1
|
Wang J, Fang X, Wu R, Liu Z, Wang G, Hu Y, Wang H, Pi J, Xu Y. Airway exposure to lithium nickel manganese cobalt oxide particles induces alterations in lung microenvironment and potential kidney and liver damage in mice. Toxicology 2025; 511:154036. [PMID: 39708921 DOI: 10.1016/j.tox.2024.154036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
With the increasing use of lithium-ion batteries, the exposure and health effects of lithium nickel manganate cobalt (NMC), a popular cathode material for the battery, have attracted widespread attention. However, the main absorption routes and target organs of NMC are unknown. This study aims to systematically investigate the main absorption routes and target organs of NMC. Male adult C57BL/6 J mice were subjected to acute exposure to NMC particles (Ni: Mn: Co = 5: 3: 2, mass median geometric diameter 9.15 μm) by intragastric administration, transdermal drug delivery, and oropharyngeal aspiration (OPA). The OPA group showed a significant increase in NMC metal levels in organs and blood compared to the other exposure routes. After OPA treatment (0.5 or 2 mg, once per day, 3 days), significantly increased metal levels were found in the lung, liver and kidney, but there was no dose-response effect. In the lung, obvious inflammation, and significant elevation of white blood cells, neutrophils and eosinophils in bronchoalveolar lavage fluid were observed, all of which showed a dose-response effect. Reduced urine output and renal tubular cell loss, as well as dysregulated metabolic and immune functions as indicated by the hepatic transcriptome, were observed in NMC-exposed mice. Respiratory exposure is the main exposure route of NMC. Short-term respiratory exposure to NMC results in potential damage to the kidney and liver in addition to severe inflammation in the lung.
Collapse
Affiliation(s)
- Junyi Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Xin Fang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Ruirui Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Ziyu Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Gang Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Yuxin Hu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Huihui Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Jingbo Pi
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China
| | - Yuanyuan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, PR China.
| |
Collapse
|
2
|
Dürig J, Calcagni M, Buschmann J. Transition metals in angiogenesis - A narrative review. Mater Today Bio 2023; 22:100757. [PMID: 37593220 PMCID: PMC10430620 DOI: 10.1016/j.mtbio.2023.100757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
The aim of this paper is to offer a narrative review of the literature regarding the influence of transition metals on angiogenesis, excluding lanthanides and actinides. To our knowledge there are not any reviews up to date offering such a summary, which inclined us to write this paper. Angiogenesis describes the process of blood vessel formation, which is an essential requirement for human growth and development. When the complex interplay between pro- and antiangiogenic mediators falls out of balance, angiogenesis can quickly become harmful. As it is so fundamental, both its inhibition and enhancement take part in various diseases, making it a target for therapeutic treatments. Current methods come with limitations, therefore, novel agents are constantly being researched, with metal agents offering promising results. Various transition metals have already been investigated in-depth, with studies indicating both pro- and antiangiogenic properties, respectively. The transition metals are being applied in various formulations, such as nanoparticles, complexes, or scaffold materials. Albeit the increasing attention this field is receiving, there remain many unanswered questions, mostly regarding the molecular mechanisms behind the observed effects. Notably, approximately half of all the transition metals have not yet been investigated regarding potential angiogenic effects. Considering the promising results which have already been established, it should be of great interest to begin investigating the remaining elements whilst also further analyzing the established effects.
Collapse
Affiliation(s)
- Johannes Dürig
- University of Zürich, Faculty of Medicine, Pestalozzistrasse 3, 8032, Zurich, Switzerland
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Maurizio Calcagni
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johanna Buschmann
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
3
|
|
4
|
Badhe RV, Akinfosile O, Bijukumar D, Barba M, Mathew MT. Systemic toxicity eliciting metal ion levels from metallic implants and orthopedic devices - A mini review. Toxicol Lett 2021; 350:213-224. [PMID: 34252509 DOI: 10.1016/j.toxlet.2021.07.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 06/19/2021] [Accepted: 07/07/2021] [Indexed: 12/28/2022]
Abstract
The metal/metal alloy-based implants and prostheses are in use for over a century, and the rejections, revisions, and metal particle-based toxicities were reported concurrently. Complications developed due to metal ions, metal debris, and organo-metallic particles in orthopedic patients have been a growing concern in recent years. It was reported that local and systemic toxicity caused by such released products from the implants is one of the major reasons for implant rejection and revision. Even though the description of environmental metal toxicants and safety limits for their exposure to humans were well established in the literature, an effort was not adequately performed in the case of implant-based metal toxicology. Since the metal ion concentration in serum acts as a possible indicator of the systemic toxicity, this review summarizes the reported human serum safe limits, toxic limits, and concentration range (μg/L, ppb, etc.) for mild to severe symptoms of six (cardiac, hepatic, neuro, nephron, dermal and endocrine) systemic toxicities for twelve most commonly used metallic implants. It also covers the widely used metal ion quantification techniques and systemic toxicity treatments reported.
Collapse
Affiliation(s)
- Ravindra V Badhe
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA
| | - Obakanyin Akinfosile
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA
| | - Divya Bijukumar
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA
| | | | - Mathew T Mathew
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA.
| |
Collapse
|
5
|
Zhu W, Liu Y, Zhang W, Fan W, Wang S, Gu JH, Sun H, Liu F. Selenomethionine protects hematopoietic stem/progenitor cells against cobalt nanoparticles by stimulating antioxidant actions and DNA repair functions. Aging (Albany NY) 2021; 13:11705-11726. [PMID: 33875618 PMCID: PMC8109066 DOI: 10.18632/aging.202865] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/22/2021] [Indexed: 01/13/2023]
Abstract
Hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) can differentiate into all blood lineages to maintain hematopoiesis, wound healing, and immune functions. Recently, cobalt-chromium alloy casting implants have been used extensively in total hip replacements; however, cobalt nanoparticles (CoNPs) released from the alloy were toxic to HSCs and HPCs. We aimed to investigate the mechanism underlying the toxic effect of CoNPs on HSCs/HPCs and to determine the protective effect of selenomethionine (SeMet) against CoNPs in vitro and in vivo. Human and rat CD34+ HSCs/HPCs were isolated from cord blood and bone marrow, respectively. CoNPs decreased the viability of CD34+ HSCs/HPCs and increased apoptosis. SeMet attenuated the toxicity of CoNPs by enhancing the antioxidant ability of cells. The protective effect of SeMet was not completely abolished after adding H2O2 to abrogate the improvement of the antioxidant capacity by SeMet. SeMet and CoNPs stimulated ATM/ATR DNA damage response signals and inhibited cell proliferation. Unlike CoNPs, SeMet did not damage the DNA, and cell proliferation recovered after removing SeMet. SeMet inhibited the CoNP-induced upregulation of hypoxia inducible factor (HIF)-1α, thereby disrupting the inhibitory effect of HIF-1α on breast cancer type 1 susceptibility protein (BRCA1). Moreover, SeMet promoted BRCA1-mediated ubiquitination of cyclin B by upregulating UBE2K. Thus, SeMet enhanced cell cycle arrest and DNA repair post-CoNP exposure. Overall, SeMet protected CD34+ HSCs/HPCs against CoNPs by stimulating antioxidant activity and DNA repair.
Collapse
Affiliation(s)
- Wenfeng Zhu
- Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Yake Liu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Weinan Zhang
- Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Wentao Fan
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Siqi Wang
- Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province, China.,Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Huanjian Sun
- Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Fan Liu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
6
|
A paradox: Fe2+-containing agents decreased ROS and apoptosis induced by CoNPs in vascular endothelial cells by inhibiting HIF-1α. Biosci Rep 2021; 41:227394. [PMID: 33345265 PMCID: PMC7796189 DOI: 10.1042/bsr20203456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/27/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023] Open
Abstract
Cobalt nanoparticles (CoNPs) released from hip joint implants are known to have a toxic effect on several organs probably through increasing reactive oxygen species (ROS). Ferrous ion (Fe2+) is well-known to enhance oxidative stress by catalysing the production of ROS. However, in our pilot study, we found that Fe2+ conversely inhibited the ROS production induced by CoNPs. To elucidate the underlying mechanism, the present study treated vascular endothelial HUVEC and HMEC-1 cells with CoNPs alone or in combination with ferrous lactate [Fe(CH3CHOHCOO)2], ferrous succinate [Fe(CH2COO)2], and ferrous chloride (FeCl2). CoNP toxicity was evaluated by measuring cell viability, rate of apoptosis and lactose dehydrogenase (LDH) release, and intracellular ROS levels. Treatment with CoNPs decreased cell viability, LDH release, and ROS production and increased apoptosis. CoNPs increased hypoxia-inducible factor-1α (HIF-1α) protein level and mRNA levels of vascular endothelial growth factor (VEGF) and glucose transporter 1 (GLUT1) downstream of HIF-1α signalling. Silencing HIF-1α attenuated CoNP toxicity, as seen by recovery of cell viability, LDH release, and ROS levels and reduced apoptosis. CoNPs caused a pronounced reduction of Fe2+ in cells, but supplementation with Fe(CH3CHOHCOO)2, Fe(CH2COO)2, and FeCl2 restored Fe2+ levels and inhibited HIF-1α activation. Moreover, all three Fe2+-containing agents conferred protection from CoNPs; Fe(CH3CHOHCOO)2 and Fe(CH2COO)2 more effectively than FeCl2. In summary, the present study revealed that CoNPs exert their toxicity on human vascular endothelial cells by depleting intracellular Fe2+ level, which causes activation of HIF-1α signalling. Supplements of Fe2+, especially in the form of Fe(CH3CHOHCOO)2 and Fe(CH2COO)2, mitigated CoNP toxicity.
Collapse
|
7
|
Skalny AV, Gluhcheva Y, Ajsuvakova OP, Pavlova E, Petrova E, Rashev P, Vladov I, Shakieva RA, Aschner M, Tinkov AA. Perinatal and early-life cobalt exposure impairs essential metal metabolism in immature ICR mice. Food Chem Toxicol 2021; 149:111973. [PMID: 33421458 DOI: 10.1016/j.fct.2021.111973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 12/30/2022]
Abstract
The objective of the present study was to assess the impact of cobalt (Co) exposure on tissue distribution of iron (Fe), copper (Cu), manganese (Mn), and zinc (Zn), as well as serum hepcidin levels in immature mice (18, 25, 30 days). Pregnant mice were exposed to 75 mg/kg b.w. cobalt chloride (CoCl2 × 6H2O) with drinking water starting from 3 days before delivery and during lactation. At weaning (day 25) the offspring were separated and housed in individual cages with subsequent exposure to 75 mg/kg b.w. CoCl2 until 30 days postnatally. Evaluation of tissue metal levels was performed by an inductively coupled plasma-mass spectrometry (ICP-MS). Serum hepcidin level was assayed by enzyme linked immunosorbent assay (ELISA). Cobalt exposure resulted in a time- and tissue-dependent increase in Co levels in kidney, spleen, liver, muscle, erythrocytes, and serum on days 18, 25, and 30. In parallel with increasing Co levels, CoCl2 exposure resulted in a significant accumulation of Cu, Fe, Mn, and Zn in the studied tissues, with the effect being most pronounced in 25-day-old mice. Cobalt exposure significantly increased serum hepcidin levels only in day18 mice. The obtained data demonstrate that Co exposure may alter essential metal metabolism in vivo.
Collapse
Affiliation(s)
- Anatoly V Skalny
- Yaroslavl State University, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Moscow, 119146, Russia; KG Razumovsky Moscow State University of Technologies and Management, Moscow, Russia
| | - Yordanka Gluhcheva
- Institute of Experimental Morphology, Pathology and Anthropology with Museum - Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Olga P Ajsuvakova
- Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, Orenburg, 460000, Russia
| | - Ekaterina Pavlova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum - Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Emilia Petrova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum - Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Pavel Rashev
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov" - Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Ivelin Vladov
- Institute of Experimental Morphology, Pathology and Anthropology with Museum - Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | | | - Michael Aschner
- IM Sechenov First Moscow State Medical University, Moscow, 119146, Russia; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexey A Tinkov
- Yaroslavl State University, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Moscow, 119146, Russia.
| |
Collapse
|
8
|
Liu Y, Zhu W, Ni D, Zhou Z, Gu JH, Zhang W, Sun H, Liu F. Alpha lipoic acid antagonizes cytotoxicity of cobalt nanoparticles by inhibiting ferroptosis-like cell death. J Nanobiotechnology 2020; 18:141. [PMID: 33008409 PMCID: PMC7532644 DOI: 10.1186/s12951-020-00700-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/24/2020] [Indexed: 12/20/2022] Open
Abstract
As a main element in the hard metal industry, cobalt is one of the major components of human metal implants. Cobalt-containing implants, especially joint prostheses used for artificial joint replacement, can be corroded due to the complex physiological environment in vivo, producing a large number of nanoscale cobalt particles (Cobalt Nanoparticles, CoNPs). These CoNPs can be first accumulated around the implant to cause adverse local reactions and then enter into the blood vessels followed by reaching the liver, heart, brain, kidney, and other organs through systematic circulation, which leads to multi-system toxicity symptoms. To ensure the long-term existence of cobalt-containing implants in the body, it is urgently required to find out a safe and effective detoxification drug. Herein, we have demonstrated that CoNPs could induce the ferroptosis-like cell death through the enhancement of intracellular reactive oxygen species (ROS) level, cytoplasmic Fe2+ level, lipid peroxidation, and consumption of reduced glutathione (GSH) as well as inhibition of glutathione peroxidase 4 (GPX4) activity. Importantly, α-lipoic acid (ALA), a natural antioxidant with the capability to scavenge free radicals and chelate toxic metals, was found to efficiently alleviate the adverse effects of CoNPs. The present study illustrates a new mechanism of CoNPs mediated by ferroptosis-like cytotoxicity and discloses an effective method for the detoxification of CoNPs by employing the natural antioxidant of ALA, providing a basis for further in vivo detoxification study.![]()
Collapse
Affiliation(s)
- Yake Liu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.,Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Wenfeng Zhu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.,Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Dalong Ni
- Department of Radiology, University of Wisconsin-Madison, 11111 Highland Avenue, Madison, WI, 53705, USA
| | - Zihua Zhou
- Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Weinan Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.,Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Huanjian Sun
- Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Fan Liu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
9
|
Rasool A, Zulfajri M, Gulzar A, Hanafiah MM, Unnisa SA, Mahboob M. In vitro effects of cobalt nanoparticles on aspartate aminotransferase and alanine aminotransferase activities of wistar rats. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2020; 26:e00453. [PMID: 32368512 PMCID: PMC7184135 DOI: 10.1016/j.btre.2020.e00453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 12/22/2022]
Abstract
Cobalt nanoparticles (Co-NPs) have been extensively used in clinical practices and medical diagnosis. In this study, the potential toxicity effects of Co-NPs with special emphasis over the biochemical enzyme activities, such as aspartate aminotransferase (ASAT) and alanine aminotransferase (ALAT) in serum, liver, and kidney of Wistar rats were investigated. This toxicity measurement of nanomaterials can support the toxicological data. The biochemical enzymatic variations are powerful tools for the assessment of toxicity. ASAT and ALAT enzymes have been widely used to predict tissue-specific toxicities associated with xenobiotic. The biochemical changes induced by Co-NPs have significance in their toxicological studies because the alterations in biochemical parameters before clinical symptoms indicate either their toxicant safety or detrimental effect. Herein, Co-NPs with particle size <50 nm significantly activated ASAT and ALAT enzymes in the serum, liver, and kidney of rats at concentration-dependent order.
Collapse
Affiliation(s)
- Akhtar Rasool
- Toxicology Unit, Applied Biology Division, CSIR‐Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana India
- Department of Environmental Sciences, UCS, Osmania University, Hyderabad, 500007, Telangana, India
| | - Muhammad Zulfajri
- Department of Chemistry Education, Universitas Serambi Mekkah, Banda Aceh 23245, Aceh, Indonesia
| | - Arif Gulzar
- Key Laboratory of Material Science and Chemical Engineering, Harbin Engineering University, Heilongjiang 150001, Harbin, China
| | - Marlia Mohd Hanafiah
- Department of Earth Sciences and Environment, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, UKM Bangi 43600, Selangor, Malaysia
- Centre for Tropical Climate Change System, Institute of Climate Change, Universiti Kebangsaan Malaysia, UKM Bangi 43600, Selangor, Malaysia
| | - Syeda Azeem Unnisa
- Department of Environmental Sciences, UCS, Osmania University, Hyderabad, 500007, Telangana, India
| | - Mohammed Mahboob
- Toxicology Unit, Applied Biology Division, CSIR‐Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana India
| |
Collapse
|
10
|
Barguilla I, Barszczewska G, Annangi B, Domenech J, Velázquez A, Marcos R, Hernández A. MTH1 is involved in the toxic and carcinogenic long-term effects induced by zinc oxide and cobalt nanoparticles. Arch Toxicol 2020; 94:1973-1984. [PMID: 32377776 DOI: 10.1007/s00204-020-02737-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 04/06/2020] [Indexed: 12/26/2022]
Abstract
The nanoparticles (NPs) exposure-related oxidative stress is considered among the main causes of the toxic effects induced by these materials. However, the importance of this mechanism has been mostly explored at short term. Previous experience with cells chronically exposed to ZnO and Co NPs hinted to the existence of an adaptative mechanism contributing to the development of oncogenic features. MTH1 is a well-described enzyme expressed exclusively in cancer cells and required to avoid the detrimental consequences of its high prooxidant microenvironment. In the present work, a significantly marked overexpression was found when MTH1 levels were monitored in long-term ZnO and Co NP-exposed cells, a fact that correlates with acquired 2.5-fold and 3.75-fold resistance to the ZnO and Co NPs treatment, respectively. The forced stable inhibition of Mth1 expression by shRNA, followed by 6 additional weeks of exposure, significantly reduced this acquired resistance and sensitized cells to the oxidizing agents H2O2 and KBrO3. When the oncogenic phenotype of Mth1 knock-down cells was evaluated, we found a decrease in several oncogenic markers, including proliferation, anchorage-independent cell growth, and migration and invasion potential. Thus, MTH1 elicits here as a relevant player in the NPs-induced toxicity and carcinogenicity. This study is the first to give a mechanistic explanation for long-term NPs exposure-derived effects. We propose MTH1 as a candidate biomarker to unravel NPs potential genotoxic and carcinogenic effects, as its expression is expected to be elevated only under exposure conditions able to induce DNA damage and the acquisition of an oncogenic phenotype.
Collapse
Affiliation(s)
- Irene Barguilla
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici C, Campus de Bellaterra, 08193, Cerdanyola del Vallès (Barcelona), Spain
| | - Gabriela Barszczewska
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici C, Campus de Bellaterra, 08193, Cerdanyola del Vallès (Barcelona), Spain
| | - Balasubramanyam Annangi
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici C, Campus de Bellaterra, 08193, Cerdanyola del Vallès (Barcelona), Spain
| | - Josefa Domenech
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici C, Campus de Bellaterra, 08193, Cerdanyola del Vallès (Barcelona), Spain
| | - Antonia Velázquez
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici C, Campus de Bellaterra, 08193, Cerdanyola del Vallès (Barcelona), Spain.,CIBER Epidemiología y Salud Pública, ISCIII, Barcelona, Spain
| | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici C, Campus de Bellaterra, 08193, Cerdanyola del Vallès (Barcelona), Spain. .,CIBER Epidemiología y Salud Pública, ISCIII, Barcelona, Spain.
| | - Alba Hernández
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Edifici C, Campus de Bellaterra, 08193, Cerdanyola del Vallès (Barcelona), Spain. .,CIBER Epidemiología y Salud Pública, ISCIII, Barcelona, Spain.
| |
Collapse
|
11
|
Cornu R, Béduneau A, Martin H. Influence of nanoparticles on liver tissue and hepatic functions: A review. Toxicology 2019; 430:152344. [PMID: 31843632 DOI: 10.1016/j.tox.2019.152344] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/14/2019] [Accepted: 12/12/2019] [Indexed: 12/16/2022]
Abstract
Due to the increasing interest in nanotechnology in very large application fields, including biotechnology, electronics and food industries, humans are increasingly exposed to nanoparticles (NPs). Consequently, the question about the safety of these nanomaterials and their impact on human health is a legitimate concern. The liver is the primary organ of detoxification and is one of the tissues that is most exposed to NPs. When they reach the bloodstream, NPs are mainly internalized by liver cells. This review focuses on recent in vitro and in vivo studies addressing the effects of organic and inorganic NPs on the liver. Specifically, the impact of the NPs on hepatic enzyme activities, the inflammatory response and genotoxicity processes will be described. Depending on the physicochemical parameters of the NPs and the conditions of exposure, NPs could lead to global liver injury.
Collapse
Affiliation(s)
- Raphaël Cornu
- PEPITE EA4267, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - Arnaud Béduneau
- PEPITE EA4267, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France.
| | - Hélène Martin
- PEPITE EA4267, Univ. Bourgogne Franche-Comté, F-25000 Besançon, France.
| |
Collapse
|
12
|
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
|
13
|
Hernandez-Delgadillo R, García-Cuéllar CM, Sánchez-Pérez Y, Pineda-Aguilar N, Martínez-Martínez MA, Rangel-Padilla EE, Nakagoshi-Cepeda SE, Solís-Soto JM, Sánchez-Nájera RI, Nakagoshi-Cepeda MAA, Chellam S, Cabral-Romero C. In vitro evaluation of the antitumor effect of bismuth lipophilic nanoparticles (BisBAL NPs) on breast cancer cells. Int J Nanomedicine 2018; 13:6089-6097. [PMID: 30323596 PMCID: PMC6179729 DOI: 10.2147/ijn.s179095] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim The objective of this study was to evaluate the antitumor activity of lipophilic bismuth nanoparticles (BisBAL NPs) on breast cancer cells. Materials and methods The effect of varying concentrations of BisBAL NPs was evaluated on human MCF-7 breast cancer cells and on MCF-10A fibrocystic mammary epitheliocytes as noncancer control cells. Cell viability was evaluated with the MTT assay, plasma membrane integrity was analyzed with the calcein AM assay, genotoxicity with the comet assay, and apoptosis with the Annexin V/7-AAD assay. Results BisBAL NPs were spherical in shape (average diameter, 28 nm) and agglomerated into dense electronic clusters. BisBAL NP induced a dose-dependent growth inhibition. Most importantly, growth inhibition was higher for MCF-7 cells than for MCF-10A cells. At 1 µM BisBAL NP, MCF-7 growth inhibition was 51%, while it was 11% for MCF-10A; at 25 µM BisBAL NP, the growth inhibition was 81% for MCF-7 and 24% for MCF-10A. With respect to mechanisms of action, a 24-hour exposure of 10 and 100 µM BisBAL NP caused loss of cell membrane integrity and fragmentation of tumor cell DNA. BisBAL NPs at 10 µM were genotoxic to and caused apoptosis of breast cancer cells. Conclusion BisBAL NP-induced growth inhibition is dose dependent, and breast cancer cells are more vulnerable than noncancer breast cells. The mechanism of action of BisBAL NPs may include loss of plasma membrane integrity and a genotoxic effect on the genomic DNA of breast cancer cells.
Collapse
Affiliation(s)
- Rene Hernandez-Delgadillo
- Universidad Autónoma de Nuevo León, UANL, Facultad de Odontología, Laboratorio de Biología Molecular, Monterrey, Nuevo León, México,
| | | | | | - Nayely Pineda-Aguilar
- Centro de Investigación en Materiales Avanzados, S.C. (CIMAV), Unidad Monterrey, Nuevo León, México
| | - Marco Antonio Martínez-Martínez
- Universidad Autónoma de Nuevo León, UANL, Facultad de Odontología, Laboratorio de Biología Molecular, Monterrey, Nuevo León, México,
| | - Eyra Elvyra Rangel-Padilla
- Universidad Autónoma de Nuevo León, UANL, Facultad de Odontología, Laboratorio de Biología Molecular, Monterrey, Nuevo León, México,
| | - Sergio Eduardo Nakagoshi-Cepeda
- Universidad Autónoma de Nuevo León, UANL, Facultad de Odontología, Laboratorio de Biología Molecular, Monterrey, Nuevo León, México,
| | - Juan Manuel Solís-Soto
- Universidad Autónoma de Nuevo León, UANL, Facultad de Odontología, Laboratorio de Biología Molecular, Monterrey, Nuevo León, México,
| | - Rosa Isela Sánchez-Nájera
- Universidad Autónoma de Nuevo León, UANL, Facultad de Odontología, Laboratorio de Biología Molecular, Monterrey, Nuevo León, México,
| | | | | | - Claudio Cabral-Romero
- Universidad Autónoma de Nuevo León, UANL, Facultad de Odontología, Laboratorio de Biología Molecular, Monterrey, Nuevo León, México,
| |
Collapse
|
14
|
Chamaon K, Schönfeld P, Awiszus F, Bertrand J, Lohmann CH. Ionic cobalt but not metal particles induces ROS generation in immune cells in vitro. J Biomed Mater Res B Appl Biomater 2018; 107:1246-1253. [PMID: 30261124 DOI: 10.1002/jbm.b.34217] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022]
Abstract
Total joint replacement is one of the most successful procedures in orthopedic surgery today. However, metal implant materials undergo wear and corrosion processes. Generated particles and ions can cause a variety of cellular reactions. Cobalt-containing alloys are used frequently in implant materials. Some studies suggest that cobalt exhibits potential cytotoxic effects, for example, via generation of reactive oxygen species (ROS). To further elucidate the effects of cobalt on human cells, we determined cell viability and cytosolic and mitochondrial superoxide formation after incubation of either ions or particles with different cells. MM-6 and Jurkat cell lines were treated for 24, 48 and 72 h with either CoCrMo particles or cobalt ions (supplied as CoCl2 ). A total of 24 h exposure of both forms of cobalt did not induce cell death using terminal deoxynucleotidyl transferase (TUNEL) and trypan blue assay. Interestingly, the formation of superoxide (O2 .- ) is evoked mainly by ionic CoCl2 but not cobalt particles. Cobalt alloy particles are likely to even suppress O2 .- formation in mitochondria in both used cell lines. Furthermore, we did not observe any effect of cobalt particles on O2 .- formation in peripheral blood mononuclear cells (PBMCs) from healthy donors. We also found that the O2 - formation by CoCl2 within mitochondria is a generalized effect for all cell types used, while the formation of superoxide in cytosolic compartment is cell-type dependent. In summary, our data suggest that cobalt ions specifically induce the formation of O2 .- , whereas the cobalt particles were better tolerated. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1246-1253, 2019.
Collapse
Affiliation(s)
- Kathrin Chamaon
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Peter Schönfeld
- Institute for Biochemistry and Cell Biology, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Friedemann Awiszus
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Christoph H Lohmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| |
Collapse
|
15
|
Mirshafiee V, Sun B, Chang CH, Liao YP, Jiang W, Jiang J, Liu X, Wang X, Xia T, Nel AE. Toxicological Profiling of Metal Oxide Nanoparticles in Liver Context Reveals Pyroptosis in Kupffer Cells and Macrophages versus Apoptosis in Hepatocytes. ACS NANO 2018; 12:3836-3852. [PMID: 29543433 PMCID: PMC5946698 DOI: 10.1021/acsnano.8b01086] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The liver and the mononuclear phagocyte system are a frequent target for engineered nanomaterials, either as a result of particle uptake and spread from primary exposure sites or systemic administration of therapeutic and imaging nanoparticles. In this study, we performed a comparative analysis of the toxicological impact of 29 metal oxide nanoparticles (NPs), some commonly used in consumer products, in transformed or primary Kupffer cells (KCs) and hepatocytes. We not only observed differences between KCs and hepatocytes, but also differences in the toxicological profiles of transition-metal oxides (TMOs, e. g., Co3O4) versus rare-earth oxide (REO) NPs ( e. g., Gd2O3). While pro-oxidative TMOs induced the activation of caspases 3 and 7, resulting in apoptotic cell death in both cell types, REOs induced lysosomal damage, NLRP3 inflammasome activation, caspase 1 activation, and pyroptosis in KCs. Pyroptosis was accompanied by cell swelling, membrane blebbing, IL-1β release, and increased membrane permeability, which could be reversed by knockdown of the pore forming protein, gasdermin D. Though similar features were not seen in hepatocytes, the investigation of the cytotoxic effects of REO NPs could also be seen to affect macrophage cell lines such as J774A.1 and RAW 264.7 cells as well as bone marrow-derived macrophages. These phagocytic cell types also demonstrated features of pyroptosis and increased IL-1β production. Collectively, these findings demonstrate important mechanistic considerations that can be used for safety evaluation of metal oxides, including commercial products that are developed from these materials.
Collapse
Affiliation(s)
- Vahid Mirshafiee
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
| | - Bingbing Sun
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, 2 Linggong Rd., Dalian 116024, China
| | - Chong Hyun Chang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Yu Pei Liao
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
| | - Wen Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Jinhong Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Xiangsheng Liu
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
| | - Xiang Wang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Tian Xia
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
- Address correspondence to: ;
| | - André E. Nel
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, California 90095, United States
- Address correspondence to: ;
| |
Collapse
|