1
|
Monika P, Krishna RH, Hussain Z, Nandhini K, Pandurangi SJ, Malek T, Kumar SG. Antimicrobial hybrid coatings: A review on applications of nano ZnO based materials for biomedical applications. BIOMATERIALS ADVANCES 2025; 172:214246. [PMID: 40037050 DOI: 10.1016/j.bioadv.2025.214246] [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: 12/12/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
The extreme survivability of infectious microorganisms on various surfaces prompts for the risk of disease transmissions, posing a perilous concern for global health. Thus, the treatment of these pathogenic microorganisms using the nanomaterials functionalized with antimicrobial coatings reaps relevant scope in the ongoing trend of research. Driven by their admirable biocompatibility, cost-effectiveness, and minimal toxicity, ZnO nanoparticles (ZnO-NPs) based antimicrobial hybrid coatings have emerged as a robust material to prevent the growth of infectious microorganisms on various surfaces, which in turn boosted their applications in the area of biomedical sciences. In this context, the current review focuses on the synthesis of ZnO-NPs based hybrid coatings using different polymers and inorganic materials for effective utilization in biomedical domains including dentistry, orthopedics, implantable medical devices and wound healing. The synergistic effect of ZnO-NPs hybrids with remarkable antibacterial, antifungal and antiviral property has been discussed. Finally, we highlight the future potential of ZnO-NPs based antimicrobial hybrid coatings for potential clinical translation.
Collapse
Affiliation(s)
- Prakash Monika
- Department of Biotechnology, M.S. Ramaiah Institute of Technology, MSR Nagar, MSRIT Post, Bangalore 560054, India.
| | - R Hari Krishna
- Department of Chemistry, M.S. Ramaiah Institute of Technology, MSR Nagar, MSRIT Post, Bangalore 560054, India; Centre for Bio and Energy Materials Innovation, M.S. Ramaiah Institute of Technology, MSR Nagar, MSRIT Post, Bangalore 560054, India.
| | - Zayaan Hussain
- Department of Biotechnology, M.S. Ramaiah Institute of Technology, MSR Nagar, MSRIT Post, Bangalore 560054, India
| | - Krithika Nandhini
- Department of Biotechnology, M.S. Ramaiah Institute of Technology, MSR Nagar, MSRIT Post, Bangalore 560054, India
| | - Samhitha J Pandurangi
- Department of Biotechnology, M.S. Ramaiah Institute of Technology, MSR Nagar, MSRIT Post, Bangalore 560054, India
| | - Tausif Malek
- Department of Biotechnology, M.S. Ramaiah Institute of Technology, MSR Nagar, MSRIT Post, Bangalore 560054, India
| | - S Girish Kumar
- Department of Chemistry and Centre for Nanomaterials and Devices, RV College of Engineering, Bangalore 560059, India.
| |
Collapse
|
2
|
Zheng C, Zhang L, Sun Y, Ma Y, Zhang Y. Alveolar epithelial cell dysfunction and epithelial-mesenchymal transition in pulmonary fibrosis pathogenesis. Front Mol Biosci 2025; 12:1564176. [PMID: 40343260 PMCID: PMC12058482 DOI: 10.3389/fmolb.2025.1564176] [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: 01/21/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Pulmonary fibrosis (PF) is a progressive and lethal interstitial lung disease characterized by aberrant scar formation and destruction of alveolar architecture. Dysfunctional alveolar epithelial cells (AECs) play a central role in initiating PF, where chronic injury triggers apoptosis and disrupts epithelial homeostasis, leading to epithelial-mesenchymal transition (EMT). This dynamic reprogramming process causes AECs to shed epithelial markers and adopt a mesenchymal phenotype, fueling fibroblast activation and pathological extracellular matrix (ECM) deposition. This review systematically explores the multi-layered mechanisms driving AECs dysfunction and EMT, focusing on core signaling axes such as transforming growth factor-β (TGF-β)/Smad, WNT/β-catenin, NF-κB-BRD4, and nuclear factor erythroid 2-related factor 2 (Nrf2), which regulate EMT and fibroblast-ECM interactions. It also highlights emerging regulators, including metabolic reprogramming, exosomal miRNA trafficking, and immune-epithelial interactions. Furthermore, understanding these mechanisms is essential for developing targeted therapeutic strategies to modulate these pathways and halt or reverse fibrosis progression, offering critical insights into potential clinical treatments for PF.
Collapse
Affiliation(s)
- Caopei Zheng
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yuqing Sun
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| |
Collapse
|
3
|
Cho H, Choi M, Park JM, Choi J, Kwon YJ, Lee SM, Kang HC. Autophagic Cell Survival-to-Death Switch Induced by Cisplatin and Zn(II) Dual-Loaded Dispersity-Tunable Zwitterionic Hybrid Polyion Nanocomplex. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23774-23785. [PMID: 40226983 DOI: 10.1021/acsami.5c04242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Engineered nanomaterials, with their tunable size and surface characteristics, have played a pivotal role in biomedical delivery systems by modulating specific cellular processes such as autophagy and apoptosis, thereby influencing cellular homeostasis and cytotoxicity. Recently, zwitterionic poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC) has gained attention for its enhanced biocompatibility and antifouling properties, presenting potential as a promising alternative to poly(ethylene glycol) (PEG) in various biomedical applications. This study investigates the relationship between metal-chelated nanomaterial structures and their biochemical activity using core-corona nanoparticles (NPs) composed of a metallic pharmacophore core and a zwitterionic PMPC polymer shell. Two NP systems were designed, both containing aquated cisplatin (cis-[Pt(NH3)2(H2O)2]2+) and a Zn(II) adjuvant, with identical surface structures but contrasting core stabilities, leading to distinct drug release profiles: (1) labile NPs with pH-responsive drug release and variable size dispersity under endosomal conditions and (2) inert NPs characterized by slow drug release and low dispersity due to stable heterometallic Lewis complex formation at the core. The labile NP combinations exhibited strong drug synergism in cytotoxicity through apoptosis and ferroptosis, with cisplatin driving the primary pharmacological effects, while Zn-NPs modulated prosurvival autophagy toward autophagic cell death, which demonstrated the potential of Zn-NPs as a synergistic adjuvant in cisplatin delivery. In contrast, Pt/Zn-coloaded inert NPs exhibited controlled drug release and cytotoxicity, showing approximately 8-11 times higher IC50 values than those of labile NP combinations, depending on the combination ratio. These results demonstrate the critical role of the NP structure in modulating cytotoxicity and autophagy, providing insights for the rational design of effective drug delivery systems.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Korea
- Department of Biopharmaceutical Science, Soonchunhyang University, Asan, Chungcheongnam-do 31538, Korea
| | - Minho Choi
- Department of Chemistry, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Korea
| | - Jeong-Min Park
- Department of Chemistry, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Korea
| | - Jiye Choi
- Department of Chemistry, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Korea
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, California 92697, United States
| | - Sang-Min Lee
- Department of Chemistry, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Korea
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Korea
- Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Korea
| |
Collapse
|
4
|
Wang W, Li Z, Lyu C, Wang T, Han C, Cui S, Wang J, Xu R. Mechanism of a Novel Complex: Zinc Oxide Nanoparticles-Luteolin to Promote Ferroptosis in Human Acute Myeloid Leukemia Cells in Vitro. Int J Nanomedicine 2025; 20:4035-4050. [PMID: 40191047 PMCID: PMC11972579 DOI: 10.2147/ijn.s509007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Purpose Acute myeloid leukemia (AML) is a hematological malignancy. Zinc oxide nanoparticles (ZnO NPs) and Luteolin are commonly used to fight cancer. In this study, we synthesized a new complex: zinc oxide nanoparticles-luteolin (ZnONPs-Lut) and aimed to investigate its effects on cell death in the AML cell line (MOLM-13) in vitro and to elucidate the underlying mechanisms. Methods We assessed cell viability, quantified changes in gene expression using real-time quantitative PCR (qRT-PCR), and measured changes in ferrous (Fe2+) content, glutathione (GSH) content, malondialdehyde (MDA) content, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) levels following treatment with different concentrations of MOLM-13 cells with different concentrations of ZnONPs-Lut. Western blotting was used to detect the protein expression levels of ACSL4, GPX4, FTH1, and SLC7A11, while the cell morphology was observed by transmission electron microscopy (TEM). Meanwhile, the effect of Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, on the expression of the aforementioned ferroptosis-related proteins and cell morphology was evaluated. Results The results showed that ZnONPs-Lut was able to significantly inhibit the proliferation of MOLM-13 cells in a time- and dose-dependent manner. Additionally, it increased the concentrations of Fe2+ and MDA, reduced the expression levels of GSH and MMP, and induced ROS generation. Furthermore, it also enhanced the expression of ACSL4 protein while decreasing the expression of GPX4, FTH1, and SLC7A11 proteins. Notably, Fer-1 was able to significantly restrain the changes in protein levels and mitochondrial morphology damage induced by ZnONPs-Lut after its action on cells. Conclusion ZnONPs-Lut inhibits the proliferation of MOLM-13 cells, likely through promoting the cellular ferroptosis signaling pathway. These findings suggest that ZnONPs-Lut could be a potential therapeutic approach for AML.
Collapse
Affiliation(s)
- Wenhao Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Zonghong Li
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Chunyi Lyu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Teng Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Chen Han
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Siyuan Cui
- Department of Hematology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, People’s Republic of China
| | - Jinxin Wang
- Department of Hematology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, People’s Republic of China
| | - Ruirong Xu
- Department of Hematology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, People’s Republic of China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Jinan, People’s Republic of China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| |
Collapse
|
5
|
Rana SVS. Mechanistic paradigms of immunotoxicity, triggered by nanoparticles - a review. Toxicol Mech Methods 2025; 35:262-278. [PMID: 39585654 DOI: 10.1080/15376516.2024.2431687] [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: 07/22/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Nanoparticles (NPs) possess the ability to penetrate cells and elicit a rapid and targeted immune response, influenced by their distinct physicochemical properties. These particles can engage with both micro and macromolecules, thereby impacting various downstream signaling pathways that may lead to cell death. This review provides a comprehensive overview of the primary mechanisms contributing to the immunotoxicity of both organic and inorganic nanoparticles. The effects of carbon-based nanomaterials (CNMs), including single-walled carbon nanotubes, multi-walled carbon nanotubes, graphene, and metal oxide nanoparticles, on various immune cell types such as macrophages, neutrophils, monocytes, dendritic cells (DCs), antigen-presenting cells (APCs), and RAW 264.7 cells are examined. The immune responses discussed encompass inflammation, oxidative stress, autophagy, and apoptosis. Additionally, the roles of pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α, and IFN-γ, along with JAK/STAT signaling pathways, are highlighted. The interaction of NPs with oxidative stress pathways, including MAPK signaling and Nrf2/ARE signaling, is also explored. Furthermore, the mechanisms by which nanoparticles induce damage to organelles such as lysosomes, the endoplasmic reticulum, exosomes, and Golgi bodies within the immune system are addressed. The review also emphasizes the genotoxic and epigenetic mechanisms associated with the immunotoxicity of NPs. Recent advancements regarding the immunotherapeutic potential of engineered NPs are reported. The roles of autophagy and apoptosis in the immunotoxicity of NPs merit further investigation. In conclusion, understanding how engineered nanoparticles modulate immune responses may facilitate the prevention and treatment of human diseases, including cancer and autoimmune disorders.
Collapse
Affiliation(s)
- S V S Rana
- Department of Toxicology, Chaudhary Charan Singh University, Meerut, India
| |
Collapse
|
6
|
Liu N, Zhang B, Lin N. Review on the role of autophagy in the toxicity of nanoparticles and the signaling pathways involved. Chem Biol Interact 2025; 406:111356. [PMID: 39701490 DOI: 10.1016/j.cbi.2024.111356] [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: 10/27/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
As the development of nanotechnology, the application of nanoproducts and the advancement of nanomedicine, the contact of nanoparticles (NPs) with human body is becoming increasingly prevalent. This escalation elevates the risk of NPs exposure for workers, consumers, researchers, and both aquatic and terrestrial organisms throughout the production, usage, and disposal stages. Consequently, evaluating nanotoxicity remains critically important, though standardized assessment criteria are still lacking. The diverse and complex properties of NPs further complicate the understanding of their toxicological mechanisms. Autophagy, a fundamental cellular process, exhibits dual functions-both pro-survival and pro-death. This review offers an updated perspective on the dual roles of autophagy in nanotoxicity and examines the factors influencing autophagic responses. However, no definitive framework exists for predicting NPs-induced autophagy. Beyond the conventional autophagy pathways, the review highlights specific transcription factors activated by NPs and explores metabolic reprogramming. Particular attention is given to NPs-induced selective autophagy, including mitophagy, ER-phagy, ferritinophagy, lysophagy, and lipophagy. Additionally, the review investigates autophagy's involvement in NPs-mediated biological processes such as ferroptosis, inflammation, macrophage polarization, epithelial-mesenchymal transition, tumor cell proliferation and drug resistance, as well as liver and kidney injury, neurotoxicity, and other diseases. In summary, this review presents a novel update on selective autophagy-mediated nanotoxicity and elucidates the broader interactions of autophagy in NPs-induced biological processes. Collectively, these insights offer valuable strategies for mitigating nanotoxicity through autophagy modulation and advancing the development of NPs in biomedical applications.
Collapse
Affiliation(s)
- Na Liu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| |
Collapse
|
7
|
Cao J, Yang Y, Liu X, Huang Y, Xie Q, Kadushkin A, Nedelko M, Wu D, Aquilina NJ, Li X, Cai X, Li R. Deciphering key nano-bio interface descriptors to predict nanoparticle-induced lung fibrosis. Part Fibre Toxicol 2025; 22:1. [PMID: 39810232 PMCID: PMC11731361 DOI: 10.1186/s12989-024-00616-3] [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: 08/16/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The advancement of nanotechnology underscores the imperative need for establishing in silico predictive models to assess safety, particularly in the context of chronic respiratory afflictions such as lung fibrosis, a pathogenic transformation that is irreversible. While the compilation of predictive descriptors is pivotal for in silico model development, key features specifically tailored for predicting lung fibrosis remain elusive. This study aimed to uncover the essential predictive descriptors governing nanoparticle-induced pulmonary fibrosis. METHODS We conducted a comprehensive analysis of the trajectory of metal oxide nanoparticles (MeONPs) within pulmonary systems. Two biological media (simulated lung fluid and phagolysosomal simulated fluid) and two cell lines (macrophages and epithelial cells) were meticulously chosen to scrutinize MeONP behaviors. Their interactions with MeONPs, also referred to as nano-bio interactions, can lead to alterations in the properties of the MeONPs as well as specific cellular responses. Physicochemical properties of MeONPs were assessed in biological media. The impact of MeONPs on cell membranes, lysosomes, mitochondria, and cytoplasmic components was evaluated using fluorescent probes, colorimetric enzyme substrates, and ELISA. The fibrogenic potential of MeONPs in mouse lungs was assessed by examining collagen deposition and growth factor release. Random forest classification was employed for analyzing in chemico, in vitro and in vivo data to identify predictive descriptors. RESULTS The nano-bio interactions induced diverse changes in the 4 characteristics of MeONPs and had variable effects on the 14 cellular functions, which were quantitatively evaluated in chemico and in vitro. Among these 18 quantitative features, seven features were found to play key roles in predicting the pro-fibrogenic potential of MeONPs. Notably, IL-1β was identified as the most important feature, contributing 27.8% to the model's prediction. Mitochondrial activity (specifically NADH levels) in macrophages followed closely with a contribution of 17.6%. The remaining five key features include TGF-β1 release and NADH levels in epithelial cells, dissolution in lysosomal simulated fluids, zeta potential, and the hydrodynamic size of MeONPs. CONCLUSIONS The pro-fibrogenic potential of MeONPs can be predicted by combination of key features at nano-bio interfaces, simulating their behavior and interactions within the lung environment. Among the 18 quantitative features, a combination of seven in chemico and in vitro descriptors could be leveraged to predict lung fibrosis in animals. Our findings offer crucial insights for developing in silico predictive models for nano-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Jiayu Cao
- School of Public Health, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yuhui Yang
- School of Public Health, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xi Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yang Huang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Dalian Key Laboratory on Chemicals Risk Control and Pollution Prevention Technology, School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, China
| | - Qianqian Xie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Aliaksei Kadushkin
- Department of Biological Chemistry, Belarusian State Medical University, Minsk, 220089, Belarus
| | - Mikhail Nedelko
- B.I. Stepanov Institute of Physics of National Academy of Sciences of Belarus, 68Nezalezhnasti Ave, Minsk, 220072, Belarus
| | - Di Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Noel J Aquilina
- Department of Chemistry, University of Malta, Msida, 2080, MSD, Malta
| | - Xuehua Li
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Dalian Key Laboratory on Chemicals Risk Control and Pollution Prevention Technology, School of Environmental Science and Technology, Dalian University of Technology, Dalian, 116024, China
| | - Xiaoming Cai
- School of Public Health, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical School, Soochow University, Suzhou, Jiangsu, 215123, China.
- CEET, Nanotechnology Centre, VSB-Technical University of Ostrava, 17 listopadu, Ostrava, 2172-15, 70800, Czech Republic.
| |
Collapse
|
8
|
Chen R, Luo S, Zhang Y, Mao L, Diao J, Cheng S, Zou Z, Chen C, Qin X, Jiang X, Zhang J. LC3B-regulated autophagy mitigates zinc oxide nanoparticle-induced epithelial cell dysfunction and acute lung injury. Toxicol Sci 2025; 203:105-117. [PMID: 39509325 DOI: 10.1093/toxsci/kfae146] [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: 11/15/2024] Open
Abstract
Zinc oxide nanoparticles (ZnONPs) are widely utilized across various industries, raising concerns about their potential toxicity, especially in the respiratory system. This study explores the role of autophagy, regulated by microtubule-associated protein 1A/1B-light chain 3B (LC3B), in ZnONPs-induced toxicity using both in vivo (LC3B knockout mice) and in vitro (BEAS-2B cells) models. Our findings demonstrate that LC3B-regulated autophagy mitigates ZnONPs-induced epithelial cell dysfunction and acute lung injury. In the absence of LC3B, oxidative stress, inflammation, and intracellular zinc accumulation are exacerbated, resulting in mitochondrial dysfunction and epithelial cell death. In vitro, LC3B knockdown disrupted zinc ion transporter expression and impaired mitophagic flux in BEAS-2B cells. Treatment with zinc ion chelators alleviated these toxic effects, confirming that free zinc ions play a critical role in driving ZnONPs toxicity. These findings highlight that targeting autophagy and maintaining zinc homeostasis could offer therapeutic strategies to reduce ZnONPs-induced lung damage.
Collapse
Affiliation(s)
- Ruonan Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Sen Luo
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yunxiao Zhang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing 400016, People's Republic of China
| | - Jun Diao
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Shuqun Cheng
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing 400016, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing 400016, People's Republic of China
| |
Collapse
|
9
|
El-Saadony MT, Fang G, Yan S, Alkafaas SS, El Nasharty MA, Khedr SA, Hussien AM, Ghosh S, Dladla M, Elkafas SS, Ibrahim EH, Salem HM, Mosa WFA, Ahmed AE, Mohammed DM, Korma SA, El-Tarabily MK, Saad AM, El-Tarabily KA, AbuQamar SF. Green Synthesis of Zinc Oxide Nanoparticles: Preparation, Characterization, and Biomedical Applications - A Review. Int J Nanomedicine 2024; 19:12889-12937. [PMID: 39651353 PMCID: PMC11624689 DOI: 10.2147/ijn.s487188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 12/11/2024] Open
Abstract
Over the last decade, biomedical nanomaterials have garnered significant attention due to their remarkable biological properties and diverse applications in biomedicine. Metal oxide nanoparticles (NPs) are particularly notable for their wide range of medicinal uses, including antibacterial, anticancer, biosensing, cell imaging, and drug/gene delivery. Among these, zinc oxide (ZnO) NPs stand out for their versatility and effectiveness. Recently, ZnO NPs have become a primary material in various sectors, such as pharmaceutical, cosmetic, antimicrobials, construction, textile, and automotive industries. ZnO NPs can generate reactive oxygen species and induce cellular apoptosis, thus underpinning their potent anticancer and antibacterial properties. To meet the growing demand, numerous synthetic approaches have been developed to produce ZnO NPs. However, traditional manufacturing processes often involve significant economic and environmental costs, prompting a search for more sustainable alternatives. Intriguingly, biological synthesis methods utilizing plants, plant extracts, or microorganisms have emerged as ideal for producing ZnO NPs. These green production techniques offer numerous medicinal, economic, environmental, and health benefits. This review highlights the latest advancements in the green synthesis of ZnO NPs and their biomedical applications, showcasing their potential to revolutionize the field with eco-friendly and cost-effective solutions.
Collapse
Affiliation(s)
- Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Guihong Fang
- School of Public Health, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
- Qionghai People’s Hospital, Qionghai, Hainan, 571400, People’s Republic of China
| | - Si Yan
- Qionghai People’s Hospital, Qionghai, Hainan, 571400, People’s Republic of China
| | - Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud A El Nasharty
- Department of Chemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta, 31733, Egypt
| | - Aya Misbah Hussien
- Biotechnology Department at Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21531, Egypt
| | - Soumya Ghosh
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman
| | - Mthokozisi Dladla
- Human Molecular Biology Unit (School of Biomedical Sciences), Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Shebin El Kom, Menofia, 32511, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Essam H Ibrahim
- Biology Department, Faculty of Science, King Khalid University, Abha, 61413, Saudi Arabia
- Blood Products Quality Control and Research Department, National Organization for Research and Control of Biologicals, Cairo, 12611, Egypt
| | - Heba Mohammed Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Walid F A Mosa
- Plant Production Department (Horticulture-Pomology), Faculty of Agriculture, Saba Basha, Alexandria University, Alexandria, 21531, Egypt
| | - Ahmed Ezzat Ahmed
- Biology Department, Faculty of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Sameh A Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | | | - Ahmed M Saad
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Synan F AbuQamar
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| |
Collapse
|
10
|
Zhang P, Zhong D, Yu Y, Wang L, Li Y, Liang Y, Shi Y, Duan M, Li B, Niu H, Xu Y. Integration of STING activation and COX-2 inhibition via steric-hindrance effect tuned nanoreactors for cancer chemoimmunotherapy. Biomaterials 2024; 311:122695. [PMID: 38954960 DOI: 10.1016/j.biomaterials.2024.122695] [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: 04/17/2024] [Revised: 06/01/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
Integrating immunotherapy with nanomaterials-based chemotherapy presents a promising avenue for amplifying antitumor outcomes. Nevertheless, the suppressive tumor immune microenvironment (TIME) and the upregulation of cyclooxygenase-2 (COX-2) induced by chemotherapy can hinder the efficacy of the chemoimmunotherapy. This study presents a TIME-reshaping strategy by developing a steric-hindrance effect tuned zinc-based metal-organic framework (MOF), designated as CZFNPs. This nanoreactor is engineered by in situ loading of the COX-2 inhibitor, C-phycocyanin (CPC), into the framework building blocks, while simultaneously weakening the stability of the MOF. Consequently, CZFNPs achieve rapid pH-responsive release of zinc ions (Zn2+) and CPC upon specific transport to tumor cells overexpressing folate receptors. Accordingly, Zn2+ can induce reactive oxygen species (ROS)-mediated cytotoxicity therapy while synchronize with mitochondrial DNA (mtDNA) release, which stimulates mtDNA/cGAS-STING pathway-mediated innate immunity. The CPC suppresses the chemotherapy-induced overexpression of COX-2, thus cooperatively reprogramming the suppressive TIME and boosting the antitumor immune response. In xenograft tumor models, the CZFNPs system effectively modulates STING and COX-2 expression, converting "cold" tumors into "hot" tumors, thereby resulting in ≈ 4-fold tumor regression relative to ZIF-8 treatment alone. This approach offers a potent strategy for enhancing the efficacy of combined nanomaterial-based chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Di Zhong
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Yongbo Yu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Lupeng Wang
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Yifan Li
- Department of Breast Center of the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong Province, China
| | - Ye Liang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yanfeng Shi
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Meilin Duan
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, Shandong Province, China.
| | - Haitao Niu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Yuanhong Xu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China; Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
11
|
Liu X, Li J, Zhu L, Huang J, Zhang Q, Wang J, Xie J, Dong Q, Zou Z, Huang G, Gu Q, Wang J, Li J. Mechanistic insights into zinc oxide nanoparticles induced embryotoxicity via H3K9me3 modulation. Biomaterials 2024; 311:122679. [PMID: 38943823 DOI: 10.1016/j.biomaterials.2024.122679] [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: 02/08/2024] [Revised: 06/05/2024] [Accepted: 06/23/2024] [Indexed: 07/01/2024]
Abstract
The widespread application of nanoparticles (NPs) in various fields has raised health concerns, especially in reproductive health. Our research has shown zinc oxide nanoparticles (ZnONPs) exhibit the most significant toxicity to pre-implantation embryos in mice compared to other common NPs. In patients undergoing assisted reproduction technology (ART), a significant negative correlation was observed between Zn concentration and clinical outcomes. Therefore, this study explores the impact of ZnONPs exposure on pre-implantation embryonic development and its underlying mechanisms. We revealed that both in vivo and in vitro exposure to ZnONPs impairs pre-implantation embryonic development. Moreover, ZnONPs were found to reduce the pluripotency of mouse embryonic stem cells (mESCs), as evidenced by teratoma and diploid chimera assays. Employing multi-omics approaches, including RNA-Seq, CUT&Tag, and ATAC-seq, the embryotoxicity mechanisms of ZnONPs were elucidated. The findings indicate that ZnONPs elevate H3K9me3 levels, leading to increased heterochromatin and consequent inhibition of gene expression related to development and pluripotency. Notably, Chaetocin, a H3K9me3 inhibitor, sucessfully reversed the embryotoxicity effects induced by ZnONPs. Additionally, the direct interaction between ZnONPs and H3K9me3 was verified through pull-down and immunoprecipitation assays. Collectively, these findings offer new insights into the epigenetic mechanisms of ZnONPs toxicity, enhancing our understanding of their impact on human reproductive health.
Collapse
Affiliation(s)
- Xuemei Liu
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China
| | - Jie Li
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ling Zhu
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China
| | - Jiayu Huang
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Zhang
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China
| | - Jianwu Wang
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Juan Xie
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China
| | - Qiang Dong
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Guoning Huang
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China.
| | - Qi Gu
- State Key Laboratory of Membrane Biology and the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Jianyu Wang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Jingyu Li
- Chongqing Key Laboratory of Human Embryo Engineering and Precision Medicine, Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing, China.
| |
Collapse
|
12
|
Ijatuyi TT, Lawal AO, Akinjiyan MO, Ojo FM, Koledoye OF, Agboola OO, Dahunsi DT, Folorunso IM, Elekofehinti OO. Effects of Bryophyllum pinnatum on Dysfunctional Autophagy in Rats Lungs Exposed to Zinc Oxide Nanoparticles. Int Immunopharmacol 2024; 141:113005. [PMID: 39213874 DOI: 10.1016/j.intimp.2024.113005] [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: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Lung inflammation as a result of exposure to toxicants is a major pathological problem. Autophagy (AP) is a process of cell self-digestion and can be disrupted by environmental toxicants, leading to oxidative stress, inflammation and cellular damage. Bryophyllum pinnatum (Lam.) Oken has been used in folklore medicine to manage pathological abnormalities, including inflammation, but mechanisms remain unclear. This work investigated the effects of Bryophyllum pinnatum ethanol leaf extract (BP) on dysfunctional AP in the lungs of Wistar rats exposed to zinc oxide nanoparticles (ZONPs). The experimental rats were orally administered ZONPs for seven days (10 mg/kg). Some exposed rats were post-treated with BP (62.5 and 125 mg/kg) through oral gavage. Oxidative stress, inflammation, and apoptotic and autophagic parameters were assessed using biochemical assay and gene expression methods. Several indices of pulmonary damage were also evaluated. PCR analysis suggested that ZONP downregulated the expression of pro-autophagy-related genes (Beclin 2, ATG5, DAPK, and FOXP3) and upregulated the expression of the TNF-alpha, NF-Kb, LC3 and Bcl2 genes. In contrast, BP significantly (p < 0.0001) reversed ZONP-induced pulmonary toxicity and oxidative stress. It reduced MDA levels and increased SOD, CAT, GSH and GPxD activities. BP significantly (p < 0.0001) downregulated the expressions of proinflammatory genes (IL-6 and JNK) and upregulated the expressions of IL-10, CAT and SOD genes in ZONP-exposed rats. BP restored the lung's histoarchitectural structure after ZNOP-induced distortion. The results suggested that BP has antioxidant and anti-inflammatory properties, and could effectively restore ZNOP-induced dysfunctional AP in the lungs of Wistar rats.
Collapse
Affiliation(s)
- Taiwo Tolulope Ijatuyi
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria
| | - Akeem Olalekan Lawal
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria; Precision Molecular Laboratory, Akure, Ondo State, Nigeria
| | - Moses Orimoloye Akinjiyan
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria; Medical Biochemistry Department, School of Basic Medical Sciences, Federal University of Technology, Akure, Ondo State, Nigeria.
| | - Funmilayo Mercy Ojo
- Medical Biochemistry Department, School of Basic Medical Sciences, Federal University of Technology, Akure, Ondo State, Nigeria
| | - Omowumi Funmilayo Koledoye
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria
| | - Olaoluwa Oladimeji Agboola
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria
| | - Damilola Timothy Dahunsi
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria
| | - Ibukun Mary Folorunso
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria; Precision Molecular Laboratory, Akure, Ondo State, Nigeria
| | - Olusola Olalekan Elekofehinti
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, School of Life Sciences, Federal University of Technology, Akure, P.M.B. 704, Akure, Ondo-State, Nigeria
| |
Collapse
|
13
|
Aschner M, Skalny AV, Lu R, Martins AC, Tsatsakis A, Miroshnikov SA, Santamaria A, Tinkov AA. Molecular mechanisms of zinc oxide nanoparticles neurotoxicity. Chem Biol Interact 2024; 403:111245. [PMID: 39278458 DOI: 10.1016/j.cbi.2024.111245] [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: 07/31/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Zinc oxide nanoparticles (ZnONPs) are widely used in industry and biomedicine. A growing body of evidence demonstrates that ZnONPs exposure may possess toxic effects to a variety of tissues, including brain. Therefore, the objective of the present review was to summarize existing evidence on neurotoxic effects of ZnONPs and discuss the underlying molecular mechanisms. The existing laboratory data demonstrate that both in laboratory rodents and other animals ZnONPs exposure results in a significant accumulation of Zn in brain and nervous tissues, especially following long-term exposure. As a result, overexposure to ZnONPs causes oxidative stress and cell death, both in neurons and glial cells, by induction of apoptosis, necrosis and ferroptosis. In addition, ZnONPs may induce neuroinflammation through the activation of nuclear factor kappa B (NF-κB), extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK), and lipoxygenase (LOX) signaling pathways. ZnONPs exposure is associated with altered cholinergic, dopaminergic, serotoninergic, as well as glutamatergic and γ-aminobutyric acid (GABA)-ergic neurotransmission, thus contributing to impaired neuronal signal transduction. Cytoskeletal alterations, as well as impaired autophagy and mitophagy also contribute to ZnONPs-induced brain damage. It has been posited that some of the adverse effects of ZnONPs in brain are mediated by altered microRNA expression and dysregulation of gut-brain axis. Furthermore, in vivo studies have demonstrated that ZnONPs exposure induced anxiety, motor and cognitive deficits, as well as adverse neurodevelopmental outcome. At the same time, the relevance of ZnONPs-induced neurotoxicity and its contribution to pathogenesis of neurological diseases in humans are still unclear. Further studies aimed at estimation of hazards of ZnONPs to human brain health and the underlying molecular mechanisms are warranted.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Anatoly V Skalny
- Institute of Bioelementology, Orenburg State University, Orenburg, 460018, Russia; Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, 700 13, Heraklion, Greece
| | - Sergey A Miroshnikov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460018, Russia
| | - Abel Santamaria
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico; Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Alexey A Tinkov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460018, Russia; Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia; Laboratory of Molecular Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia.
| |
Collapse
|
14
|
Xu Z, Wu Y, Hu J, Mei Z, Zhao Y, Yang K, Shi Y, Xu X. Recent advances in nanoadjuvant-triggered STING activation for enhanced cancer immunotherapy. Heliyon 2024; 10:e38900. [PMID: 39640775 PMCID: PMC11620084 DOI: 10.1016/j.heliyon.2024.e38900] [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: 04/30/2024] [Revised: 07/22/2024] [Accepted: 10/01/2024] [Indexed: 12/07/2024] Open
Abstract
The development of effective cancer treatments is a popular in contemporary medical research. Immunotherapy, the fourth most common cancer treatment method, relies on activating autoimmune function to eradicate tumors and exhibits advantages such as a good curative effect and few side effects. In recent years, tumor vaccines that activate the stimulator of interferon genes (STING) pathway are being actively researched in the field of immunotherapy; however, their application is still limited because of the rapid clearance rate of tumor-related lymph nodes and low efficiency of antigen presentation. The rise of nanomedicine has provided new opportunities for solving these problems. By preparing materials with adjuvant effects nanoparticles, the small size of nanoparticles can be exploited to enable the entry of vaccines into tumor-related lymph nodes to accurately deliver STING agonists and activate the immune response. Based on this, this paper reviews various types of nano-adjuvants based on metals, platinum chemotherapy drugs, camptothecin derivatives, deoxyribonucleic acid, etc. and highlights the transformation prospects of these nano-adjuvants in tumor vaccines to provide a reference for promoting the development of nano-medicine and tumor vaccinology.
Collapse
Affiliation(s)
- Zicong Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yihong Wu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Junjie Hu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Zhaozhao Mei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yutong Zhao
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, PR China
| | - Keda Yang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, PR China
| | - Xiaoling Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| |
Collapse
|
15
|
Carrillo-Romero J, Mentxaka G, García-Salvador A, Katsumiti A, Carregal-Romero S, Goñi-de-Cerio F. Assessing the Toxicity of Metal- and Carbon-Based Nanomaterials In Vitro: Impact on Respiratory, Intestinal, Skin, and Immune Cell Lines. Int J Mol Sci 2024; 25:10910. [PMID: 39456693 PMCID: PMC11507852 DOI: 10.3390/ijms252010910] [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: 09/14/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The field of nanotechnology has experienced exponential growth, with the unique properties of nanomaterials (NMs) being employed to enhance a wide range of products across diverse industrial sectors. This study examines the toxicity of metal- and carbon-based NMs, with a particular focus on titanium dioxide (TiO2), zinc oxide (ZnO), silica (SiO2), cerium oxide (CeO2), silver (Ag), and multi-walled carbon nanotubes (MWCNTs). The potential health risks associated with increased human exposure to these NMs and their effect on the respiratory, gastrointestinal, dermal, and immune systems were evaluated using in vitro assays. Physicochemical characterisation of the NMs was carried out, and in vitro assays were performed to assess the cytotoxicity, genotoxicity, reactive oxygen species (ROS) production, apoptosis/necrosis, and inflammation in cell lines representative of the systems evaluated (3T3, Caco-2, HepG2, A549, and THP-1 cell lines). The results obtained show that 3T3 and A549 cells exhibit high cytotoxicity and ROS production after exposure to ZnO NMs. Caco-2 and HepG2 cell lines show cytotoxicity when exposed to ZnO and Ag NMs and oxidative stress induced by SiO2 and MWCNTs. THP-1 cell line shows increased cytotoxicity and a pro-inflammatory response upon exposure to SiO2. This study emphasises the importance of conducting comprehensive toxicological assessments of NMs given their physicochemical interactions with biological systems. Therefore, it is of key importance to develop robust and specific methodologies for the assessment of their potential health risks.
Collapse
Affiliation(s)
- Juliana Carrillo-Romero
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain; (J.C.-R.); (G.M.); (A.G.-S.); (A.K.)
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 San Sebastián, Spain;
| | - Gartze Mentxaka
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain; (J.C.-R.); (G.M.); (A.G.-S.); (A.K.)
| | - Adrián García-Salvador
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain; (J.C.-R.); (G.M.); (A.G.-S.); (A.K.)
| | - Alberto Katsumiti
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain; (J.C.-R.); (G.M.); (A.G.-S.); (A.K.)
| | - Susana Carregal-Romero
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 San Sebastián, Spain;
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Felipe Goñi-de-Cerio
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain; (J.C.-R.); (G.M.); (A.G.-S.); (A.K.)
| |
Collapse
|
16
|
Nazemof N, Breznan D, Dirieh Y, Blais E, Johnston LJ, Tayabali AF, Gomes J, Kumarathasan P. Cytotoxic Potencies of Zinc Oxide Nanoforms in A549 and J774 Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1601. [PMID: 39404328 PMCID: PMC11482475 DOI: 10.3390/nano14191601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Zinc oxide nanoparticles (NPs) are used in a wide range of consumer products and in biomedical applications, resulting in an increased production of these materials with potential for exposure, thus causing human health concerns. Although there are many reports on the size-related toxicity of ZnO NPs, the toxicity of different nanoforms of this chemical, toxicity mechanisms, and potency determinants need clarification to support health risk characterization. A set of well-characterized ZnO nanoforms (e.g., uncoated ca. 30, 45, and 53 nm; coated with silicon oil, stearic acid, and (3-aminopropyl) triethoxysilane) were screened for in vitro cytotoxicity in two cell types, human lung epithelial cells (A549), and mouse monocyte/macrophage (J774) cells. ZnO (bulk) and ZnCl2 served as reference particles. Cytotoxicity was examined 24 h post-exposure by measuring CTB (viability), ATP (energy metabolism), and %LDH released (membrane integrity). Cellular oxidative stress (GSH-GSSG) and secreted proteins (targeted multiplex assay) were analyzed. Zinc oxide nanoform type-, dose-, and cell type-specific cytotoxic responses were seen, along with cellular oxidative stress. Cell-secreted protein profiles suggested ZnO NP exposure-related perturbations in signaling pathways relevant to inflammation/cell injury and corresponding biological processes, namely reactive oxygen species generation and apoptosis/necrosis, for some nanoforms, consistent with cellular oxidative stress and ATP status. The size, surface area, agglomeration state and metal contents of these ZnO nanoforms appeared to be physicochemical determinants of particle potencies. These findings warrant further research on high-content "OMICs" to validate and resolve toxicity pathways related to exposure to nanoforms to advance health risk-assessment efforts and to inform on safer materials.
Collapse
Affiliation(s)
- Nazila Nazemof
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada; (N.N.); (J.G.)
| | - Dalibor Breznan
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - Yasmine Dirieh
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - Erica Blais
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - Linda J. Johnston
- Metrology Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada;
| | - Azam F. Tayabali
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - James Gomes
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada; (N.N.); (J.G.)
| | - Premkumari Kumarathasan
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada; (N.N.); (J.G.)
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| |
Collapse
|
17
|
Chen Y, Wang X, Zhou J, Wang G, Gao T, Wei H, Che Y, Li T, Zhang Z, Wang S, Hu L, Lu R. Association of seminal plasma zinc levels with human semen quality and its toxic effects on sperm motility. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116889. [PMID: 39186840 DOI: 10.1016/j.ecoenv.2024.116889] [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: 04/20/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Infertility has become one of the most common chronic diseases among reproductive- aged individuals, and male factors account for about 50 %. Zinc is a trace element that is essential for sperm quality and male reproductive system. Although several current studies with small sample sizes have investigated this relationship, the conclusions have been still controversial. Here, using a large population sample involved 25,915 participants from Changzhou Maternity and Child Health Care Hospital, we revealed an inverted "U"-shaped trend between seminal plasma zinc concentrations and sperm motility PR/PR + NP (PR, progressive motility; NP, non-progressive motility). The results showed that the highest values of sperm PR/PR + NP observed in the group with intermediate concentrations of zinc (Group 2, 0.25-2.11 mmol/L). The mean values were 43.17 ± 19.03 % and 56.64 ± 20.28 %, respectively. And the lowest values came out in the highest zinc levels group (Group 4, > 3.04 mmol/L). In vitro cell experiments also showed that zinc caused dose-dependent cytotoxicity for GC-2 cells at a threshold value. Furthermore, RNA-seq analysis demonstrated that high concentrations of zinc exerted toxic effects on GC-2 cells through immune injury. Taken together, our findings suggested that moderate amounts of zinc are crucial for human reproduction and excessive concentrations may have adverse effects on male fertility.
Collapse
Affiliation(s)
- Yu Chen
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Xiaoyu Wang
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jing Zhou
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Ganlin Wang
- Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Tingting Gao
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Hai Wei
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Yurui Che
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Tianfu Li
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Zhenglin Zhang
- Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Shuxian Wang
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Lingmin Hu
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China.
| | - Renjie Lu
- Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China.
| |
Collapse
|
18
|
Fernández-Bertólez N, Alba-González A, Touzani A, Ramos-Pan L, Méndez J, Reis AT, Quelle-Regaldie A, Sánchez L, Folgueira M, Laffon B, Valdiglesias V. Toxicity of zinc oxide nanoparticles: Cellular and behavioural effects. CHEMOSPHERE 2024; 363:142993. [PMID: 39097108 DOI: 10.1016/j.chemosphere.2024.142993] [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: 03/01/2024] [Revised: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Due to their extensive use, the release of zinc oxide nanoparticles (ZnO NP) into the environment is increasing and may lead to unintended risk to both human health and ecosystems. Access of ZnO NP to the brain has been demonstrated, so their potential toxicity on the nervous system is a matter of particular concern. Although evaluation of ZnO NP toxicity has been reported in several previous studies, the specific effects on the nervous system are not completely understood and, particularly, effects on genetic material and on organism behaviour are poorly addressed. We evaluated the potential toxic effects of ZnO NP in vitro and in vivo, and the role of zinc ions (Zn2+) in these effects. In vitro, the ability of ZnO NP to be internalized by A172 glial cells was verified, and the cytotoxic and genotoxic effects of ZnO NP or the released Zn2+ ions were addressed by means of vital dye exclusion and comet assay, respectively. In vivo, behavioural alterations were evaluated in zebrafish embryos using a total locomotion assay. ZnO NP induced decreases in viability of A172 cells after 24 h of exposure and genetic damage after 3 and 24 h. The involvement of the Zn2+ ions released from the NP in genotoxicity was confirmed. ZnO NP exposure also resulted in decreased locomotor activity of zebrafish embryos, with a clear role of released Zn2+ ions in this effect. These findings support the toxic potential of ZnO NP showing, for the first time, genetic effects on glial cells and proving the intervention of Zn2+ ions.
Collapse
Affiliation(s)
- Natalia Fernández-Bertólez
- Universidade da Coruña, Grupo NanoToxGen, Centro Interdisciplinar de Química e Bioloxía-CICA, Departamento de Biología, Facultad de Ciencias, Campus A Zapateira s/n, 15071, A Coruña, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, As Xubias, 15006, A Coruña, Spain
| | - Anabel Alba-González
- Universidade da Coruña, Grupo NEUROVER, Centro Interdisciplinar de Química e Bioloxía-CICA, Rúa As Carballeiras, 15071, A Coruña, Spain
| | - Assia Touzani
- Universidade da Coruña, Grupo NanoToxGen, Centro Interdisciplinar de Química e Bioloxía-CICA, Departamento de Biología, Facultad de Ciencias, Campus A Zapateira s/n, 15071, A Coruña, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, As Xubias, 15006, A Coruña, Spain
| | - Lucía Ramos-Pan
- Universidade da Coruña, Grupo NanoToxGen, Centro Interdisciplinar de Química e Bioloxía-CICA, Departamento de Biología, Facultad de Ciencias, Campus A Zapateira s/n, 15071, A Coruña, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, As Xubias, 15006, A Coruña, Spain
| | - Josefina Méndez
- Universidade da Coruña, Grupo NanoToxGen, Centro Interdisciplinar de Química e Bioloxía-CICA, Departamento de Biología, Facultad de Ciencias, Campus A Zapateira s/n, 15071, A Coruña, Spain
| | - Ana Teresa Reis
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas 135, 4050-600, Porto, Portugal; Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Rua das Taipas 135, 4050-600, Porto, Portugal; Environmental Health Department, National Institute of Health, Rua Alexandre Herculano, 321, 4000-055, Porto, Portugal
| | - Ana Quelle-Regaldie
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary Science, University of Santiago de Compostela, 27002, Lugo, Spain; Translational Research for Neurological Diseases, Institut Imagine, INSERM UMR 1163, Université Paris Cité, F-75015, Paris, France
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary Science, University of Santiago de Compostela, 27002, Lugo, Spain
| | - Mónica Folgueira
- Universidade da Coruña, Grupo NEUROVER, Centro Interdisciplinar de Química e Bioloxía-CICA, Rúa As Carballeiras, 15071, A Coruña, Spain
| | - Blanca Laffon
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, As Xubias, 15006, A Coruña, Spain; Universidade da Coruña, Grupo DICOMOSA, Centro Interdisciplinar de Química e Bioloxía-CICA, Departamento de Psicología, Facultad de Ciencias de la Educación, Campus Elviña s/n, 15071, A Coruña, Spain.
| | - Vanessa Valdiglesias
- Universidade da Coruña, Grupo NanoToxGen, Centro Interdisciplinar de Química e Bioloxía-CICA, Departamento de Biología, Facultad de Ciencias, Campus A Zapateira s/n, 15071, A Coruña, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, As Xubias, 15006, A Coruña, Spain
| |
Collapse
|
19
|
Zhou X, Medina-Ramirez IE, Su G, Liu Y, Yan B. All Roads Lead to Rome: Comparing Nanoparticle- and Small Molecule-Driven Cell Autophagy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310966. [PMID: 38616767 DOI: 10.1002/smll.202310966] [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: 12/05/2023] [Revised: 03/27/2024] [Indexed: 04/16/2024]
Abstract
Autophagy, vital for removing cellular waste, is triggered differently by small molecules and nanoparticles. Small molecules, like rapamycin, non-selectively activate autophagy by inhibiting the mTOR pathway, which is essential for cell regulation. This can clear damaged components but may cause cytotoxicity with prolonged use. Nanoparticles, however, induce autophagy, often causing oxidative stress, through broader cellular interactions and can lead to a targeted form known as "xenophagy." Their impact varies with their properties but can be harnessed therapeutically. In this review, the autophagy induced by nanoparticles is explored and small molecules across four dimensions: the mechanisms behind autophagy induction, the outcomes of such induction, the toxicological effects on cellular autophagy, and the therapeutic potential of employing autophagy triggered by nanoparticles or small molecules. Although small molecules and nanoparticles each induce autophagy through different pathways and lead to diverse effects, both represent invaluable tools in cell biology, nanomedicine, and drug discovery, offering unique insights and therapeutic opportunities.
Collapse
Affiliation(s)
- Xiaofei Zhou
- College of Science & Technology, Hebei Agricultural University, Baoding, 071001, China
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, 071100, China
| | - Iliana E Medina-Ramirez
- Department of Chemistry, Universidad Autónoma de Aguascalientes, Av Universidad 940, Aguascalientes, Aguascalientes, México
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 10024, China
| | - Bing Yan
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, China
| |
Collapse
|
20
|
Zhang H, Chen Q, Han H, Guo C, Jiang X, Xia Y, Zhang Y, Zhou L, Zhang J, Tian X, Mao L, Qiu J, Zou Z, Chen C. SUMOylation modification of FTO facilitates oxidative damage response of arsenic by IGF2BP3 in an m6A-dependent manner. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134440. [PMID: 38723480 DOI: 10.1016/j.jhazmat.2024.134440] [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: 02/02/2024] [Revised: 04/09/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024]
Abstract
N6-methyladenosine (m6A) is the most common form of internal post-transcriptional methylation observed in eukaryotic mRNAs. The abnormally increased level of m6A within the cells can be catalyzed by specific demethylase fat mass and obesity-associated protein (FTO) and stay in a dynamic and reversible state. However, whether and how FTO regulates oxidative damage via m6A modification remain largely unclear. Herein, by using both in vitro and in vivo models of oxidative damage induced by arsenic, we demonstrated for the first time that exposure to arsenic caused a significant increase in SUMOylation of FTO protein, and FTO SUMOylation at lysine (K)- 216 site promoted the down-regulation of FTO expression in arsenic target organ lung, and therefore, remarkably elevating the oxidative damage via an m6A-dependent pathway by its specific m6A reader insulin-like growth factor-2 mRNA-binding protein-3 (IGF2BP3). Consequently, these findings not only reveal a novel mechanism underlying FTO-mediated oxidative damage from the perspective of m6A, but also imply that regulation of FTO SUMOylation may serve as potential approach for treatment of oxidative damage.
Collapse
Affiliation(s)
- Hongyang Zhang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Qian Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Huifang Han
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Changxin Guo
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yunxiao Zhang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Lixiao Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
21
|
Ji Y, Chen L, Wang Y, Zhang J, Yu Y, Wang M, Wang X, Liu W, Yan B, Xiao L, Song X, Lv C, Chen L. Realistic Nanoplastics Induced Pulmonary Damage via the Crosstalk of Ferritinophagy and Mitochondrial Dysfunction. ACS NANO 2024; 18:16790-16807. [PMID: 38869479 DOI: 10.1021/acsnano.4c02335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The smaller size fraction of plastics may be more substantially existing and detrimental than larger-sized particles. However, reports on nanoplastics (NPs), especially their airborne occurrences and potential health hazards to the respiratory system, are scarce. Previous studies limit the understanding of their real respiratory effects, since sphere-type polystyrene (PS) nanoparticles differ from NPs occurring in nature with respect to their physicochemical properties. Here, we employ a mechanical breakdown method, producing NPs directly from bulk plastic, preserving NP properties in nature. We report that among four relatively high abundance NP materials PS, polyethylene terephthalate (PET), polyvinyl chloride (PVC), and polyethylene (PE) with a size of 100 nm, PVC induced slightly more severe lung toxicity profiles compared to the other plastics. The lung cytotoxicity of NPs is higher than that of commercial PS NPs and comparable to natural particles silicon dioxide (SiO2) and anatase titanium dioxide (TiO2). Mechanistically, BH3-interacting domain death agonist (Bid) transactivation-mediated mitochondrial dysfunction and nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy or ferroptosis are likely common mechanisms of NPs regardless of their chemical composition. This study provides relatively comprehensive data for evaluating the risk of atmospheric NPs to lung health.
Collapse
Affiliation(s)
- Yunxia Ji
- Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| | - Libang Chen
- Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Yunqing Wang
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Jinjin Zhang
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| | - Yue Yu
- Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Meirong Wang
- Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Xiaoyan Wang
- Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Weili Liu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| | - Bing Yan
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Liang Xiao
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Xiaodong Song
- Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Changjun Lv
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| | - Lingxin Chen
- Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
22
|
Liu Y, Wang Y, Zhang J, Peng Q, Wang X, Xiao X, Shi K. Nanotherapeutics targeting autophagy regulation for improved cancer therapy. Acta Pharm Sin B 2024; 14:2447-2474. [PMID: 38828133 PMCID: PMC11143539 DOI: 10.1016/j.apsb.2024.03.019] [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: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/29/2024] [Indexed: 06/05/2024] Open
Abstract
The clinical efficacy of current cancer therapies falls short, and there is a pressing demand to integrate new targets with conventional therapies. Autophagy, a highly conserved self-degradation process, has received considerable attention as an emerging therapeutic target for cancer. With the rapid development of nanomedicine, nanomaterials have been widely utilized in cancer therapy due to their unrivaled delivery performance. Hence, considering the potential benefits of integrating autophagy and nanotechnology in cancer therapy, we outline the latest advances in autophagy-based nanotherapeutics. Based on a brief background related to autophagy and nanotherapeutics and their impact on tumor progression, the feasibility of autophagy-based nanotherapeutics for cancer treatment is demonstrated. Further, emerging nanotherapeutics developed to modulate autophagy are reviewed from the perspective of cell signaling pathways, including modulation of the mammalian target of rapamycin (mTOR) pathway, autophagy-related (ATG) and its complex expression, reactive oxygen species (ROS) and mitophagy, interference with autophagosome-lysosome fusion, and inhibition of hypoxia-mediated autophagy. In addition, combination therapies in which nano-autophagy modulation is combined with chemotherapy, phototherapy, and immunotherapy are also described. Finally, the prospects and challenges of autophagy-based nanotherapeutics for efficient cancer treatment are envisioned.
Collapse
Affiliation(s)
- Yunmeng Liu
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Yaxin Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Jincheng Zhang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Qikai Peng
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xingdong Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xiyue Xiao
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Kai Shi
- College of Pharmacy, Nankai University, Tianjin 300350, China
| |
Collapse
|
23
|
Feng Y, Zhao X, Ruan Z, Li Z, Mo H, Lu F, Shi D. Zinc improves the developmental ability of bovine in vitro fertilization embryos through its antioxidative action. Theriogenology 2024; 221:47-58. [PMID: 38554613 DOI: 10.1016/j.theriogenology.2024.03.013] [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: 12/23/2023] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Zinc, an essential trace mineral, exerts a pivotal influence in various biological processes. Through zinc concentration analysis, we found that the zinc concentration in the bovine embryo in vitro culture (IVC) medium was significantly lower than that in bovine follicular fluid. Therefore, this study explored the impact of zinc sulfate on IVC bovine embryo development and investigated the underlying mechanism. The results revealed a significant decline in zygote cleavage and blastocyst development rates when zinc deficiency was induced using zinc chelator N, N, N', N'-Tetrakis (2-pyridylmethyl) ethylenediamine (TPEN) in culture medium during embryo in vitro culture. The influence of zinc-deficiency was time-dependent. Conversely, supplementing 0.8 μg/mL zinc sulfate to culture medium (CM) increased the cleavage and blastocyst formation rate significantly. Moreover, this supplementation reduced reactive oxygen species (ROS) levels, elevated the glutathione (GSH) levels in blastocysts, upregulated the mRNA expression of antioxidase-related genes, and activated the Nrf2-Keap1-ARE signaling pathways. Furthermore, 0.8 μg/mL zinc sulfate enhanced mitochondrial membrane potential, maintained DNA stability, and enhanced the quality of bovine (in vitro fertilization) IVF blastocysts. In conclusion, the addition of 0.8 μg/mL zinc sulfate to CM could enhance the antioxidant capacity, activates the Nrf2-Keap1-ARE signaling pathways, augment mitochondrial membrane potential, and stabilizes DNA, ultimately improving blastocyst quality and in vitro bovine embryo development.
Collapse
Affiliation(s)
- Yun Feng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning, 530005, China
| | - Xin Zhao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning, 530005, China; Reproductive Medicine Center, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, China
| | - Ziyun Ruan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning, 530005, China
| | - Zhengda Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning, 530005, China
| | - Hongfang Mo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning, 530005, China
| | - Fenghua Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning, 530005, China.
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
24
|
Bautista-Pérez R, Cano-Martínez A, Herrera-Rodríguez MA, Ramos-Godinez MDP, Pérez Reyes OL, Chirino YI, Rodríguez Serrano ZJ, López-Marure R. Oral Exposure to Titanium Dioxide E171 and Zinc Oxide Nanoparticles Induces Multi-Organ Damage in Rats: Role of Ceramide. Int J Mol Sci 2024; 25:5881. [PMID: 38892068 PMCID: PMC11172338 DOI: 10.3390/ijms25115881] [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: 04/10/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Food-grade titanium dioxide (E171) and zinc oxide nanoparticles (ZnO NPs) are common food additives for human consumption. We examined multi-organ toxicity of both compounds on Wistar rats orally exposed for 90 days. Rats were divided into three groups: (1) control (saline solution), (2) E171-exposed, and (3) ZnO NPs-exposed. Histological examination was performed with hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM). Ceramide (Cer), 3-nitrotyrosine (NT), and lysosome-associated membrane protein 2 (LAMP-2) were detected by immunofluorescence. Relevant histological changes were observed: disorganization, inflammatory cell infiltration, and mitochondrial damage. Increased levels of Cer, NT, and LAMP-2 were observed in the liver, kidney, and brain of E171- and ZnO NPs-exposed rats, and in rat hearts exposed to ZnO NPs. E171 up-regulated Cer and NT levels in the aorta and heart, while ZnO NPs up-regulated them in the aorta. Both NPs increased LAMP-2 expression in the intestine. In conclusion, chronic oral exposure to metallic NPs causes multi-organ injury, reflecting how these food additives pose a threat to human health. Our results suggest how complex interplay between ROS, Cer, LAMP-2, and NT may modulate organ function during NP damage.
Collapse
Affiliation(s)
- Rocío Bautista-Pérez
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.)
| | | | | | - Olga Lidia Pérez Reyes
- Departamento de Patología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Yolanda Irasema Chirino
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Zariá José Rodríguez Serrano
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.)
| | - Rebeca López-Marure
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (A.C.-M.)
| |
Collapse
|
25
|
Florance I, Cordani M, Pashootan P, Moosavi MA, Zarrabi A, Chandrasekaran N. The impact of nanomaterials on autophagy across health and disease conditions. Cell Mol Life Sci 2024; 81:184. [PMID: 38630152 PMCID: PMC11024050 DOI: 10.1007/s00018-024-05199-y] [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: 11/02/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 04/19/2024]
Abstract
Autophagy, a catabolic process integral to cellular homeostasis, is constitutively active under physiological and stress conditions. The role of autophagy as a cellular defense response becomes particularly evident upon exposure to nanomaterials (NMs), especially environmental nanoparticles (NPs) and nanoplastics (nPs). This has positioned autophagy modulation at the forefront of nanotechnology-based therapeutic interventions. While NMs can exploit autophagy to enhance therapeutic outcomes, they can also trigger it as a pro-survival response against NP-induced toxicity. Conversely, a heightened autophagy response may also lead to regulated cell death (RCD), in particular autophagic cell death, upon NP exposure. Thus, the relationship between NMs and autophagy exhibits a dual nature with therapeutic and environmental interventions. Recognizing and decoding these intricate patterns are essential for pioneering next-generation autophagy-regulating NMs. This review delves into the present-day therapeutic potential of autophagy-modulating NMs, shedding light on their status in clinical trials, intervention of autophagy in the therapeutic applications of NMs, discusses the potency of autophagy for application as early indicator of NM toxicity.
Collapse
Affiliation(s)
- Ida Florance
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| | - Parya Pashootan
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O Box 14965/161, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O Box 14965/161, Tehran, Iran
| | - 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
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, Taiwan
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
26
|
Li C, Liu R, Xiong Z, Bao X, Liang S, Zeng H, Jin W, Gong Q, Liu L, Guo J. Ferroptosis: a potential target for the treatment of atherosclerosis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:331-344. [PMID: 38327187 PMCID: PMC10984869 DOI: 10.3724/abbs.2024016] [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: 11/06/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Atherosclerosis (AS), the main contributor to acute cardiovascular events, such as myocardial infarction and ischemic stroke, is characterized by necrotic core formation and plaque instability induced by cell death. The mechanisms of cell death in AS have recently been identified and elucidated. Ferroptosis, a novel iron-dependent form of cell death, has been proven to participate in atherosclerotic progression by increasing endothelial reactive oxygen species (ROS) levels and lipid peroxidation. Furthermore, accumulated intracellular iron activates various signaling pathways or risk factors for AS, such as abnormal lipid metabolism, oxidative stress, and inflammation, which can eventually lead to the disordered function of macrophages, vascular smooth muscle cells, and vascular endothelial cells. However, the molecular pathways through which ferroptosis affects AS development and progression are not entirely understood. This review systematically summarizes the interactions between AS and ferroptosis and provides a feasible approach for inhibiting AS progression from the perspective of ferroptosis.
Collapse
Affiliation(s)
- Chengyi Li
- School of MedicineYangtze UniversityJingzhou434020China
| | - Ran Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Zhenyu Xiong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Xue Bao
- School of MedicineYangtze UniversityJingzhou434020China
| | - Sijia Liang
- Department of PharmacologyZhongshan School of MedicineSun Yat-Sen UniversityGuangzhou510120China
| | - Haotian Zeng
- Department of GastroenterologyShenzhen People’s HospitalThe Second Clinical Medical CollegeJinan UniversityShenzhen518000China
| | - Wei Jin
- Department of Second Ward of General PediatricsSuizhou Central HospitalHubei University of MedicineSuizhou441300China
| | - Quan Gong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Lian Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Jiawei Guo
- School of MedicineYangtze UniversityJingzhou434020China
| |
Collapse
|
27
|
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
|
28
|
Zhuo LB, Liu YM, Jiang Y, Yan Z. Zinc oxide nanoparticles induce acute lung injury via oxidative stress-mediated mitochondrial damage and NLRP3 inflammasome activation: In vitro and in vivo studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122950. [PMID: 37979646 DOI: 10.1016/j.envpol.2023.122950] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
The widespread application of zinc oxide nanoparticles (ZnO-NPs) brings convenience to our lives while also renders threats to public health and ecological environment. The lung has been recognized as a primary target of ZnO-NPs, however, the detrimental effects and mechanism of ZnO-NPs on the respiratory system have not been thoroughly characterized so far. To investigate the effect of ZnO-NPs on acute lung injury (ALI), Sprague Dawley rats were intratracheally instilled with ZnO-NPs suspension at doses of 1, 2, and 4 mg/kg/day for 3 consecutive days. Our study revealed that ZnO-NPs induced ALI in rats characterized by increased airway resistance, excessive inflammatory response and lung histological damage. In addition, we identified several molecular biomarkers related to the potential mechanism of ZnO-NP-induced ALI, including oxidative stress, mitochondrial damage, and NLRP3 inflammasome activation. The results of in vitro experiments showed that the viability of A549 cells decreased with the increase in ZnO-NPs concentration. Meanwhile, it was also found that ZnO-NP treatment induced the production of ROS, the decrease in mitochondrial membrane potential and activation of NLRP3 inflammasome in A549 cells. Furthermore, to explore the underlying molecular mechanisms of ZnO-NP-induced ALI, N-acetyl-L-cysteine (a ROS scavenger), Cyclosporin A (an inhibitor for mitochondrial depolarization) and Glibenclamide (an inhibitor for NLRP3 inflammasome activity) were used to pre-treat A549 cells before ZnO-NPs stimulation in the in vitro experiments, respectively. The results from this study suggested that ZnO-NP-induced ROS production triggered the accumulation of damaged mitochondria and assembly of NLRP3 inflammatory complex, leading to maturation and release of IL-1β. Moreover, ZnO-NP-induced NLRP3 inflammasome activation was partly mediated by mitochondrial damage. Taken together, our study suggested that ZnO-NPs induced ALI through oxidative stress-mediated mitochondrial damage and NLRP3 inflammasome activation and provided insight into the mechanisms of ZnO-NPs-induced ALI.
Collapse
Affiliation(s)
- Lai-Bao Zhuo
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yu-Mei Liu
- International School of Public Health and One Health, Hainan Medical University, Haikou, China
| | - Yuhan Jiang
- Department of Built Environment, North Carolina A&T State University, Greensboro, NC, 27411, United States
| | - Zhen Yan
- International School of Public Health and One Health, Hainan Medical University, Haikou, China; School of Public Health, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
29
|
Liu D, Ding J, Li Z, Lu Y. Pachymic acid (PA) inhibits ferroptosis of cardiomyocytes via activation of the AMPK in mice with ischemia/reperfusion-induced myocardial injury. Cell Biol Int 2024; 48:46-59. [PMID: 37750505 DOI: 10.1002/cbin.12090] [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: 07/18/2022] [Revised: 08/02/2023] [Accepted: 09/02/2023] [Indexed: 09/27/2023]
Abstract
Pachymic acid (PA) is a lanostane-type triterpenoid with various pharmacological effects. However, little is known about the effect of PA on myocardial infarction (MI) induced by ischemia/reperfusion (I/R). In this study, we aimed to investigate the protective effect of PA and its underlying mechanism. A cellular MI model was established by oxygen-glucose deprivation and reperfusion (OGD/R) treatment in HL-1 cardiomyocytes, and we found that OGD/R treatment decreased cell viability and glutathione peroxide (GSH-Px) activity, increased Fe2+ concentration and lactate dehydrogenase (LDH) activity, promoted malondialdehyde (MDA) and reactive oxygen species (ROS) production, and inhibited the expression of ferroptosis marker proteins SLC7A11 and GPX4 in a time-dependent manner. OGD/R-induced HL-1 cells were pretreated with different concentrations of PA (0, 20, 40, 60 μg/mL) for 24 h, and toxicological experiments showed that 150 μg/mL PA decreased cell viability, while low concentrations of PA had no toxic effect on cells. 20 μg/mL PA reversed the inhibitory effect of OGD/R on cell viability, reduced MDA and ROS production, and Fe2+ accumulation, increased GSH-Px activity and the expression of SLC7A11 and GPX4, and decreased LDH activity, especially at 60 μg/mL PA. Meanwhile, PA promoted the phosphorylation of IRS-1, AKT, and AMPK proteins in a dose-dependent manner. AICAR, an AMPK activator, inhibited ferroptosis, while STO-609, an AMPK inhibitor, largely abolished the effect of PA on OGD/R-induced ferroptosis of HL-1 cells. In addition, PA inhibited ferroptosis and myocardial I/R injury in wild-type mice and AMPK knockout (AMPK-/- ) mice. Collectively, PA inhibited ferroptosis of cardiomyocytes through activating of the AMPK pathway, thereby alleviating myocardial I/R injury in mice.
Collapse
Affiliation(s)
- Dongmin Liu
- Cardiovascular Department I, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jiru Ding
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenzhen Li
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Youquan Lu
- Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
30
|
Yu S, Wang X, Zhang R, Chen R, Ma L. A review on the potential risks and mechanisms of heavy metal exposure to Chronic Obstructive Pulmonary Disease. Biochem Biophys Res Commun 2023; 684:149124. [PMID: 37897914 DOI: 10.1016/j.bbrc.2023.149124] [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: 08/15/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a chronic disease that affects patients as well as the health and economic stability of society as a whole. At the same time, heavy metal pollution is widely recognized as having a possible impact on the environment and human health. Therefore, these diseases have become important global public health issues. In recent years, researchers have shown great interest in the potential association between heavy metal exposure and the development of COPD, and there has been a substantial increase in the number of related studies. However, we still face the challenge of developing a comprehensive and integrated understanding of this complex association. Therefore, this review aimed to evaluate the existing epidemiological studies to clarify the association between heavy metal exposure and COPD. In addition, we will discuss the biological mechanisms between the two to better understand the multiple molecular pathways and possible mechanisms of action involved, and provide additional insights for the subsequent identification of potential strategies to prevent and control the effects of heavy metal exposure on the development of COPD in individuals and populations.
Collapse
Affiliation(s)
- Shuxia Yu
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Xiaoxia Wang
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Rongxuan Zhang
- Department of Respiratory, The Second People's Hospital of Lanzhou City, 730030, China
| | - Rentong Chen
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Li Ma
- School of Public Health, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
31
|
Yang J, He Y, Zhang M, Liang C, Li T, Ji T, Zu M, Ma X, Zhang Z, Liang C, Zhang Q, Chen Y, Hou L. Programmed initiation and enhancement of cGAS/STING pathway for tumour immunotherapy via tailor-designed ZnFe 2O 4-based nanosystem. EXPLORATION (BEIJING, CHINA) 2023; 3:20230061. [PMID: 38264691 PMCID: PMC10742191 DOI: 10.1002/exp.20230061] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/25/2023] [Indexed: 01/25/2024]
Abstract
The cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/stimulator of interferon genes (STING) signalling pathway has been a promising target for anticancer immunity, but rationally activating and enhancing this pathway in tumour cells is critical. Herein, a glutathione sensitive ZnFe2O4-based nanosystem is developed to programmatically initiate and enhance the STING signalling pathway in tumour cells. The prepared ZnFe2O4 nanoparticles were coated with cancer cell membrane (CCM), which enabled the nanosystem target tumour cells. In tumour cells, ZnFe2O4 nanoparticles could be disintegrated by responding to high level glutathione, and the released Fe3+ generated reactive oxygen species to induce the DNA leakage into the cytoplasm to stimulate cGAS. Then Zn2+ promoted cGAS-DNA phase separation to intensify the cGAS enzymatic activity. In addition, the low dose encapsulation of paclitaxel (PTX) acting as an antimitotic agent (ZnFe2O4-PTX@CCM) ensured the sustained activation of cGAS/STING pathway. The in vitro and in vivo results confirmed that ZnFe2O4-PTX@CCM elevated the cGAS/STING activity, promoted dendritic cell maturation, increased cytotoxic T lymphocyte and natural killer cells infiltration, eventually inhibiting the tumour progress and postoperative recurrence. This study provided feasible references on constructing STING activation nanosystem for tumour immunotherapy.
Collapse
Affiliation(s)
- Jing Yang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Yuping He
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Meng Zhang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Chenglin Liang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Tongtong Li
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Tianjiao Ji
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Mali Zu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Xu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyNational Center for Nanoscience and TechnologyBeijingChina
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingChina
| | - Zhenzhong Zhang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Chun Liang
- Department of Plastic and Reconstructive SurgeryChinese PLA General HospitalBeijingChina
| | - Qixu Zhang
- Department of Plastic SurgeryUniversity of Texas MD Anderson Cancer CenterTexasUSA
| | - Youbai Chen
- Department of Plastic and Reconstructive SurgeryChinese PLA General HospitalBeijingChina
| | - Lin Hou
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
32
|
Qiao D, Zhang T, Tang M. Autophagy regulation by inorganic, organic, and organic/inorganic hybrid nanoparticles: Organelle damage, regulation factors, and potential pathways. J Biochem Mol Toxicol 2023; 37:e23429. [PMID: 37409715 DOI: 10.1002/jbt.23429] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 03/30/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023]
Abstract
The rapid development of nanotechnology requires a more thorough understanding of the potential health effects caused by nanoparticles (NPs). As a programmed cell death, autophagy is one of the biological effects induced by NPs, which maintain intracellular homeostasis by degrading damaged organelles and removing aggregates of defective proteins through lysosomes. Currently, autophagy has been shown to be associated with the development of several diseases. A significant number of research have demonstrated that most NPs can regulate autophagy, and their regulation of autophagy is divided into induction and blockade. Studying the autophagy regulation by NPs will facilitate a more comprehensive understanding of the toxicity of NPs. In this review, we will illustrate the effects of different types of NPs on autophagy, including inorganic NPs, organic NPs, and organic/inorganic hybrid NPs. The potential mechanisms by which NPs regulate autophagy are highlighted, including organelle damage, oxidative stress, inducible factors, and multiple signaling pathways. In addition, we list the factors influencing NPs-regulated autophagy. This review may provide basic information for the safety assessment of NPs.
Collapse
Affiliation(s)
- Dong Qiao
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
33
|
Lekki-Porębski SA, Rakowski M, Grzelak A. Free zinc ions, as a major factor of ZnONP toxicity, disrupts free radical homeostasis in CCRF-CEM cells. Biochim Biophys Acta Gen Subj 2023; 1867:130447. [PMID: 37619691 DOI: 10.1016/j.bbagen.2023.130447] [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: 03/20/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Nanotechnology has become a ubiquitous part of our everyday life. Besides the already-known nanoparticles (NPs), plenty of new nanomaterials are being synthesized every day. Here, we explain the mechanism of the zinc oxide nanoparticles (ZnONPs) cytotoxicity in a cellular model of acute lymphoblastic leukaemia (CCRF-CEM). To do so, we investigated both possible hypotheses about the ZnONPs mechanism of toxicity: a free zinc ions release and/or reactive oxygen species (ROS) generation. Presented here results show that: Our results support the hypothesis that the mechanism of ZnONPs cytotoxicity is based on the release of free zinc ions. Nevertheless, both previously quoted hypotheses incompletely described the mechanism of action of ZnONPs. In this paper, we show that the mechanism of cytotoxicity of ZnONPs is based on the induction of reductive stress in CCRF-CEM cells, which is caused by free zinc ions released from ZnONPs. Therefore, the increase of oxidative stress markers is most likely a secondary response of the cells towards the Zn2+. These results provide a crucial expansion of the zinc ion hypothesis and thus explain the biphasic cellular response of CCRF-CEM cells treated with ZnONPs.
Collapse
Affiliation(s)
- S A Lekki-Porębski
- Cytometry Laboratory, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; The Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, 90-237 Lodz, Poland.
| | - M Rakowski
- Cytometry Laboratory, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; The Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, 90-237 Lodz, Poland
| | - A Grzelak
- Cytometry Laboratory, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| |
Collapse
|
34
|
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
|
35
|
Chen JL, Wu X, Yin D, Jia XH, Chen X, Gu ZY, Zhu XM. Autophagy inhibitors for cancer therapy: Small molecules and nanomedicines. Pharmacol Ther 2023; 249:108485. [PMID: 37406740 DOI: 10.1016/j.pharmthera.2023.108485] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/27/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Autophagy is a conserved process in which the cytosolic materials are degraded and eventually recycled for cellular metabolism to maintain homeostasis. The dichotomous role of autophagy in pathogenesis is complicated. Accumulating reports have suggested that cytoprotective autophagy is responsible for tumor growth and progression. Autophagy inhibitors, such as chloroquine (CQ) and hydroxychloroquine (HCQ), are promising for treating malignancies or overcoming drug resistance in chemotherapy. With the rapid development of nanotechnology, nanomaterials also show autophagy-inhibitory effects or are reported as the carriers delivering autophagy inhibitors. In this review, we summarize the small-molecule compounds and nanomaterials inhibiting autophagic flux as well as the mechanisms involved. The nanocarrier-based drug delivery systems for autophagy inhibitors and their distinct advantages are also described. The progress of autophagy inhibitors for clinical applications is finally introduced, and their future perspectives are discussed.
Collapse
Affiliation(s)
- Jian-Li Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xuan Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Dan Yin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xiao-Hui Jia
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xu Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Ze-Yun Gu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xiao-Ming Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China.
| |
Collapse
|
36
|
Li Z, Yin X, Lyu C, Wang T, Wang W, Zhang J, Wang J, Wang Z, Han C, Zhang R, Guo D, Xu R. Zinc oxide nanoparticles induce toxicity in diffuse large B-cell lymphoma cell line U2932 via activating PINK1/Parkin-mediated mitophagy. Biomed Pharmacother 2023; 164:114988. [PMID: 37307677 DOI: 10.1016/j.biopha.2023.114988] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common type of lymphoma. Zinc oxide (ZnO) nanoparticles have excellent anti-tumor properties in the biomedical field. The present study aimed to explore the underlying mechanism by which ZnO nanoparticles induce toxicity in DLBCL cells (U2932) via the PINK1/Parkin-mediated mitophagy pathway. After U2932 cells were exposed to various concentrations of ZnO nanoparticles, the cell survival rate, reactive oxygen species (ROS) generation, cell cycle arrest, and changes in the expression of PINK1, Parkin, P62, and LC3 were monitored. Moreover, we investigated monodansylcadaverine (MDC) fluorescence intensity and autophagosome and further validated the results using the autophagy inhibitor 3-methyladenine (3-MA). The results showed that ZnO nanoparticles could effectively inhibit the proliferation of U2932 cells and induce cell cycle arrest at the G0/G1 phases. Moreover, ZnO nanoparticles significantly increased ROS production, MDC fluorescence intensity, autophagosome formation, and the expression of PINK1, Parkin, and LC3, and decreased the expression of P62 in U2932 cells. In contrast, the autophagy level was reduced after the intervention of the 3-MA. Overall, ZnO nanoparticles can trigger PINK1/Parkin-mediated mitophagy signaling in U2932 cells, which may be a potential therapeutic approach for DLBCL.
Collapse
Affiliation(s)
- Zonghong Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Xuewei Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chunyi Lyu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Teng Wang
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Jinan 250014, China
| | - Wenhao Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Jiachen Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Jinxin Wang
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan 250014, Shandong Province, China
| | - Zhenzhen Wang
- Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan 250014, Shandong Province, China
| | - Chen Han
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Ruixue Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, No. 48#, Yingxiongshan Road, Jinan, Shandong Province, China.
| | - Ruirong Xu
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Jinan 250014, China; Department of Hematology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369#, Jingshi Road, Jinan 250014, Shandong Province, China; Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
37
|
Ning J, Pei Z, Wang M, Hu H, Chen M, Liu Q, Wu M, Yang P, Geng Z, Zheng J, Du Z, Hu W, Wang Q, Pang Y, Bao L, Niu Y, Leng S, Zhang R. Site-specific Atg13 methylation-mediated autophagy regulates epithelial inflammation in PM2.5-induced pulmonary fibrosis. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131791. [PMID: 37295326 DOI: 10.1016/j.jhazmat.2023.131791] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/02/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
Fine particulate matters (PM2.5) increased the risk of pulmonary fibrosis. However, the regulatory mechanisms of lung epithelium in pulmonary fibrosis remained elusive. Here we developed PM2.5-exposure lung epithelial cells and mice models to investigate the role of autophagy in lung epithelia mediating inflammation and pulmonary fibrosis. PM2.5 exposure induced autophagy in lung epithelial cells and then drove pulmonary fibrosis by activation of NF-κB/NLRP3 signaling pathway. PM2.5-downregulated ALKBH5 protein expression promotes m6A modification of Atg13 mRNA at site 767 in lung epithelial cells. Atg13-mediated ULK complex positively regulated autophagy and inflammation in epithelial cells with PM2.5 treatment. Knockout of ALKBH5 in mice further accelerated ULK complex-regulated autophagy, inflammation and pulmonary fibrosis. Thus, our results highlighted that site-specific m6A methylation on Atg13 mRNA regulated epithelial inflammation-driven pulmonary fibrosis in an autophagy-dependent manner upon PM2.5 exposure, and it provided target intervention strategies towards PM2.5-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Jie Ning
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Zijie Pei
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China
| | - Mengruo Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Huaifang Hu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Meiyu Chen
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Mengqi Wu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Peihao Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Zihan Geng
- Department of Occupation Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jie Zheng
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Zhe Du
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Wentao Hu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Qian Wang
- Experimental Center, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Lei Bao
- Department of Occupation Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yujie Niu
- Department of Occupation Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, PR China
| | - Shuguang Leng
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA; Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, PR China.
| |
Collapse
|
38
|
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
|
39
|
Bi J, Mo C, Li S, Huang M, Lin Y, Yuan P, Liu Z, Jia B, Xu S. Immunotoxicity of metal and metal oxide nanoparticles: from toxic mechanisms to metabolism and outcomes. Biomater Sci 2023. [PMID: 37161951 DOI: 10.1039/d3bm00271c] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The influence of metal and metal oxide nanomaterials on various fields since their discovery has been remarkable. They have unique properties, and therefore, have been employed in specific applications, including biomedicine. However, their potential health risks cannot be ignored. Several studies have shown that exposure to metal and metal oxide nanoparticles can lead to immunotoxicity. Different types of metals and metal oxide nanoparticles may have a negative impact on the immune system through various mechanisms, such as inflammation, oxidative stress, autophagy, and apoptosis. As an essential factor in determining the function and fate of immune cells, immunometabolism may also be an essential target for these nanoparticles to exert immunotoxic effects in vivo. In addition, the biodegradation and metabolic outcomes of metal and metal oxide nanoparticles are also important considerations in assessing their immunotoxic effects. Herein, we focus on the cellular mechanism of the immunotoxic effects and toxic effects of different types of metal and metal oxide nanoparticles, as well as the metabolism and outcomes of these nanoparticles in vivo. Also, we discuss the relationship between the possible regulatory effect of nanoparticles on immunometabolism and their immunotoxic effects. Finally, we present perspectives on the future research and development direction of metal and metal oxide nanomaterials to promote scientific research on the health risks of nanomaterials and reduce their adverse effects on human health.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Siwei Li
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Mingshu Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yunhe Lin
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
40
|
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
|
41
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
42
|
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
|
43
|
Luo D, Wang X, Luo X, Wu S. Low-dose of zeolitic imidazolate framework-8 nanoparticle cause energy metabolism disorder through lysosome-mitochondria dysfunction. Toxicology 2023; 489:153473. [PMID: 36870412 DOI: 10.1016/j.tox.2023.153473] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Understanding the underlying interaction between nanoparticle and organelles is conclusive to the nanotoxicology. According to existing literatures, lysosome is a crucial target of the nanoparticle carrier. Meanwhile, mitochondria could provide the essential energy for nanopaticles entering/exiting the cell. Based on the investigation of lysosome-mitochondria connection, we decoded the effects of low-dose ZIF-8 on energy metabolism, which are still largely obscure beforehand. In this research, low-dose ZIF-8 NPs were utilized to explore the effects on vascular endothelial cells, the first cells exposed to NPs during intravenous injection. Consequently, ZIF-8 could damage the energy metabolism, mainly manifested as mitochondrial fission, the decreased ATP production, and lysosomal dysfuction, which would subsequently affect the cell survival, proliferation and protein expression. This study highlights the fundamental understanding for exploring the regulation of nanoscale ZIF-8 in biological processes and its further application in biomedical field.
Collapse
Affiliation(s)
- Dan Luo
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojiao Wang
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Xin Luo
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Sisi Wu
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
44
|
Anand AS, Jain K, Chauhan A, Prasad DN, Kohli E. Zinc oxide nanoparticles trigger dysfunction of mitochondrial respiratory complexes and repair dynamics in human alveolar cells. Toxicol Ind Health 2023; 39:127-137. [PMID: 36680355 DOI: 10.1177/07482337231152956] [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] [Indexed: 01/22/2023]
Abstract
Zinc oxide nanoparticles (ZnO NP) are commonly used engineered NPs with extensive usage in consumer products, thus leading to direct exposure to humans. The direct route of exposure is through inhalation. Once inhaled, these particles accumulate in the lungs, increasing the chances of respiratory tract illness through cellular organelle damage. Zinc oxide nanoparticle-treated lung cells are reported to display cytotoxicity, increase DNA damage, and induce oxidative stress. The current study focused on the effects of ZnO NPs on mitochondrial dynamics (fission and fusion) in human lung epithelial cells (A549). The lung cells were exposed to ZnO NPs at 50 and 100 μg/ml concentrations, and their mitochondrial dynamics were assessed to understand the effects of the NPs. Treatment with ZnO NPs reduced the activity of mitochondrial complex I and complex III and altered mitochondrial structural and functional characteristics in a concentration-dependent manner. Zinc oxide nanoparticles exposure showed an increase in small and round-shaped mitochondria. The expression of various fission proteins (Drp1 and Fis1) and fusion proteins (Mfn1, Mfn2, and OPA1) was altered upon exposure to ZnO NPs. Our studies showed dysfunction of the mitochondria induced by ZnO NPs. In fibroblast mitochondrial dynamics, fission symbolizes threshold damage. In this paper, we have shown that the mitochondrial fission phenotype increased upon exposure to ZnO NPs. The paper emphasizes that these particles enter mitochondria, triggering a stress response that results in the removal of mitochondria via fission. It provides relevant data for safety guidelines to ensure the safer use of these particles.
Collapse
Affiliation(s)
- Avnika Singh Anand
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Ministry of Defence, Timarpur, Delhi, India
| | - Khushbu Jain
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Ministry of Defence, Timarpur, Delhi, India
| | - Amitabh Chauhan
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Ministry of Defence, Timarpur, Delhi, India
| | - Dipti N Prasad
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Ministry of Defence, Timarpur, Delhi, India
| | - Ekta Kohli
- Neurobiology Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Ministry of Defence, Timarpur, Delhi, India
| |
Collapse
|
45
|
Sun S, Yu M, Yu L, Huang W, Zhu M, Fu Y, Yan L, Wang Q, Ji X, Zhao J, Wu M. Nrf2 silencing amplifies DNA photooxidative damage to activate the STING pathway for synergistic tumor immunotherapy. Biomaterials 2023; 296:122068. [PMID: 36868032 DOI: 10.1016/j.biomaterials.2023.122068] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/02/2023]
Abstract
Photodynamic therapy (PDT)-mediated antitumor immune response depends on oxidative stress intensity and subsequent immunogenic cell death (ICD) in tumor cells, yet the inherent antioxidant system restricts reactive oxygen species (ROS)-associated oxidative damage, which is highly correlated with the upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) and the downstream products, such as glutathione (GSH). Herein, to overcome this dilemma, we designed a versatile nanoadjuvant (RI@Z-P) to enhance the sensitivity of tumor cells to oxidative stress via Nrf2-specific small interfering RNA (siNrf2). The constructed RI@Z-P could significantly amplify photooxidative stress and achieve robust DNA oxidative damage, activating the stimulator of interferon genes (STING)-dependent immune-sensing to produce interferon-β (IFN-β). Additionally, RI@Z-P together with laser irradiation reinforced tumor immunogenicity by exposing or releasing damage-associated molecular patterns (DAMPs), showing the prominent adjuvant effect for promoting dendritic cell (DC) maturation and T-lymphocyte activation and even alleviating the immunosuppressive microenvironment to some extent.
Collapse
Affiliation(s)
- Shengjie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Liu Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Wenxin Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Meishu Zhu
- Department of Burn and Plastic Surgery, Department of Wound Repair, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yanan Fu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lingchen Yan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qiang Wang
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China.
| | - Jing Zhao
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Meiying Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
46
|
Cytotoxic and Bactericidal Effects of Inhalable Ciprofloxacin-Loaded Poly(2-ethyl-2-oxazoline) Nanoparticles with Traces of Zinc Oxide. Int J Mol Sci 2023; 24:ijms24054532. [PMID: 36901963 PMCID: PMC10002581 DOI: 10.3390/ijms24054532] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
The bactericidal effects of inhalable ciprofloxacin (CIP) loaded-poly(2-ethyl-2-oxazoline) (PEtOx) nanoparticles (NPs) with traces of zinc oxide (ZnO) were investigated against clinical strains of the respiratory pathogens Staphylococcus aureus and Pseudomonas aeruginosa. CIP-loaded PEtOx NPs retained their bactericidal activity within the formulations compared to free CIP drugs against these two pathogens, and bactericidal effects were enhanced with the inclusion of ZnO. PEtOx polymer and ZnO NPs did not show bactericidal activity alone or in combination against these pathogens. The formulations were tested to determine the cytotoxic and proinflammatory effects on airway epithelial cells derived from healthy donors (NHBE), donors with chronic obstructive pulmonary disease (COPD, DHBE), and a cell line derived from adults with cystic fibrosis (CFBE41o-) and macrophages from healthy adult controls (HCs), and those with either COPD or CF. NHBE cells demonstrated maximum cell viability (66%) against CIP-loaded PEtOx NPs with the half maximal inhibitory concentration (IC50) value of 50.7 mg/mL. CIP-loaded PEtOx NPs were more toxic to epithelial cells from donors with respiratory diseases than NHBEs, with respective IC50 values of 0.103 mg/mL for DHBEs and 0.514 mg/mL for CFBE41o- cells. However, high concentrations of CIP-loaded PEtOx NPs were toxic to macrophages, with respective IC50 values of 0.002 mg/mL for HC macrophages and 0.021 mg/mL for CF-like macrophages. PEtOx NPs, ZnO NPs, and ZnO-PEtOx NPs with no drug were not cytotoxic to any cells investigated. The in vitro digestibility of PEtOx and its NPs was investigated in simulated lung fluid (SLF) (pH 7.4). The analysed samples were characterized using Fourier transform infrared spectroscopy (ATR-FTIR), scanning electron microscopy (SEM), and UV-Vis spectroscopy. Digestion of PEtOx NPs commenced one week following incubation and was completely digested after four weeks; however, the original PEtOx was not digested after six weeks of incubation. The outcome of this study revealed that PEtOx polymer could be considered an efficient drug delivery carrier in respiratory linings, and CIP-loaded PEtOx NPs with traces of ZnO could be a promising addition to inhalable treatments against resistant bacteria with reduced toxicity.
Collapse
|
47
|
Lu JT, Xiao MK, Feng YY, Wang XY, Qiu LL, Chai YR, Wang TY, Jia YL. Apilimod enhances specific productivity in recombinant CHO cells through cell cycle arrest and mediation of autophagy. Biotechnol J 2023; 18:e2200147. [PMID: 36478399 DOI: 10.1002/biot.202200147] [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: 03/23/2022] [Revised: 08/02/2022] [Accepted: 09/06/2022] [Indexed: 12/12/2022]
Abstract
Chinese hamster ovary (CHO) cells are expected to acquire the ability to produce higher recombinant therapeutic protein levels using various strategies. Genetic engineering targeting the cell cycle and autophagy pathways in the regulation of cell death in CHO cell cultures has received attention for enhancing the production of therapeutic proteins. In this study, we examined the small-molecule compound apilimod, which was found to have a positive influence on recombinant protein expression in CHO cells. This was confirmed by selective blocking of the cell cycle at the G0/G1 phase. Apilimod treatment resulted in decreased expression of cyclin-dependent kinase 3 (CDK3) and Cyclin C and increased expression of cyclin-dependent kinase suppressor p27Kip1, which are critical regulators of G1 cell cycle progression and important targets controlling cell proliferation. Furthermore, total transcription factor EB (TFEB) was lower in apilimod-treated CHO cells than in control cells, resulting in decreased lysosome biogenesis and autophagy with apilimod treatment. These multiple effects demonstrate the potential of apilimod for development as a novel enhancer for the production of recombinant proteins in CHO cell engineering.
Collapse
Affiliation(s)
- Jiang-Tao Lu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| | - Meng-Ke Xiao
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China.,School of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Ying-Ying Feng
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiao-Yin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China.,School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Le-Le Qiu
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yu-Rong Chai
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China.,School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yan-Long Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, Henan, China.,Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
48
|
Kim B, Kim G, Jeon S, Cho WS, Jeon HP, Jung J. Zinc oxide nanoparticles trigger autophagy-mediated cell death through activating lysosomal TRPML1 in normal kidney cells. Toxicol Rep 2023; 10:529-536. [PMID: 37152410 PMCID: PMC10160241 DOI: 10.1016/j.toxrep.2023.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/09/2023] Open
Abstract
Zinc oxide nanoparticles (ZnO NPs) have been widely used in various materials including sunscreens, cosmetics, over-the-counter topical skin products, and pigments. As traces of the used ZnO NPs have been found in the kidney, it is crucial to uncover their potential risks. The aim of this study is to elucidate detrimental effects of ZnO NPs and the molecular mechanism behind their renal toxicity. Cytotoxic effects were measured by MTT assay after HK2 cells were exposed to ZnO NPs for 24 h and IC50 value was determined. ROS and intracellular Zn2+ levels were detected by flow cytometry, and localization of Zn2+ and lysosome was determined by confocal microscopy. Occurrence of autophagy and detection of autophagic flux were determined by Western blot and confocal microscopy, respectively. We performed unpaired student t test for two groups, and one-way ANOVA with Tukey's post hoc for over three groups. ZnO NPs induced cell death in human renal proximal tubule epithelial cells, HK2. Cytosolic Zn2+ caused autophagy-mediated cell death rather than apoptosis. Cytosolic Zn2+ processed in lysosome was released by TRPML1, and inhibition of TRPML1 significantly decreased autophagic flux and cell death. The findings of this study suggest that ZnO NPs strongly induce autophagy-mediated cell death in human kidney cells. Controlling TRPML1 can be potentially used to prevent the kidney from ZnO NPs-induced toxicity.
Collapse
Affiliation(s)
- Boyun Kim
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
| | - Gaeun Kim
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
- Graduate School of Chemical Safety Management, Kyungsung University, Busan, the Republic of Korea
| | - Soyeon Jeon
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, the Republic of Korea
| | - Wan-Seob Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, the Republic of Korea
| | - Hyun Pyo Jeon
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
- Graduate School of Chemical Safety Management, Kyungsung University, Busan, the Republic of Korea
- Correspondence to: Department of SmartBio, College of Life and Health Science, Kyungsung University, 309 Suyeong-ro Room 507-2, Nam-gu, Busan 48434, the Republic of Korea.
| | - Jewon Jung
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
- Graduate School of Chemical Safety Management, Kyungsung University, Busan, the Republic of Korea
- Correspondence to: Department of SmartBio, College of Life and Health Science, Kyungsung University, 309 Suyeong-ro Room 507-2, Nam-gu, Busan 48434, the Republic of Korea.
| |
Collapse
|
49
|
Inhibition of cGAS ameliorates acute lung injury triggered by zinc oxide nanoparticles. Toxicol Lett 2022; 373:62-75. [PMID: 36368621 DOI: 10.1016/j.toxlet.2022.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE Zinc oxide nanoparticles (ZnONPs) have been widely used in various industrial and biomedical fields. Occupational or accidental inhalation exposure to ZnONPs might lead to acute lung injury (ALI). Cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are critical for the initiation and expansion of inflammation and contribute to tissue injury; however, the role and mechanism of the cGAS-STING pathway in ALI-induced by ZnONPs are unclear. METHODS Male C57BL/6 J mice were intratracheally injected with ZnONPs (0.6 mg/kg) or mock. The mice were euthanized and the degree of lung injury was determined 3 days after the instillation of ZnONPs. The BEAS-2B cell line was used as a cell model to investigate the cytotoxicity of ZnONPs in vitro. RESULTS We found that ZnONPs inhalation induced ALI in mice, manifested by exacerbated lung pathological changes, mitochondrial damage, oxidative stress and inflammation. Interestingly, cGAS and STING were activated in the lung tissues of the mice and BEAS-2B lung epithelial cells treated with ZnONPs. More importantly, we illustrated that the cGAS inhibitor RU.521 inhibited the activation of the cGAS-STING pathway, further decreased oxidative stress and inflammation, and led to ameliorated lung injury in mice treated with ZnONPs. CONCLUSION This study demonstrated that ZnONPs trigger the activation of the cGAS-STING pathway, which plays an important role in ZnONPs-induced ALI. Inhibition of cGAS with RU.521 mitigates the oxidative stress induced by ZnONPs, suggesting that targeting the cGAS-STING pathway may be a feasible strategy to ameliorate the pulmonary injury caused by nanoparticles.
Collapse
|
50
|
Kong J, Zhang J, Shen M, Zhang S, Shen P, Ren C. Preparation of manganese(II) oxide doped zinc oxide nanocomposites with improved antibacterial activity via ROS. Chem Phys Lett 2022. [DOI: 10.1016/j.cplett.2022.140053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|