1
|
Hussain A, Saeed A. Hazardous or Advantageous: Uncovering the Roles of Heavy Metals and Humic Substances in Shilajit (Phyto-mineral) with Emphasis on Heavy Metals Toxicity and Their Detoxification Mechanisms. Biol Trace Elem Res 2024; 202:5794-5814. [PMID: 38393486 DOI: 10.1007/s12011-024-04109-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Shilajit is a phyto-mineral diffusion and semi-solid matter used as traditional medicine with extraordinary health benefits. This study provides a comprehensive data on Shilajit with emphasis on heavy metal profile, associated toxicities, and metal detoxification mechanisms by humic substances present in Shilajit. Data was searched across papers and traditional books using Google Scholar, PubMed, Science Direct, Medline, SciELO, Web of Science, and Scopus as key scientific databases. Findings showed that Shilajit is distributed in almost 20 regions of the world with uses against 20 health problems as traditional medicine. With various humic substances, almost 11 biological activities were reported in Shilajit. This phyto-mineral diffusion possesses around 65 heavy metals including the toxic heavy metals like Cu, Al, Pb, As, Cd, and Hg. However, humic substances in Shilajit actively detoxify around 12 heavy metals. The recommended levels of heavy metals by WHO and FDA in herbal drugs is 0.20 and 0.30 ppm for Cd, 1 ppm for Hg, 10.00 ppm for As and Pb, 20 ppm for Cu, and 50 ppm for Zn. The levels of reported metals in Shilajit were found to be lower than the permissible limits set by WHO and FDA, except in few studies where exceeded levels were reported. Shilajit consumption without knowing permissible levels of metals is not safe and could pose serious health problems. Although the humic substances and few metals in Shilajit are beneficial in terms of chelating toxic heavy metals, the data on metal detoxification still needs to be clarified.
Collapse
Affiliation(s)
- Adil Hussain
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research (PCSIR) Laboratories Complex, Ferozepur Road, Lahore, 54600, Punjab, Pakistan.
| | - Asma Saeed
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research (PCSIR) Laboratories Complex, Ferozepur Road, Lahore, 54600, Punjab, Pakistan
| |
Collapse
|
2
|
Schoofs H, Schmit J, Rink L. Zinc Toxicity: Understanding the Limits. Molecules 2024; 29:3130. [PMID: 38999082 PMCID: PMC11243279 DOI: 10.3390/molecules29133130] [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: 05/27/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Zinc, a vital trace element, holds significant importance in numerous physiological processes within the body. It participates in over 300 enzymatic reactions, metabolic functions, regulation of gene expression, apoptosis and immune modulation, thereby demonstrating its essential role in maintaining overall health and well-being. While zinc deficiency is associated with significant health risks, an excess of this trace element can also lead to harmful effects. According to the World Health Organization (WHO), 6.7 to 15 mg per day are referred to be the dietary reference value. An excess of the recommended daily intake may result in symptoms such as anemia, neutropenia and zinc-induced copper deficiency. The European Food Safety Authority (EFSA) defines the tolerable upper intake level (UL) as 25 mg per day, whereas the Food and Drug Administration (FDA) allows 40 mg per day. This review will summarize the current knowledge regarding the calculation of UL and other health risks associated with zinc. For example, zinc intake is not limited to oral consumption; other routes, such as inhalation or topical application, may also pose risks of zinc intoxication.
Collapse
Affiliation(s)
- Hannah Schoofs
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52074 Aachen, Germany
| | - Joyce Schmit
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52074 Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
3
|
Amin R, Pandey R, Vaishali K, Acharya V, Sinha MK, Kumar N. Therapeutic Approaches for the Treatment of Interstitial Lung Disease: An Exploratory Review on Molecular Mechanisms. Mini Rev Med Chem 2024; 24:618-633. [PMID: 37587813 DOI: 10.2174/1389557523666230816090112] [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: 01/12/2023] [Revised: 05/04/2023] [Accepted: 06/09/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Interstitial Lung Diseases (ILDs) are characterized by shortness of breath caused by alveolar wall inflammation and/or fibrosis. OBJECTIVE Our review aims to study the depth of various variants of ILD, diagnostic procedures, pathophysiology, molecular dysfunction and regulation, subject and objective assessment techniques, pharmacological intervention, exercise training and various modes of delivery for rehabilitation. METHOD Articles are reviewed from PubMed and Scopus and search engines. RESULTS ILD is a rapidly progressing disease with a high mortality rate. Each variant has its own set of causal agents and expression patterns. Patients often find it challenging to self-manage due to persistent symptoms and a rapid rate of worsening. The present review elaborated on the pathophysiology, risk factors, molecular mechanisms, diagnostics, and therapeutic approaches for ILD will guide future requirements in the quest for innovative and tailored ILD therapies at the molecular and cellular levels. CONCLUSION The review highlights the rationale for conventional and novel therapeutic approaches for better management of ILD.
Collapse
Affiliation(s)
- Revati Amin
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, India
| | - Ruchi Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar, 844102, India
| | - K Vaishali
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, India
| | - Vishak Acharya
- Department of Pulmonary Medicine, Kasturba Medical College, Manipal Academy of Higher Education, Mangalore, India
| | - Mukesh Kumar Sinha
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar, 844102, India
| |
Collapse
|
4
|
Kadhim AA, Abbas NR, Kadhum HH, Albukhaty S, Jabir MS, Naji AM, Hamzah SS, Mohammed MKA, Al-Karagoly H. Investigating the Effects of Biogenic Zinc Oxide Nanoparticles Produced Using Papaver somniferum Extract on Oxidative Stress, Cytotoxicity, and the Induction of Apoptosis in the THP-1 Cell Line. Biol Trace Elem Res 2023; 201:4697-4709. [PMID: 36662347 DOI: 10.1007/s12011-023-03574-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/14/2023] [Indexed: 01/21/2023]
Abstract
This study investigated the effect of novel zinc oxide nanoparticles (ZnO NPs) biosynthesized employing Papaver somniferum leaf on oxidative stress, necrosis, and apoptosis in the leukemia cancer THP-1 cell. The obtained ZnO was examined using SEM, AFM, and TEM microscopy, which revealed an irregular spherical morphology with a size ranging from 20 to 30 nm, and the UV-vis absorbance revealed a strong absorption peak in the range of 360-370, nm confirming the production of ZnO NPs. THP-1 cells were subjected to an MTT, an EdU proliferation, a lactate dehydrogenase release tests, a reactive oxygen species (ROS) induction experiment, a DAPI staining detection assay, and a flow cytometric analysis for Annexin V to measure the effects of ZnO NPs on cancer cell growth inhibition, apoptosis, and necrosis. Our results show that ZnO NPs inhibit THP-1 line in a concentration-dependent pattern. It was observed that ZnO NPs triggered necrosis (cell death) and apoptosis in the cell line. ZnO NPs massively improved the formation of intracellular ROS, which is crucial in deactivating the development of leukemic cells. In conclusion, ZnO nanoparticles synthesized using Papaver somniferum extract have the ability to inhibit proliferation leukemic cancer cells, making them potential anticancer agents.
Collapse
Affiliation(s)
- Afraa Ali Kadhim
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq
| | | | | | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan, 62001, Iraq
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Majid S Jabir
- Division of Biotechnology, Applied Science Department, University of Technology, Baghdad, Iraq
| | - Amel Muhson Naji
- Department of Optics Techniques, Dijlah University College, Al-Masafi Street, Baghdad, 00964, Iraq
| | - Sawsan S Hamzah
- Dentistry Department, Al-Farahidi University, Baghdad, 00964, Iraq
| | - Mustafa K A Mohammed
- Radiological Techniques Department, Al-Mustaqbal University College, 51001, Hillah, , Babylon, Iraq.
| | - Hassan Al-Karagoly
- Department of Internal and Preventive Medicine, College of Veterinary Medicine, University of Al-Qadisiyah, Al Diwaniyah, Iraq
| |
Collapse
|
5
|
Cary C, Stapleton P. Determinants and mechanisms of inorganic nanoparticle translocation across mammalian biological barriers. Arch Toxicol 2023; 97:2111-2131. [PMID: 37303009 PMCID: PMC10540313 DOI: 10.1007/s00204-023-03528-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/22/2023] [Indexed: 06/13/2023]
Abstract
Biological barriers protect delicate internal tissues from exposures to and interactions with hazardous materials. Primary anatomical barriers prevent external agents from reaching systemic circulation and include the pulmonary, gastrointestinal, and dermal barriers. Secondary barriers include the blood-brain, blood-testis, and placental barriers. The tissues protected by secondary barriers are particularly sensitive to agents in systemic circulation. Neurons of the brain cannot regenerate and therefore must have limited interaction with cytotoxic agents. In the testis, the delicate process of spermatogenesis requires a specific milieu distinct from the blood. The placenta protects the developing fetus from compounds in the maternal circulation that would impair limb or organ development. Many biological barriers are semi-permeable, allowing only materials or chemicals, with a specific set of properties, that easily pass through or between cells. Nanoparticles (particles less than 100 nm) have recently drawn specific concern due to the possibility of biological barrier translocation and contact with distal tissues. Current evidence suggests that nanoparticles translocate across both primary and secondary barriers. It is known that the physicochemical properties of nanoparticles can affect biological interactions, and it has been shown that nanoparticles can breach primary and some secondary barriers. However, the mechanism by which nanoparticles cross biological barriers has yet to be determined. Therefore, the purpose of this review is to summarize how different nanoparticle physicochemical properties interact with biological barriers and barrier products to govern translocation.
Collapse
Affiliation(s)
- Chelsea Cary
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Phoebe Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
6
|
Sengupta A, Dorn A, Jamshidi M, Schwob M, Hassan W, De Maddalena LL, Hugi A, Stucki AO, Dorn P, Marti TM, Wisser O, Stucki JD, Krebs T, Hobi N, Guenat OT. A multiplex inhalation platform to model in situ like aerosol delivery in a breathing lung-on-chip. Front Pharmacol 2023; 14:1114739. [PMID: 36959848 PMCID: PMC10029733 DOI: 10.3389/fphar.2023.1114739] [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: 12/02/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
Prolonged exposure to environmental respirable toxicants can lead to the development and worsening of severe respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD) and fibrosis. The limited number of FDA-approved inhaled drugs for these serious lung conditions has led to a shift from in vivo towards the use of alternative in vitro human-relevant models to better predict the toxicity of inhaled particles in preclinical research. While there are several inhalation exposure models for the upper airways, the fragile and dynamic nature of the alveolar microenvironment has limited the development of reproducible exposure models for the distal lung. Here, we present a mechanistic approach using a new generation of exposure systems, the Cloud α AX12. This novel in vitro inhalation tool consists of a cloud-based exposure chamber (VITROCELL) that integrates the breathing AXLung-on-chip system (AlveoliX). The ultrathin and porous membrane of the AX12 plate was used to create a complex multicellular model that enables key physiological culture conditions: the air-liquid interface (ALI) and the three-dimensional cyclic stretch (CS). Human-relevant cellular models were established for a) the distal alveolar-capillary interface using primary cell-derived immortalized alveolar epithelial cells (AXiAECs), macrophages (THP-1) and endothelial (HLMVEC) cells, and b) the upper-airways using Calu3 cells. Primary human alveolar epithelial cells (AXhAEpCs) were used to validate the toxicity results obtained from the immortalized cell lines. To mimic in vivo relevant aerosol exposures with the Cloud α AX12, three different models were established using: a) titanium dioxide (TiO2) and zinc oxide nanoparticles b) polyhexamethylene guanidine a toxic chemical and c) an anti-inflammatory inhaled corticosteroid, fluticasone propionate (FL). Our results suggest an important synergistic effect on the air-blood barrier sensitivity, cytotoxicity and inflammation, when air-liquid interface and cyclic stretch culture conditions are combined. To the best of our knowledge, this is the first time that an in vitro inhalation exposure system for the distal lung has been described with a breathing lung-on-chip technology. The Cloud α AX12 model thus represents a state-of-the-art pre-clinical tool to study inhalation toxicity risks, drug safety and efficacy.
Collapse
Affiliation(s)
- Arunima Sengupta
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Aurélien Dorn
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- AlveoliX AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Mohammad Jamshidi
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Magali Schwob
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Widad Hassan
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | | | - Andreas Hugi
- AlveoliX AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Andreas O. Stucki
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- *Correspondence: Andreas O. Stucki,
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Thomas M. Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | | | | | - Nina Hobi
- AlveoliX AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Olivier T. Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
7
|
Zheng Z, Liu P, Zhang X, Jingguo xin, Yongjie wang, Zou X, Mei X, Zhang S, Zhang S. Strategies to improve bioactive and antibacterial properties of polyetheretherketone (PEEK) for use as orthopedic implants. Mater Today Bio 2022; 16:100402. [PMID: 36105676 PMCID: PMC9466655 DOI: 10.1016/j.mtbio.2022.100402] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/26/2022] Open
Abstract
Polyetheretherketone (PEEK) has gradually become the mainstream material for preparing orthopedic implants due to its similar elastic modulus to human bone, high strength, excellent wear resistance, radiolucency, and biocompatibility. Since the 1990s, PEEK has increasingly been used in orthopedics. Yet, the widespread application of PEEK is limited by its bio-inertness, hydrophobicity, and susceptibility to microbial infections. Further enhancing the osteogenic properties of PEEK-based implants remains a difficult task. This article reviews some modification methods of PEEK in the last five years, including surface modification of PEEK or incorporating materials into the PEEK matrix. For surface modification, PEEK can be modified by chemical treatment, physical treatment, or surface coating with bioactive substances. For PEEK composite material, adding bioactive filler into PEEK through the melting blending method or 3D printing technology can increase the biological activity of PEEK. In addition, some modification methods such as sulfonation treatment of PEEK or grafting antibacterial substances on PEEK can enhance the antibacterial performance of PEEK. These strategies aim to improve the bioactive and antibacterial properties of the modified PEEK. The researchers believe that these modifications could provide valuable guidance on the future design of PEEK orthopedic implants.
Collapse
|
8
|
Chemistry and lung toxicity of particulate matter emitted from firearms. Sci Rep 2022; 12:20722. [PMID: 36456643 PMCID: PMC9715551 DOI: 10.1038/s41598-022-24856-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Smoke emissions produced by firearms contain hazardous chemicals, but little is known if their properties change depending on firearm and ammunition type and whether such changes affect toxicity outcomes. Pulmonary toxicity was assessed in mice exposed by oropharyngeal aspiration to six different types of smoke-related particulate matter (PM) samples; (1) handgun PM, (2) rifle PM, (3) copper (Cu) particles (a surrogate for Cu in the rifle PM) with and without the Cu chelator penicillamine, (4) water-soluble components of the rifle PM, (5) soluble components with removal of metal ions, and (6) insoluble components of the rifle PM. Gun firing smoke PM was in the respirable size range but the chemical composition varied with high levels of Pb in the handgun and Cu in the rifle smoke. The handgun PM did not induce appreciable lung toxicity at 4 and 24 h post-exposure while the rifle PM significantly increased lung inflammation and reduced lung function. The same levels of pure Cu particles alone and the soluble components from the rifle fire PM increased neutrophil numbers but did not cause appreciable cellular damage or lung function changes when compared to the negative (saline) control. Penicillamine treated rifle PM or Cu, slightly reduced lung inflammation and injury but did not improve the lung function decrements. Chelation of the soluble metal ions from the rifle fire PM neutralized the lung toxicity while the insoluble components induced the lung toxicity to the same degree as the rifle PM. The results show that different firearm types can generate contrasting chemical spectra in their emissions and that the rifle PM effects were mostly driven by water-insoluble components containing high levels of Cu. These findings provide better knowledge of hazardous substances in gun firing smoke and their potential toxicological profile.
Collapse
|
9
|
A A, X J, V A, P V M. L-Cysteine capped zinc oxide nanoparticles induced cellular response on adenocarcinomic human alveolar basal epithelial cells using a conventional and organ-on-a-chip approach. Colloids Surf B Biointerfaces 2022; 211:112300. [PMID: 34974288 DOI: 10.1016/j.colsurfb.2021.112300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/16/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
Zinc oxide nanoparticles (ZnO NPs) are among the well-characterized nanomaterials with multifaceted biomedical applications, including biomedical imaging, drug delivery, and pharmaceutical preparations. The high surface charge of ZnO NPs leads to the agglomeration of the particles. Therefore, surface coating with a suitable ligand can increase colloidal stability. In this present study, in-vitro responses of ZnO NPs capped with a sulfur-containing amino acid, L-cysteine (Cys-ZnO NPs), on A549 cells was investigated. Fourier Transform Infrared Spectroscopy (FTIR) studies were carried out to confirm the capping of ZnO NPs with L-cysteine. Cytotoxic studies using A549 cells demonstrated reduced cytotoxicity in comparison with already reported pristine Zinc Oxide nanoparticles. The cellular uptake is confirmed by fluorescent cytometry. However, a higher concentration (160 µg/mL) of Cys-ZnO NPs led to apoptotic cell death marked by nuclear condensation, mitochondrial membrane depolarization, actin filament condensation, lysosomal damage LDH leakage, intracellular ROS production, blebbing, upregulation of Bax and downregulation of Bcl-2 gene expression. Cys-ZnO NPs treatment was also carried out in cells cultured in a microfluidic lung-on-a-chip device under a physiologically relevant flow rate. The study concluded that the microfluidic-based lung-on-a-chip culture resulted in reduced cell death compared to the conventional condition.
Collapse
Affiliation(s)
- Arathi A
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India
| | - Joseph X
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India
| | - Akhil V
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India
| | - Mohanan P V
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India.
| |
Collapse
|
10
|
Lucas D, Frachon I, Barnier A, Edy P, Tissot V, Dewitte JD, Lodde B. Pneumopathie atypique chez un soudeur : sidérose pulmonaire ? Rev Mal Respir 2022; 39:170-174. [DOI: 10.1016/j.rmr.2021.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 12/06/2021] [Indexed: 10/19/2022]
|
11
|
Medici S, Peana M, Pelucelli A, Zoroddu MA. An updated overview on metal nanoparticles toxicity. Semin Cancer Biol 2021; 76:17-26. [PMID: 34182143 DOI: 10.1016/j.semcancer.2021.06.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Although thousands of different nanoparticles (NPs) have been identified and synthesized to date, well-defined, consistent guidelines to control their exposure and evaluate their potential toxicity have yet to be fully established. As potential applications of nanotechnology in numerous fields multiply, there is an increased awareness of the issue of nanomaterials' toxicity among scientists and producers managing them. An updated inventory of customer products containing NPs estimates that they currently number over 5.000; ten years ago, they were one fifth of this. More often than not, products bear no information regarding the presence of NPs in the indicated list of ingredients or components. Consumers are therefore largely unaware of the extent to which nanomaterials have entered our lives, let alone their potential risks. Moreover, the lack of certainties with regard to the safe use of NPs is curbing their applications in the biomedical field, especially in the diagnosis and treatment of cancer, where they are performing outstandingly but are not yet being exploited as much as they could. The production of radical oxygen species is a predominant mechanism leading to metal NPs-driven carcinogenesis. The release of particularly reactive metal ions capable of crossing cell membranes has also been implicated in NPs toxicity. In this review we discuss the origin, behavior and biological toxicity of different metal NPs with the aim of rationalizing related health hazards and calling attention to toxicological concerns involved in their increasingly widespread use.
Collapse
Affiliation(s)
- Serenella Medici
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy.
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy.
| | - Alessio Pelucelli
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | | |
Collapse
|
12
|
Nanoparticles as a Tool in Neuro-Oncology Theranostics. Pharmaceutics 2021; 13:pharmaceutics13070948. [PMID: 34202660 PMCID: PMC8309086 DOI: 10.3390/pharmaceutics13070948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 11/17/2022] Open
Abstract
The rapid growth of nanotechnology and the development of novel nanomaterials with unique physicochemical characteristics provides potential for the utility of nanomaterials in theranostics, including neuroimaging, for identifying neurodegenerative changes or central nervous system malignancy. Here we present a systematic and thorough review of the current evidence pertaining to the imaging characteristics of various nanomaterials, their associated toxicity profiles, and mechanisms for enhancing tropism in an effort to demonstrate the utility of nanoparticles as an imaging tool in neuro-oncology. Particular attention is given to carbon-based and metal oxide nanoparticles and their theranostic utility in MRI, CT, photoacoustic imaging, PET imaging, fluorescent and NIR fluorescent imaging, and SPECT imaging.
Collapse
|
13
|
Elfsmark L, Ekstrand-Hammarström B, Forsgren N, Lejon C, Hägglund L, Wingfors H. Characterization of toxicological effects of complex nano-sized metal particles using in vitro human cell and whole blood model systems. J Appl Toxicol 2021; 42:203-215. [PMID: 34050537 DOI: 10.1002/jat.4202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 04/27/2021] [Accepted: 05/10/2021] [Indexed: 11/07/2022]
Abstract
Metal oxide fumes form at high temperatures, for instance, during welding or firing ammunition. Inhalation exposure to high levels of airborne metal oxide particles can cause metal fume fever, cardiovascular effects, and lung damage in humans, but the associated underlying pathological mechanisms are still not fully understood. Using human alveolar epithelial cells, vascular endothelial cells, and whole blood model systems, we aimed to elucidate the short-term effects of well-characterized metal particles emitted while firing pistol ammunition. Human lung epithelial cells exposed to gunshot smoke particles (0.1-50 μg/ml) produced reactive oxygen species (ROS) and pro-inflammatory cytokines (interleukin 8 (IL-8), granulocyte-macrophage colony-stimulating factor (GM-CSF)) that activate and recruit immune cells. Particles comprising high copper (Cu) and zinc (Zn) content activated human endothelial cells via a non-ROS-mediated mechanism that triggered immune activation (IL-8, GM-CSF), leukocyte adhesion to the endothelium (soluble intercellular adhesion molecule 1 (sICAM-1)), and secretion of regulators of the acute-phase protein synthesis (interleukin 6 (IL-6)). In human whole blood, metal oxides in gunshot smoke demonstrated intrinsic properties that activated platelets (release of soluble cluster of differentiation 40 ligand (sCD40L), platelet-derived growth factor B-chain homodimer(PDGF-BB), and vascular endothelial growth factor A (VEGF-A)) and blood coagulation and induced concomitant release of pro-inflammatory cytokines from blood leukocytes that further orchestrate thrombogenesis. The model systems applied provide useful tools for health risk assessment of particle exposures, but more studies are needed to further elucidate the mechanisms of metal fume fever and to evaluate the potential risk of long-term cardiovascular diseases.
Collapse
Affiliation(s)
- Linda Elfsmark
- CBRN Defence and Security, Swedish Defence Research Agency, Umeå, Sweden
| | | | - Nina Forsgren
- CBRN Defence and Security, Swedish Defence Research Agency, Umeå, Sweden
| | - Christian Lejon
- CBRN Defence and Security, Swedish Defence Research Agency, Umeå, Sweden
| | - Lars Hägglund
- CBRN Defence and Security, Swedish Defence Research Agency, Umeå, Sweden
| | - Håkan Wingfors
- CBRN Defence and Security, Swedish Defence Research Agency, Umeå, Sweden
| |
Collapse
|
14
|
Wu J, Lai X, Cui G, Chen Q, Liu J, Kang Y, Zhang Y, Feng X, Hu C, Shao L. Dual effects of JNK activation in blood-milk barrier damage induced by zinc oxide nanoparticles. JOURNAL OF HAZARDOUS MATERIALS 2020; 399:122809. [PMID: 32937690 DOI: 10.1016/j.jhazmat.2020.122809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/10/2020] [Accepted: 04/25/2020] [Indexed: 06/11/2023]
Abstract
Zinc oxide nanoparticles (ZnO-NPs) have been extensively applied in our daily life. Humans are at high risk of being exposed to ZnO-NPs, which induce potentially adverse health effects. Although a growing number of studies have investigated the toxic effects of ZnO-NPs, the available data concerning ZnO-NP interactions with the blood-milk barrier (BMB) remain highly limited. Herein, we systematically investigated the damage to BMB integrity induced by ZnO-NPs and the mechanisms involved. ZnO-NPs that were intravenously injected into lactating dams accumulated in the mammary gland and entered into the breast milk, inducing disruption to BMB integrity and changes in the tight junction (TJ) and adherens junction (AJ) components. Furthermore, using an in vitro BMB model composed of EpH4-Ev cells, we verified that ZnO-NP-triggered ROS generation and the activation of MKK4 and JNK are the main mechanism of cell-cell junction damage. More interestingly, JNK activation played different roles in inducing changes in the TJ and AJ complex, and these effects did not need to activate the downstream c-Jun. These data provide more information for understanding ZnO-NP interactions with the BMB and raise concern for the daily use and the intravenous use of ZnO-NPs by lactating mothers.
Collapse
Affiliation(s)
- Junrong Wu
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China
| | - Xuan Lai
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guangman Cui
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiyue Chen
- Stomatological Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoli Feng
- Stomatological Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chen Hu
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China.
| |
Collapse
|
15
|
Bhunia AK, Jha PK, Saha S. Optical and Structural Characterization of ZnO Nanoparticles for Binding Analysis with Semen Sample by Isothermal Titration Calorimetry. BIONANOSCIENCE 2020. [DOI: 10.1007/s12668-020-00788-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
16
|
Zhou M, Xiao L, Yang S, Wang B, Shi T, Tan A, Wang X, Mu G, Chen W. Cross-sectional and longitudinal associations between urinary zinc and lung function among urban adults in China. Thorax 2020; 75:771-779. [DOI: 10.1136/thoraxjnl-2019-213909] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 05/10/2020] [Accepted: 05/27/2020] [Indexed: 12/26/2022]
Abstract
BackgroundExposure to zinc was suggested to be associated with pulmonary damage, but whether zinc exposure affects lung function remains unclear.ObjectivesTo quantify the association between urinary zinc and lung function and explore the potential mechanisms.MethodsUrinary zinc and lung function were measured in 3917 adults from the Wuhan-Zhuhai cohort and were repeated after 3 years of follow-up. Indicators of systemic inflammation (C reactive protein), lung epithelium integrity (club cell secretory protein-16) and oxidative damage (8-hydroxy-2′-deoxyguanosine and 8-isoprostane) were measured at baseline. Linear mixed models were used to estimate the exposure–response relationship between urinary zinc and lung function. Mediation analyses were conducted to assess mediating roles of inflammation and oxidative damage in above relationships.ResultsEach 1-unit increase in log-transformed urinary zinc values was associated with a 35.72 mL decrease in forced vital capacity (FVC) and a 24.89 mL decrease in forced expiratory volume in 1 s (FEV1) in the baseline analyses. In the follow-up analyses, there was a negative association between urinary zinc and FVC among participants with persistent high urinary zinc levels, with an estimated change of −93.31 mL (95% CI −178.47 to −8.14). Furthermore, urinary zinc was positively associated with restrictive ventilatory impairment. The mediation analyses suggested that C reactive protein mediated 8.62% and 8.71% of the associations of urinary zinc with FVC and FEV1, respectively.ConclusionUrinary zinc was negatively associated with lung function, and the systemic inflammation may be one of the underlying mechanisms.
Collapse
|
17
|
Yu Z, Li Q, Wang J, Yu Y, Wang Y, Zhou Q, Li P. Reactive Oxygen Species-Related Nanoparticle Toxicity in the Biomedical Field. NANOSCALE RESEARCH LETTERS 2020; 15:115. [PMID: 32436107 PMCID: PMC7239959 DOI: 10.1186/s11671-020-03344-7] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/10/2020] [Indexed: 05/19/2023]
Abstract
The unique physicochemical characteristics of nanoparticles have recently gained increasing attention in a diverse set of applications, particularly in the biomedical field. However, concerns about the potential toxicological effects of nanoparticles remain, as they have a higher tendency to generate excessive amounts of reactive oxygen species (ROS). Due to the strong oxidation potential, the excess ROS induced by nanoparticles can result in the damage of biomolecules and organelle structures and lead to protein oxidative carbonylation, lipid peroxidation, DNA/RNA breakage, and membrane structure destruction, which further cause necrosis, apoptosis, or even mutagenesis. This review aims to give a summary of the mechanisms and responsible for ROS generation by nanoparticles at the cellular level and provide insights into the mechanics of ROS-mediated biotoxicity. We summarize the literature on nanoparticle toxicity and suggest strategies to optimize nanoparticles for biomedical applications.
Collapse
Affiliation(s)
- Zhongjie Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Qi Li
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
| | - Jing Wang
- Oral Research Center, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Yali Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Qihui Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China.
- Center for Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
18
|
Singh N, Das MK, Ramteke A, R. P. Oxidative stress mediated hepatotoxicity induced by ZNP and modulatory role of fruit extract on male Wistar rat. Toxicol Rep 2020; 7:492-500. [PMID: 32309148 PMCID: PMC7155234 DOI: 10.1016/j.toxrep.2020.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 03/17/2020] [Accepted: 03/21/2020] [Indexed: 01/24/2023] Open
Abstract
Zinc oxide nanoparticles (ZNP) are being used in various fields viz cosmetics industry as UV protectants, in the food packaging industry due to their anti-bacterial properties, in agriculture as micronutrients, etc. Increased applications of ZNPs in our day to day life, leading to the contamination of the surrounding environment posing a direct or indirect health risk. Various reports suggest that fruits and vegetables are a rich source of phytochemicals having antioxidant properties which help in neutralizing ROS generated on metal toxicity of the body. The present study focuses to study the ameliorative effect of apple (Pyrus malus) extract (E) on ZNP induced toxicity. Therefore, animals were grouped, six in each, exposed to various doses of ZNP (50 and 250 mg/kg), ZNP (50 and 250 mg/kg)+E. The studied parameters was: food intake, water intake, antioxidants assay, zinc accumulation, and histological alterations and was compared to control. Investigation revealed that ZNP induces toxicity as revealed by the alteration in the studied parameter, whereas those exposed to ZNP along with Pyrus malus fruit extract try to reduce the toxicity induced by nanoparticles but at low doses only. This ameliorative effect of fruit extract might be due to the presence of antioxidants scavenging the free radicals generated by ZNPs suggesting that antioxidant-rich fruit may have a protective role and have the potential to reduce the nanoparticles mediated oxidative stress.
Collapse
Affiliation(s)
- Neelu Singh
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Monoj Kumar Das
- Cancer Genetics and Chemoprevention Research Group, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028 Assam, India
| | - Anand Ramteke
- Cancer Genetics and Chemoprevention Research Group, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028 Assam, India
| | - Paulraj R.
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
- Corresponding author.
| |
Collapse
|
19
|
Keerthana S, Kumar A. Potential risks and benefits of zinc oxide nanoparticles: a systematic review. Crit Rev Toxicol 2020; 50:47-71. [PMID: 32186437 DOI: 10.1080/10408444.2020.1726282] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- S. Keerthana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| | - A. Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow, Uttar Pradesh, India
| |
Collapse
|
20
|
Nazir S, Rabbani A, Mehmood K, Maqbool F, Shah GM, Khan MF, Sajid M. Antileishmanial activity and cytotoxicity of ZnO-based nano-formulations. Int J Nanomedicine 2019; 14:7809-7822. [PMID: 31576125 PMCID: PMC6767875 DOI: 10.2147/ijn.s203351] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 07/17/2019] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Nanoparticles (NPs) can be toxic due to their nano-range sizes. Zinc oxide (ZnO) has good biocompatibility and is commercially used in cosmetics. Moreover, ZnO NPs have potential biomedical uses, but their safety remains unclear. METHODS A range of doped ZnO NPs was evaluated for antileishmanial activity and in vitro toxicity in brine shrimp and human macrophages, and N-doped ZnO NPs were evaluated for in vivo toxicity in male BALB/C mice. N-doped ZnO NPs were administered via two routes: intra-peritoneal injection and topically as a paste. The dosages were 10, 50, and 100 mg/kg/day for 14 days. RESULTS Topical administration was safe at all dosages, but intra-peritoneal injection displayed toxicity at higher doses, namely, 50 and 100 mg/kg/day. The pathological results for the i.p. dose groups were mild to severe degenerative changes in parenchyma cells, increases in Kupffer cells, disappearance of hepatic plates, increases in cell size, ballooning, cytoplasmic changes, and nuclear pyknosis in the liver. Kidney histology was also altered in the i.p. administration group (dose 100 mg/kg/day), with inflammatory changes in the focal area. We associate pathological abnormalities with the presence of doped ZnO NPs at the diseased site, which was verified by PIXE analysis of the liver and kidney samples of the treated and untreated mice groups. CONCLUSION The toxicity of the doped ZnO NPs can serve as an essential determinant for the effects of ZnO NPs on environmental toxicity and can be used for guidelines for safer use of ZnO-based nanomaterials in topical treatment of leishmaniasis and other biomedical applications.
Collapse
Affiliation(s)
- Samina Nazir
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Atiya Rabbani
- Department of Biochemistry, Hazara University, Mansehra, Pakistan
| | - Khalid Mehmood
- Medical Centre, Quaid-e-Azam University, Islamabad, Pakistan
| | - Farhana Maqbool
- Department of Microbiology, Hazara University, Mansehra, Pakistan
| | | | | | - Muhammad Sajid
- Department of Biochemistry, Hazara University, Mansehra, Pakistan
| |
Collapse
|
21
|
Redox interactions and genotoxicity of metal-based nanoparticles: A comprehensive review. Chem Biol Interact 2019; 312:108814. [PMID: 31509734 DOI: 10.1016/j.cbi.2019.108814] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/11/2019] [Accepted: 09/05/2019] [Indexed: 12/25/2022]
Abstract
Nanotechnology is a growing science that may provide several new applications for medicine, food preservation, diagnostic technologies, and sanitation. Despite its beneficial applications, there are several questions related to the safety of nanomaterials for human use. The development of nanotechnology is associated with some concerns because of the increased risk of carcinogenesis following exposure to nanomaterials. The increased levels of reactive oxygen species (ROS) that are due to exposure to nanoparticles (NPs) are primarily responsible for the genotoxicity of metal NPs. Not all, but most metal NPs are able to directly produce free radicals through the release of metal ions and through interactions with water molecules. Furthermore, the increased production of free radicals and the cell death caused by metal NPs can stimulate reduction/oxidation (redox) reactions, leading to the continuous endogenous production of ROS in a positive feedback loop. The overexpression of inflammatory mediators, such as NF-kB and STATs, the mitochondrial malfunction and the increased intracellular calcium levels mediate the chronic oxidative stress that occurs after exposure to metal NPs. In this paper, we review the genotoxicity of different types of metal NPs and the redox mechanisms that amplify the toxicity of these NPs.
Collapse
|
22
|
Gong G, Jiao Y, Pan Q, Tang H, An Y, Yuan A, Wang K, Huang C, Dai W, Lu Y, Wang S, Zhang J, Su H. Antitumor Effect and Toxicity of an Albumin-Paclitaxel Nanocarrier System Constructed via Controllable Alkali-Induced Conformational Changes. ACS Biomater Sci Eng 2019; 5:1895-1906. [PMID: 33405563 DOI: 10.1021/acsbiomaterials.9b00312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Guangming Gong
- Department of Pharmaceutics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Yongjun Jiao
- Institute of Pathogenic Microbiology, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, Jiangsu 210009, China
| | - Qinqin Pan
- HLA Laboratory, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Hao Tang
- Department of Pharmaceutics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Yanli An
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu 210009, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Kaikai Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Canping Huang
- Research Department, Ringpu Bio-Tech, Tianjin, China
| | - Weimin Dai
- Administrative Committee of Taizhou Medical Hi-tech Zone, China Medical City, Taizhou, Zhejiang 225316, P.R. China
| | - Ying Lu
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Shudong Wang
- Department of Pharmaceutics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Jian Zhang
- National Laboratory of Solid State, Microstructure and Department of Physics, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Hua Su
- Department of Pharmaceutics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| |
Collapse
|
23
|
The pro-inflammatory stimulus of zinc- and copper-containing welding fumes in whole blood assay via protein tyrosine phosphatase 1B inhibition. Sci Rep 2019; 9:1315. [PMID: 30718726 PMCID: PMC6362009 DOI: 10.1038/s41598-018-37803-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
An asymptomatic systemic inflammation after exposure to zinc- and copper-containing welding fumes has been described as mild form of metal fume fever in recent studies. Since chronic systemic inflammation leads to a higher cardiovascular risk, examining the inflammation with the underlying pathomechanism is necessary to estimate and hopefully prevent long-term effects of welding. We established a whole blood assay to investigate the effects of zinc- and copper-containing welding fume particles on the blood immune response. Increased levels of IL-6, IL-8, TNFα and IL-1β determined after 24 hours of exposure indicated an acute systemic inflammatory reaction. In vitro increases of IL-6 were comparable to in vivo increases of serum IL-6 levels in a study with welding fume exposure of human subjects. Inhibition of PTP1B was identified as one pathway responsible for the effects of zinc- and copper-containing welding fumes and therefore welding fume fever. In conclusion, the whole blood assay is a reliable and feasible method to investigate effects of zinc- and copper-containing welding fumes on the immune system and as a surrogate for systemic inflammation and welding fume fever. Future research can utilize whole blood assays to reduce and partially replace human exposure studies for further investigations of welding fume fever.
Collapse
|
24
|
Singh S. Zinc oxide nanoparticles impacts: cytotoxicity, genotoxicity, developmental toxicity, and neurotoxicity. Toxicol Mech Methods 2019; 29:300-311. [DOI: 10.1080/15376516.2018.1553221] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Science and Education, Raebareli, India
| |
Collapse
|
25
|
Krabbe J, Esser A, Kanzler S, Braunschweig T, Kintsler S, Spillner J, Schröder T, Kalverkamp S, Balakirski G, Gerhards B, Rieg AD, Kraus T, Brand P, Martin C. The effects of zinc- and copper-containing welding fumes on murine, rat and human precision-cut lung slices. J Trace Elem Med Biol 2018; 49:192-201. [PMID: 29551464 DOI: 10.1016/j.jtemb.2018.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 01/14/2023]
Abstract
Recently, the pro-inflammatory effects of metal inert gas brazing welding fumes containing zinc and copper have been demonstrated in humans. Here, murine, rat and human precision cut lung slices (PCLS) were incubated in welding fume containing media with 0.1, 1, 10 and 100 μg/ml for 24 or 48 h. 24 h incubation were determined either by incubation for the total time or for only 6 h followed by a 18 h post-incubation phase. Cytotoxicity, proliferation and DNA repair rates, and cytokine levels were determined. Welding fume particle concentrations of 0.1 and 1 μg/ml showed no toxic effects on PCLS of all three species, while for 10 and 100 μg/ml a concentration-dependent toxicity occurred. Proliferation and DNA repair rates were reduced for all tested concentrations and incubation times. Additionally, the cytokine levels in the supernatants were markedly reduced, while after 6 h of exposure with 18 h of post-incubation time a trend towards increased cytokine levels occurred. PCLS are a reliable and feasible method to assess and offer a prediction of toxic effects of welding fume particles on human lungs. Rat PCLS showed similar responses compared to human PCLS and are suitable for further evaluation of toxic effects exerted by welding fume particles.
Collapse
Affiliation(s)
- Julia Krabbe
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; Institute of Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany.
| | - André Esser
- Institute of Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Stephanie Kanzler
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Till Braunschweig
- Institute of Pathology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Svetlana Kintsler
- Institute of Pathology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Jan Spillner
- Departement of Thoracic and Cardiovascular Surgery, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Thomas Schröder
- Department of Surgery, Luisenhospital Aachen, Boxgraben 99, 52064 Aachen, Germany
| | - Sebastian Kalverkamp
- Departement of Thoracic and Cardiovascular Surgery, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Galina Balakirski
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; Departement of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Benjamin Gerhards
- ISF- Welding and Joining Institute, RWTH Aachen University, Pontstraße 49, 52062 Aachen, Germany
| | - Annette D Rieg
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Thomas Kraus
- Institute of Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Peter Brand
- Institute of Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| |
Collapse
|
26
|
Abstract
The aging of the human population has resulted in an unprecedented increase in the incidence and prevalence of age-related diseases, including those of the lung. Idiopathic pulmonary fibrosis is a disease of aging, and is characterized by a progressive decline in lung function and high mortality. Recent studies suggest that mitochondrial dysfunction, which can accompany aging phenotypes, may contribute to the pathogenesis of idiopathic pulmonary fibrosis. In this review, we explore current evidence for mitochondrial dysfunction in alveolar epithelial cells, fibroblasts, and immune cells that participate in the fibrotic process. Further, the fates of these cell populations and the potential to target mitochondrial dysfunction as a therapeutic strategy are discussed.
Collapse
|
27
|
Sipos A, Kim KJ, Chow RH, Flodby P, Borok Z, Crandall ED. Alveolar epithelial cell processing of nanoparticles activates autophagy and lysosomal exocytosis. Am J Physiol Lung Cell Mol Physiol 2018; 315:L286-L300. [PMID: 29722567 DOI: 10.1152/ajplung.00108.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Using confocal microscopy, we quantitatively assessed uptake, processing, and egress of near-infrared (NIR)-labeled carboxylated polystyrene nanoparticles (PNP) in live alveolar epithelial cells (AEC) during interactions with primary rat AEC monolayers (RAECM). PNP fluorescence intensity (content) and colocalization with intracellular vesicles in a cell were determined over the entire cell volume via z stacking. Isotropic cuvette-based microfluorimetry was used to determine PNP concentration ([PNP]) from anisotropic measurements of PNP content assessed by confocal microscopy. Results showed that PNP uptake kinetics and steady-state intracellular content decreased as diameter increased from 20 to 200 nm. For 20-nm PNP, uptake rate and steady-state intracellular content increased with increased apical [PNP] but were unaffected by inhibition of endocytic pathways. Intracellular PNP increasingly colocalized with autophagosomes and/or lysosomes over time. PNP egress exhibited fast Ca2+ concentration-dependent release and a slower diffusion-like process. Inhibition of microtubule polymerization curtailed rapid PNP egress, resulting in elevated vesicular and intracellular PNP content. Interference with autophagosome formation led to slower PNP uptake and markedly decreased steady-state intracellular content. At steady state, cytosolic [PNP] was higher than apical [PNP], and vesicular [PNP] (~80% of intracellular PNP content) exceeded both cytosolic and intracellular [PNP]. These data are consistent with the following hypotheses: 1) autophagic processing of nanoparticles is essential for maintenance of AEC integrity; 2) altered autophagy and/or lysosomal exocytosis may lead to AEC injury; and 3) intracellular [PNP] in AEC can be regulated, suggesting strategies for enhancement of nanoparticle-driven AEC gene/drug delivery and/or amelioration of AEC nanoparticle-related cellular toxicity.
Collapse
Affiliation(s)
- Arnold Sipos
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Kwang-Jin Kim
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California , Los Angeles, California.,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California , Los Angeles, California
| | - Robert H Chow
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California , Los Angeles, California.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Per Flodby
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zea Borok
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Edward D Crandall
- Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California , Los Angeles, California.,Will Rogers Institute Pulmonary Research Center, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California.,Department of Pathology, Keck School of Medicine, University of Southern California , Los Angeles, California.,Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California , Los Angeles, California
| |
Collapse
|
28
|
Jeong J, Kim SH, Lee S, Lee DK, Han Y, Jeon S, Cho WS. Differential Contribution of Constituent Metal Ions to the Cytotoxic Effects of Fast-Dissolving Metal-Oxide Nanoparticles. Front Pharmacol 2018; 9:15. [PMID: 29403385 PMCID: PMC5786562 DOI: 10.3389/fphar.2018.00015] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/05/2018] [Indexed: 12/04/2022] Open
Abstract
The main mechanism of toxicity for fast-dissolving nanoparticles (NPs) is relatively simple as it originates from the intrinsic toxicity of their constituent elements rather than complicated surface reactivity. However, there is little information about the compared toxicity of fast-dissolving NP and its constituent ion, which is essential for understanding the mechanism of NP toxicity and the development of a structure-toxicity relationship (STR) model. Herein, we selected three types of fast-dissolving metal-oxide NPs (CoO, CuO, and ZnO) and constituent metal chlorides (CoCl2, CuCl2, and ZnCl2) to compare dose-response curves between NP and its constituent metal. These materials were treated relevant cell lines for inhalation setting (i.e., differentiated THP-1 cells for macrophages and A549 cells for alveolar epithelial cells) and cytotoxicity as an endpoint was evaluated at 24 h post-incubation. The results showed that CoO and CuO NPs in both cell types showed similar patterns of dose-response curves and cytotoxic potential compared to that of their respective metal chloride. On the other hand, ZnO NPs in both cell types showed a completely different dose-response curve compared to that of ZnCl2: ZnO NPs showed modest slope and much less potential for cytotoxicity compared to that of ZnCl2. These results imply that fast-dissolving metal-oxide NPs are not always have similar dose-response curves and toxic potentials compared to their constituent metal chlorides and this may be due to the differential mechanism of intracellular uptake of these substances and their interaction with intracellular detoxification molecules. Further investigations are needed for the use of toxic potential of metal ions as a predicting factors of fast-dissolving NPs toxicity. In addition, chelating agent specific for dissolved metal ions can be applied for the treatment of these fast-dissolving NPs.
Collapse
Affiliation(s)
- Jiyoung Jeong
- Laboratory of Toxicology, Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| | - Sung-Hyun Kim
- Laboratory of Toxicology, Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| | - Seonghan Lee
- Laboratory of Toxicology, Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| | - Dong-Keon Lee
- Laboratory of Toxicology, Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| | - Youngju Han
- Laboratory of Toxicology, Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| | - Soyeon Jeon
- Laboratory of Toxicology, Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| | - Wan-Seob Cho
- Laboratory of Toxicology, Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| |
Collapse
|
29
|
Wang B, Zhang J, Chen C, Xu G, Qin X, Hong Y, Bose DD, Qiu F, Zou Z. The size of zinc oxide nanoparticles controls its toxicity through impairing autophagic flux in A549 lung epithelial cells. Toxicol Lett 2017; 285:51-59. [PMID: 29289694 DOI: 10.1016/j.toxlet.2017.12.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
Zinc oxide nanoparticles (ZnONPs) widely used in various products, have been concerned with its impact on human health, in particular, on the risk of pulmonary toxicity. Our previous study indicated that ZnONPs could harness autophagy and impair the autophagic flux, which was positively linked to ZnONPs-induced toxicity. The objective of this study was to investigate whether ZnONPs-induced impairment of autophagic flux and cell death in lung epithelial cells is related to the size of ZnONPs. We demonstrate that ZnONPs with the average size of 50 nm could induce toxic effects in A549 lung epithelial cells, including accumulation of autophagosomes (the elevation of LC3B-II/LC3B-I ratio), impaired autophagic flux (the increase of p62 expression), the release of intracellular zinc ions (the increase of FluoZin-3 signal and ZnT1 mRNA expression), mitochondrial damage (the decrease of TMRE signal), lysosomal dysfunction (the aberrant expression of LAMP-2), oxidative stress (the increase of DCFH-DA signal and HO-1 expression) and cell death. Interestingly, ZnONPs with the average size of 200 nm failed to induce autophagy-mediated toxicity. Taken together, our results indicate that the size of ZnONPs is closely correlated with its toxicity, which is probably mediated by induction of impaired autophagic flux. This finding provides an insight into better understating of ZnONPs-associated toxicity, and mitigating the risk to humans and allowing the safer application.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing 400016, China; Post-doctoral Research Stations of Nursing Science, School of Nursing, Chongqing Medical University, Chongqing 400016, China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yueling Hong
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Diptiman D Bose
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
30
|
Branica G, Mladinić M, Omanović D, Želježić D. An alternative approach to studying the effects of ZnO nanoparticles in cultured human lymphocytes: combining electrochemistry and genotoxicity tests. Arh Hig Rada Toksikol 2017; 67:277-288. [PMID: 28033099 DOI: 10.1515/aiht-2016-67-2910] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/01/2016] [Indexed: 11/15/2022] Open
Abstract
Nanoparticle use has increased radically raising concern about possible adverse effects in humans. Zinc oxide nanoparticles (ZnO NPs) are among the most common nanomaterials in consumer and medical products. Several studies indicate problems with their safe use. The aim of our study was to see at which levels ZnO NPs start to produce adverse cytogenetic effects in human lymphocytes as an early attempt toward establishing safety limits for ZnO NP exposure in humans. We assessed the genotoxic effects of low ZnO NP concentrations (1.0, 2.5, 5, and 7.5 μg mL-1) in lymphocyte cultures over 14 days of exposure. We also tested whether low and high-density lymphocytes differed in their ability to accumulate ZnO NPs in these experimental conditions. Primary DNA damage (measured with the alkaline comet assay) increased with nanoparticle concentration in unseparated and high density lymphocytes. The same happened with the fragmentation of TP53 (measured with the comet-FISH). Nanoparticle accumulation was significant only with the two highest concentrations, regardless of lymphocyte density. High-density lymphocytes had significantly more intracellular Zn2+ than light-density ones. Our results suggest that exposure to ZnO NPs in concentrations above 5 μg mL-1 increases cytogenetic damage and intracellular Zn2+ levels in lymphocytes.
Collapse
|
31
|
Liang X, Zhang D, Liu W, Yan Y, Zhou F, Wu W, Yan Z. Reactive oxygen species trigger NF-κB-mediated NLRP3 inflammasome activation induced by zinc oxide nanoparticles in A549 cells. Toxicol Ind Health 2017; 33:737-745. [DOI: 10.1177/0748233717712409] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inhaled zinc oxide nanoparticles (ZnO-NPs) induce lung inflammation associated with oxidative stress. The NLRP3 inflammasome plays a pivotal role in the development of lung inflammation. However, the underlying effects of the NLRP3 inflammasome on ZnO-NPs-induced inflammation remain obscure. In the present study, reactive oxygen species (ROS) generation, expression of NLRP3, caspase-1 p10, and cytokines release of interleukin (IL)-1β and IL-18 were determined after A549 cells were exposed to ZnO-NPs. The ROS scavenger N-acetyl-L-cysteine (NAC), nuclear factor kappa B (NF-κB inhibitor BAY11-7082, and NLRP3 inhibitor glibenclamide (GEL) were used to explore the mechanism of NLRP3 inflammasome activation-induced by ZnO-NPs. ZnO-NPs stimulation induced ROS generation and NF-κB p65 phosphorylation. Similarly, the expression of NLRP3 and caspase-1 p10 and the release of IL-1β and IL-18 were significantly increased after ZnO-NPs treatment, which indicated that the NLRP3 inflammasome was activated by ZnO-NPs. Meanwhile, NAC pretreatment inhibited ZnO-NPs-induced activation of NF-κB and NLRP3 inflammasome. The NF-κB inhibitor BAY11-7082 did not affect ROS production but significantly reduced the NLRP3 inflammasome activation induced by ZnO-NPs. Furthermore, the ability of ZnO-NPs to increase the production of IL-1β and IL-18 was significantly inhibited by GEL. The ZnO-NPs induced the activation of the NLRP3 inflammasome in A549 cells, which might be via a ROS-NF-κB-NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Xiao Liang
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Di Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wenjia Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yingjie Yan
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Fang Zhou
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Zhen Yan
- College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Bengalli R, Gualtieri M, Capasso L, Urani C, Camatini M. Impact of zinc oxide nanoparticles on an in vitro model of the human air-blood barrier. Toxicol Lett 2017; 279:22-32. [DOI: 10.1016/j.toxlet.2017.07.877] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/04/2017] [Accepted: 07/09/2017] [Indexed: 01/10/2023]
|
33
|
Fukui H, Iwahashi H, Nishio K, Hagihara Y, Yoshida Y, Horie M. Ascorbic acid prevents zinc oxide nanoparticle-induced intracellular oxidative stress and inflammatory responses. Toxicol Ind Health 2017; 33:687-695. [PMID: 28854869 DOI: 10.1177/0748233717707361] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Exposure to zinc oxide nanoparticles (ZnO NPs) promotes acute pulmonary toxicity through oxidative stress and inflammation. Furthermore, dissolved zinc from ZnO NPs induces the formation of intracellular reactive oxygen species (ROS). We previously reported that supplemental ascorbic acid (AA) inhibits ZnO NP-induced acute pulmonary toxicity in a rat model; however, the mechanism of this action remains unclear. Therefore, we investigated the effects of AA on ZnO NP-induced cytotoxicity in human lung carcinoma A549 cells. AA was found to suppress intracellular production of ROS, and thus reduce the subsequent inflammation of ZnO NPs. However, intracellular Zn2+ concentrations were higher in AA-treated cells than in AA-untreated cells. AA was found to react with Zn2+ but not with the ZnO NPs themselves. These results suggest the possibility that AA-chelated extracellular Zn2+ and the Zn-AA complex was readily taken up into cell. Even if the intracellular Zn2+ level was high, cytotoxicity might be reduced because the Zn-AA complex was stable. Co-treatment of AA to A549 inhibited ROS production and subsequent intracellular inflammatory responses. These results are consistent with those previously reported from an in vivo model. Thus, two possibilities can be considered about the cytotoxicity-reducing the effect of AA: antioxidant efficacy and chelating effect.
Collapse
Affiliation(s)
- Hiroko Fukui
- 1 United Graduate School of Agricultural Science, Gifu University, Yanagido, Gifu City, Japan
| | - Hitoshi Iwahashi
- 1 United Graduate School of Agricultural Science, Gifu University, Yanagido, Gifu City, Japan
| | - Keiko Nishio
- 2 Health Research Institute (HRI), National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka, Japan
| | - Yoshihisa Hagihara
- 2 Health Research Institute (HRI), National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka, Japan
| | | | | |
Collapse
|
34
|
Zhang J, Qin X, Wang B, Xu G, Qin Z, Wang J, Wu L, Ju X, Bose DD, Qiu F, Zhou H, Zou Z. Zinc oxide nanoparticles harness autophagy to induce cell death in lung epithelial cells. Cell Death Dis 2017; 8:e2954. [PMID: 28749469 PMCID: PMC5550878 DOI: 10.1038/cddis.2017.337] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/06/2017] [Accepted: 06/19/2017] [Indexed: 12/25/2022]
Abstract
Although zinc oxide nanoparticles (ZnONPs) are widely used, they have raised concerns of toxicity in humans. Previous studies have indicated that reactive oxygen species (ROS) and autophagy are involved in the cytotoxicity of ZnONPs, but the regulatory mechanisms between autophagy and ROS remain to be elucidated. Herein, we comprehensively investigated the regulatory mechanism of autophagy and the link between autophagy and ROS in ZnONPs-treated lung epithelial cells. We demonstrated that ZnONPs could induce autophagy, and this process could enhance the dissolution of ZnONPs in lysosomes to release zinc ions. Sequentially, zinc ions released from ZnONPs were able to damage not only lysosomes, leading to impaired autophagic flux, but also mitochondria. Impaired autophagic flux resulted in the accumulation of damaged mitochondria, which could generate excessive ROS to cause cell death. We further demonstrated that the inhibition of autophagy by either pharmacological inhibitors or small interfering RNA (siRNA)-mediated knockdown of Beclin-1 and AMP-activated protein kinase could ameliorate ZnONPs-induced cell death. Moreover, we found that lysosomal-associated membrane protein 1/2 (LAMP-1/2), which were the most abundant highly glycosylated protein in late endosomes/lysosomes, exhibited aberrant expression pattern upon treatment with ZnONPs. Intriguingly, LAMP-2 knockdown, but not LAMP-1 knockdown, could exacerbate the ROS generation and cell death induced by ZnONPs treatment. Meanwhile, LAMP-2 overexpression alleviated ZnONPs-induced cell death, suggesting that LAMP-2 was linked to this toxic phenotype induced by ZnONPs. Our results indicate that autophagic dysfunction could contribute to excessive ROS generation upon treatment with ZnONPs in lung epithelial cells, suggesting that modulating the autophagy process would minimize ZnONPs-associated toxicity.
Collapse
Affiliation(s)
- Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Ge Xu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Zhexue Qin
- Department of Cardiology, XinQiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jian Wang
- National Center for Science and Technology Evaluation (NCSTE), Beijing 100081, China
| | - Lanxiang Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xiangwu Ju
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, and Department of Biochemistry and Molecular Biology, Peking Union Medical College, Tsinghua University, Beijing 100005, China
| | - Diptiman D Bose
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy, Western New England University, Springfield, MA 01119, USA
| | - Feng Qiu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Honghao Zhou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
35
|
Oxidative Stress-Induced DNA Damage by Manganese Dioxide Nanoparticles in Human Neuronal Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5478790. [PMID: 28596964 PMCID: PMC5449756 DOI: 10.1155/2017/5478790] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/13/2017] [Accepted: 04/10/2017] [Indexed: 01/08/2023]
Abstract
Metal nanoparticles have been extensively used in industry as well as in biomedical application. In this work, we have evaluated the toxic potential of manganese dioxide (MnO2) nanoparticles (MNPs) on human neuronal (SH-SY5Y) cells. Cellular toxicity due to MNPs (0, 10, 30, and 60 μg/ml) on the SH-SY5Y cell was observed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and neutral red uptake (NRU) tests. MNPs produced reactive oxygen species (ROS) and declined in mitochondrial membrane potential in the SH-SY5Y cell in dose and duration dependent manner. Moreover, lipid peroxide (LPO), superoxide dismutase (SOD), and catalase (CAT) activities were increased and glutathione was reduced in dose and time dependent manner. A significant upgrade in Hoechst 33342 fluorescence intensity (chromosome condensation) and phosphatidylserine translocation (apoptotic cell) was visualized in cells treated with MNPs for 48 h. On the other hand, caspase-3 activity was increased due to MNPs in SH-SY5Y cells. DNA strand breaks were determined by alkaline single cell gel electrophoresis assay (Comet Assay) and maximum fragmentation of DNA produced due to MNPs (60 μg/ml) for 48 hours. This result provides a basic mechanism of induction of apoptosis and toxicity by MNPs in SH-SY5Y cells.
Collapse
|
36
|
Park EJ, Jeong U, Yoon C, Kim Y. Comparison of distribution and toxicity of different types of zinc-based nanoparticles. ENVIRONMENTAL TOXICOLOGY 2017; 32:1363-1374. [PMID: 27510841 DOI: 10.1002/tox.22330] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/14/2016] [Accepted: 07/17/2016] [Indexed: 06/06/2023]
Abstract
Zinc-based nanoparticles (Zn-NPs), mainly zinc oxide (ZnO) NPs, have promising application in a wide area, but their potential harmful effects on environment and human health have been continuously raised together with their high dissolution rate. In this study, we coated the surface of ZnO NPs with phosphate (ZnP NPs) and sulfide (ZnS NPs) which have very low solubility in water, administered orally (0.5 and 1 mg/kg) to mice for 28 days, and then compared their biodistribution and toxicity. As expected, ZnO NPs were rapidly ionized in an artificial gastric fluid. On the other hand, ZnO NPs were more particlized in an artificial intestinal fluid than ZnP and ZnS NPs. After repeated dosing, all three types of Zn-NPs the most distributed in the spleen and thymus and altered the level of redox reaction-related metal ions in the tissues. We also found that three types of Zn-NPs clearly disturb tissue ion homeostasis and influence immune regulation function. However, there were no remarkable difference in distribution and toxicity following repeated exposure of three types of Zn-NPs, although Na+ and K+ level in the spleen and thymus were notably higher in mice exposed to ZnO NPs compared to ZnP and ZnS NPs. Taken together, we suggest that all three types of Zn-NPs may influence human health by disrupting homeostasis of trace elements and ions in the tissues. In addition, the surface transformation of ZnO NPs with phosphate and sulfide may not attenuate toxicity due to the higher particlization rate of ZnO NPs in the intestine, at least in part. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1363-1374, 2017.
Collapse
Affiliation(s)
- Eun-Jung Park
- Myunggok Eye Research Institute, Konyang University, Daejeon, 302-718, Korea
| | - Uiseok Jeong
- Department of Chemical Engineering, Kwangwoon University, Seoul, 139-701, Korea
| | - Cheolho Yoon
- Seoul Center, Korea Basic Science Institute, Seoul, 126-16, Korea
| | - Younghun Kim
- Department of Chemical Engineering, Kwangwoon University, Seoul, 139-701, Korea
| |
Collapse
|
37
|
Abukabda AB, Stapleton PA, Nurkiewicz TR. Metal Nanomaterial Toxicity Variations Within the Vascular System. Curr Environ Health Rep 2016; 3:379-391. [PMID: 27686080 PMCID: PMC5112123 DOI: 10.1007/s40572-016-0112-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Engineered nanomaterials (ENM) are anthropogenic materials with at least one dimension less than 100 nm. Their ubiquitous employment in biomedical and industrial applications in the absence of full toxicological assessments raises significant concerns over their safety on human health. This is a significant concern, especially for metal and metal oxide ENM as they may possess the greatest potential to impair human health. A large body of literature has developed that reflects adverse systemic effects associated with exposure to these materials, but an integrated mechanistic framework for how ENM exposure influences morbidity remains elusive. This may be due in large part to the tremendous diversity of existing ENM and the rate at which novel ENM are produced. In this review, the influence of specific ENM physicochemical characteristics and hemodynamic factors on cardiovascular toxicity is discussed. Additionally, the toxicity of metallic and metal oxide ENM is presented in the context of the cardiovascular system and its discrete anatomical and functional components. Finally, future directions and understudied topics are presented. While it is clear that the nanotechnology boom has increased our interest in ENM toxicity, it is also evident that the field of cardiovascular nanotoxicology remains in its infancy and continued, expansive research is necessary in order to determine the mechanisms via which ENM exposure contributes to cardiovascular morbidity.
Collapse
Affiliation(s)
- Alaeddin B. Abukabda
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Phoebe A. Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| | - Timothy R. Nurkiewicz
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
38
|
Thomson EM, Breznan D, Karthikeyan S, MacKinnon-Roy C, Vuong NQ, Dabek-Zlotorzynska E, Celo V, Charland JP, Kumarathasan P, Brook JR, Vincent R. Contrasting biological potency of particulate matter collected at sites impacted by distinct industrial sources. Part Fibre Toxicol 2016; 13:65. [PMID: 27906031 PMCID: PMC5134226 DOI: 10.1186/s12989-016-0176-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/23/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Industrial sources contribute a significant proportion of anthropogenic particulate matter (PM) emissions, producing particles of varying composition that may differentially impact health. This study investigated the in vitro toxicity of ambient PM collected near industrial sites in relation to particle size and composition. METHODS Size-fractionated particles (ultrafine, PM0.1-2.5, PM2.5-10, PM>10) were collected in the vicinity of steel, copper, aluminium, and petrochemical industrial sites. Human lung epithelial-like A549 and murine macrophage-like J774A.1 cells were exposed for 24 h to particle suspensions (0, 30, 100, 300 μg/cm2). Particle potency was assessed using cytotoxic (resazurin reduction, lactate dehydrogenase (LDH) release) and inflammatory (cytokine release) assays, and regressed against composition (metals, polycyclic aromatic hydrocarbons (PAHs), endotoxin). RESULTS Coarse (PM2.5-10, PM>10) particle fractions were composed primarily of iron and aluminium; in contrast, ultrafine and fine (PM0.1-2.5) fractions displayed considerable variability in metal composition (especially water-soluble metals) across collection sites consistent with source contributions. Semi-volatile and PM-associated PAHs were enriched in the fine and coarse fractions collected near metal industry. Cell responses to exposure at equivalent mass concentrations displayed striking differences among sites (SITE x SIZE and SITE x DOSE interactions, p < 0.05), suggesting that particle composition, in addition to size, impacted particle toxicity. While both J774A.1 and A549 cells exhibited clear particle size-dependent effects, site-dependent differences were more pronounced in J774A.1 cells, suggesting greater sensitivity to particle composition. Plotting particle potency according to cytotoxic and inflammatory response grouped particles by size and site, and showed that particles of similar composition tended to cluster together. Cytotoxic effects in J774A.1 cells correlated with metal and PAH content, while inflammatory responses were associated primarily with endotoxin content in coarse particles. CONCLUSIONS Industrial sources produce particulate emissions with varying chemical composition that differ in their in vitro potency in relation to particle size and the levels of specific constituents.
Collapse
Affiliation(s)
- Errol M Thomson
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada.
| | - Dalibor Breznan
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada
| | - Subramanian Karthikeyan
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada
| | - Christine MacKinnon-Roy
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada
| | - Ngoc Q Vuong
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada
| | - Ewa Dabek-Zlotorzynska
- Analysis and Air Quality Section, Air Quality Research Division, Atmospheric Science and Technology Directorate, Environment and Climate Change Canada, Ottawa, ON, K1A 0H3, Canada
| | - Valbona Celo
- Analysis and Air Quality Section, Air Quality Research Division, Atmospheric Science and Technology Directorate, Environment and Climate Change Canada, Ottawa, ON, K1A 0H3, Canada
| | - Jean-Pierre Charland
- Analysis and Air Quality Section, Air Quality Research Division, Atmospheric Science and Technology Directorate, Environment and Climate Change Canada, Ottawa, ON, K1A 0H3, Canada
| | - Prem Kumarathasan
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada
| | - Jeffrey R Brook
- Air Quality Processes Research Section, Air Quality Research Division, Atmospheric Science and Technology Directorate, Environment and Climate Change Canada, Toronto, ON, M3H 5T4, Canada
| | - Renaud Vincent
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, K1A 0K9, Canada.
| |
Collapse
|
39
|
Evaluation of Pulmonary Toxicity of Zinc Oxide Nanoparticles Following Inhalation and Intratracheal Instillation. Int J Mol Sci 2016; 17:ijms17081241. [PMID: 27490535 PMCID: PMC5000639 DOI: 10.3390/ijms17081241] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/07/2016] [Accepted: 07/22/2016] [Indexed: 11/24/2022] Open
Abstract
We conducted inhalation and intratracheal instillation studies of zinc oxide (ZnO) nanoparticles in order to examine their pulmonary toxicity. F344 rats were received intratracheal instillation at 0.2 or 1 mg of ZnO nanoparticles with a primary diameter of 35 nm that were well-dispersed in distilled water. Cell analysis and chemokines in bronchoalveolar lavage fluid (BALF) were analyzed at three days, one week, one month, three months, and six months after the instillation. As the inhalation study, rats were exposed to a concentration of inhaled ZnO nanoparticles (2 and 10 mg/m3) for four weeks (6 h/day, 5 days/week). The same endpoints as in the intratracheal instillation study were analyzed at three days, one month, and three months after the end of the exposure. In the intratracheal instillation study, both the 0.2 and the 1.0 mg ZnO groups had a transient increase in the total cell and neutrophil count in the BALF and in the expression of cytokine-induced neutrophil chemoattractant (CINC)-1, CINC-2, chemokine for neutrophil, and heme oxygenase-1 (HO-1), an oxidative stress marker, in the BALF. In the inhalation study, transient increases in total cell and neutrophil count, CINC-1,-2 and HO-1 in the BALF were observed in the high concentration groups. Neither of the studies of ZnO nanoparticles showed persistent inflammation in the rat lung, suggesting that well-dispersed ZnO nanoparticles have low toxicity.
Collapse
|
40
|
Zhao X, Takabayashi F, Ibuki Y. Coexposure to silver nanoparticles and ultraviolet A synergistically enhances the phosphorylation of histone H2AX. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 162:213-222. [PMID: 27383448 DOI: 10.1016/j.jphotobiol.2016.06.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/25/2016] [Indexed: 11/19/2022]
Abstract
Owing to the wide application of silver nanoparticles (AgNPs), the assessment of health risks associated with their use is of great importance. In this study, we revealed that the potential genotoxicity of AgNPs was enhanced by ultraviolet A (UVA) exposure. Three cultured cell lines were treated with AgNPs, followed by exposure to UVA. AgNPs induced phosphorylation of histone H2AX (γ-H2AX) following the formation of DNA double-strand breaks (DSBs), which was synergistically enhanced by UVA exposure. Enhanced γ-H2AX was observed only in cell lines that positively took up AgNPs, and microsized Ag particles, which were difficult to incorporate into cells, showed no γ-H2AX. Incorporation of AgNPs was not increased by UVA exposure. AgNO3 treatment followed by UVA exposure also induced a marked increase in γ-H2AX, indicating that the enhanced γ-H2AX was attributed to Ag ions released from AgNPs. Ag ions reacted with the -SH group of antioxidant molecules, such as glutathione, and induced intracellular oxidative conditions. 8-Hydroxy-2'-deoxyguanosine was formed in the cells treated with AgNPs, which was augmented by UVA irradiation, suggesting that intracellular oxidation caused oxidative DNA damage, leading to the enhanced formation of DSBs and γ-H2AX. Ag has been considered a safe metal; however, our results provide important insights into the influence of sunlight on the genotoxic potency of AgNPs.
Collapse
Affiliation(s)
- Xiaoxu Zhao
- Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga, Shizuoka 422-8526, Japan
| | - Fumiyo Takabayashi
- School of Nursing, University of Shizuoka, 52-1 Yada, Suruga, Shizuoka 422-8526, Japan
| | - Yuko Ibuki
- Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga, Shizuoka 422-8526, Japan.
| |
Collapse
|
41
|
Liu J, Feng X, Wei L, Chen L, Song B, Shao L. The toxicology of ion-shedding zinc oxide nanoparticles. Crit Rev Toxicol 2016; 46:348-84. [DOI: 10.3109/10408444.2015.1137864] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
42
|
Cao Z, Liu D, Zhang Q, Sun X, Li Y. Aluminum Chloride Induces Osteoblasts Apoptosis via Disrupting Calcium Homeostasis and Activating Ca(2+)/CaMKII Signal Pathway. Biol Trace Elem Res 2016; 169:247-53. [PMID: 26138010 DOI: 10.1007/s12011-015-0417-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/17/2015] [Indexed: 12/16/2022]
Abstract
Aluminum promotes osteoblast (OB) apoptosis. Apoptosis is induced by the disordered calcium homeostasis. Therefore, to investigate the relationship between Al-induced OB apoptosis and calcium homeostasis, calvarium OBs from neonatal rats (3-4 days) were cultured and exposed to 0.048-mg/mL Al(3+) or 0.048-mg/mL Al(3+) combined with 5 μM BAPTA-AM (OBs were pretreated with 5 μM BAPTA-AM for 1 h, then added 0.048 mg/mL Al(3+)), respectively. Then OB apoptosis rate, intracellular calcium ions concentration ([Ca(2+)]i), mRNA expression level of calmodulin (CaM), and protein expression levels of CaM and p-CaMKII in OBs were examined. The result showed that AlCl3 increased OB apoptosis rate, and [Ca(2+)]i and p-CaMKII expression levels and decreased CaM expression levels, whereas BAPTA-AM relieved the effects. These results proved that AlCl3 induced OB apoptosis by disrupting the intracellular Ca(2+) homeostasis and activating the Ca(2+)/CaMKII signal pathway. Our findings can provide new insights for revealing the apoptosis mechanism of OBs exposed to AlCl3.
Collapse
Affiliation(s)
- Zheng Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Dawei Liu
- Heilongjiang Province Hospital, Harbin, 150036, China
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Qiuyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xudong Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yanfei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
43
|
Pan CH, Chuang KJ, Chen JK, Hsiao TC, Lai CH, Jones TP, BéruBé KA, Hong GB, Ho KF, Chuang HC. Characterization of pulmonary protein profiles in response to zinc oxide nanoparticles in mice: a 24-hour and 28-day follow-up study. Int J Nanomedicine 2015; 10:4705-16. [PMID: 26251593 PMCID: PMC4524458 DOI: 10.2147/ijn.s82979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Although zinc oxide nanoparticles (ZnONPs) are recognized to cause systemic disorders, little is known about the mechanisms that underlie the time-dependent differences that occur after exposure. The objective of this study was to investigate the mechanistic differences at 24 hours and 28 days after the exposure of BALB/c mice to ZnONPs via intratracheal instillation. An isobaric tag for the relative and absolute quantitation coupled with liquid chromatography/tandem mass spectrometry was used to identify the differential protein expression, biological processes, molecular functions, and pathways. A total of 18 and 14 proteins displayed significant changes in the lung tissues at 24 hours and 28 days after exposure, respectively, with the most striking changes being observed for S100-A9 protein. Metabolic processes and catalytic activity were the main biological processes and molecular functions, respectively, in the responses at the 24-hour and 28-day follow-up times. The glycolysis/gluconeogenesis pathway was continuously downregulated from 24 hours to 28 days, whereas detoxification pathways were activated at the 28-day time-point after exposure. A comprehensive understanding of the potential time-dependent effects of exposure to ZnONPs was provided, which highlights the metabolic mechanisms that may be important in the responses to ZnONP.
Collapse
Affiliation(s)
- Chih-Hong Pan
- Institute of Occupational Safety and Health, Council of Labor Affairs, Executive Yuan, Taiwan ; School of Public Health, National Defense Medical Center, Taipei Medical University, Taipei, Taiwan
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health and Nutrition, Taipei Medical University, Taipei, Taiwan ; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Central University, Taoyuan, Taiwan
| | - Ching-Huang Lai
- School of Public Health, National Defense Medical Center, Taipei Medical University, Taipei, Taiwan
| | - Tim P Jones
- School of Earth and Ocean Sciences, Cardiff University, Cardiff, Wales, UK
| | - Kelly A BéruBé
- School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Gui-Bing Hong
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan
| | - Kin-Fai Ho
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, People's Republic of China ; Shenzhen Municipal Key Laboratory for Health Risk Analysis, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan ; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
44
|
Stan MS, Sima C, Cinteza LO, Dinischiotu A. Silicon-based quantum dots induce inflammation in human lung cells and disrupt extracellular matrix homeostasis. FEBS J 2015; 282:2914-29. [PMID: 26032556 DOI: 10.1111/febs.13330] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/11/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022]
Abstract
Quantum dots (QDs) are nanocrystalline semiconductor materials that have been tested for biological applications such as cancer therapy, cellular imaging and drug delivery, despite the serious lack of information of their effects on mammalian cells. The present study aimed to evaluate the potential of Si/SiO2 QDs to induce an inflammatory response in MRC-5 human lung fibroblasts. Cells were exposed to different concentrations of Si/SiO2 QDs (25-200 μg·mL(-1)) for 24, 48, 72 and 96 h. The results obtained showed that uptake of QDs was dependent on biocorona formation and the stability of nanoparticles in various biological media (minimum essential medium without or with 10% fetal bovine serum). The cell membrane damage indicated by the increase in lactate dehydrogenase release after exposure to QDs was dose- and time-dependent. The level of lysosomes increased proportionally with the concentration of QDs, whereas an accumulation of autophagosomes was also observed. Cellular morphology was affected, as shown by the disruption of actin filaments. The enhanced release of nitric oxide and the increase in interleukin-6 and interleukin-8 protein expression suggested that nanoparticles triggered an inflammatory response in MRC-5 cells. QDs decreased the protein expression and enzymatic activity of matrix metalloproteinase (MMP)-2 and MMP-9 and also MMP-1 caseinase activity, whereas the protein levels of MMP-1 and tissue inhibitor of metalloproteinase-1 increased. The present study reveals for the first time that silicon-based QDs are able to generate inflammation in lung cells and cause an imbalance in extracellular matrix turnover through a differential regulation of MMPs and tissue inhibitor of metalloproteinase-1 protein expression.
Collapse
Affiliation(s)
- Miruna-Silvia Stan
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Romania
| | - Cornelia Sima
- National Institute for Laser, Plasma and Radiation Physics, Bucharest-Magurele, Romania
| | | | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Romania
| |
Collapse
|
45
|
Tada-Oikawa S, Ichihara G, Suzuki Y, Izuoka K, Wu W, Yamada Y, Mishima T, Ichihara S. Zn(II) released from zinc oxide nano/micro particles suppresses vasculogenesis in human endothelial colony-forming cells. Toxicol Rep 2015; 2:692-701. [PMID: 28962405 PMCID: PMC5598154 DOI: 10.1016/j.toxrep.2015.04.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 11/30/2022] Open
Abstract
Zinc oxide (ZnO) nanoparticles have been widely used in industry, cosmetics, and biomedicine. Recent studies suggested that these nanoparticles could have a major impact on the cardiovascular system. Endothelial progenitor cells (EPCs) contribute to postnatal endothelial repair and regeneration. The present study dissected the effects of ZnO nanoparticles on vasculogenesis using human endothelial colony forming cells (ECFCs), which participate in post-natal vasculogenesis. Two types of ZnO particles were used (nano and micro), in addition to zinc chloride solutions with zinc ion concentrations equal to those in ZnO nanoparticles. Twenty-four-hour exposure induced cytotoxicity in a dose-dependent manner and increased ECFCs apoptosis in all groups. The exposure also reduced the functional capacity of ECFCs on Matrix gel to form tubules, compared with the control cells. These effects were associated with downregulation of expression of vascular endothelial growth factor receptor, VEGFR2 and CXC chemokine receptor, CXCR4. The results suggest that ZnO nanoparticles suppress vasculogenesis from ECFCs through downregulation of the expression of receptors related to vasculogenesis. These effects are based the concentration of released Zn(II).
Collapse
Affiliation(s)
- Saeko Tada-Oikawa
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo Univeristy of Science, Noda, Japan
| | - Yuka Suzuki
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Kiyora Izuoka
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Wenting Wu
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiji Yamada
- Life Science Research Center, Mie University, Tsu, Japan
| | - Takashi Mishima
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
- Life Science Research Center, Mie University, Tsu, Japan
| |
Collapse
|
46
|
|
47
|
Investigating the immunomodulatory nature of zinc oxide nanoparticles at sub-cytotoxic levels in vitro and after intranasal instillation in vivo. J Nanobiotechnology 2015; 13:6. [PMID: 25645871 PMCID: PMC4324663 DOI: 10.1186/s12951-015-0067-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/16/2015] [Indexed: 12/27/2022] Open
Abstract
Background This study evaluates the time-dependent pro-inflammatory response of the model human lung epithelial cells (A549) to industrially relevant zinc oxide nanoparticles (ZnO NPs). In terms of toxicity, ZnO-NPs are categorised into the group of high toxicity nanomaterials. However information on pro-inflammatory potential of these NPs at sub-toxic concentrations is limited. Understanding how cellular defense mechanisms function in the presence of sub-cytotoxic concentrations of these NPs is vital. Moreover, there is an urgent need for additional in vivo studies addressing pulmonary toxicity due to accidental inhalation of ZnO NPs. Results Exposure to sub-cytotoxic ZnO NP concentrations (20 μg/mL) induced significant up-regulation of mRNA for the pro-inflammatory cytokine IL-8 and redox stress marker heme oxygenase-1, along with increased release of IL-8. The highest pro-inflammatory response was recorded after 4 to 6 hr exposure to ZnO NPs over a 24 hr period. Pre-treatment of A549 cells with the sulfhydryl antioxidant N-acetyl cysteine (at 5 mM) resulted in significant reduction of the up-regulation of inflammatory markers, confirming the role of reactive oxygen species in the observed immunomodulatory effects, independent of cytotoxicity. Furthermore, we report for the first time that, intranasal instillation of a single dose (5 mg/kg) of pristine or surfactant-dispersed ZnO NPs can cause pulmonary inflammation, already after 24 hr in a murine model. This was confirmed by up-regulation of eotaxin mRNA in the lung tissue and release of pro-inflammatory cytokines in the sera of mice exposed to ZnO NPs. Conclusion Our study highlights that even at sub-cytotoxic doses ZnO NPs can stimulate a strong inflammatory and antioxidant response in A549 cells. ZnO NP mediated cytotoxicity may be the outcome of failure of cellular redox machinery to contain excessive ROS formation. Moreover exposure to a single but relatively high dose of ZnO NPs via intranasal instillation may provoke acute pulmonary inflammatory reactions in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0067-7) contains supplementary material, which is available to authorized users.
Collapse
|
48
|
Roy R, Das M, Dwivedi PD. Toxicological mode of action of ZnO nanoparticles: Impact on immune cells. Mol Immunol 2015; 63:184-92. [DOI: 10.1016/j.molimm.2014.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/01/2014] [Accepted: 08/03/2014] [Indexed: 10/24/2022]
|
49
|
Fukui H, Iwahashi H, Endoh S, Nishio K, Yoshida Y, Hagihara Y, Horie M. Ascorbic acid attenuates acute pulmonary oxidative stress and inflammation caused by zinc oxide nanoparticles. J Occup Health 2015; 57:118-25. [PMID: 25735507 DOI: 10.1539/joh.14-0161-oa] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES It is known that inhalation of zinc oxide nanoparticles (ZnO NPs) induces acute pulmonary dysfunction, including oxidative stress, inflammation, and injury, but there are no reports on how to prevent these adverse effects. We have previously reported that the pulmonary symptoms caused by ZnO NPs were associated with oxidative stress; in the present study, we therefore investigated the use of ascorbic acid (AA), which is known as vitamin C, to prevent these toxic effects. METHODS A ZnO NP dispersion was introduced into rat lungs by intratracheal injection, and thereafter a 1% aqueous AA solution was given as drinking water. Bronchoalveolar lavage fluid was collected at 1 day and 1 week after injection, and lactate dehydrogenase (LDH) activity, heme oxygenase-1 (HO-1), and interleukin-6 (IL-6) levels were measured. In addition, expression of the chemokine cytokine-induced neutrophil chemoattractants (CINCs), HO-1, and metallothionein-1 (MT-1) genes in the lungs were determined. RESULTS Acute oxidative stress induced by ZnO NPs was suppressed by supplying AA. Increases in LDH activity and IL-6 concentration were also suppressed by AA, as was the expression of the CINC-1, CINC-3, and HO-1 genes. CONCLUSIONS Oral intake of AA prevents acute pulmonary oxidative stress and inflammation caused by ZnO NPs. Intake of AA after unanticipated exposure to ZnO NPs is possibly the first effective treatment for the acute pulmonary dysfunction they cause.
Collapse
Affiliation(s)
- Hiroko Fukui
- Health Research Institute (HRI), National Institute of Advanced Industrial Science and Technology (AIST)
| | | | | | | | | | | | | |
Collapse
|
50
|
Wu W, Ichihara G, Hashimoto N, Hasegawa Y, Hayashi Y, Tada-Oikawa S, Suzuki Y, Chang J, Kato M, D'Alessandro-Gabazza CN, Gabazza EC, Ichihara S. Synergistic effect of bolus exposure to zinc oxide nanoparticles on bleomycin-induced secretion of pro-fibrotic cytokines without lasting fibrotic changes in murine lungs. Int J Mol Sci 2014; 16:660-76. [PMID: 25561223 PMCID: PMC4307267 DOI: 10.3390/ijms16010660] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/16/2014] [Indexed: 01/20/2023] Open
Abstract
Zinc oxide (ZnO) nanoparticles are widely used in various products, and the safety evaluation of this manufactured material is important. The present study investigated the inflammatory and fibrotic effects of pulmonary exposure to ZnO nanoparticles in a mouse model of pulmonary fibrosis. Pulmonary fibrosis was induced by constant subcutaneous infusion of bleomycin (BLM). Female C57BL/6Jcl mice were divided into BLM-treated and non-treated groups. In each treatment group, 0, 10, 20 or 30 µg of ZnO nanoparticles were delivered into the lungs through pharyngeal aspiration. Bronchoalveolar lavage fluid (BALF) and the lungs were sampled at Day 10 or 14 after administration. Pulmonary exposure by a single bolus of ZnO nanoparticles resulted in severe, but transient inflammatory infiltration and thickening of the alveolar septa in the lungs, along with the increase of total and differential cell counts in BLAF. The BALF level of interleukin (IL)-1β and transforming growth factor (TGF)-β was increased at Day 10 and 14, respectively. At Day 10, the synergistic effect of BLM and ZnO exposure was detected on IL-1β and monocyte chemotactic protein (MCP)-1 in BALF. The present study demonstrated the synergistic effect of pulmonary exposure to ZnO nanoparticles and subcutaneous infusion of BLM on the secretion of pro-fibrotic cytokines in the lungs.
Collapse
Affiliation(s)
- Wenting Wu
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo University of Science, Noda 278-8510, Japan.
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Yoshinori Hasegawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Yasuhiko Hayashi
- Department of Electrical and Electronic Engineering, Okayama University, Okayama 700-8530, Japan.
| | - Saeko Tada-Oikawa
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan.
| | - Yuka Suzuki
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan.
| | - Jie Chang
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | | | - Esteban C Gabazza
- Department of Immunology, Mie University School of Medicine, Tsu 514-8507, Japan.
| | - Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan.
| |
Collapse
|