1
|
Kusi D, Sun Y, Liu C. Advances in Manganese-based nanomaterials for cancer therapy via regulating Non-Ferrous ferroptosis. Int J Pharm 2025; 669:125101. [PMID: 39706379 DOI: 10.1016/j.ijpharm.2024.125101] [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: 09/19/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Ferroptosis, a regulated form of cell death distinct from apoptosis, was first identified in 2012 and is characterized by iron-dependent lipid peroxidation driven by reactive oxygen species (ROS). Since its discovery, ferroptosis has been linked to various diseases, with recent studies highlighting its potential in cancer therapy, particularly for targeting cancer cells that are resistant to traditional treatments like chemotherapy and radiotherapy. While iron has historically been central to ferroptosis, emerging evidence indicates that non-ferrous ions, especially manganese (Mn), also play a crucial role in modulating this process. Mn-based nanomaterials have shown significant promise in cancer treatment by enhancing ROS production, depleting antioxidant defenses, and inducing ferroptosis. Additionally, these materials offer advantages in tumor imaging, immunotherapy, and catalyzing the Fenton-like reactions essential for ferroptosis. This review delves into the mechanisms of Mn-induced ferroptosis, focusing on recent advancements in Mn-based nanomaterials and their applications in chemodynamic therapy and immunotherapy. By leveraging non-ferrous ion-mediated ferroptosis, these approaches provide a novel avenue for cancer treatment. Furthermore, this review explores the potential role of Mn-based nanomaterials in the lipid metabolism pathways involved in ferroptosis and highlights the advantages of Mn ions over other metals in promoting ferroptosis. These insights offer new perspectives for the development of tumor therapies centered on Mn-based nanomaterials.
Collapse
Affiliation(s)
- Dipa Kusi
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Yan Sun
- Department of Cardiology, Zhejiang Rongjun Hospital, Jiaxing 314001 PR China.
| | - Chenguang Liu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.
| |
Collapse
|
2
|
Zheng X, Wang Q, Xu X, Huang X, Chen J, Huo X. Associations of insulin sensitivity and immune inflammatory responses with child blood lead (Pb) and PM 2.5 exposure at an e-waste recycling area during the COVID-19 lockdown. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 46:296. [PMID: 38980420 DOI: 10.1007/s10653-024-02066-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024]
Abstract
Fine particular matter (PM2.5) and lead (Pb) exposure can induce insulin resistance, elevating the likelihood of diabetes onset. Nonetheless, the underlying mechanism remains ambiguous. Consequently, we assessed the association of PM2.5 and Pb exposure with insulin resistance and inflammation biomarkers in children. A total of 235 children aged 3-7 years in a kindergarten in e-waste recycling areas were enrolled before and during the Corona Virus Disease 2019 (COVID-19) lockdown. Daily PM2.5 data was collected and used to calculate the individual PM2.5 daily exposure dose (DED-PM2.5). Concentrations of whole blood Pb, fasting blood glucose, serum insulin, and high mobility group box 1 (HMGB1) in serum were measured. Compared with that before COVID-19, the COVID-19 lockdown group had lower DED-PM2.5 and blood Pb, higher serum HMGB1, and lower blood glucose and homeostasis model assessment of insulin resistance (HOMA-IR) index. Decreased DED-PM2.5 and blood Pb levels were linked to decreased levels of fasting blood glucose and increased serum HMGB1 in all children. Increased serum HMGB1 levels were linked to reduced levels of blood glucose and HOMA-IR. Due to the implementation of COVID-19 prevention and control measures, e-waste dismantling activities and exposure levels of PM2.5 and Pb declined, which probably reduced the association of PM2.5 and Pb on insulin sensitivity and diabetes risk, but a high level of risk of chronic low-grade inflammation remained. Our findings add new evidence for the associations among PM2.5 and Pb exposure, systemic inflammation and insulin resistance, which could be a possible explanation for diabetes related to environmental exposure.
Collapse
Affiliation(s)
- Xiangbin Zheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, 855 East Xingye Avenue, Guangzhou, 511443, Guangdong, China
- Center for Reproductive Medicine, Clinical Research Center, Shantou Central Hospital, Shantou, 515041, Guangdong, China
| | - Qihua Wang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, 855 East Xingye Avenue, Guangzhou, 511443, Guangdong, China
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xiaofan Huang
- Center for Reproductive Medicine, Clinical Research Center, Shantou Central Hospital, Shantou, 515041, Guangdong, China
| | - Jiaxue Chen
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, 855 East Xingye Avenue, Guangzhou, 511443, Guangdong, China.
| |
Collapse
|
3
|
Nguyen HD, Kim MS. The Effects of a Mixture of Cadmium, Lead, and Mercury on Metabolic Syndrome and Its Components, as well as Cognitive Impairment: Genes, MicroRNAs, Transcription Factors, and Sponge Relationships : The Effects of a Mixture of Cadmium, Lead, and Mercury on Metabolic Syndrome and Its Components, as well as Cognitive Impairment: Genes, MicroRNAs, Transcription Factors, and Sponge Relationships. Biol Trace Elem Res 2023; 201:2200-2221. [PMID: 35798913 DOI: 10.1007/s12011-022-03343-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/21/2022] [Indexed: 01/11/2023]
Abstract
Converging evidence indicates heavy metal-induced genes, transcription factors (TFs), and microRNAs (miRNAs) are critical pathological components of metabolic syndrome (MetS) and cognitive impairment. Thus, our goals are to identify the interaction of mixed heavy metals (cadmium + lead + mercury) with genes, TFs, and miRNAs involved in MetS and its components, as well as cognitive impairment development. The most commonly retrieved genes for each disease were different, but essential biological pathways such as oxidative stress, altered lipoprotein metabolism, fluid shear stress and atherosclerosis, apoptosis, the IL-6 signaling pathway, and Alzheimer's disease were highlighted. The genes CASP3, BAX, BCL2, IL6, TNF, APOE, HMOX1, and IGF were found to be mutually affected by the heavy metal mixture studied, suggesting the importance of apoptosis, inflammation, lipid, heme, and glucose metabolism in MetS and cognitive impairment, as well as the potentiality of targeting these genes in prospective therapeutic intervention for these diseases. EGR2, ATF3, and NFE2L2 were noted as the most key TFs implicated in the etiology of MetS and its components, as well as cognitive impairment. We also found six miRNAs induced by studied heavy metals were the mutual miRNAs linked to MetS, its components, and cognitive impairment. In particular, we used miRNAsong to construct and verify a miRNA sponge sequence for these miRNAs. These sponges are promising molecules for the treatment of MetS and its components, as well as cognitive impairment.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922, Republic of Korea.
| |
Collapse
|
4
|
Ji Y, Hu Q, Ma G, Yu A, Zhao L, Zhang X, Zhao R. Selenium biofortification in Pleurotus eryngii and its effect on lead adsorption of gut microbiota via in vitro fermentation. Food Chem 2022; 396:133664. [PMID: 35841676 DOI: 10.1016/j.foodchem.2022.133664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/15/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022]
Abstract
It is of great significance to develop safe and efficient dietary selenium sources to improve lead toxicity. In this study, selenate, selenite, SeMet and Se-yeast were supplied to investigate the Se biofortification and bioaccessibility in Pleurotus eryngii. The effects of Se-enriched P. eryngii on lead binding bacteria were investigated via in vitro fermentation. With 40 mg/kg Se in the substrate, the total Se contents of P. eryngii treated with selenite and Se-yeast were 145.22 ± 8.00 mg/kg and 90.01 ± 7.01 mg/kg, respectively. Compared with selenite, Se-yeast treatment significantly increased the organic Se proportion in P. eryngii (SeCys2 2.85 ± 0.17%, MeSeCys 2.33 ± 0.21% and SeMet 78.19 ± 1.58%), which led to higher bioaccessibility. With 1 mg/L lead treatment during in vitro fermentation, Se-enriched P. eryngii promoted the growth of Desulfovibrio, which contributed to the increase of gut microbiota lead adsorption. Se-enriched P. eryngii cultivated with Se-yeast could be used as dietary Se sources for lead toxicity improvement.
Collapse
Affiliation(s)
- Yang Ji
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Qiuhui Hu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, People's Republic of China.
| | - Gaoxing Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, People's Republic of China
| | - Anqi Yu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, People's Republic of China
| | - Liyan Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Xueli Zhang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Ruiqiu Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
5
|
Chen Y, Mao G, Zhang Z, Zhao T, Feng W, Yang L, Wu X. The protective effect of C3G against Pb-induced learning and memory impairments through cAMP-PKA-CREB signaling pathway in rat hippocampus. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
6
|
Ferroptosis as a mechanism of non-ferrous metal toxicity. Arch Toxicol 2022; 96:2391-2417. [PMID: 35727353 DOI: 10.1007/s00204-022-03317-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
Abstract
Ferroptosis is a recently discovered form of regulated cell death, implicated in multiple pathologies. Given that the toxicity elicited by some metals is linked to alterations in iron metabolism and induction of oxidative stress and lipid peroxidation, ferroptosis might be involved in such toxicity. Although direct evidence is insufficient, certain pioneering studies have demonstrated a crosstalk between metal toxicity and ferroptosis. Specifically, the mechanisms underlying metal-induced ferroptosis include induction of ferritinophagy, increased DMT-1 and TfR cellular iron uptake, mitochondrial dysfunction and mitochondrial reactive oxygen species (mitoROS) generation, inhibition of Xc-system and glutathione peroxidase 4 (GPX4) activity, altogether resulting in oxidative stress and lipid peroxidation. In addition, there is direct evidence of the role of ferroptosis in the toxicity of arsenic, cadmium, zinc, manganese, copper, and aluminum exposure. In contrast, findings on the impact of cobalt and nickel on ferroptosis are scant and nearly lacking altogether for mercury and especially lead. Other gaps in the field include limited studies on the role of metal speciation in ferroptosis and the critical cellular targets. Although further detailed studies are required, it seems reasonable to propose even at this early stage that ferroptosis may play a significant role in metal toxicity, and its modulation may be considered as a potential therapeutic tool for the amelioration of metal toxicity.
Collapse
|
7
|
Yamashita R, Takahashi Y, Takashima K, Okano H, Ojiro R, Tang Q, Kikuchi S, Kobayashi M, Ogawa B, Jin M, Kubota R, Ikarashi Y, Yoshida T, Shibutani M. Induction of cellular senescence as a late effect and BDNF-TrkB signaling-mediated ameliorating effect on disruption of hippocampal neurogenesis after developmental exposure to lead acetate in rats. Toxicology 2021; 456:152782. [PMID: 33862172 DOI: 10.1016/j.tox.2021.152782] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 03/24/2021] [Accepted: 04/11/2021] [Indexed: 12/27/2022]
Abstract
Lead (Pb) exposure causes cognitive deficits in children. The present study investigated the effect of developmental exposure to Pb acetate (PbAc) on postnatal hippocampal neurogenesis. Pregnant rats were administered drinking water containing 0, 2000, or 4000 ppm PbAc from gestational day 6 until day 21 post-delivery (weaning), and offspring were maintained without PbAc exposure until adulthood on postnatal day (PND) 77. There was a dose-related accumulation of Pb in the offspring brain at weaning, while Pb was mainly excreted in adulthood. In the hippocampus, metallothionein I/II immunoreactive (+) glia were increased through adulthood as a neuroprotective response to accumulated Pb, accompanied by increased astrocyte and microglia numbers in adulthood, suggesting sustained neural damage. Gene expression changes suggested elevated oxidative stress at weaning and suppression of the antioxidant system in adulthood, as well as continued neuroinflammatory responses. At weaning, granule cell apoptosis was increased and numbers of type-3 neural progenitor cells (NPCs) were decreased. By contrast, type-2a and type-2b NPCs were increased, suggesting suppressed differentiation to type-3 NPCs. In adulthood, there were increased numbers of immature granule cells. In the hilus of the dentate gyrus, somatostatin+ interneurons were increased at weaning, while calbindin-D-29K+ interneurons were increased throughout adulthood, suggesting a strengthened interneuron regulatory system against the suppressed differentiation at weaning. In the dentate gyrus, Bdnf, Ntrk2, and Chrna7 gene expression were upregulated and numbers of hilar TrkB+ interneurons increased at weaning. These findings suggest activation of BDNF-TrkB signaling to increase somatostatin+ interneurons and promote cholinergic signaling, thus increasing later production of immature granule cells. In adulthood, Pcna and Apex1 gene expression were downregulated and Chek1 and cyclin-dependent kinase inhibitor expression were upregulated. Furthermore, there was an increase in γ-H2AX+ SGZ cells, suggesting induction of cellular senescence of SGZ cells due to Pb genotoxicity.
Collapse
Affiliation(s)
- Risako Yamashita
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Yasunori Takahashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Kazumi Takashima
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Hiromu Okano
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Ryota Ojiro
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Qian Tang
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Satomi Kikuchi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Mio Kobayashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Bunichiro Ogawa
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Meilan Jin
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, BeiBei District, Chongqing, 400715, PR China.
| | - Reiji Kubota
- Division of Environmental Chemistry, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-shi, Kawasaki-ku, Kanagawa, 210-9501, Japan.
| | - Yoshiaki Ikarashi
- Division of Environmental Chemistry, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-shi, Kawasaki-ku, Kanagawa, 210-9501, Japan.
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| |
Collapse
|
8
|
Tian ZK, Zhang YJ, Feng ZJ, Jiang H, Cheng C, Sun JM, Liu CM. Nephroprotective effect of gastrodin against lead-induced oxidative stress and inflammation in mice by the GSH, Trx, Nrf2 antioxidant system, and the HMGB1 pathway. Toxicol Res (Camb) 2021; 10:249-263. [PMID: 33884175 DOI: 10.1093/toxres/tfab003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/02/2021] [Accepted: 01/11/2021] [Indexed: 01/07/2023] Open
Abstract
Gastrodin (GAS), the main phenolic glycoside derivative from Gastrodiaelata Blume, has several bio-activities. However, the molecular mechanisms of these protective actions currently remain unclear. This study aimed to investigate the mechanisms of GAS on lead (Pb)-induced oxidative stress and inflammation in the kidneys and primary kidney mesangial cells. Results indicated that GAS improved Pb-induced renal dysfunction and morphological changes in mice. GAS ameliorated Pb-induced inflammation in kidneys by reducing the TNF-α and IL-6 levels. GAS inhibited Pb-induced oxidative stress by regulating the glutathione, thioredoxin (Trx), and Nrf2 antioxidant systems. Furthermore, GAS supplementation increased the activation of SOD, GPx, HO-1, and NQO1 in the kidneys. GAS decreased the expression levels of HMGB1, TLR4, RAGE, MyD88, and NF-κB. These results were further confirmed in primary kidney mesangial cells. Collectively, this study demonstrated that GAS alleviated Pb-induced kidney oxidative stress and inflammation by regulating the antioxidant systems and the Nrf2 signaling pathway. Highlights Gastrodin ameliorated Pb-induced kidney injury in mice.Gastrodin inhibited oxidative stress and inflammation in kidneys.Gastrodin activated the GSH, Trx and Nrf2 antioxidant system in kidneys.Gastrodin inhibited the activities of HMGB1. RAGE, TLR4, and MyD88.
Collapse
Affiliation(s)
- Zhi-Kai Tian
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan New Area, Xuzhou, Jiangsu 221116, P. R. China
| | - Yu-Jia Zhang
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan New Area, Xuzhou, Jiangsu 221116, P. R. China
| | - Zhao-Jun Feng
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan New Area, Xuzhou, Jiangsu 221116, P. R. China
| | - Hong Jiang
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan New Area, Xuzhou, Jiangsu 221116, P. R. China
| | - Chao Cheng
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan New Area, Xuzhou, Jiangsu 221116, P. R. China
| | - Jian-Mei Sun
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan New Area, Xuzhou, Jiangsu 221116, P. R. China
| | - Chan-Min Liu
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tongshan New Area, Xuzhou, Jiangsu 221116, P. R. China
| |
Collapse
|
9
|
Zhu G, Zhu Q, Zhang W, Hui C, Li Y, Yang M, Pang S, Li Y, Xue G, Chen H. Mitochondrial uncoupling protein 2 is regulated through heterogeneous nuclear ribonucleoprotein K in lead exposure models. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:1-16. [PMID: 33576715 DOI: 10.1080/26896583.2020.1854596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Synaptic plasticity plays an important role in learning and memory in the developing hippocampus. However, the precise molecular mechanism in lead exposure models remains to be studied. UCP2, an inner mitochondrial anion carrier, regulates synaptic plasticity through uncoupling neurons. And hnRNP K, an RNA binding protein, plays a role in modulating the expression of transcripts coding synaptic plasticity. We aim to investigate whether lead exposure affects UCP2 and hnRNP K expression levels. The Sprague-Dawley rats were exposed to different lead acetate concentrations (0 g/l, 0.5 g/l, 2.0 g/l) during gestational and lactational periods. PC12 cells were also exposed to different lead acetate concentrations (0 μM, 1 μM and 100 μM). We found that the expression levels of UCP2 and hnRNP K had significant declines in the lead exposure rat hippocampus and PC12 cells. Furthermore, the up-regulation of hnRNP K expression level could reverse the expression level of UCP2 in lead exposure models. In conclusion, these results suggest that lead exposure can reduce the expression level of UCP2 which is mediated by decreasing the expression level of hnRNP K.
Collapse
Affiliation(s)
- Gaochun Zhu
- Department of Anatomy, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Qian Zhu
- Department of Anatomy, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Wei Zhang
- Department of Anatomy, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Chen Hui
- Department of Anatomy, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Yuwen Li
- Queen Mary College, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Meiyuan Yang
- Department of Anatomy, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Shimin Pang
- Second Clinical College, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Yaobing Li
- Department of Anatomy, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Guoyong Xue
- Department of Anatomy, School of Medicine, Nanchang University, Nanchang, P.R. China
| | - Hongping Chen
- Department of Histology and Embryology, School of Medicine, Nanchang University, Nanchang, P.R. China
| |
Collapse
|
10
|
Li B, Peng X, Li H, Chen F, Chen Y, Zhang Y, Le K. The performance of the alarmin HMGB1 in pediatric diseases: From lab to clinic. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:8-30. [PMID: 33140586 PMCID: PMC7860603 DOI: 10.1002/iid3.370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/10/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The ubiquitously expressed nonhistone nuclear protein high-mobility group box protein 1 (HMGB1) has different functions related to posttranslational modifications and cellular localization. In the nucleus, HMGB1 modulates gene transcription, replication and DNA repair as well as determines chromosomal architecture. When the post-transcriptional modified HMGB1 is released into the extracellular space, it triggers several physiological and pathological responses and initiates innate immunity through interacting with its reciprocal receptors (i.e., TLR4/2 and RAGE). The effect of HMGB1-mediated inflammatory activation on different systems has received increasing attention. HMGB1 is now considered to be an alarmin and participates in multiple inflammation-related diseases. In addition, HMGB1 also affects the occurrence and progression of tumors. However, most studies involving HMGB1 have been focused on adults or mature animals. Due to differences in disease characteristics between children and adults, it is necessary to clarify the role of HMGB1 in pediatric diseases. METHODS AND RESULTS Through systematic database retrieval, this review aimed to first elaborate the characteristics of HMGB1 under physiological and pathological conditions and then discuss the clinical significance of HMGB1 in the pediatric diseases according to different systems. CONCLUSIONS HMGB1 plays an important role in a variety of pediatric diseases and may be used as a diagnostic biomarker and therapeutic target for new strategies for the prevention and treatment of pediatric diseases.
Collapse
Affiliation(s)
- Bo Li
- Department of Cardiology, Children's Hospital of Hebei Province Affiliated to Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xin Peng
- Department of Otolaryngology, The Affiliated Children's Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - He Li
- Department of Urology Surgery, Qilu Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Fei Chen
- Department of Child Health Care, Qilu Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Yuxia Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, and Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, and Rehabilitation Centre, Children's Hospital, Chongqing Medical University, Chongqing, Yuzhong, China
| | - Yingqian Zhang
- Department of Cardiology, Children's Hospital of Hebei Province Affiliated to Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
11
|
Cai QL, Peng DJ, Lin-Zhao, Chen JW, Yong-Li, Luo HL, Ou SY, Huang ML, Jiang YM. Impact of Lead Exposure on Thyroid Status and IQ Performance among School-age Children Living Nearby a Lead-Zinc Mine in China. Neurotoxicology 2020; 82:177-185. [PMID: 33115663 DOI: 10.1016/j.neuro.2020.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 08/31/2020] [Accepted: 10/22/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Lead exposure is one of the most concerning public health problems worldwide, particularly among children. Yet the impact of chronic lead exposure on the thyroid status and related intelligence quotient performance among school-age children remained elusive. OBJECTIVE The aim of this study was to evaluate the influence of lead exposure on the thyroid hormones, amino acid neurotransmitters balances, and intelligence quotient (IQ) among school-age children living nearby a lead-zinc mining site. Other factors such as rice lead levels, mothers' smoking behavior, and diet intake were also investigated. METHODS A total of 255 children aged 7-12 years old were recruited in this study. Blood lead level (BLL), thyroid hormones including free triiodothyronine (FT3), free thyroxine (FT4) and thyroid stimulating hormone (TSH), and amino acid neurotransmitters such as glutamate (Glu), glutamine (Gln), and γ-aminobutyric acid (GABA) were measured using graphite furnace atomic absorption spectroscopy (GFAAS), chemiluminescence immunoassay, high performance liquid chromatography (HPLC). Raven's standard progressive matrices (SPM) and the questionnaire were used to determine IQ and collect related influence factors. RESULTS The average BLL of children was 84.8 μg/L. The occurrence of lead intoxication (defined as the BLL ≥ 100 μg/L) was 31.8%. Serum TSH levels and IQ of lead-intoxicated children were significantly lower than those without lead toxicity. The GABA level of girls with the lead intoxication was higher than those with no lead-exposed group. Correlation analyses revealed that BLL were inversely associated with the serum TSH levels (R= -0.186, p < 0.05), but positively related with IQ grades (R = 0.147, p < 0.05). Moreover, BLL and Glu were inversely correlated with IQ. In addition, this study revealed four factors that may contribute to the incidence of lead intoxication among children, including the frequency of mother smoking (OR = 3.587, p < 0.05) and drinking un-boiled stagnant tap water (OR = 3.716, p < 0.05); eating fresh fruits and vegetables (OR = 0.323, p < 0.05) and soy products regularly (OR = 0.181, p < 0.05) may protect against lead intoxication. CONCLUSION Lead exposure affects the serum TSH, GABA levels and IQ of school-aged children. Developing good living habits, improving environment, increasing the intake of high-quality protein and fresh vegetable and fruit may improve the condition of lead intoxication.
Collapse
Affiliation(s)
- Qiu-Ling Cai
- Department of Maternal, Child and Adolescent Health, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Dong-Jie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Lin-Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jing-Wen Chen
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Public Health Office, Wuhan First Hospital, Hubei 430022, China
| | - Yong-Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Wanzhou District Health Committee, Chongqing 404000, China
| | - Hai-Lan Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Center for Disease Control and Prevention, Nanning 530021, Guangxi, China
| | - Shi-Yan Ou
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Ming-Li Huang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China.
| |
Collapse
|
12
|
Fan Y, Zhao X, Yu J, Xie J, Li C, Liu D, Tang C, Wang C. Lead-induced oxidative damage in rats/mice: A meta-analysis. J Trace Elem Med Biol 2020; 58:126443. [PMID: 31841831 DOI: 10.1016/j.jtemb.2019.126443] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Lead (Pb) is ubiquitous in the environment and is an environmental genotoxic metal. Pb accumulation in the body could cause the oxidative stress. OBJECTIVE This meta-analysis aimed to perform a systematic evaluation of the extent of oxidative damage in rats/mice induced by lead. METHODS All relevant articles in English or Chinese were retrieved from Embase, PubMed, Web of Science, Medline, China National Knowledge Infrastructure, and Chinese Biological Medicine databases from their inception date until July 22, 2018. RESULTS A total of 108 eligible articles were included in this study. The indicators of oxidative stress included malondialdehyde (MDA), glutathione disulfide (GSSG), reactive oxygen species (ROS), hydrogen peroxide (H2O2), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), reduced glutathione (GSH), superoxide dismutase (SOD), and glutathione-s-transferase (GST). The meta-analysis showed that lead significantly increased oxidants levels, such as MDA, GSSG, ROS, and H2O2 (P < 0.05), and significantly reduced the level of antioxidants, such as CAT, GPx, GR, GSH, SOD, and GST (P < 0.05). The intraperitoneal mode was more effective than water drinking mode in reducing the levels of CAT, GPx, GSH, and SOD (P < 0.05). Other factors that influenced the overall oxidative stress, including species of animals, type of tissues, and intervention dosage and time, were comprehensively evaluated. CONCLUSION The results of meta-analysis indicated that mice were more sensitive to lead than rats, and intraperitoneal mode was an effective intervention mean. High doses and long periods of lead treatment can cause serious oxidative damage. Moreover, testicular was more vulnerable to lead than other tissues. These results provided scientific evidence for preventing and treating lead toxicity.
Collapse
Affiliation(s)
- Yongsheng Fan
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China
| | - Xue Zhao
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China
| | - Jun Yu
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China
| | - Jie Xie
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China
| | - Cong Li
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China
| | - Duanya Liu
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China
| | - Caoli Tang
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China
| | - Chunhong Wang
- Department of Toxicology, School of Public Health, Wuhan University, DongHu Road 115, Wuhan 430071, China; Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
13
|
Khalid M, Abdollahi M. Epigenetic modifications associated with pathophysiological effects of lead exposure. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:235-287. [PMID: 31402779 DOI: 10.1080/10590501.2019.1640581] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lead (Pb) exposure during different stages of development has demonstrated dose, duration, sex, and tissue-specific pathophysiological outcomes due to altered epigenetic regulation via (a) DNA methylation, (b) histone modifications, (c) miRNAs, and (d) chromatin accessibility. Pb-induced alteration of epigenetic regulation causes neurotoxic and extra-neurotoxic pathophysiological outcomes. Neurotoxic effects of Pb include dysfunction of memory and learning, behavioral disorder, attention deficit hyperactivity disorder, autism spectrum disorder, aging, Alzheimer's disease, tauopathy, and neurodegeneration. Extra-neurotoxic effects of Pb include altered body weight, metabolic disorder, cardiovascular disorders, hematopoietic disorder, and reproductive impairment. Pb exposure either early in life or at any stage of development results in undesirable pathophysiological outcomes that tends to sustain and maintain for a lifetime.
Collapse
Affiliation(s)
- Madiha Khalid
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Pang S, Li Y, Chen W, Li Y, Yang M, Zhao L, Shen Q, Cheng N, Wang Y, Lin X, Ma J, Wu H, Zhu G. Pb exposure reduces the expression of SNX6 and Homer1 in offspring rats and PC12 cells. Toxicology 2019; 416:23-29. [PMID: 30738087 DOI: 10.1016/j.tox.2019.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 11/25/2022]
Abstract
Lead (Pb) is a widespread environmental heavy metal toxicant and chronic Pb exposure can have irreversible effects on memory and cognitive function, which is closely related to dendritic spines. Studies have shown that SNX6 and Homer1 can regulate the growth of dendritic spines. We aimed to investigate the effect of Pb exposure on the dendritic spines in hippocampus, the expression of SNX6 and Homer1 in rats and PC12 cells. The animals were randomly divided to three groups: control group, low lead group and high lead group. PC12 cells were divided into 3 groups: 0 μM, 1 μM and 100 μM Pb acetate. The results showed that the Pb levels in blood and hippocampus of all exposure groups were significantly higher than that of the control group. The morphology of dendritic spines in hippocampus after Pb treatment was changed and the density of dendritic spines was reduced. The expression of SNX6 and Homer1 was decreased in Pb exposed groups compared with the control group. Furthermore, up-regulation of SNX6 expression could reverse the down-regulation of Pb exposure on Homer1. These results indicate that Pb exposure can reduce the expression of SNX6 and lead to a decrease in Homer1 expression, which affects the changes in dendritic spines causing learning and memory impairment.
Collapse
Affiliation(s)
- Shimin Pang
- Second Clinical Collage, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Yanshu Li
- Jiangxi Supervision and Inspection Center for Medical Devices, Nanchang 330029, PR China
| | - Wei Chen
- Laboratory Animal Science Center, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Yaobin Li
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Meiyuan Yang
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Lijuan Zhao
- Second Clinical Collage, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Qiwei Shen
- Second Clinical Collage, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Nuo Cheng
- Queen Marry Collage, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Ying Wang
- Queen Marry Collage, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Xuequn Lin
- Faculty of Nursing, Nanchang Insitude of technology, Nanchang 330006, PR China
| | - Jianmin Ma
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Honghao Wu
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China
| | - Gaochun Zhu
- Department of Anatomy, School of Medicine, Nanchang University, BaYi Road 461, Nanchang 330006, PR China.
| |
Collapse
|