancements in the applications of different metal-based NPs as PSs or delivery systems for optimized PDT in cancer treatment. Furthermore, it mechanistically highlights the contribution of RCD pathways during PDT with metal NPs and how these forms of cell death can improve specific PDT regimens in cancer therapy.
Collapse
Affiliation(s)
- Parya Pashootan
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Saadati
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Hossein Fahimi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; National Institute for Infectious Diseases L. Spallanzani IRCCS, Rome, Italy
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkey; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077, India
| | - Marco Cordani
- Departament of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran.
| |
Collapse
|
64
|
Li Y, Du X, Kong X, Fang Y, He Z, Liu D, Wu H, Ji J, Yang X, Ye L, Zhai G. A pro-death autophagy-based nanoplatform for enhancing antitumour efficacy with improved immune responses. Eur J Med Chem 2024; 263:115952. [PMID: 37992519 DOI: 10.1016/j.ejmech.2023.115952] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023]
Abstract
Due to the pro-survival effect of mild autophagy, the therapeutic effect of chemo-immunotherapy is unsatisfactory. In addition, the adverse tumour microenvironment (TME), including the lack of antigen presentation, the deficiency of oxygen supply and immunosuppressive cells, results in immune escape and metastasis. Herein, a novel nanoplatform (CS-3BP/PA@DOX) based on the autophagy cascade is proposed for the first time to deliver the chemotherapeutic doxorubicin (DOX) and respiration inhibitor 3-bromopyruvic acid (3BP) to overcome the above obstacles. CS-3BP/PA@DOX exerts a synergistic therapeutic effect to initiate pro-death autophagy and facilitate the antigen presentation process by combining DOX chemotherapy and starvation therapy with 3BP. Additionally, CS-3BP/PA@DOX remodelled the immunosuppressive TME by alleviating hypoxia, damaging dense ECM, and downregulating PD-L1 to enhance antitumour immunity. 3BP was found to promote GSH depletion by inhibiting respiration for the first time, which reduces the chemical resistance of cancer and increases the sensitivity of cells to ROS, providing a new therapeutic direction of 3BP for antitumour treatment. Collectively, this study offers an opportunity to magnify pro-death autophagy, augment antitumour efficacy, facilitate anti-metastatic effects, and boost immune responses.
Collapse
Affiliation(s)
- Yingying Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiyou Du
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, PR China.
| | - Xinru Kong
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yuelin Fang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Zhijing He
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Dongzhu Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Hang Wu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Jianbo Ji
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaoye Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lei Ye
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Guangxi Zhai
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
65
|
Li Y, Li J, Li M, Sun J, Shang X, Ma Y. Biological mechanism of ZnO nanomaterials. J Appl Toxicol 2024; 44:107-117. [PMID: 37518903 DOI: 10.1002/jat.4522] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Modern nanotechnology has made zinc oxide nanomaterials (ZnO NMts) multifunctional, stable, and low cost, enabling them to be widely used in commercial and biomedical fields. With its wide application, the risk of human direct contact and their release into the environment also increases. This review aims to summarize the toxicity studies of ZnO NMts in vivo, including neurotoxicity, inhalation toxicity, and reproductive toxicity. The antibacterial and antiviral mechanisms of ZnO NMts in vitro and the toxicity to eukaryotic cells were summarized. The summary found that it was mainly related to reactive oxygen species (ROS) produced by oxidative stress. It also discusses the potential harm to body and the favorable prospects of the widespread use of antibacterial and antiviral in the future medical field. The review also emphasizes that the dosage and use method of ZnO NMts will be the focus of future biomedical research.
Collapse
Affiliation(s)
- Yuanyuan Li
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, China
| | - Jingjing Li
- College of Pharmacy, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Mei Li
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, China
| | - Jiwen Sun
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, China
| | - Xiaofen Shang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, China
| | - Yonghua Ma
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, China
| |
Collapse
|
66
|
Dehghan S, Kheshtchin N, Hassannezhad S, Soleimani M. Cell death classification: A new insight based on molecular mechanisms. Exp Cell Res 2023; 433:113860. [PMID: 38013091 DOI: 10.1016/j.yexcr.2023.113860] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Cells tend to disintegrate themselves or are forced to undergo such destructive processes in critical circumstances. This complex cellular function necessitates various mechanisms and molecular pathways in order to be executed. The very nature of cell death is essentially important and vital for maintaining homeostasis, thus any type of disturbing occurrence might lead to different sorts of diseases and dysfunctions. Cell death has various modalities and yet, every now and then, a new type of this elegant procedure gets to be discovered. The diversity of cell death compels the need for a universal organizing system in order to facilitate further studies, therapeutic strategies and the invention of new methods of research. Considering all that, we attempted to review most of the known cell death mechanisms and sort them all into one arranging system that operates under a simple but subtle decision-making (If \ Else) order as a sorting algorithm, in which it decides to place and sort an input data (a type of cell death) into its proper set, then a subset and finally a group of cell death. By proposing this algorithm, the authors hope it may solve the problems regarding newer and/or undiscovered types of cell death and facilitate research and therapeutic applications of cell death.
Collapse
Affiliation(s)
- Sepehr Dehghan
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Nasim Kheshtchin
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Soleimani
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|
67
|
Ding R, Li Y, Yu Y, Sun Z, Duan J. Prospects and hazards of silica nanoparticles: Biological impacts and implicated mechanisms. Biotechnol Adv 2023; 69:108277. [PMID: 37923235 DOI: 10.1016/j.biotechadv.2023.108277] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
With the thrive of nanotechnology, silica nanoparticles (SiNPs) have been extensively adopted in the agriculture, food, cosmetic, and even biomedical industries. Due to the mass production and use, SiNPs inevitably entered the environment, resulting in ecological toxicity and even posing a threat to human health. Although considerable investigations have been conducted to assess the toxicity of SiNPs, the correlation between SiNPs exposure and consequent health risks remains ambiguous. Since the biological impacts of SiNPs can differ from their design and application, the toxicity assessment for SiNPs may be extremely difficult. This review discussed the application of SiNPs in different fields, especially their biomedical use, and documented their potential release pathways into the environment. Meanwhile, the current process of assessing SiNPs-related toxicity on various model organisms and cell lines was also detailed, thus estimating the health threats posed by SiNPs exposure. Finally, the potential toxic mechanisms of SiNPs were also elaborated based on results obtained from both in vivo and in vitro trials. This review generally summarizes the biological effects of SiNPs, which will build up a comprehensive perspective of the application and toxicity of SiNPs.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
68
|
Toscano F, Torres-Arias M. Nanoparticles cellular uptake, trafficking, activation, toxicity and in vitro evaluation. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100073. [PMID: 38020531 PMCID: PMC10663637 DOI: 10.1016/j.crimmu.2023.100073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 12/01/2023] Open
Abstract
Nanoparticles (NPs) physicochemical properties, such as size, shape, surface chemistry, charge, etc., play a critical role in biological systems interactions, which include NPs' cellular uptake, trafficking, activation, and toxicity. Although nano-bio interactions are multifaceted and complex, their assessment is essential for future therapeutic and diagnostic use since being carriers that deliver specific molecules (i.e., active pharmaceutical ingredients and imaging agents) in intracellular sites. The journey of NPs begins by reaching the plasma membrane and entering the cell mainly through endocytosis. After vesicles pinch off the cell membrane, the intracellular trafficking is mediated by a network of cellular endosomes which direct NPs to the different cellular components. Otherwise, NPs or their contents are released into the cytoplasm. In both cases, NPs can pass undetected or be recognized by the cell leading to a pro or anti-inflammatory response. Indeed, the cell response mostly depends on cell type and NPs physicochemical properties. The principal mechanism by which NPs activate the cell response is RONS production. Other mechanism includes signaling pathways modulation related to metabolic and enzymatic reactions, cell transduction, and immune modulation. Hence, the underlying mechanisms of cellular and subcellular interactions in vitro should be performed to provide insights into NPs' effect. This information helps us to improve their synthesis and design to maximize the clinical benefits while minimizing side effects. Most in vitro tests to evaluate NPs' effect in cells were developed focusing on cell dysfunctions, cytotoxicity, genotoxicity, immunogenicity, and cell death.
Collapse
Affiliation(s)
- Fernanda Toscano
- Departamento de Ciencias de la Vida y la Agricultura, Carrera de Ingeniería en Biotecnología, Laboratorio de Inmunología y Virología, GISAH, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
| | - Marbel Torres-Arias
- Departamento de Ciencias de la Vida y la Agricultura, Carrera de Ingeniería en Biotecnología, Laboratorio de Inmunología y Virología, GISAH, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
- Centro de Nanociencia y Nanotecnología, Universidad de las Fuerzas Armadas ESPE, Av. Gral. Rumiñahui s/n, P.O. Box 171-5-231B, Sangolquí, Ecuador
| |
Collapse
|
69
|
Golchin A, Maleki M, Alemi F, Malakoti F, Yousefi B. Autophagy-targeted nanoparticles in breast carcinoma: A systematic review. Cell Biol Int 2023; 47:1767-1781. [PMID: 37671447 DOI: 10.1002/cbin.12081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/10/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023]
Abstract
Breast cancer is a commonly known cancer type and the leading cause of cancer death among females. One of the unresolved problems in cancer treatment is the increased resistance of the tumor to existing treatments, which is a direct result of apoptotic defects. Calculating an alternative to cell death (autophagy) may be the ultimate solution to maximizing cancer cell death. Our aim in this study was to investigate the potential of free nanoparticles (un-drug-loaded) in the induction or inhibition of autophagy and consider this effect on the therapy process. When the studies met the inclusion criteria, the full texts of all relevant articles were carefully examined and classified. Of the 25 articles included in the analysis, carried out on MCF-7, MDA-MB-231, MDA-MB-231-TXSA, MDA-MB-468, SUM1315, and 4T1 cell lines. Twenty in vitro studies and five in vivo/in vitro studies applied five different autophagy tests: Acridine orange, western blot, Cyto-ID Autophagy Detection Kit, confocal microscope, and quantitative polymerase chain reaction. Nanoparticles (NPs) in the basic format, including Ag, Au, Y2 O3 , Se, ZnO, CuO, Al, Fe, vanadium pentoxide, and liposomes, were prepared in the included articles. Three behaviors of NPs related to autophagy were seen: induction, inhibition, and no action. Screened and presented data suggest that most of the involved free NPs (metallic NPs) in this systematic review had reactive oxygen species-mediated pathways with autophagy induction (36%). Also, PI3K/Akt/mTOR and MAPK/ERK signaling pathways were mentioned in just four studies (16%). An impressive percentage of studies (31%) did not examine the NP-related autophagy pathway.
Collapse
Affiliation(s)
- Asal Golchin
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoumeh Maleki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
70
|
Vanharen M, Mahbeer T, Léveillé A, Méthot A, Samountry P, Girard D. Impact of gold nanoparticles (AuNPs) in human neutrophils in vitro and in leukocytes attraction in vivo: A sex-based analysis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 104:104319. [PMID: 37984677 DOI: 10.1016/j.etap.2023.104319] [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: 06/07/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Some differences exist between the male and female immune systems. Despite this, a sex-based analysis is not frequently performed in most studies. Knowing that inflammation is a common undesired effect observed resulting from nanoparticle (NP) exposure, we investigate here how gold NPs with a primary size of 20 (AuNP20) and 70 nm (AuNP70) will alter the biology of polymorphonuclear neutrophil cells (PMNs) isolated from men and women as well as their potential pro-inflammatory effect in vivo in male and female mice. We found that AuNP20 significantly delay apoptosis only in PMN isolated from men. The production of interleukin (IL)- 8 by PMNs was increased by both AuNPs regardless of sex although significance was only observed in AuNP20-induced PMNs. Using the murine air pouch model of inflammation, AuNPs did not induce a neutrophilic infiltration regardless of sex. In conclusion, AuNPs could differently alter the biology of PMNs according to sex.
Collapse
Affiliation(s)
- Marion Vanharen
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Thomas Mahbeer
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Alexanne Léveillé
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Audrey Méthot
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Phonsiri Samountry
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Denis Girard
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada.
| |
Collapse
|
71
|
Xu X, Xu S, Wan J, Wang D, Pang X, Gao Y, Ni N, Chen D, Sun X. Disturbing cytoskeleton by engineered nanomaterials for enhanced cancer therapeutics. Bioact Mater 2023; 29:50-71. [PMID: 37621771 PMCID: PMC10444958 DOI: 10.1016/j.bioactmat.2023.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 08/26/2023] Open
Abstract
Cytoskeleton plays a significant role in the shape change, migration, movement, adhesion, cytokinesis, and phagocytosis of tumor cells. In clinical practice, some anti-cancer drugs achieve cytoskeletal therapeutic effects by acting on different cytoskeletal protein components. However, in the absence of cell-specific targeting, unnecessary cytoskeletal recombination in organisms would be disastrous, which would also bring about severe side effects during anticancer process. Nanomedicine have been proven to be superior to some small molecule drugs in cancer treatment due to better stability and targeting, and lower side effects. Therefore, this review summarized the recent developments of various nanomaterials disturbing cytoskeleton for enhanced cancer therapeutics, including carbon, noble metals, metal oxides, black phosphorus, calcium, silicon, polymers, peptides, and metal-organic frameworks, etc. A comprehensive analysis of the characteristics of cytoskeleton therapy as well as the future prospects and challenges towards clinical application were also discussed. We aim to drive on this emerging topic through refreshing perspectives based on our own work and what we have also learnt from others. This review will help researchers quickly understand relevant cytoskeletal therapeutic information to further advance the development of cancer nanomedicine.
Collapse
Affiliation(s)
- Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shanbin Xu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jipeng Wan
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Diqing Wang
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Xinlong Pang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yuan Gao
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore
| | - Dawei Chen
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xiao Sun
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| |
Collapse
|
72
|
He ZH, Zou JT, Chen X, Gong JS, Chen Y, Jin L, Liu YW, Rao SS, Yin H, Tan YJ, Wang Z, Du W, Li HM, Qian YX, Wang ZX, Wang YY, Wan TF, Luo Y, Zhu H, Chen CY, Xie H. Ångstrom-scale silver particles ameliorate collagen-induced and K/BxN-transfer arthritis in mice via the suppression of inflammation and osteoclastogenesis. Inflamm Res 2023; 72:2053-2072. [PMID: 37816881 DOI: 10.1007/s00011-023-01778-0] [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: 05/04/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 10/12/2023] Open
Abstract
OBJECTIVE Nanoparticles (NPs) hold a great promise in combating rheumatoid arthritis, but are often compromised by their toxicities because the currently used NPs are usually synthesized by chemical methods. Our group has previously fabricated Ångstrom-scale silver particles (AgÅPs) and demonstrated the anti-tumor and anti-sepsis efficacy of fructose-coated AgÅPs (F-AgÅPs). This study aimed to uncover the efficacy and mechanisms of F-AgÅPs for arthritis therapy. METHODS We evaluated the efficacy of F-AgÅPs in collagen-induced arthritis (CIA) mice. We also compared the capacities of F-AgÅPs, the commercial AgNPs, and the clinical drug methotrexate (MTX) in protecting against K/BxN serum-transfer arthritis (STA) mice. Moreover, we evaluated the effects of F-AgÅPs and AgNPs on inflammation, osteoclast formation, synoviocytes migration, and matrix metalloproteinases (MMPs) production in vitro and in vivo. Meanwhile, the toxicities of F-AgÅPs and AgNPs in vitro and in vivo were also tested. RESULTS F-AgÅPs significantly prevented bone erosion, synovitis, and cartilage damage, attenuated rheumatic pain, and improved the impaired motor function in mouse models of CIA or STA, the anti-rheumatic effects of which were comparable or stronger than AgNPs and MTX. Further studies revealed that F-AgÅPs exhibited similar or greater inhibitory abilities than AgNPs to suppress inflammation, osteoclast formation, synoviocytes migration, and MMPs production. No obvious toxicities were observed in vitro and in vivo after F-AgÅPs treatment. CONCLUSIONS F-AgÅPs can effectively alleviate arthritis without notable toxicities and their anti-arthritic effects are associated with the inhibition of inflammation, osteoclastogenesis, synoviocytes migration, and MMPs production. Our study suggests the prospect of F-AgÅPs as an efficient and low-toxicity agent for arthritis therapy.
Collapse
Affiliation(s)
- Ze-Hui He
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Jing-Tao Zou
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Xia Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiang-Shan Gong
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Ya Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Ling Jin
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yi-Wei Liu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Shan-Shan Rao
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Yin
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yi-Juan Tan
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Zun Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Nursing, Central South University, Changsha, Hunan, China
| | - Wei Du
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hong-Ming Li
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yu-Xuan Qian
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Zhen-Xing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Yi Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Teng-Fei Wan
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Yi Luo
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Hao Zhu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China
- Angmedicine Research Center, Central south university, Changsha, China
| | - Chun-Yuan Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China.
- Angmedicine Research Center, Central south university, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, China.
- Angmedicine Research Center, Central south university, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
73
|
Lin YW, Lin TT, Chen CH, Wang RH, Lin YH, Tseng TY, Zhuang YJ, Tang SY, Lin YC, Pang JY, Chakravarthy RD, Lin HC, Tzou SC, Chao JI. Enhancing Efficacy of Albumin-Bound Paclitaxel for Human Lung and Colorectal Cancers through Autophagy Receptor Sequestosome 1 (SQSTM1)/p62-Mediated Nanodrug Delivery and Cancer therapy. ACS NANO 2023; 17:19033-19051. [PMID: 37737568 DOI: 10.1021/acsnano.3c04739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Selective autophagy is a defense mechanism by which foreign pathogens and abnormal substances are processed to maintain cellular homeostasis. Sequestosome 1 (SQSTM1)/p62, a vital selective autophagy receptor, recruits ubiquitinated cargo to form autophagosomes for lysosomal degradation. Nab-PTX is an albumin-bound paclitaxel nanoparticle used in clinical cancer therapy. However, the role of SQSTM1 in regulating the delivery and efficacy of nanodrugs remains unclear. Here we showed that SQSTM1 plays a crucial role in Nab-PTX drug delivery and efficacy in human lung and colorectal cancers. Nab-PTX induces SQSTM1 phosphorylation at Ser403, which facilitates its incorporation into the selective autophagy of nanoparticles, known as nanoparticulophagy. Nab-PTX increased LC3-II protein expression, which triggered autophagosome formation. SQSTM1 enhanced Nab-PTX recognition to form autophagosomes, which were delivered to lysosomes for albumin degradation, thereby releasing PTX to induce mitotic catastrophe and apoptosis. Knockout of SQSTM1 downregulated Nab-PTX-induced mitotic catastrophe, apoptosis, and tumor inhibition in vitro and in vivo and inhibited Nab-PTX-induced caspase 3 activation via a p53-independent pathway. Ectopic expression of SQSTM1 by transfection of an SQSTM1-GFP vector restored the drug efficacy of Nab-PTX. Importantly, SQSTM1 is highly expressed in advanced lung and colorectal tumors and is associated with poor overall survival in clinical patients. Targeting SQSTM1 may provide an important strategy to improve nanodrug efficacy in clinical cancer therapy. This study demonstrates the enhanced efficacy of Nab-PTX for human lung and colorectal cancers via SQSTM1-mediated nanodrug delivery.
Collapse
Affiliation(s)
- Yu-Wei Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Tzu-Ting Lin
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Chien-Hung Chen
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Rou-Hsin Wang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Ya-Hui Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Tzu-Yen Tseng
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Yan-Jun Zhuang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Sheng-Yueh Tang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Yen-Cheng Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Jiun-Yu Pang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Rajan Deepan Chakravarthy
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Shey-Cherng Tzou
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Jui-I Chao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center For Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| |
Collapse
|
74
|
Mehta P, Shende P. Dual role of autophagy for advancements from conventional to new delivery systems in cancer. Biochim Biophys Acta Gen Subj 2023; 1867:130430. [PMID: 37506854 DOI: 10.1016/j.bbagen.2023.130430] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Autophagy, a programmed cell-lysis mechanism, holds significant promise in the prevention and treatment of a wide range of conditions, including cancer, Alzheimer's, and Parkinson's disease. The successful utilization of autophagy modulation for therapeutic purposes hinges upon accurately determining the role of autophagy in disease progression, whether it acts as a cytotoxic or cytoprotective factor. This critical knowledge empowers scientists to effectively manipulate tumor sensitivity to anti-cancer therapies through autophagy modulation, while also circumventing drug resistance. However, conventional therapies face limitations such as low bioavailability, poor solubility, and a lack of controlled release mechanisms, hindering their clinical applicability. In this regard, innovative nanoplatforms including organic and inorganic systems have emerged as promising solutions to offer stimuli-responsive, theranostic-controlled drug delivery systems with active targeting and improved solubility. The review article explores a variety of organic nanoplatforms, such as lipid-based, polymer-based, and DNA-based systems, which incorporate autophagy-inhibiting drugs like hydroxychloroquine. By inhibiting the glycolytic pathway and depriving cells of essential nutrients, these platforms exhibit tumor-suppressive effects in advanced forms of cancer such as leukemia, colon cancer, and glioblastoma. Furthermore, metal-based, metal-oxide-based, silica-based, and quantum dot-based nanoplatforms selectively induce autophagy in tumors, leading to extensive cancer cell destruction. Additionally, this article discusses the current clinical status of autophagy-modulating drugs for cancer therapy with valuable insights of progress and potential of such approaches.
Collapse
Affiliation(s)
- Parth Mehta
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, India.
| |
Collapse
|
75
|
Arnst J, Jing Z, Cohen C, Ha SW, Viggeswarapu M, Beck GR. Bioactive silica nanoparticles target autophagy, NF-κB, and MAPK pathways to inhibit osteoclastogenesis. Biomaterials 2023; 301:122238. [PMID: 37441901 PMCID: PMC10530178 DOI: 10.1016/j.biomaterials.2023.122238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023]
Abstract
Spherical 50 nm silica-based nanoparticles (SiNPs) promote healthy bone homeostasis and maintenance by supporting bone forming osteoblast lineage cells while simultaneously inhibiting the differentiation of bone resorbing osteoclasts. Previous work demonstrated that an intraperitoneal injection of SiNPs in healthy mice - both young and old - increased bone density and quality, suggesting the possibility that SiNPs represent a dual action therapeutic. However, the underlying mechanisms governing the osteoclast response to SiNPs have yet to be fully explored and defined. Therefore, the goals of this study were to investigate the cellular and molecular mechanisms by which SiNPs inhibit osteoclastogenesis. SiNPs strongly inhibited RANKL-induced osteoclast differentiation within the first hours and concomitantly inhibited early transcriptional regulators such as Nfatc1. SiNPs simultaneously stimulated expression of autophagy related genes p62 and LC3β dependent on ERK1/2 signaling pathway. Intriguingly, SiNPs were found to stimulate autophagosome formation while inhibiting the autophagic flux necessary for RANKL-stimulated osteoclast differentiation, resulting in the inhibition of both the canonical and non-canonical NF-κB signaling pathways and stabilizing TRAF3. These results suggest a model in which SiNPs inhibit osteoclastogenesis by inhibiting the autophagic machinery and RANKL-dependent functionality. This mechanism of action defines a novel therapeutic strategy for inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Jamie Arnst
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Zhaocheng Jing
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA; The Second Hospital of Shandong University, Department of Orthopedics, Jinan, Shandong, 250033, China
| | - Cameron Cohen
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Shin-Woo Ha
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Manjula Viggeswarapu
- The Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - George R Beck
- The Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, 30033, USA; Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA; The Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
76
|
Wu Y, Li M, Chen L, Xu L, Xu Y, Zhong Y. Utilizing a Combination of Network Pharmacology and Experimental Validation to Unravel the Mechanism by Which Kuanxiongzhuyu Decoction Ameliorates Myocardial Infarction Damage. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1740. [PMID: 37893458 PMCID: PMC10608708 DOI: 10.3390/medicina59101740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: With the growing incidence and disability associated with myocardial infarction (MI), there is an increasing focus on cardiac rehabilitation post-MI. Kuanxiongzhuyu decoction (KXZY), a traditional Chinese herbal formula, has been used in the rehabilitation of patients after MI. However, the chemical composition, protective effects, and underlying mechanism of KXZY remain unclear. Materials and Methods: In this study, the compounds in KXZY were identified using a high-performance liquid chromatography-mass spectrometry (HPLC-MS) analytical method. Based on the compounds identified in the KXZY, we predictively selected the potential targets of MI and then constructed a protein-protein interaction (PPI) network to identify the key targets. Furthermore, the DAVID database was used for the GO and KEGG analyses, and molecular docking was used to verify the key targets. Finally, the cardioprotective effects and mechanism of KXZY were investigated in post-MI mice. Results: A total of 193 chemical compounds of KXZY were identified by HPLC-MS. In total, 228 potential targets were obtained by the prediction analysis. The functional enrichment studies and PPI network showed that the targets were largely associated with AKT-pathway-related apoptosis. The molecular docking verified that isoguanosine and adenosine exhibited excellent binding to the AKT. In vivo, KXZY significantly alleviated cardiac dysfunction and suppressed AKT phosphorylation. Furthermore, KXZY significantly increased the expression of the antiapoptotic proteins Bcl-2 and Bcl-xl and decreased the expression of the proapoptotic protein BAD. Conclusions: In conclusion, the network pharmacological and experimental evidence suggests that KXZY manifests anti-cardiac dysfunction behavior by alleviating cardiomyocyte apoptosis via the AKT pathway in MI and, thus, holds promising therapeutic potential.
Collapse
Affiliation(s)
- Yihao Wu
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (Y.W.); (M.L.); (L.C.); (L.X.)
| | - Miaofu Li
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (Y.W.); (M.L.); (L.C.); (L.X.)
| | - Liuying Chen
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (Y.W.); (M.L.); (L.C.); (L.X.)
| | - Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (Y.W.); (M.L.); (L.C.); (L.X.)
- Translational Medicine Research Center, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (Y.W.); (M.L.); (L.C.); (L.X.)
| | - Yigang Zhong
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; (Y.W.); (M.L.); (L.C.); (L.X.)
| |
Collapse
|
77
|
Veranič P, Križaj I. Interaction of Nanomaterials with Cells and Tissues. Int J Mol Sci 2023; 24:13667. [PMID: 37686473 PMCID: PMC10488087 DOI: 10.3390/ijms241713667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Nanomaterials have gained enormous importance in biomedicine in recent years, both in basic and applied sciences [...].
Collapse
Affiliation(s)
- Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
78
|
Abdelkader Y, Perez-Davalos L, LeDuc R, Zahedi RP, Labouta HI. Omics approaches for the assessment of biological responses to nanoparticles. Adv Drug Deliv Rev 2023; 200:114992. [PMID: 37414362 DOI: 10.1016/j.addr.2023.114992] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/08/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Nanotechnology has enabled the development of innovative therapeutics, diagnostics, and drug delivery systems. Nanoparticles (NPs) can influence gene expression, protein synthesis, cell cycle, metabolism, and other subcellular processes. While conventional methods have limitations in characterizing responses to NPs, omics approaches can analyze complete sets of molecular entities that change upon exposure to NPs. This review discusses key omics approaches, namely transcriptomics, proteomics, metabolomics, lipidomics and multi-omics, applied to the assessment of biological responses to NPs. Fundamental concepts and analytical methods used for each approach are presented, as well as good practices for omics experiments. Bioinformatics tools are essential to analyze, interpret and visualize large omics data, and to correlate observations in different molecular layers. The authors envision that conducting interdisciplinary multi-omics analyses in future nanomedicine studies will reveal integrated cell responses to NPs at different omics levels, and the incorporation of omics into the evaluation of targeted delivery, efficacy, and safety will improve the development of nanomedicine therapies.
Collapse
Affiliation(s)
- Yasmin Abdelkader
- Unity Health Toronto - St. Michael's Hospital, University of Toronto, 209 Victoria St., Toronto, Ontario M5B 1T8, Canada; College of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Av. W, Winnipeg, Manitoba R3E 0T5, Canada; Department of Cell Biology, Biotechnology Research Institute, National Research Centre, 33 El Buhouth St., Cairo 12622, Egypt
| | - Luis Perez-Davalos
- Unity Health Toronto - St. Michael's Hospital, University of Toronto, 209 Victoria St., Toronto, Ontario M5B 1T8, Canada; College of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Av. W, Winnipeg, Manitoba R3E 0T5, Canada
| | - Richard LeDuc
- Children's Hospital Research Institute of Manitoba, 513 - 715 McDermot Av. W, Winnipeg, Manitoba R3E 3P4, Canada; Department of Biochemistry and Medical Genetics, University of Manitoba, 745 Bannatyne Av., Winnipeg, Manitoba R3E 0J9, Canada
| | - Rene P Zahedi
- Department of Biochemistry and Medical Genetics, University of Manitoba, 745 Bannatyne Av., Winnipeg, Manitoba R3E 0J9, Canada; Department of Internal Medicine, 715 McDermot Av., Winnipeg, Manitoba R3E 3P4, Canada; Manitoba Centre for Proteomics and Systems Biology, 799 JBRC, 715 McDermot Av., Winnipeg, Manitoba R3E 3P4, Canada; CancerCare Manitoba Research Institute, 675 McDermot Av., Manitoba R3E 0V9, Canada
| | - Hagar I Labouta
- Unity Health Toronto - St. Michael's Hospital, University of Toronto, 209 Victoria St., Toronto, Ontario M5B 1T8, Canada; College of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Av. W, Winnipeg, Manitoba R3E 0T5, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada; Faculty of Pharmacy, Alexandria University, 1 Khartoum Square, Azarita, Alexandria, Egypt, 21521.
| |
Collapse
|
79
|
Domingo-Diez J, Souiade L, Manzaneda-González V, Sánchez-Díez M, Megias D, Guerrero-Martínez A, Ramírez-Castillejo C, Serrano-Olmedo J, Ramos-Gómez M. Effectiveness of Gold Nanorods of Different Sizes in Photothermal Therapy to Eliminate Melanoma and Glioblastoma Cells. Int J Mol Sci 2023; 24:13306. [PMID: 37686114 PMCID: PMC10488215 DOI: 10.3390/ijms241713306] [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: 08/03/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Gold nanorods are the most commonly used nanoparticles in photothermal therapy for cancer treatment due to their high efficiency in converting light into heat. This study aimed to investigate the efficacy of gold nanorods of different sizes (large and small) in eliminating two types of cancer cell: melanoma and glioblastoma cells. After establishing the optimal concentration of nanoparticles and determining the appropriate time and power of laser irradiation, photothermal therapy was applied to melanoma and glioblastoma cells, resulting in the highly efficient elimination of both cell types. The efficiency of the PTT was evaluated using several methods, including biochemical analysis, fluorescence microscopy, and flow cytometry. The dehydrogenase activity, as well as calcein-propidium iodide and Annexin V staining, were employed to determine the cell viability and the type of cell death triggered by the PTT. The melanoma cells exhibited greater resistance to photothermal therapy, but this resistance was overcome by irradiating cells at physiological temperatures. Our findings revealed that the predominant cell-death pathway activated by the photothermal therapy mediated by gold nanorods was apoptosis. This is advantageous as the presence of apoptotic cells can stimulate antitumoral immunity in vivo. Considering the high efficacy of these gold nanorods in photothermal therapy, large nanoparticles could be useful for biofunctionalization purposes. Large nanorods offer a greater surface area for attaching biomolecules, thereby promoting high sensitivity and specificity in recognizing target cancer cells. Additionally, large nanoparticles could also be beneficial for theranostic applications, involving both therapy and diagnosis, due to their superior detection sensitivity.
Collapse
Affiliation(s)
- Javier Domingo-Diez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
| | - Lilia Souiade
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
| | - Vanesa Manzaneda-González
- Departamento de Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain (A.G.-M.)
| | - Marta Sánchez-Díez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Grupo de Sistemas Complejos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Diego Megias
- Advanced Optical Microscopy Unit, UCCTs, Instituto de Salud Carlos III (ISCIII), 28222 Madrid, Spain
| | - Andrés Guerrero-Martínez
- Departamento de Química Física, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain (A.G.-M.)
| | - Carmen Ramírez-Castillejo
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Grupo de Sistemas Complejos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Departamento Biotecnología-B.V. ETSIAAB, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Departamento de Oncología, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Javier Serrano-Olmedo
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Centro de Investigación Biomédica en Red para Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Milagros Ramos-Gómez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (J.D.-D.); (M.S.-D.); (C.R.-C.)
- Centro de Investigación Biomédica en Red para Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Experimental Neurology Unit, Center for Biomedical Technology, Universidad Politécnica de Madrid, Campus de Montegancedo s/n, Pozuelo de Alarcón, 28223 Madrid, Spain
| |
Collapse
|
80
|
Mozafari N, Mozafari N, Dehshahri A, Azadi A. Knowledge Gaps in Generating Cell-Based Drug Delivery Systems and a Possible Meeting with Artificial Intelligence. Mol Pharm 2023; 20:3757-3778. [PMID: 37428824 DOI: 10.1021/acs.molpharmaceut.3c00162] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Cell-based drug delivery systems are new strategies in targeted delivery in which cells or cell-membrane-derived systems are used as carriers and release their cargo in a controlled manner. Recently, great attention has been directed to cells as carrier systems for treating several diseases. There are various challenges in the development of cell-based drug delivery systems. The prediction of the properties of these platforms is a prerequisite step in their development to reduce undesirable effects. Integrating nanotechnology and artificial intelligence leads to more innovative technologies. Artificial intelligence quickly mines data and makes decisions more quickly and accurately. Machine learning as a subset of the broader artificial intelligence has been used in nanomedicine to design safer nanomaterials. Here, how challenges of developing cell-based drug delivery systems can be solved with potential predictive models of artificial intelligence and machine learning is portrayed. The most famous cell-based drug delivery systems and their challenges are described. Last but not least, artificial intelligence and most of its types used in nanomedicine are highlighted. The present Review has shown the challenges of developing cells or their derivatives as carriers and how they can be used with potential predictive models of artificial intelligence and machine learning.
Collapse
Affiliation(s)
- Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Niloofar Mozafari
- Design and System Operations Department, Regional Information Center for Science and Technology, 71946 94171 Shiraz, Iran
| | - Ali Dehshahri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
- Pharmaceutical Sciences Research Centre, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
- Pharmaceutical Sciences Research Centre, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| |
Collapse
|
81
|
Yuan J, Mo Y, Zhang Y, Zhang Y, Zhang Q. Nickel nanoparticles induce autophagy and apoptosis via HIF-1α/mTOR signaling in human bronchial epithelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121670. [PMID: 37080518 PMCID: PMC10231338 DOI: 10.1016/j.envpol.2023.121670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
With the rapid development of nanotechnology, the potential adverse health effects of nanoparticles have been caught more attention and become global concerns. However, the underlying mechanisms in metal nanoparticle-induced toxic effects are still largely obscure. In this study, we investigated whether exposure to nickel nanoparticles (Nano-Ni) and titanium dioxide nanoparticles (Nano-TiO2) would alter autophagy and apoptosis levels in normal human bronchial epithelial BEAS-2B cells and the underlying mechanisms involved in this process. Our results showed that the expressions of autophagy- and apoptosis-associated proteins were dysregulated in cells exposed to Nano-Ni. However, exposure to the same doses of Nano-TiO2 had no significant effects on these proteins. In addition, exposure to Nano-Ni, but not Nano-TiO2, led to nuclear accumulation of HIF-1α and decreased phosphorylation of mTOR in BEAS-2B cells. Inhibition of HIF-1α by CAY10585 abolished Nano-Ni-induced decreased phosphorylation of mTOR, while activation of mTOR by MHY1485 did not affect Nano-Ni-induced nuclear accumulation of HIF-1α. Furthermore, both HIF-1α inhibition and mTOR activation abolished Nano-Ni-induced autophagy but enhanced Nano-Ni-induced apoptosis. Blockage of autophagic flux by Bafilomycin A1 exacerbated Nano-Ni-induced apoptosis, while activation of autophagy by Rapamycin effectively rescued Nano-Ni-induced apoptosis. In conclusion, our results demonstrated that Nano-Ni exposure caused increased levels of autophagy and apoptosis via the HIF-1α/mTOR signaling axis. Nano-Ni-induced autophagy has a protective role against Nano-Ni-induced apoptosis. These findings provide us with further insight into Nano-Ni-induced toxicity.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yue Zhang
- Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
82
|
Jain A, Bhattacharya S. Recent advances in nanomedicine preparative methods and their therapeutic potential for colorectal cancer: a critical review. Front Oncol 2023; 13:1211603. [PMID: 37427139 PMCID: PMC10325729 DOI: 10.3389/fonc.2023.1211603] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy that affects a large percentage of the global population. The conventional treatments for CRC have a number of limitations. Nanoparticles have emerged as a promising cancer treatment method due to their ability to directly target cancer cells and regulate drug release, thereby enhancing therapeutic efficacy and minimizing side effects. This compilation examines the use of nanoparticles as drug delivery systems for CRC treatment. Different nanomaterials can be used to administer anticancer drugs, including polymeric nanoparticles, gold nanoparticles, liposomes, and solid lipid nanoparticles. In addition, we discuss recent developments in nanoparticle preparation techniques, such as solvent evaporation, salting-out, ion gelation, and nanoprecipitation. These methods have demonstrated high efficacy in penetrating epithelial cells, a prerequisite for effective drug delivery. This article focuses on the various targeting mechanisms utilized by CRC-targeted nanoparticles and their recent advancements in this field. In addition, the review offers descriptive information regarding numerous nano-preparative procedures for colorectal cancer treatments. We also discuss the outlook for innovative therapeutic techniques in the management of CRC, including the potential application of nanoparticles for targeted drug delivery. The review concludes with a discussion of current nanotechnology patents and clinical studies used to target and diagnose CRC. The results of this investigation suggest that nanoparticles have great potential as a method of drug delivery for the treatment of colorectal cancer.
Collapse
|
83
|
Çiçek S. α-tocopherol ameliorates copper II oxide nanoparticles-induced cytotoxic, biochemical, apoptotic, and genotoxic damages in the rainbow trout gonad cells-2 (RTG-2) culture. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 101:104168. [PMID: 37295739 DOI: 10.1016/j.etap.2023.104168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023]
Abstract
We investigated the effects of α-tocopherol on oxidative stress-caused damage caused by copper II oxide nanoparticles (CuO NPs) on Oncorhynchus mykiss gonadal cells (RTG-2) for 24 and 48 h. α-Tocopherol reversed the cell death and alterations in the expressions of genes such as sod1, gpx1a, gpx4b, and igf2 caused by CuO NPs; it also supported the expressions of cat, igf1, and gapdh genes caused by CuO NPs for 24 h and promoted alterations in the expressions of the sod2, gh1, and igf1 genes for 48 h. Additionally, α-tocopherol reversed the caspase 3/7 activity increased by CuO NPs for 24 h and supported it's decrease for 48 h. α-Tocopherol supported the increase in tail DNA (%) affected by CuO NPs for 24 h and reversed it for 48 h. Therefore, α-tocopherol may have the potential to protect against cellular alterations induced by CuO NPs in a time-dependent manner.
Collapse
Affiliation(s)
- Semra Çiçek
- Department of Animal Biotechnology, Faculty of Agriculture, Atatürk University, Erzurum 25240, Turkey.
| |
Collapse
|
84
|
Rodolpho JMDA, Godoy KFD, Brassolatti P, Fragelli BDDL, Camillo L, Castro CAD, Assis M, Speglich C, Longo E, Anibal FDF. Carbon Black CB-EDA Nanoparticles in Macrophages: Changes in the Oxidative Stress Pathway and in Apoptosis Signaling. Biomedicines 2023; 11:1643. [PMID: 37371738 DOI: 10.3390/biomedicines11061643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 06/29/2023] Open
Abstract
The influence of black carbon nanoparticles on J774.A1 murine cells was investigated with the objective of exploring the cytotoxicity of black carbon functionalized with ethylenediamine CB-EDA. The results showed that CB-EDA has a cytotoxic profile for J774.A1 macrophages in a time- and dose-dependent manner. When phagocytosed by the macrophage, CB-EDA triggers a mechanism that leads to apoptosis. In this process, there is an increase in oxidative stress pathways due to the activation of nitric oxide and then ROS. This causes an imbalance in redox function and a disruption of membrane integrity that occurs due to high levels of LDH, in addition to favoring the release of the pro-inflammatory cytokines IL-6, IL-12, and tumor necrosis factor (TNF) in an attempt to modulate the cell. However, these stimuli are not sufficient to repair the cell and the level of mitochondrial integrity is affected, causing a decrease in cell viability. This mechanism may be correlated with the activation of the caspasse-3 pathway, which, when compromised, cleaves and induces cells death via apoptosis, either through early or late apoptosis. In view of this, the potential for cell damage was investigated by analyzing the oxidative and inflammatory profile in the macrophage lineage J774.A1 and identifying potential mechanisms and metabolic pathways connected to these processes when cells were exposed to NP CB-EDA for both 24 h and 48 h.
Collapse
Affiliation(s)
- Joice Margareth de Almeida Rodolpho
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Krissia Franco de Godoy
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Patricia Brassolatti
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Bruna Dias de Lima Fragelli
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Luciana Camillo
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Cynthia Aparecida de Castro
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Marcelo Assis
- Department of Analytical and Physical Chemistry, University Jaume I (UJI), 12006 Castelló, Spain
- Centro de Desenvolvimento de Materiais Funcionais, Departamento de Química, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Carlos Speglich
- Centro de Pesquisa Leopoldo Américo Miguez de Mello CENPES/Petrobras, Rio de Janeiro 21941-915, Brazil
| | - Elson Longo
- Centro de Desenvolvimento de Materiais Funcionais, Departamento de Química, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Fernanda de Freitas Anibal
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| |
Collapse
|
85
|
Skvortsov AN, Ilyechova EY, Puchkova LV. Chemical background of silver nanoparticles interfering with mammalian copper metabolism. JOURNAL OF HAZARDOUS MATERIALS 2023; 451:131093. [PMID: 36905906 DOI: 10.1016/j.jhazmat.2023.131093] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
The rapidly increasing application of silver nanoparticles (AgNPs) boosts their release into the environment, which raises a reasonable alarm for ecologists and health specialists. This is manifested as increased research devoted to the influence of AgNPs on physiological and cellular processes in various model systems, including mammals. The topic of the present paper is the ability of silver to interfere with copper metabolism, the potential health effects of this interference, and the danger of low silver concentrations to humans. The chemical properties of ionic and nanoparticle silver, supporting the possibility of silver release by AgNPs in extracellular and intracellular compartments of mammals, are discussed. The possibility of justified use of silver for the treatment of some severe diseases, including tumors and viral infections, based on the specific molecular mechanisms of the decrease in copper status by silver ions released from AgNPs is also discussed.
Collapse
Affiliation(s)
- Alexey N Skvortsov
- Graduate School of Biomedical Systems and Technologies, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Saint Petersburg 195251, Russia; Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg 194064, Russia
| | - Ekaterina Yu Ilyechova
- Graduate School of Biomedical Systems and Technologies, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Saint Petersburg 195251, Russia; Department of Molecular Genetics, Institute of Experimental Medicine of the Russian Academy of Sciences, Saint Petersburg 197376, Russia; Research Center of Advanced Functional Materials and Laser Communication Systems (RC AFMLCS), ITMO University, Saint Petersburg 197101, Russia.
| | - Ludmila V Puchkova
- Graduate School of Biomedical Systems and Technologies, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Saint Petersburg 195251, Russia; Department of Molecular Genetics, Institute of Experimental Medicine of the Russian Academy of Sciences, Saint Petersburg 197376, Russia; Research Center of Advanced Functional Materials and Laser Communication Systems (RC AFMLCS), ITMO University, Saint Petersburg 197101, Russia
| |
Collapse
|
86
|
Zhang Y, Zhang Y, Lei Y, Wu J, Kang Y, Zheng S, Shao L. MDM2 upregulation induces mitophagy deficiency via Mic60 ubiquitination in fetal microglial inflammation and consequently neuronal DNA damage caused by exposure to ZnO-NPs during pregnancy. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131750. [PMID: 37315416 DOI: 10.1016/j.jhazmat.2023.131750] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/15/2023] [Accepted: 05/29/2023] [Indexed: 06/16/2023]
Abstract
During pregnancy, the human body is quite vulnerable to external stimuli. Zinc oxide nanoparticles (ZnO-NPs) are widely used in daily life, and they enter the human body via environmental or biomedical exposure, thus having potential risks. Although accumulating studies have demonstrated the toxic effects of ZnO-NPs, few studies have addressed the effect of prenatal ZnO-NP exposure on fetal brain tissue development. Here, we systematically studied ZnO-NP-induced fetal brain damage and the underlying mechanism. Using in vivo and in vitro assays, we found that ZnO-NPs could cross the underdeveloped bloodbrain barrier and enter fetal brain tissue, where they could be endocytosed by microglia. ZnO-NP exposure impaired mitochondrial function and induced autophagosome overaccumulation by downregulation of Mic60, thus inducing microglial inflammation. Mechanistically, ZnO-NPs increased Mic60 ubiquitination by activating MDM2, resulting in imbalanced mitochondrial homeostasis. Inhibition of Mic60 ubiquitination by MDM2 silencing significantly attenuated the mitochondrial damage induced by ZnO-NPs, thereby preventing autophagosome overaccumulation and reducing ZnO-NP-mediated inflammation and neuronal DNA damage. Our results demonstrate that ZnO-NPs are likely to disrupt mitochondrial homeostasis, inducing abnormal autophagic flux and microglial inflammation and secondary neuronal damage in the fetus. We hope the information provided in our study will improve the understanding of the effects of prenatal ZnO-NP exposure on fetal brain tissue development and draw more attention to the daily use of and therapeutic exposure to ZnO-NPs among pregnant women.
Collapse
Affiliation(s)
- Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China
| | - Yulin Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China
| | - Ye Lei
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing 100853 China
| | - Junrong Wu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China
| | - Yiyuan Kang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China
| | - Shuo Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China.
| |
Collapse
|
87
|
Alzahrani B, Elderdery AY, Alsrhani A, Alzerwi NAN, Althobiti MM, Elkhalifa AME, Rayzah M, Idrees B, Kumar SS, Mok PL. Sodium alginate encapsulated iron oxide decorated with thymoquinone nanocomposite induces apoptosis in human breast cancer cells via PI3K-Akt-mTOR pathway. Int J Biol Macromol 2023:125054. [PMID: 37245766 DOI: 10.1016/j.ijbiomac.2023.125054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 05/30/2023]
Abstract
The present study investigated the cytotoxicity and proapoptotic properties of iron oxide-sodium-alginate-thymoquinone nanocomposites against breast cancer MDA-MB-231 cells in vitro and in silico. This study used chemical synthesis to formulate the nanocomposite. Electron microscopies such as scanning (SEM) and transmission (TEM), Fourier transform infrared (FT-IR), Ultraviolet-Visible, Photoluminescence spectroscopy, selected area (electron) diffraction (SAED), energy dispersive X-ray analysis (EDX), and X-ray diffraction studies (XRD) were used to characterize the synthesized ISAT-NCs and the average size of them was found to be 55 nm. To evaluate the cytotoxic, antiproliferative, and apoptotic potentials of ISAT-NCs on MDA-MB-231 cells, MTT assays, FACS-based cell cycle studies, annexin-V-PI staining, ELISA, and qRT-PCR were used. PI3K-Akt-mTOR receptors and thymoquinone were predicted using in-silico docking studies. Cell proliferation is reduced in MDA-MB-231 cells due to ISAT-NC cytotoxicity. As a result of FACS analysis, ISAT-NCs had nuclear damage, ROS production, and elevated annexin-V levels, which resulted in cell cycle arrest in the S phase. The ISAT-NCs in MDA-MB-231 cells were found to downregulate PI3K-Akt-mTOR regulatory pathways in the presence of inhibitors of PI3K-Akt-mTOR, showing that these regulatory pathways are involved in apoptotic cell death. We also predicted the molecular interaction between thymoquinone and PI3K-Akt-mTOR receptor proteins using in-silico docking studies which also support PI3K-Akt-mTOR signaling inhibition by ISAT-NCs in MDA-MB-231 cells. As a result of this study, we can conclude that ISAT-NCs inhibit the PI3K-Akt-mTOR pathway in breast cancer cell lines, causing apoptotic cell death.
Collapse
Affiliation(s)
- Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Abozer Y Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia.
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Nasser A N Alzerwi
- Department of Surgery, College of Medicine, Majmaah University, P. O. Box 66, Al-Majmaah 11952, Ri-yadh, Saudi Arabia
| | - Maryam Musleh Althobiti
- Department of Clinical Laboratory Science, College of Applied Medical Science, Shaqra, Saudi Arabia
| | - Ahmed M E Elkhalifa
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Musaed Rayzah
- Department of Surgery, College of Medicine, Majmaah University, P. O. Box 66, Al-Majmaah 11952, Ri-yadh, Saudi Arabia
| | - Bandar Idrees
- Department of Surgery, Prince Sultan Military Medical City in Riyadh, Makkah Al Mukarramah Rd, As Sulimaniyah, Saudi Arabia
| | - Suresh S Kumar
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Pooi Ling Mok
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
88
|
Pu Y, Ke H, Wu C, Xu S, Xiao Y, Han L, Lyv G, Li S. Superparamagnetic iron oxide nanoparticles target BxPC-3 cells and silence MUC4 for theranostics of pancreatic cancer. Biochim Biophys Acta Gen Subj 2023:130383. [PMID: 37236323 DOI: 10.1016/j.bbagen.2023.130383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Abstract
PURPOSE Superparamagnetic iron oxide nanoparticles (SPION) are excellent magnetic resonance imaging (MRI) contrast agents. Mucin 4 (MUC4) acts as pancreatic cancer (PC) tumor antigen and influences PC progression. Small interfering RNAs (siRNAs) are used as a gene-silencing tool to treat a variety of diseases. METHODS We designed a therapeutic probe based on polyetherimide-superparamagnetic iron oxide nanoparticles (PEI-SPION) combined with siRNA nanoprobes (PEI-SPION-siRNA) to assess the contrast in MRI. The biocompatibility of the nanocomposite, and silencing of MUC4 were characterized and evaluated. RESULTS The prepared molecular probe had a particle size of 61.7 ± 18.5 nmand a surface of 46.7 ± 0.8mVand showed good biocompatibility in vitro and T2 relaxation efficiency. It can also load and protect siRNA. PEI-SPION-siRNA showed a good silencing effect on MUC4. CONCLUSION PEI-SPION-siRNA may be beneficial as a novel theranostic tool for PC.
Collapse
Affiliation(s)
- Yu Pu
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China; Department of Medical Imaging Key Laboratory of Sichuan Province, Affiliated Hospital of North Sichuan Medical College. No. 234, Fujiang Road, Shunqing District, Nanchong City 637000, People's Republic of China; Department of Medicine, Quanzhou Medical College, No. 2 Anji Road, Luojiang District, Quanzhou 362000, People's Republic of China
| | - Helin Ke
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Changqiang Wu
- Department of Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College. No. 55, Dongshun Road, Gaoping District, Nanchong City 637100, People's Republic of China
| | - Shaodan Xu
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Yang Xiao
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Lina Han
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China
| | - Guorong Lyv
- Department of Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College. No. 55, Dongshun Road, Gaoping District, Nanchong City 637100, People's Republic of China.
| | - Shilin Li
- Department of Ultrasonography, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, People's Republic of China.
| |
Collapse
|
89
|
Zheng G, Zhang J, Zhang X, Zhang Z, Liu S, Zhang S, Zhang C. Implications of ferroptosis in silver nanoparticle-induced cytotoxicity of macrophages. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 259:115057. [PMID: 37229872 DOI: 10.1016/j.ecoenv.2023.115057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 05/27/2023]
Abstract
Metal nanoparticles (NPs) are widely used in daily life and commercial activities owing to their unique physicochemical properties. Consequently, there is an increasing risk of daily and occupational exposure to metal NPs, which raises concerns regarding their health hazards. Programmed cell deaths (PCDs) have been clarified to be involved in metal NP-induced cytotoxicity, including apoptosis, autophagy, and pyroptosis. However, whether and how ferroptosis, a newly recognized PCD, contributes to metal NP-induced cell death remain unclear. In this study, we investigated the ferroptotic effects of two representative metal NPs, silver nanoparticles (AgNPs) and gold nanoparticles (AuNPs), on macrophages in vitro. Our results revealed that AgNPs, rather than AuNPs, induced non-apoptotic PCD, accompanied by lipid peroxidation and iron homeostasis disorders, which are two hallmarks of ferroptosis, in macrophages. Treatment with a ferroptosis inhibitor (ferrostatin-1) and iron chelator (deferoxamine) reversed AgNP-induced PCD, corroborating the induction of ferroptosis upon exposure to AgNPs. Moreover, our results revealed that smaller AgNPs elicited greater ferroptotic effects on macrophages than larger ones. Importantly, ferroptosis in AgNP-treated macrophages was mainly triggered by AgNPs per se rather than by Ag ions. Overall, our study highlights the ferroptotic effects elicited by AgNPs in macrophages, which will promote the understanding of their cytotoxic effects and facilitate the safer design of metal nanoproducts.
Collapse
Affiliation(s)
- Guangzhe Zheng
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xuan Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Zhihong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Sijin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Changwen Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
90
|
Wu J, Huang X, Xiao Z, Wang Q, Mu L, Yang S, Miao S, Chen J, Deng X, Deng C, Li H. Nano-Pt induced mitochondria-dependent apoptosis and cytoprotective autophagy in human NSCLC cells. Colloids Surf B Biointerfaces 2023; 227:113344. [PMID: 37257302 DOI: 10.1016/j.colsurfb.2023.113344] [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] [Received: 02/01/2023] [Revised: 05/01/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023]
Abstract
Given that currently used classical chemotherapeutic drugs lack the ideal therapeutic effect and produce severe side effects, platinum nanomaterials (Pt-NMs) have gradually gained attention, and their antitumor effect has been initially explored. However, the specific mechanisms underlying the action of Pt-NMs in non-small cell lung cancer (NSCLC) cells remain unclear. Moreover, the interaction between Pt-NMs and autophagy in inducing apoptosis of NSCLC cells remains unexplored. In this study, we explored the anti-NSCLC effect of amine-caged Pt nanoclusters (Nano-Pt) using cell cycle, migration, proliferation, apoptosis, and autophagy assays. We found that Nano-Pt significantly inhibited cell viability, reduced migration ability, caused DNA damage, induced S phase (period of DNA synthesis in the cell cycle) arrest, and promoted apoptosis in NSCLC cells. Nano-Pt also reduced mitochondrial membrane potential (MMP), increased permeability transition, and promoted apoptosis by upregulating Bax and PARP expression. Nano-Pt-induced apoptosis was accompanied by protective autophagy, which could be enhanced by autophagy inhibitors. Our findings on the biological behavior and the interaction between autophagy and apoptosis can provide the clear anti-NSCLC molecular mechanism of Nano-Pt, which have a promising potential for the development of novel Pt-based antitumor chemotherapy drugs with excellent curative efficacy and fewer side effects.
Collapse
Affiliation(s)
- Jie Wu
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450055, China
| | - Xin Huang
- Department of Light Chemical Engineering, College of Textiles, Zhongyuan University of Technology, Zhengzhou 450007, China
| | - Zhongqing Xiao
- Department of Respiratory and Critical Care Medicine, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, China
| | - Qi Wang
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450055, China
| | - Liufan Mu
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450055, China
| | - Shanshan Yang
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450055, China
| | - Shaoyi Miao
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450055, China
| | - Jing Chen
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450055, China
| | - Xinjie Deng
- Department of Light Chemical Engineering, College of Textiles, Zhongyuan University of Technology, Zhengzhou 450007, China
| | - Chaoyang Deng
- Department of Light Chemical Engineering, College of Textiles, Zhongyuan University of Technology, Zhengzhou 450007, China
| | - Hongyun Li
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450055, China.
| |
Collapse
|
91
|
Liang C, Jiang Q, Liu Z, Yang J, Zhang J, Zhang S, Xin W. Effect of Sublethal Concentrations of Metal Nanomaterials on Cell Energy Metabolism. TOXICS 2023; 11:toxics11050453. [PMID: 37235267 DOI: 10.3390/toxics11050453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
Metallic nanomaterials (MNMs) are widely used in the medical field because of their photocatalytic, optical, electrical, electronic, antibacterial, and bactericidal properties. Despite the advantages of MNMs, there is a lack of complete understanding of their toxicological behavior and their interactions with cellular mechanisms that determine cell fate. Most of the existing studies are acute toxicity studies with high doses, which is not conducive to understanding the toxic effects and mechanisms of homeostasis-dependent organelles, such as mitochondria, which are involved in many cellular functions. In this study, four types of MNMs were used to investigate the effects of metallic nanomaterials on mitochondrial function and structure. We first characterized the four MNMs and selected the appropriate sublethal concentration for application in cells. Mitochondrial characterization, energy metabolism, mitochondrial damage, mitochondrial complex activity, and expression levels were evaluated using various biological methods. The results showed that the four types of MNMs greatly inhibited mitochondrial function and cell energy metabolism and that the material entering the mitochondria damaged the mitochondrial structure. Additionally, the complex activity of mitochondrial electron transport chains is critical for assessing the mitochondrial toxicity of MNMs, which may serve as an early warning of MNM-induced mitochondrial dysfunction and cytotoxicity.
Collapse
Affiliation(s)
- Chaoshuai Liang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Qiuyao Jiang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250000, China
| | - Zhenzhen Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250000, China
| | - Jian Yang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250000, China
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250000, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250000, China
| | - Wei Xin
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250000, China
| |
Collapse
|
92
|
Bai Y, Li FF, Zhang Y, Ding YB. Silicon dioxide nanoparticles compromise decidualization via autophagy impairment to possibly cause embryo resorption. Toxicol Lett 2023; 381:72-82. [PMID: 37169230 DOI: 10.1016/j.toxlet.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/15/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
The wide application of silicon dioxide nanoparticles (SiO2NPs) has raised concerns about their harmful effects on reproduction. The purpose of this research was to investigate the toxic effects and the possible mechanisms by which SiO2NPs affect decidualization and pregnancy progression. We found that SiO2NPs could inhibit decidualization, both in mice and in human endometrial stromal cells (HESCs). Embryo resorption was also evident in mice treated with SiO2NPs. When HESCs were treated with SiO2NPs, decidualization was inhibited and there was an increase in intracellular lysosomes and autophagosomes as well as the blockage of autophagic flux. Interestingly, a reduction of autophagosome accumulation via 3-methyladenine (3MA) significantly restored the decidualization of HESCs. In summary, our results indicate that SiO2NPs can affect embryo survival by impairing decidualization through a dysfunctional autophagic process.
Collapse
Affiliation(s)
- Ying Bai
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Fang-Fang Li
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Yi Zhang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, P. R. China.
| |
Collapse
|
93
|
Chang X, Niu S, Shang M, Li J, Guo M, Zhang W, Sun Z, Li Y, Zhang R, Shen X, Tang M, Xue Y. ROS-Drp1-mediated mitochondria fission contributes to hippocampal HT22 cell apoptosis induced by silver nanoparticles. Redox Biol 2023; 63:102739. [PMID: 37187014 DOI: 10.1016/j.redox.2023.102739] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023] Open
Abstract
Silver nanoparticles (AgNPs) have widely used in industrial and medical applications for their excellent antibacterial activities. AgNPs can penetrate into the brain and cause neuronal death, but limited evidence focused on toxic effects and mechanic study in hippocampal neuron. This study aimed to investigate the molecular mechanisms of mitochondrial damage and apoptosis in mouse hippocampal HT22 cells and further to explore role of reactive oxygen species (ROS) and GTPase dynamin-related protein 1 (Drp1) in AgNPs-induced neurotoxicity. Our results showed that acute exposure to AgNPs at low doses (2-8 μg/mL) increased ROS generation, decreased mitochondrial membrane potential (MMP) and ATP synthesis in HT22 cells. In addition, AgNPs promoted mitochondrial fragmentation and mitochondria-dependent apoptosis via excessive mitochondrial fission/fusion by 8 μg/mL AgNPs treatment for 24 h. The mechanism was involved in increased protein expression of Drp1, mitochondrial fission protein 1 (Fis1), mitofusin 1/2 (Mfn1/2) and inhibited optic atrophy 1 (OPA1), and mainly mediated by phosphorylation of Drp1 Ser616. The AgNPs-induced mitochondrial impairment and apoptosis was mainly due to their particle-specific effect rather than silver ions release. Furthermore Drp1-mediated mitochondrial fission contributed to mitochondria-dependent apoptosis induced by AgNPs, all aforementioned changes were significantly rescued by N-acetyl-l-cysteine (NAC) and Mdivi-1 except for OPA1 protein expression. Hence, our results provide a novel neurotoxic mechanism to AgNPs-induced neurotoxicity and revealed that the mechanism of mitochondria-dependent apoptosis in HT22 cells was mediated by excessive activation of ROS-Drp1-mitochondrial fission axis. These findings can deepen current evidences on neurotoxicological evaluation of AgNPs and aid in guiding their proper applications in different areas, especially in biomedical use.
Collapse
Affiliation(s)
- Xiaoru Chang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Mengting Shang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Jiangyan Li
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Menghao Guo
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Wenli Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Zuoyi Sun
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yunjing Li
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Rui Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xin Shen
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
94
|
Wang Z, He Y, Liu S, Xu X, Song Y, Zhang L, An X. Toxic effects of zinc oxide nanoparticles as a food additive in goat mammary epithelial cells. Food Res Int 2023; 167:112682. [PMID: 37087259 DOI: 10.1016/j.foodres.2023.112682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/22/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023]
Abstract
Zinc oxide nanoparticles (ZnO NPs) have recently been used as food preservatives and additives because of their good antibacterial and nutritional functions. This study performed RNA-seq analyses to evaluate the potential toxicity of ZnO NPs on goat mammary epithelial cells (GMECs) in vitro. Our results suggested that the ZnO NP treatment significantly reduced GMEC viability in a time- and dose-dependent manner. Transcriptomic analysis showed that ZnO NP exposure changed the expression levels of more than 500 genes in GMECs, including various biological pathways. We observed that decreased mitochondrial membrane potential caused mitochondrial dysfunction. Further study indicated that the treatment of cells with ZnO NPs resulted in the accumulation of reactive oxygen species (ROS), which led to oxidative stress. Meanwhile, the expression of genes (TNFα, TNFR1, FADD, Caspase 8 and Caspase 6) associated with the death receptor pathway was upregulated, which indicated the death receptor-mediated extrinsic apoptosis pathway was activated. Moreover, the expression levels of Bax, Cytc, Caspase 3 and Caspase 9 were upregulated, while the expression levels of Bcl2 were downregulated, which indicated mitochondria-mediated intrinsic apoptosis pathway was activated. More notably, ZnO NP exposure increased the expression levels of ER stress-related genes (PERK, ATF4, eIF2α and CHOP) and proteins (p-PERK, p-eIF2α, PERK and CHOP). Furthermore, gene ontology (GO) terms and genes related to autophagy were altered, suggesting that exposure to ZnO NPs might activate autophagy in GMECs. In summary, our findings showed that ZnO NPs could exert significant toxic effects on GMECs through multiple mechanisms. These pathways are related to each other and influence each other to participate in ZnO NPs-induced the damage of GMECs. Thus, their safe use in the feed and food industry should be considered. Meanwhile, RNA-seq might represent a new method of assessing the toxicity mechanisms of nanomaterials.
Collapse
|
95
|
Tkachenko A, Onishchenko A, Myasoedov V, Yefimova S, Havranek O. Assessing regulated cell death modalities as an efficient tool for in vitro nanotoxicity screening: a review. Nanotoxicology 2023; 17:218-248. [PMID: 37083543 DOI: 10.1080/17435390.2023.2203239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Nanomedicine is a fast-growing field of nanotechnology. One of the major obstacles for a wider use of nanomaterials for medical application is the lack of standardized toxicity screening protocols for assessing the safety of newly synthesized nanomaterials. In this review, we focus on less frequently studied nanomaterials-induced regulated cell death (RCD) modalities, including eryptosis, necroptosis, pyroptosis, and ferroptosis, as a tool for in vitro nanomaterials safety evaluation. We summarize the latest insights into the mechanisms that mediate these RCDs in response to nanomaterials exposure. Comprehensive data from reviewed studies suggest that ROS (reactive oxygen species) overproduction and ROS-mediated pathways play a central role in nanomaterials-induced RCDs activation. On the other hand, studies also suggest that individual properties of nanomaterials, including size, shape, or surface charge, could determine specific toxicity pathways with consequent RCD induction as well. We anticipate that the evaluation of RCDs can become one of the mechanism-based screening methods in nanotoxicology. In addition to the toxicity assessment, evaluation of necroptosis-, pyroptosis-, and ferroptosis-promoting capacity of nanomaterials could simultaneously provide useful information for specific medical applications as could be their anti-tumor potential. Moreover, a detailed understanding of molecular mechanisms driving nanomaterials-mediated induction of immunogenic RCDs will substantially aid novel anti-tumor nanodrugs development.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Anatolii Onishchenko
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Valeriy Myasoedov
- Department of Medical Biology, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Svetlana Yefimova
- Institute for Scintillation Materials, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
96
|
Benavides RAS, Leiro-Vidal JM, Rodriguez-Gonzalez JA, Ares-Pena FJ, López-Martín E. The HL-60 human promyelocytic cell line constitutes an effective in vitro model for evaluating toxicity, oxidative stress and necrosis/apoptosis after exposure to black carbon particles and 2.45 GHz radio frequency. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 867:161475. [PMID: 36632900 DOI: 10.1016/j.scitotenv.2023.161475] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
The cellular and molecular mechanisms by which atmospheric pollution from particulate matter and/or electromagnetic fields (EMFs) may prove harmful to human health have not been extensively researched. We analyzed whether the combined action of EMFs and black carbon (BC) particles induced cell damage and a pro-apoptotic response in the HL-60 promyelocytic cell line when exposed to 2.45 GHz radio frequency (RF) radiation in a gigahertz transverse electromagnetic (GTEM) chamber at sub-thermal specific absorption rate (SAR) levels. RF and BC induced moderately significant levels of cell damage in the first 8 or 24 h for all exposure times/doses and much greater damage after 48 h irradiation and the higher dose of BC. We observed a clear antiproliferative effect that increased with RF exposure time and BC dose. Oxidative stress or ROS production increased with time (24 or 48 h of radiation), BC dose and the combination of both. Significant differences between the proportion of damaged and healthy cells were observed in all groups. Both radiation and BC participated separately and jointly in triggering necrosis and apoptosis in a programmed way. Oxidative-antioxidant action activated mitochondrial anti-apoptotic BCL2a gene expression after 24 h irradiation and exposure to BC. After irradiation of the cells for 48 h, expression of FASR cell death receptors was activated, precipitating the onset of pro-apoptotic phenomena and expression and intracellular activity of caspase-3 in the mitochondrial pathways, all of which can lead to cell death. Our results indicate that the interaction between BC and RF modifies the immune response in the human promyelocytic cell line and that these cells had two fates mediated by different pathways: necrosis and mitochondria-caspase dependent apoptosis. The findings may be important in regard to antimicrobial, inflammatory and autoimmune responses in humans.
Collapse
Affiliation(s)
- Rosa Ana Sueiro Benavides
- Institute of Research in Biological and Chemical Analysis, IAQBUS, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - José Manuel Leiro-Vidal
- Institute of Research in Biological and Chemical Analysis, IAQBUS, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - J Antonio Rodriguez-Gonzalez
- Department of Applied Physics, Santiago de Compostela School of Physics, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Francisco J Ares-Pena
- Department of Applied Physics, Santiago de Compostela School of Physics, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Elena López-Martín
- Department of Morphological Sciences, Santiago de Compostela School of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
97
|
Sipos A, Kim KJ, Sioutas C, Crandall ED. Kinetics of autophagic activity in nanoparticle-exposed lung adenocarcinoma (A549) cells. AUTOPHAGY REPORTS 2023; 2:2186568. [PMID: 37520337 PMCID: PMC10373127 DOI: 10.1080/27694127.2023.2186568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 08/01/2023]
Abstract
Autophagy, a homeostatic mechanism, is crucial in maintaining normal cellular function. Although dysregulation of autophagic processes is recognized in certain diseases, it is unknown how maintenance of cellular homeostasis might be affected by the kinetics of autophagic activity in response to various stimuli. In this study, we assessed those kinetics in lung adenocarcinoma (A549) cells in response to exposure to nanoparticles (NP) and/or Rapamycin. Since NP are known to induce autophagy, we wished to determine if this phenomenon could be a driver of the harmful effects seen in lung tissues exposed to air pollution. A549 cells were loaded with a fluorescent marker (DAPRed) that labels autophagosomes and autolysosomes. Autophagic activity was assessed based on the fluorescence intensity of DAPRed measured over the entire cell volume of live single cells using confocal laser scanning microscopy (CLSM). Autophagic activity over time was determined during exposure of A549 cells to single agents (50 nM Rapamycin; 80 μg/mL, 20 nm carboxylated polystyrene NP (PNP); or, 1 μg/mL ambient ultrafine particles (UFP) (<180 nm)), or double agents (Rapamycin + PNP or Rapamycin + UFP; concomitant and sequential), known to stimulate autophagy. Autophagic activity increased in all experimental modalities, including both single agent and double agent exposures, and reached a steady state in all cases ~2 times control from ~8 to 24 hrs, suggesting the presence of an upper limit to autophagic capacity. These results are consistent with the hypothesis that environmental stressors might exert their harmful effects, at least in part, by limiting available autophagic response to additional stimulation, thereby making nanoparticle-exposed cells more susceptible to secondary injury due to autophagic overload.
Collapse
Affiliation(s)
- Arnold Sipos
- Will Rogers Institute Pulmonary Research Center and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kwang-Jin Kim
- Will Rogers Institute Pulmonary Research Center and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neurosciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Constantinos Sioutas
- Sonny Astani Department of Civil and Environmental Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Edward D. Crandall
- Will Rogers Institute Pulmonary Research Center and Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
98
|
Samrot AV, Noel Richard Prakash LX. Nanoparticles Induced Oxidative Damage in Reproductive System and Role of Antioxidants on the Induced Toxicity. Life (Basel) 2023; 13:life13030767. [PMID: 36983922 PMCID: PMC10059981 DOI: 10.3390/life13030767] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
Nanotechnology is used in a variety of scientific, medical, and research domains. It is significant to mention that there are negative and severe repercussions of nanotechnology on both individuals and the environment. The toxic effect of nanoparticles exerted on living beings is termed as nanotoxicity. Nanoparticles are synthesized by various methods such as chemical, biological, physical, etc. These nanoparticles’ nanotoxicity has been observed to vary depending on the synthesis process, precursors, size of the particles, etc. Nanoparticles can enter the cell in different ways and can cause cytotoxic effects. In this review, the toxicity caused in the reproductive system and the role of the antioxidants against the nanotoxicity are briefly explained.
Collapse
Affiliation(s)
- Antony V. Samrot
- School of Bioscience, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jalan SP2, Bandar Saujana Putra, Jenjarom 42610, Malaysia
- Correspondence:
| | - Lawrence Xavier Noel Richard Prakash
- Department of Biotechnology, School of Bio and Chemical Engineering Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India;
| |
Collapse
|
99
|
Miletić M, Vilotić A, Korićanac L, Žakula J, Krivokuća MJ, Dohčević-Mitrović Z, Aškrabić S. Spectroscopic signature of ZnO NP-induced cell death modalities assessed by non-negative PCA. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 288:122180. [PMID: 36470088 DOI: 10.1016/j.saa.2022.122180] [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: 08/03/2022] [Revised: 11/10/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Selective cytotoxicity of ZnO nanoparticles among different cell types and cancer and non-cancerous cells has been demonstrated earlier. In the view of anticancer potential of ZnO nanoparticles and their presence in numerous industrial products, it is of great importance to carefully evaluate their effects and mechanisms of action in both cancerous and healthy cells. In this paper, the effects of ZnO nanoparticles on cancerous HeLa and non-cancerous MRC-5 cells are investigated by studying the changes in the vibrational properties of the cells using Raman spectroscopy. Both types of cells were incubated with ZnO nanoparticles of average size 40 nm in the doses from the range 10-40 µg/ml for the period of 48 h, after which Raman spectra were collected. Raman modes' intensity ratios I1659/I1444, I2855/I2933 and I1337/I1305 were determined as spectral markers of the cytotoxic effect of ZnO in both cell types. Non-negative principal component analysis was used instead of standard one for analysis and detection of spectral features characteristic for nanoparticle-treated cells. The first several non-negative loading vectors obtained in this analysis coincided remarkably well with the Raman spectra of particular biomolecules, showing increase of lipid and decrease of nucleic acids and protein content. Our study pointed out that Raman spectral markers of lipid unsaturation, especially I1270/I1300, are relevant for tracing the cytotoxic effect of ZnO nanoparticles on both cancerous and non-cancerous cells. The change of these spectral markers is correlated to the dose of applied nanoparticles and to the degree of cellular damage. Furthermore, great similarity of spectral features of increasing lipids to spectral features of phosphatidylserine, one of the main apoptotic markers, was recognized in treated cells. Finally, the results strongly indicated that the degree of lipid saturation, presented in the cells, plays an important role in the interaction of cells with nanoparticles.
Collapse
Affiliation(s)
- Mirjana Miletić
- Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia.
| | - Aleksandra Vilotić
- Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, University of Belgrade, Banatska 31b, 11080 Belgrade, Serbia
| | - Lela Korićanac
- Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Jelena Žakula
- Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Milica Jovanović Krivokuća
- Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, University of Belgrade, Banatska 31b, 11080 Belgrade, Serbia
| | | | - Sonja Aškrabić
- Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia.
| |
Collapse
|
100
|
Nasarudin NA, Razali M, Goh V, Chai WL, Muchtar A. Expression of Interleukin-1β and Histological Changes of the Three-Dimensional Oral Mucosal Model in Response to Yttria-Stabilized Nanozirconia. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2027. [PMID: 36903142 PMCID: PMC10003861 DOI: 10.3390/ma16052027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Over the years, advancement in ceramic-based dental restorative materials has led to the development of monolithic zirconia with increased translucency. The monolithic zirconia fabricated from nano-sized zirconia powders is shown to be superior in physical properties and more translucent for anterior dental restorations. Most in vitro studies on monolithic zirconia have focused mainly on the effect of surface treatment or the wear of the material, while the nanotoxicity of this material is yet to be explored. Hence, this research aimed to assess the biocompatibility of yttria-stabilized nanozirconia (3-YZP) on the three-dimensional oral mucosal models (3D-OMM). The 3D-OMMs were constructed using human gingival fibroblast (HGF) and immortalized human oral keratinocyte cell line (OKF6/TERT-2), co-cultured on an acellular dermal matrix. On day 12, the tissue models were exposed to 3-YZP (test) and inCoris TZI (IC) (reference material). The growth media were collected at 24 and 48 h of exposure to materials and assessed for IL-1β released. The 3D-OMMs were fixed with 10% formalin for the histopathological assessments. The concentration of the IL-1β was not statistically different between the two materials for 24 and 48 h of exposure (p = 0.892). Histologically, stratification of epithelial cells was formed without evidence of cytotoxic damage and the epithelial thickness measured was the same for all model tissues. The excellent biocompatibility of nanozirconia, as evidenced by the multiple endpoint analyses of the 3D-OMM, may indicate the potential of its clinical application as a restorative material.
Collapse
Affiliation(s)
- Naziratul Adirah Nasarudin
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Masfueh Razali
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Victor Goh
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Wen Lin Chai
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Andanastuti Muchtar
- Department of Mechanical and Manufacturing Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|