1
|
Huang H, Li G, Guo S, Li K, Li W, Zhou Q, He Z, Yang X, Liu L, Wei Q. RNA Methylation and Transcriptome Analysis Reveal Key Regulatory Pathways Related to Cadmium-Induced Liver Damage. Chem Res Toxicol 2025; 38:717-732. [PMID: 40135526 DOI: 10.1021/acs.chemrestox.4c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Cadmium (Cd) is a prevalent environmental and industrial contaminant that causes significant damage to liver function. However, the role of m6A methylation─a critical epigenetic modification─in Cd-induced liver injury remains poorly understood. This study aimed to investigate the effects of m6A methylation in Cd-induced liver damage. A mouse model of Cd-induced liver injury was established, and exposure to CdCl2 (20 mg/kg) for 90 days resulted in reduced m6A methylation levels. Using methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-Seq), we characterized the m6A methylation profiles in both control and Cd-exposed groups. A total of 8355 unique m6A peaks and 1,101 unique m6A-modified genes were identified. Among these, 673 genes exhibited differential m6A methylated modifications, including 463 hyper-methylated and 210 hypo-methylated genes. Conjoint analysis of MeRIP-seq and RNA-Seq data unveiled genes that showed both differential methylation and expression. These genes were significantly enriched in the AGE-RAGE and PI3K-Akt signaling pathway. Through bioinformatics screening, five key genes (Il-1β, Ccl2, Tlr2, Itgax, and Ccr2) were identified, and expression validation indicated that Itgax and Ccr2 may play pivotal roles in Cd-induced liver injury. Notably, elevated expression of methyltransferase-like 14 (METTL14) was observed in both in vivo and in vitro models. Inhibition of Mettl14 can regulate Cd-induced liver inflammation through m6A-dependent regulation of Ccr2 expression. Collectively, our findings highlight the crucial role of Mettl14 and Ccr2 in Cd-induced liver injury, providing novel insights into the epigenetic mechanisms underlying liver diseases and potential biomarkers for diagnosis and therapy.
Collapse
Affiliation(s)
- Hao Huang
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| | - Guoliang Li
- Guangdong Provincial Key Laboratory of Occupational Disease Prevention and Treatment, Department of Toxicology, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou 510300, China
| | - Sihui Guo
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| | - Kaile Li
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| | - Wei Li
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| | - Qinwen Zhou
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| | - Zhini He
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| | - Xingfen Yang
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| | - Lili Liu
- Guangdong Provincial Key Laboratory of Occupational Disease Prevention and Treatment, Department of Toxicology, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou 510300, China
| | - Qinzhi Wei
- School of Public Health, Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
2
|
Li X, Liu L, Lou H, Dong X, Hao S, Sun Z, Dou Z, Li H, Zhao W, Sun X, Liu X, Zhang Y, Yang B. Cardiomyocyte-specific long noncoding RNA Trdn-as induces mitochondrial calcium overload by promoting the m 6A modification of calsequestrin 2 in diabetic cardiomyopathy. Front Med 2025; 19:329-346. [PMID: 39821729 DOI: 10.1007/s11684-024-1102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/26/2024] [Indexed: 01/19/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a medical condition characterized by cardiac remodeling and dysfunction in individuals with diabetes mellitus. Sarcoplasmic reticulum (SR) and mitochondrial Ca2+ overload in cardiomyocytes have been recognized as biological hallmarks in DCM; however, the specific factors underlying these abnormalities remain largely unknown. In this study, we aimed to investigate the role of a cardiac-specific long noncoding RNA, D830005E20Rik (Trdn-as), in DCM. Our results revealed the remarkably upregulation of Trdn-as in the hearts of the DCM mice and cardiomyocytes treated with high glucose (HG). Knocking down Trdn-as in cardiac tissues significantly improved cardiac dysfunction and remodeling in the DCM mice. Conversely, Trdn-as overexpression resulted in cardiac damage resembling that observed in the DCM mice. At the cellular level, Trdn-as induced Ca2+ overload in the SR and mitochondria, leading to mitochondrial dysfunction. RNA-seq and bioinformatics analyses identified calsequestrin 2 (Casq2), a primary calcium-binding protein in the junctional SR, as a potential target of Trdn-as. Further investigations revealed that Trdn-as facilitated the recruitment of METTL14 to the Casq2 mRNA, thereby enhancing the m6A modification of Casq2. This modification increased the stability of Casq2 mRNA and subsequently led to increased protein expression. When Casq2 was knocked down, the promoting effects of Trdn-as on Ca2+ overload and mitochondrial damage were mitigated. These findings provide valuable insights into the pathogenesis of DCM and suggest Trdn-as as a potential therapeutic target for this condition.
Collapse
Affiliation(s)
- Xiaohan Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Ling Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Han Lou
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Xinxin Dong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Shengxin Hao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Zeqi Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Zijia Dou
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Huimin Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Wenjie Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Xiuxiu Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China.
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China.
| | - Baofeng Yang
- Department of Pharmacology (SKLFZCD, State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China.
| |
Collapse
|
3
|
Lin J, Hong H, Liu S, Liang Z, Zheng Q, Luo K, Li J, Du Z, Yu J, Yang L, Deng P, Pi H, Yu Z, Yuan W, Zhou Z. Aflatoxin B1 exposure induces Alzheimer's disease like pathology by disrupting redox homeostasis and activating ferroptotic signals in C57BL/6 J mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 970:179049. [PMID: 40054237 DOI: 10.1016/j.scitotenv.2025.179049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Aflatoxin B1 (AFB1) is one of the most toxic mycotoxins with neurotoxicity. Human exposure to AFB1 via contaminated foodstuffs has been linked to the risk of cognitive impairment, which may contribute to the progression of Alzheimer's disease (AD). However, the mechanism underlying the pathogenesis of AD in relation to AFB1 exposure is not clear. Herein, C57BL/6 J mice were exposed to 1.5 mg/L AFB1 in drinking water for 8 weeks. It was found that AFB1 damaged blood-brain barrier function, accumulated in the brain, and led to cognitive impairments and AD-like pathology in the hippocampus. Impaired cognitive function was indicated by the significant alterations in Morris' water maze and Y-maze tests at 8 weeks after AFB1 exposure. Concurrently, AD-like pathology was evinced by a marked neuronal loss and the up-regulated AD related gene and protein expressions in the hippocampus. AFB1 exposure remarkably disrupted redox homeostasis and induced ferroptosis both in the hippocampus at 8 weeks after AFB1 exposure and in cultured hippocampal neuron in vitro as indicated by the suppressions on SOD and CAT activities, the down-regulation of Slc7a11/Gpx4 expressions, the decline in GSH content, the increase in MDA and the lipid peroxidation. AFB1 exposure also increased Fe2+ content significantly at 8 weeks after exposure. In addition, we demonstrated that ferroptosis inhibition by Fer-1 obviously alleviated AFB1 neurotoxicity in HT22 cells. These results revealed an unknown pivotal role of ferroptosis in AFB1 neurotoxicity in relation to AD pathogenesis and emphasized the importance to reduce the health risk of AFB1 exposure as an etiology of AD in humans.
Collapse
Affiliation(s)
- Jinxian Lin
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Huihui Hong
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Sicheng Liu
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Zhengwei Liang
- Department of Otolaryngology, Chongqing General Hospital, Chongqing University, China
| | - Qixue Zheng
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Kun Luo
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Jiayi Li
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Zhulin Du
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Jinping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Wei Yuan
- Department of Otolaryngology, Chongqing General Hospital, Chongqing University, China.
| | - Zhou Zhou
- Department of Environmental Medicine, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
4
|
Wang Z, Wang PS, Yang C. Dysregulation of Long Non-coding RNAs-the Novel lnc in Metal Toxicity and Carcinogenesis. Curr Environ Health Rep 2024; 12:3. [PMID: 39715843 PMCID: PMC11755759 DOI: 10.1007/s40572-024-00468-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 12/25/2024]
Abstract
PURPOSE OF REVIEW Metals are common environmental pollutants. Acute and chronic exposures to non-essential toxic metals or excessive essential metals cause various diseases including cancer in humans. However, the underlying mechanisms have not been well understood. Long non-coding RNAs (lncRNAs) refer to RNA transcripts that have more than 200 nucleotides but do not have significant protein coding capacities. While lncRNAs were once considered transcription noise, they have become increasingly recognized as crucial players in various physiological and pathogenesis processes. The goal of this article is to review and discuss recent studies that show important roles of lncRNA dysregulations in metal toxicity and carcinogenesis. RECENT FINDINGS Recent studies showed that metal exposures dysregulate expression of lncRNAs in cultured cells, animals and humas. However, only a few studies determined the mechanisms of how metal exposure dysregulated expression of lncRNAs. The majority of the studies reported the association of abnormally expressed lncRNAs with various toxic effects of metal exposures, only limited studies established causal relationships demonstrating causal roles of dysregulated lncRNAs in metal toxicity and carcinogenesis. Mechanistically, most studies reported that dysregulated lncRNAs functioned as microRNA sponges to regulate gene expression, much less studies explored other mechanisms of lncRNA actions. It is evident that metal exposures dysregulate expression of lncRNAs, which may serve as novel mediators in metal toxicity and carcinogenesis. Further studies are needed to establish dysregulated lncRNAs as potential diagnostic biomarkers and therapeutic targets for metal exposure-associated diseases.
Collapse
Affiliation(s)
- Zhishan Wang
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 101 Nicolls Road, Stony Brook, NY, 11794, USA
| | - Po-Shun Wang
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Chengfeng Yang
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA.
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 101 Nicolls Road, Stony Brook, NY, 11794, USA.
| |
Collapse
|
5
|
Wu L, Xin Y, Zhang J, Yang X, Chen T, Niu P. Associations between Metals, Serum Folate, and Cognitive Function in the Elderly: Mixture and Mediation Analyses. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2024; 2:865-874. [PMID: 39722838 PMCID: PMC11667285 DOI: 10.1021/envhealth.4c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 12/28/2024]
Abstract
Exposure to metals may potentially impact cognitive health in the elderly; however, the evidence remains ambiguous. The specific role of serum folate in this relationship is also unclear. We aimed to evaluate the individual and joint impact of metals on cognition in the elderly from the United States and explore the potential mediating effect of serum folate. Data from the NHANES 2011-2014 were used, with inductively coupled plasma mass spectrometry (ICP-MS) employed to measure blood metal concentrations. Cognitive function was assessed using tests for immediate, delayed, and working memory: Immediate Recall test (IRT), the Delayed Recall test (DRT), the Animal Fluency test (AFT), and the Digit Symbol Substitution test (DSST). Generalized linear regression models (GLMs), Bayesian kernel machine regression model (BKMR), and quantile g-computation (QG-C) models were used to assess associations between metals (lead, cadmium, mercury, selenium, manganese) and cognition, with mediation analyses examining serum folate's involvement in metal effects. This study included 2002 participants aged ≥60. GLMs revealed the negative association between cadmium and the z-scores of IRT (β: -0.17,95% CI: -0.30, -0.04) and DSST (β: -0.15,95% CI: -0.27, -0.04), with negative effects also observed in the BKMR and QG-C models. Selenium displayed significantly positive association with cognition across various statistical models, including GLMs, QG-C, and BKMR. Serum folate played a mediating role in the effects of cadmium and selenium exposure on DSST z-scores, with a proportion of mediation of 17% and 10%, respectively. Our study assessed the impact of metal mixtures on cognition in the elderly population, finding that high selenium level was strongly associated with better cognitive performance, while cadmium was associated with lower cognitive function scores. Serum folate might partially mediate the association between cadmium, selenium, and DSST z-scores.
Collapse
Affiliation(s)
- Luli Wu
- Department
of Occupational Health and Environmental Health, School of Public
Health, Capital Medical University, 100069 Beijing, China
- Beijing
Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, 100069 Beijing, China
| | - Ye Xin
- Department
of Occupational Health and Environmental Health, School of Public
Health, Capital Medical University, 100069 Beijing, China
- Beijing
Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, 100069 Beijing, China
| | - Junrou Zhang
- Department
of Occupational Health and Environmental Health, School of Public
Health, Capital Medical University, 100069 Beijing, China
- Beijing
Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, 100069 Beijing, China
| | - Xin Yang
- Department
of Occupational Health and Environmental Health, School of Public
Health, Capital Medical University, 100069 Beijing, China
- Beijing
Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, 100069 Beijing, China
| | - Tian Chen
- Department
of Occupational Health and Environmental Health, School of Public
Health, Capital Medical University, 100069 Beijing, China
- Beijing
Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, 100069 Beijing, China
| | - Piye Niu
- Department
of Occupational Health and Environmental Health, School of Public
Health, Capital Medical University, 100069 Beijing, China
- Beijing
Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, 100069 Beijing, China
| |
Collapse
|
6
|
Hao R, Li F, Sun-Waterhouse D, Li D. The roles of MicroRNAs in cadmium toxicity and in the protection offered by plant food-derived dietary phenolic bioactive substances against cadmium-induced toxicity. Food Chem 2024; 460:140483. [PMID: 39032304 DOI: 10.1016/j.foodchem.2024.140483] [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: 05/13/2024] [Revised: 07/11/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Cadmium, a harmful food contaminant, poses severe health risks. There are ongoing efforts to reduce cadmium pollution and alleviate its toxicity, including plant-based dietary intervention. This review hypothesizes that microRNAs (miRNAs), as regulatory eukaryotic transcripts, play crucial roles in modulating cadmium-induced organ damage, and plant food-derived bioactive compounds provide protective effects via miRNA-mediated mechanisms. The review reveals that there are interplays between certain miRNAs and plant food-derived dietary bioactive substances when these bioactives, especially phenolics, counteract cadmium toxicity through regulating physiologic and pathologic events (including oxidative stress, apoptosis, autophagy and inflammation). The review discusses common miRNA-associated physiologic/pathologic events and signal pathways shared by the cadmium toxicity and dietary intervention processes. This paper identifies the existing knowledge gaps and potential future work (e.g. joint actions between miRNAs and other noncoding RNAs in the fights against cadmium). The insights provided by this review can improve food safety strategies and public health outcomes.
Collapse
Affiliation(s)
- Rili Hao
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Feng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Dongxiao Sun-Waterhouse
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China.
| | - Dapeng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China.
| |
Collapse
|
7
|
Zhang CY, Ou AJ, Jin L, Yang NSY, Deng P, Guan CX, Huang XT, Duan JX, Zhou Y. Cadmium exposure triggers alveolar epithelial cell pyroptosis by inducing mitochondrial oxidative stress and activating the cGAS-STING pathway. Cell Commun Signal 2024; 22:566. [PMID: 39587603 PMCID: PMC11590492 DOI: 10.1186/s12964-024-01946-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Cadmium is a ubiquitous toxic metal and environmental pollutant. More and more studies have shown that cadmium exposure can damage lung function. Alveolar epithelial cells (AECs) are structural cells that maintain the stability of lung function. The injury of AECs is an essential determinant of many lung diseases. In the lung, cadmium accumulation can cause damage to AECs. However, the specific mechanism is still unclear. This study aimed to explore the key mechanism underlying the injury of AECs caused by cadmium exposure. METHODS The main modes of death of AECs induced by cadmium exposure were evaluated in vivo and in vitro. Transcriptomic changes of AECs induced by cadmium exposure were analyzed using RNA-sequence. Mitochondrial ROS scavengers (mitoQ), voltage-dependent anion channel 1 (VDAC1) oligomer inhibitor (VBIT4), and cyclic GMP-AMP synthase (cGAS) inhibitor (RU.521) were used to assess whether cadmium exposure triggered pyroptosis of AECs by inducing mitochondrial stress to activate the cGAS-STING-NLRP3 axis. RESULTS In this study, the expression of pyroptosis-related proteins was significantly up-regulated in the cadmium-exposed AECs, while the expression of apoptosis, necroptosis, and ferroptosis-related proteins had no significant up-regulated. The pan-caspase inhibitor ZVAD-FMK significantly reduced cell death. Thus, our research indicates that pyroptosis is the primary type of AEC death exported to cadmium. Mechanistically, RNA-seq and Western Blot results showed that cadmium exposure activated the cGAS-STING pathway in AECs and promoted pyroptosis by activating the NLRP3 inflammasome. Further investigation of the mechanism found that cadmium exposure caused mitochondrial oxidative stress, which led to mtDNA leakage into the cytoplasm and activated the cGAS-STING pathway. In addition, inhibition of the cGAS-STING pathway significantly alleviated lung injury induced by cadmium exposure in mice. CONCLUSION Our study confirmed that pyroptosis of AECs was a vital mechanism of lung injury after cadmium exposure in a cGAS-STING-dependent manner, which may provide a new target for the treatment of lung diseases induced by cadmium exposure.
Collapse
Affiliation(s)
- Chen-Yu Zhang
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, 410078, Hunan, China
| | - An-Jun Ou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, 410078, Hunan, China
| | - Ling Jin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, 410078, Hunan, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, 410078, Hunan, China
| | - Ping Deng
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, 410078, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, 410078, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China
| | - Jia-Xi Duan
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China.
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, 410078, Hunan, China.
| |
Collapse
|
8
|
Li CX, Talukder M, Xu YR, Zhu SY, Wang YX, Li JL. Cadmium causes cerebral mitochondrial dysfunction through regulating mitochondrial HSF1. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 360:124677. [PMID: 39127336 DOI: 10.1016/j.envpol.2024.124677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/15/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Mitochondria, as the powerhouse of the cell, play a vital role in maintaining cellular energy homeostasis and are known to be a primary target of cadmium (Cd) toxicity. The improper targeting of proteins to mitochondria can compromise the normal functions of the mitochondria. However, the precise mechanism by which protein localization contributes to the development of mitochondrial dysfunction induced by Cd is still not fully understood. For this research, Hy-Line white variety chicks (1-day-old) were used and equally distributed into 4 groups: the Control group (fed with a basic diet), the Cd35 group (basic diet with 35 mg/kg CdCl2), the Cd70 group (basic diet with 70 mg/kg CdCl2) and the Cd140 group (basic diet with 140 mg/kg CdCl2), respectively for 90 days. It was found that Cd caused the accumulation of heat shock factor 1 (HSF1) in the mitochondria, and the overexpression of HSF1 in the mitochondria led to mitochondrial dysfunction and neuronal damage. This process is due to the mitochondrial HSF1 (mtHSF1), causing mitochondrial fission through the upregulation of dynamin-related protein 1 (Drp1) content, while inhibiting oligomer formation of single-stranded DNA-binding protein 1 (SSBP1), resulting in the mitochondrial DNA (mtDNA) deletion. The findings unveil an unforeseen role of HSF1 in triggering mitochondrial dysfunction.
Collapse
Affiliation(s)
- Chen-Xi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh
| | - Ya-Ru Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shi-Yong Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yu-Xiang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
9
|
Liu Y, Chen C, Hao Z, Shen J, Tang S, Dai C. Ellagic Acid Reduces Cadmium Exposure-Induced Apoptosis in HT22 Cells via Inhibiting Oxidative Stress and Mitochondrial Dysfunction and Activating Nrf2/HO-1 Pathway. Antioxidants (Basel) 2024; 13:1296. [PMID: 39594438 PMCID: PMC11590970 DOI: 10.3390/antiox13111296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Exposure to cadmium sulfate (CdSO4) can lead to neurotoxicity. Nevertheless, the precise molecular mechanisms underlying this phenomenon remain unclear, and effective treatment strategies are scarce. This study explored the protective effects of ellagic acid (EA), a natural polyphenolic compound, against CdSO4 exposure-induced neurotoxicity in HT22 cells and the underlying molecular mechanisms. Our findings demonstrated that exposure of HT22 cells to CdSO4 resulted in apoptosis, which was effectively reversed by EA in a dose-dependent manner. EA supplementation also decreased reactive oxygen species (ROS) and mitochondrial ROS production, reduced malondialdehyde (MDA) levels, and restored the activities of superoxide dismutase (SOD) and catalase (CAT). Additionally, EA supplementation at 5-20 μM significantly counteracted Cd-induced the loss of mitochondrial membrane potential and the decrease of ATP and reduced the ratio of Bax/Bcl-2 and cleaved-caspase-3 protein expression. Furthermore, EA supplementation resulted in the upregulation of Nrf2 and HO-1 protein and mRNAs while simultaneously downregulating the phosphorylation of JNK and p38 proteins. The pharmacological inhibition of c-Jun N-terminal kinase (JNK) partially attenuated the activation of the Nrf2/HO-1 pathway induced by CdSO4 and exacerbated its cytotoxic effects. In conclusion, our findings suggest that ethyl acetate (EA) supplementation offers protective effects against CdSO4-induced apoptosis in HT22 cells by inhibiting oxidative stress and activating the Nrf2 signaling pathway. Furthermore, the activation of the JNK pathway appears to play a protective role in CdSO4-induced apoptosis in HT22 cells.
Collapse
Affiliation(s)
- Yue Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.L.); (C.C.); (Z.H.); (J.S.)
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Chunhong Chen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.L.); (C.C.); (Z.H.); (J.S.)
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.L.); (C.C.); (Z.H.); (J.S.)
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.L.); (C.C.); (Z.H.); (J.S.)
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Shusheng Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.L.); (C.C.); (Z.H.); (J.S.)
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.L.); (C.C.); (Z.H.); (J.S.)
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| |
Collapse
|
10
|
Huang H, Lin Y, Xin J, Sun N, Zhao Z, Wang H, Duan L, Zhou Y, Liu X, Fang J, Jing B, Pan K, Zeng Y, Zeng D, Li H, Ma H, Bai Y, Wei L, Ni X. Fluoride exposure-induced gut microbiota alteration mediates colonic ferroptosis through N 6-methyladenosine (m 6A) mediated silencing of SLC7A11. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116816. [PMID: 39096685 DOI: 10.1016/j.ecoenv.2024.116816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/05/2024]
Abstract
Fluoride exposure is widespread worldwide and poses a significant threat to organisms, particularly to their gastrointestinal tracts. However, due to limited knowledge of the mechanism of fluoride induced intestinal injury, it has been challenging to develop an effective treatment. To address this issue, we used a series of molecular biology in vitro and in vivo experiments. NaF triggered m6A mediated ferroptosis to cause intestinal damage. Mechanistically, NaF exposure increased the m6A level of SLC7A11 mRNA, promoted YTHDF2 binding to m6A-modified SLC7A11 mRNA, drove the degradation of SLC7A11 mRNA, and led to a decrease in its protein expression, which eventually triggers ferroptosis. Moreover, NaF aggravated ferroptosis of the colon after antibiotics destroyed the composition of gut microbiota. 16 S rRNA sequencing and SPEC-OCCU plots, Zi-Pi relationships, and Spearman correlation coefficients verified that Lactobacillus murinus (ASV54, ASV58, and ASV82) plays a key role in the response to NaF-induced ferroptosis. Collectively, NaF-induced gut microbiota alteration mediates severe intestinal cell injury by inducing m6A modification-mediated ferroptosis. Our results highlight a key mechanism of the gut in response to NaF exposure and suggest a valuable theoretical basis for its prevention and treatment.
Collapse
Affiliation(s)
- Haonan Huang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yu Lin
- Department of Gastroenterology, Southern Medical University Hospital of Integrative Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Jinge Xin
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhifang Zhao
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Hesong Wang
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lixiao Duan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yanxi Zhou
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xingmei Liu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jing Fang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hao Li
- Plateau Brain Science Research Center, Tibet University, Lhasa, Tibet 850000, China
| | - Hailin Ma
- Plateau Brain Science Research Center, Tibet University, Lhasa, Tibet 850000, China
| | - Yang Bai
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Limin Wei
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Wen Y, Fu Z, Li J, Liu M, Wang X, Chen J, Chen Y, Wang H, Wen S, Zhang K, Deng Y. Targeting m 6A mRNA demethylase FTO alleviates manganese-induced cognitive memory deficits in mice. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:134969. [PMID: 38908185 DOI: 10.1016/j.jhazmat.2024.134969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Manganese (Mn) induced learning and memory deficits through mechanisms that are not fully understood. In this study, we discovered that the demethylase FTO was significantly downregulated in hippocampal neurons in an experimental a mouse model of Mn exposure. This decreased expression of FTO was associated with Mn-induced learning and memory impairments, as well as the dysfunction in synaptic plasticity and damage to regional neurons. The overexpression of FTO, or its positive modulation with agonists, provides protection against neurological damage and cognitive impairments. Mechanistically, FTO interacts synergistically with the reader YTHDF3 to facilitate the degradation of GRIN1 and GRIN3B through the m6A modification pathway. Additionally, Mn decreases the phosphorylation of SOX2, which specifically impairs the transcriptional regulation of FTO activity. Additionally, we found that the natural compounds artemisinin and apigenin that can bind molecularly with SOX2 and reduce Mn-induced cognitive dysfunction in mice. Our findings suggest that the SOX2-FTO-Grins axis represents a viable target for addressing Mn-induced neurotoxicity and cognitive impairments.
Collapse
Affiliation(s)
- Yi Wen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Zhushan Fu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China; Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Mingyue Liu
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xinmiao Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Jingqi Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Yue Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Haocheng Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Sihang Wen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Ke Zhang
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China; Institute of Health Professions Education Assessment and Reform, China Medical University, Shenyang, China.
| |
Collapse
|
12
|
Zhang J, Xiong YW, Zhu HL, Tan LL, Zhou H, Zheng XM, Zhang YF, Chang W, Xu DX, Wei T, Guan SZ, Wang H. Adolescent co-exposure to environmental cadmium and high-fat diet induces cognitive decline via Larp7 m6A-mediated SIRT6 inhibition. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135159. [PMID: 39002485 DOI: 10.1016/j.jhazmat.2024.135159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/07/2024] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
The effects and underlying mechanisms of adolescent exposure to combined environmental hazards on cognitive function remain unclear. Here, using a combined exposure model, we found significant cognitive decline, hippocampal neuronal damage, and neuronal senescence in mice exposed to cadmium (Cd) and high-fat diet (HFD) during adolescence. Furthermore, we observed a significant downregulation of Sirtuin 6 (SIRT6) expression in the hippocampi of co-exposed mice. UBCS039, a specific SIRT6 activator, markedly reversed the above adverse effects. Further investigation revealed that co-exposure obviously reduced the levels of La ribonucleoprotein 7 (LARP7), disrupted the interaction between LARP7 and SIRT6, ultimately decreasing SIRT6 expression in mouse hippocampal neuronal cells. Overexpression of Larp7 reversed the combined exposure-induced SIRT6 decrease and senescence in mouse hippocampal neuronal cells. Additionally, the results showed notably elevated levels of Larp7 m6A and YTH domain family protein 2 (YTHDF2) in mouse hippocampal neuronal cells treated with the combined hazards. Ythdf2 short interfering RNA, RNA immunoprecipitation, and RNA stability assays further demonstrated that YTHDF2 mediated the degradation of Larp7 mRNA under combined exposure. Collectively, adolescent co-exposure to Cd and HFD causes hippocampal senescence and cognitive decline in mice by inhibiting LARP7-mediated SIRT6 expression in an m6A-dependent manner.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yong-Wei Xiong
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China
| | - Hua-Long Zhu
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China
| | - Lu-Lu Tan
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Huan Zhou
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Xin-Mei Zheng
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yu-Feng Zhang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Wei Chang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China
| | - Tian Wei
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China.
| | - Su-Zhen Guan
- School of Public Health, Ningxia Medical University, China.
| | - Hua Wang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China.
| |
Collapse
|
13
|
Deng P, Fan T, Gao P, Peng Y, Li M, Li J, Qin M, Hao R, Wang L, Li M, Zhang L, Chen C, He M, Lu Y, Ma Q, Luo Y, Tian L, Xie J, Chen M, Xu S, Zhou Z, Yu Z, Pi H. SIRT5-Mediated Desuccinylation of RAB7A Protects Against Cadmium-Induced Alzheimer's Disease-Like Pathology by Restoring Autophagic Flux. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402030. [PMID: 38837686 PMCID: PMC11321632 DOI: 10.1002/advs.202402030] [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: 02/26/2024] [Revised: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cadmium (Cd) is a neurotoxic contaminant that induces cognitive decline similar to that observed in Alzheimer's disease (AD). Autophagic flux dysfunction is attributed to the pathogenesis of AD, and this study aimed to investigate the effect of autophagy on environmental Cd-induced AD progression and the underlying mechanism. Here, Cd exposure inhibited autophagosome-lysosome fusion and impaired lysosomal function, leading to defects in autophagic clearance and then to APP accumulation and nerve cell death. Proteomic analysis coupled with Ingenuity Pathway Analysis (IPA) identified SIRT5 as an essential molecular target in Cd-impaired autophagic flux. Mechanistically, Cd exposure hampered the expression of SIRT5, thus increasing the succinylation of RAB7A at lysine 31 and inhibiting RAB7A activity, which contributed to autophagic flux blockade. Importantly, SIRT5 overexpression led to the restoration of autophagic flux blockade, the alleviation of Aβ deposition and memory deficits, and the desuccinylation of RAB7A in Cd-exposed FAD4T mice. Additionally, SIRT5 levels decrease mainly in neurons but not in other cell clusters in the brains of AD patients according to single-nucleus RNA sequencing data from the public dataset GSE188545. This study reveals that SIRT5-catalysed RAB7A desuccinylation is an essential adaptive mechanism for the amelioration of Cd-induced autophagic flux blockade and AD-like pathogenesis.
Collapse
Affiliation(s)
- Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Tengfei Fan
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410007China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Yongchun Peng
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410007China
| | - Min Li
- Basic Medical LaboratoryGeneral Hospital of Central Theater CommandWuhan430070China
- Hubei Key Laboratory of Central Nervous System Tumour and InterventionWuhan430070China
| | - Jingdian Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Mingke Qin
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Rongrong Hao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Liting Wang
- Biomedical Analysis CenterArmy Medical UniversityChongqing400038China
| | - Min Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Shangcheng Xu
- Center of Laboratory MedicineChongqing Prevention and Treatment Center for Occupational DiseasesChongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and PoisoningChongqing400060China
| | - Zhou Zhou
- Center for Neuro IntelligenceSchool of MedicineChongqing UniversityChongqing400030China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education)Army Medical University (Third Military Medical University)Chongqing400038China
- State Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqing400038China
| |
Collapse
|
14
|
Qian B, Li TY, Zheng ZX, Zhang HY, Xu WQ, Mo SM, Cui JJ, Chen WJ, Lin YC, Lin ZN. The involvement of SigmaR1 K142 degradation mediated by ERAD in neural senescence linked with CdCl 2 exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134466. [PMID: 38718507 DOI: 10.1016/j.jhazmat.2024.134466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 05/30/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Due to its uncertain pathogenesis, there is currently no treatment available for AD. Increasing evidences have linked cellular senescence to AD, although the mechanism triggering cellular senescence in AD requires further exploration. To investigate the involvement of cellular senescence in AD, we explored the effects of cadmium chloride (CdCl2) exposure, one of the potential environmental risk factors for AD, on neuron senescence in vivo and in vitro. β-amyloid (Aβ) and tubulin-associated protein (tau) pathologies were found to be enhanced by CdCl2 exposure in the in vitro models, while p53/p21/Rb cascade-related neuronal senescence pathways were activated. Conversely, the use of melatonin, a cellular senescence inhibitor, or a cadmium ion chelator suppressed CdCl2-induced neuron senescence, along with the Aβ and tau pathologies. Mechanistically, CdCl2 exposure activated the suppressor enhancer Lin-12/Notch 1-like (SEL1L)/HMG-CoA reductase degradation 1 (HRD1)-regulated endoplasmic reticulum-associated degradation (ERAD), which enhanced the ubiquitin degradation of sigma-1 receptor (SigmaR1) by specifically recognizing its K142 site, resulting in the activation of the p53/p21/Rb pathway via the induction of Ca2+ dyshomeostasis and mitochondrial dysfunction. In the in vivo models, the administration of the SigmaR1 agonist ANAVEX2-73 rescues neurobehavioral inhibition and alleviates cellular senescence and AD-like pathology in the brain tissue of CdCl2-exposed mice. Consequently, the present study revealed a novel senescence-associated regulatory route for the SEL1L/HRD1/SigmaR1 axis that affects the pathological progression of CdCl2 exposure-associated AD. CdCl2 exposure activated SEL1L/HRD1-mediated ERAD and promoted the ubiquitinated degradation of SigmaR1, activating p53/p21/Rb pathway-regulated neuronal senescence. The results of the present study suggest that SigmaR1 may function as a neuroprotective biomarker of neuronal senescence, and pharmacological activation of SigmaR1 could be a promising intervention strategy for AD therapy.
Collapse
Affiliation(s)
- Bo Qian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ting-Yu Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhao-Xuan Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Han-Yu Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wen-Qi Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Su-Min Mo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jia-Jia Cui
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei-Jie Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yu-Chun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zhong-Ning Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
15
|
Zhao H, Yan Y, Gao Y, Wang J, Li S. Tris (2-chloroisopropyl) phosphate and Tris (nonylphenyl) phosphite Promote Human Renal Cell Apoptosis through the ERK/CEPBA/Long Non-Coding RNA Cytoskeleton Regulator Axis. TOXICS 2024; 12:452. [PMID: 39058104 PMCID: PMC11281261 DOI: 10.3390/toxics12070452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024]
Abstract
Organophosphorus compounds (OPs) are widely used and have the potential to be harmful environmental toxicants to humans. Long non-coding RNA (lncRNA) plays a crucial regulatory role in cytotoxicity. This study aimed to investigate the effects of OPs on the expression of lncRNAs in cells. The effects of the industrial OPs TNPP and TCPP on both CYTOR and cellular viability were examined in the following human renal cell lines: HEK293T and HK-2. Both TCPP and TNPP downregulated CYTOR expression, increased reactive oxygen species levels, and induced apoptosis; the upregulated expression of CYTOR resulted in a reduction in apoptosis. The results of the luciferase reporter assay and the knock-down assay indicate that CEBPA binds to the upstream promoter region of CYTOR and regulates its transcription. Furthermore, TCPP and TNPP were found to downregulate the phosphorylation of ERK in the signaling pathway that is upstream of CEBPA. These results indicate that TCPP and TNPP can decrease the level of CEBPA by reducing ERK phosphorylation; this leads to a decrease in CYTOR expression, which further promotes cellular reactive oxygen species and apoptosis. Therefore, the ERK/CEBPA/CYTOR axis is one of the pathways by which organophosphates produce cytotoxicity, leading to renal cell injury. This study presents evidence for both the abnormal expression of lncRNA that is caused by organophosphates and the regulatory function of lncRNA regarding downstream cellular viability.
Collapse
Affiliation(s)
| | | | | | | | - Sheng Li
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang 550025, China; (H.Z.); (Y.Y.); (Y.G.); (J.W.)
| |
Collapse
|
16
|
Wang J, Yin H, Li G, Wu D, Xu Y, Chen Y, Wang X, Xing Y, Zhang T, Fei D, Yang P, Fang F, Tao Y, Li X, Yu J, Yang Y, Li Z, Shi L, Zhang Z, Pan J. METTL14 promotes neuroblastoma formation by inhibiting YWHAH via an m6A-YTHDF1-dependent mechanism. Cell Death Discov 2024; 10:186. [PMID: 38649363 PMCID: PMC11035551 DOI: 10.1038/s41420-024-01959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/31/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Neuroblastoma (NB) is a common childhood tumor with a high incidence worldwide. The regulatory role of RNA N6-methyladenosine (m6A) in gene expression has attracted significant attention, and the impact of methyltransferase-like 14 (METTL14) on tumor progression has been extensively studied in various types of cancer. However, the specific influence of METTL14 on NB remains unexplored. Using data from the Target database, our study revealed significant upregulation of METTL14 expression in high-risk NB patients, with strong correlation with poor prognosis. Furthermore, we identified ETS1 and YY1 as upstream regulators that control the expression of METTL14. In vitro experiments involving the knockdown of METTL14 in NB cells demonstrated significant inhibition of cell proliferation, migration, and invasion. In addition, suppressing METTL14 inhibited NB tumorigenesis in nude mouse models. Through MeRIP-seq and RNA-seq analyses, we further discovered that YWHAH is a downstream target gene of METTL14. Mechanistically, we observed that methylated YWHAH transcripts, particularly those in the 5' UTR, were specifically recognized by the m6A "reader" protein YTHDF1, leading to the degradation of YWHAH mRNA. Moreover, the downregulation of YWHAH expression activated the PI3K/AKT signaling pathway, promoting NB cell activity. Overall, our study provides valuable insights into the oncogenic effects of METTL14 in NB cells, highlighting its role in inhibiting YWHAH expression through an m6A-YTHDF1-dependent mechanism. These findings also suggest the potential utility of a biomarker panel for prognostic prediction in NB patients.
Collapse
Affiliation(s)
- Jianwei Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Di Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yunyun Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yanling Chen
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Xiaodong Wang
- Children's Hospital of Soochow University, Suzhou, China
| | - Yujiao Xing
- Children's Hospital of Soochow University, Suzhou, China
| | - Ting Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Danhong Fei
- Department of Pediatrics, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Pengcheng Yang
- Department of Pediatric Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yanfang Tao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Juanjuan Yu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yang Yang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
17
|
Shen Y, Wei C, Taishi Y, Zhang G, Su Z, Zhao P, Wang Y, Li M, Ji Y, Sun L. Association between the circulating very long-chain saturated fatty acid and cognitive function in older adults: findings from the NHANES. BMC Public Health 2024; 24:1061. [PMID: 38627688 PMCID: PMC11022414 DOI: 10.1186/s12889-024-18478-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Age-related cognitive decline has a significant impact on the health and longevity of older adults. Circulating very long-chain saturated fatty acids (VLSFAs) may actively contribute to the improvement of cognitive function. The objective of this study was to investigate the associations between arachidic acid (20:0), docosanoic acid (22:0), tricosanoic acid (23:0), and lignoceric acid (24:0) with cognitive function in older adults. METHODS This study used a dataset derived from the 2011-2014 National Health and Nutrition Examination Survey (NHANES). A total of 806 adults (≥ 60 years) were included who underwent comprehensive cognitive testing and plasma fatty acid measurements. Multivariable linear regression, restricted cubic spline (RCS), and interaction analyses were used to assess associations between VLSFAs and cognitive function. Partial Spearman' s correlation analysis was used to examine the correlations between VLSFAs and palmitic acid (16:0), high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, total cholesterol, triglycerides, systemic inflammatory markers, and dietary nutrients. RESULTS Multivariable linear regression analysis, adjusting for sociodemographic, clinical conditions, and lifestyle factors, showed that 22:0 and 24:0 levels were positively associated with better global cognitive function (β = 0.37, 95% confidence interval [CI] = 0.01, 0.73; β = 0.73, 95% CI = 0.29, 1.2, respectively) as well as better CEARD-DR Z-score (β = 0.82, 95% CI = 0.36, 1.3 and β = 1.2, 95% CI = 0.63, 1.8, respectively). RCS analysis showed linear associations between higher 22:0 and 24:0 levels and better cognitive performance in both global cognitive function and CERAD-DR tests. CONCLUSIONS The study suggests that higher levels of 22:0 and 24:0 are associated with better global cognitive function in older adults. 22:0 and 24:0 may be important biomarkers for recognizing cognitive impairment, and supplementation with specific VLSFAs (22:0 and 24:0) may be an important intervention to improve cognitive function. Further studies are needed to elucidate the underlying biological mechanisms between VLSFAs and cognitive function.
Collapse
Affiliation(s)
- Yanxin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yezi Taishi
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhan Su
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Panpan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yongchun Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yingshi Ji
- Department of Pharmacology, Physiology and Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, 130021, Changchun, China.
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
18
|
Liu WW, Zheng SQ, Li T, Fei YF, Wang C, Zhang S, Wang F, Jiang GM, Wang H. RNA modifications in cellular metabolism: implications for metabolism-targeted therapy and immunotherapy. Signal Transduct Target Ther 2024; 9:70. [PMID: 38531882 DOI: 10.1038/s41392-024-01777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
Cellular metabolism is an intricate network satisfying bioenergetic and biosynthesis requirements of cells. Relevant studies have been constantly making inroads in our understanding of pathophysiology, and inspiring development of therapeutics. As a crucial component of epigenetics at post-transcription level, RNA modification significantly determines RNA fates, further affecting various biological processes and cellular phenotypes. To be noted, immunometabolism defines the metabolic alterations occur on immune cells in different stages and immunological contexts. In this review, we characterize the distribution features, modifying mechanisms and biological functions of 8 RNA modifications, including N6-methyladenosine (m6A), N6,2'-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), N7-methylguanosine (m7G), Pseudouridine (Ψ), adenosine-to-inosine (A-to-I) editing, which are relatively the most studied types. Then regulatory roles of these RNA modification on metabolism in diverse health and disease contexts are comprehensively described, categorized as glucose, lipid, amino acid, and mitochondrial metabolism. And we highlight the regulation of RNA modifications on immunometabolism, further influencing immune responses. Above all, we provide a thorough discussion about clinical implications of RNA modification in metabolism-targeted therapy and immunotherapy, progression of RNA modification-targeted agents, and its potential in RNA-targeted therapeutics. Eventually, we give legitimate perspectives for future researches in this field from methodological requirements, mechanistic insights, to therapeutic applications.
Collapse
Affiliation(s)
- Wei-Wei Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Clinical Medicine, Shandong University, Jinan, China
| | - Si-Qing Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Tian Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yun-Fei Fei
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Chen Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Shuang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Fei Wang
- Neurosurgical Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Guan-Min Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Hao Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
19
|
Li J, Gao P, Qin M, Wang J, Luo Y, Deng P, Hao R, Zhang L, He M, Chen C, Lu Y, Ma Q, Li M, Tan M, Wang L, Yue Y, Wang H, Tian L, Xie J, Chen M, Yu Z, Zhou Z, Pi H. Long-term cadmium exposure induces epithelial-mesenchymal transition in breast cancer cells by activating CYP1B1-mediated glutamine metabolic reprogramming in BT474 cells and MMTV-Erbb2 mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 918:170773. [PMID: 38336054 DOI: 10.1016/j.scitotenv.2024.170773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/04/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
Cadmium (Cd) exposure is known to enhance breast cancer (BC) progression. Cd promotes epithelial-mesenchymal transition (EMT) in BC cells, facilitating BC cell aggressiveness and invasion, but the underlying molecular mechanisms are unclear. Hence, transgenic MMTV-Erbb2 mice (6 weeks) were orally administered Cd (3.6 mg/L, approximately equal to 19.64 μΜ) for 23 weeks, and BC cells (BT474 cells) were exposed to Cd (0, 0.1, 1 or 10 μΜ) for 72 h to investigate the effect of Cd exposure on EMT in BC cells. Chronic Cd exposure dramatically expedited tumor metastasis to multiple organs; decreased E-cadherin density; and increased Vimentin, N-cadherin, ZEB1, and Twist density in the tumor tissues of MMTV-Erbb2 mice. Notably, transcriptomic analysis of BC tumors revealed cytochrome P450 1B1 (CYP1B1) as a key factor that regulates EMT progression in Cd-treated MMTV-Erbb2 mice. Moreover, Cd increased CYP1B1 expression in MMTV-Erbb2 mouse BC tumors and in BT474 cells, and CYP1B1 inhibition decreased Cd-induced BC cell malignancy and EMT in BT474 cells. Importantly, the promotion of EMT by CYP1B1 in Cd-treated BC cells was presumably controlled by glutamine metabolism. This study offers novel perspectives into the effect of environmental Cd exposure on driving BC progression and metastasis, and this study provides important guidance for comprehensively assessing the ecological and health risks of Cd.
Collapse
Affiliation(s)
- Jingdian Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mingke Qin
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Junhua Wang
- Nuclear Medicine Department, General Hospital of Tibet Military Area Command, Lhasa 850000, Xizang, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rongrong Hao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Min Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Miduo Tan
- Department of Breast Surgery, Central Hospital of Zhuzhou City, Central South University, Zhuzhou 412000, Hunan, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yang Yue
- Bioinformatics Center of Academy of Military Medical Sciences, Beijing 100850, China
| | - Hui Wang
- Nuclear Medicine Department, General Hospital of Tibet Military Area Command, Lhasa 850000, Xizang, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China; State key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
20
|
Yu G, Wu L, Su Q, Ji X, Zhou J, Wu S, Tang Y, Li H. Neurotoxic effects of heavy metal pollutants in the environment: Focusing on epigenetic mechanisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123563. [PMID: 38355086 DOI: 10.1016/j.envpol.2024.123563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/04/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
The pollution of heavy metals (HMs) in the environment is a significant global environmental issue, characterized by its extensive distribution, severe contamination, and profound ecological impacts. Excessive exposure to heavy metal pollutants can damage the nervous system. However, the mechanisms underlying the neurotoxicity of most heavy metals are not completely understood. Epigenetics is defined as a heritable change in gene function that can influence gene and subsequent protein expression levels without altering the DNA sequence. Growing evidence indicates that heavy metals can induce neurotoxic effects by triggering epigenetic changes and disrupting the epigenome. Compared with genetic changes, epigenetic alterations are more easily reversible. Epigenetic reprogramming techniques, drugs, and certain nutrients targeting specific epigenetic mechanisms involved in gene expression regulation are emerging as potential preventive or therapeutic tools for diseases. Therefore, this review provides a comprehensive overview of epigenetic modifications encompassing DNA/RNA methylation, histone modifications, and non-coding RNAs in the nervous system, elucidating their association with various heavy metal exposures. These primarily include manganese (Mn), mercury (Hg), lead (Pb), cobalt (Co), cadmium (Cd), nickel (Ni), sliver (Ag), toxic metalloids arsenic (As), and etc. The potential epigenetic mechanisms in the etiology, precision prevention, and target therapy of various neurodevelopmental disorders or different neurodegenerative diseases are emphasized. In addition, the current gaps in research and future areas of study are discussed. From a perspective on epigenetics, this review offers novel insights for prevention and treatment of neurotoxicity induced by heavy metal pollutants.
Collapse
Affiliation(s)
- Guangxia Yu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Lingyan Wu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Qianqian Su
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xianqi Ji
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Jinfu Zhou
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Maternity and Child Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
| | - Siying Wu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Ying Tang
- Fujian Center for Prevention and Control Occupational Diseases and Chemical Poisoning, Fuzhou 350125, China
| | - Huangyuan Li
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
21
|
Zhang Z, Li Y, Feng H, Li S, Qin Z, Li J, Chen Y, Zhang Y, Zhao Y, Yin X, Huang B, Gao Y, Shi Y, Shi H. Effects of postweaning cadmium exposure on socioemotional behaviors in adolescent male mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116089. [PMID: 38354436 DOI: 10.1016/j.ecoenv.2024.116089] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Exposure to cadmium (Cd), a toxic heavy metal classified as an environmental endocrine disruptor, can exert significant toxicity in both animals and humans. However, the potential effects of Cd exposure on socioemotional behaviors are still poorly understood, as are the underlying mechanisms. In the present study, employing a series of behavioral tests as well as 16 S rRNA sequencing analysis, we investigated the long-term effects of Cd exposure on socioemotional behaviors and their associated mechanisms in mice based on the brain-gut interaction theory. The results showed that postweaning exposure to Cd reduced the ability to resist depression, decreased social interaction, subtly altered sexual preference, and changed the composition of the gut microbiota in male mice during adolescence. These findings provided direct evidence for the deleterious effects of exposure to Cd in the postweaning period on socioemotional behaviors later in adolescence, and suggested that these effects of Cd exposure may be linked to changes in the gut microbiota.
Collapse
Affiliation(s)
- Zhengxin Zhang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Yuxin Li
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Hao Feng
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Shijun Li
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Zihan Qin
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Jiabo Li
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Yifei Chen
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Yue Zhang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Ye Zhao
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xueyong Yin
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Boya Huang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yuan Gao
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang 050017, China.
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang 050017, China; Nursing School, Hebei Medical University, Shijiazhuang 050031, China; Hebei Key Laboratory of Forensic Medicine, Hebei Province, Shijiazhuang 050017, China.
| |
Collapse
|
22
|
Lyu L, Tao Y, Abaakil K, Gu Y, Zhong G, Hu Y, Zhang Y. Novel insights into DEHP-induced zebrafish spleen damage: Cellular apoptosis, mitochondrial dysfunction, and innate immunity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169324. [PMID: 38145680 DOI: 10.1016/j.scitotenv.2023.169324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 12/27/2023]
Abstract
DEHP (Di(2-ethylhexyl) phthalate) is the most abundant phthalate component detected in environmental samples as it is widely used in the manufacturing of children's toys, medical devices and furniture. Due to its wide prevalence and propensity to accumulate in the food chain, significant concerns have risen about the safety profile of DEHP. Here, we used a zebrafish model to investigate the toxicity mechanisms of DEHP. Our results indicated that exposure to DEHP altered the ROS content in zebrafish spleen and inhibited the activities of antioxidant enzymes SOD and CAT, detoxification enzyme GSH-Px and induced histopathological damage. In addition, elucidated the mechanism of DEHP significantly promoted apoptosis and caused damage in spleen cells through the bax/bcl-2 pathway. Further genetic testing demonstrated significant alterations in mitochondrial biogenesis, fission, and fusion-related genes and suggested potential mechanistic pathways, including GM10532/m6A/FIS1 axis, the STAT3/POA1 axis, and the NFR1/TFAM axis. Serological and genomic analysis indicated that DEHP exposure activated the C3 complement cascade immune pathway and interfered with innate immune function. IBRv2 analysis proposes that innate immunity may serve as a signal indicator of early toxic responses to DEHP pollutants. This study provided comprehensive cellular and genetic data for DEHP toxicity studies and emphasized the need for future management and remediation of DEHP contamination. It also provides data to specifically support the health risk assessments of DEHP, as well as contributing to broader health and environmental research.
Collapse
Affiliation(s)
- Liang Lyu
- College of Resources and Environment, Northeast Agricultural University, Changjiang Street 600, Harbin 150030, PR China; Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, SW7 2AZ London, UK.
| | - Yue Tao
- College of Resources and Environment, Northeast Agricultural University, Changjiang Street 600, Harbin 150030, PR China
| | - Kaoutar Abaakil
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, SW7 2AZ London, UK.
| | - Yanyan Gu
- College of Resources and Environment, Northeast Agricultural University, Changjiang Street 600, Harbin 150030, PR China
| | - Guanyu Zhong
- College of Resources and Environment, Northeast Agricultural University, Changjiang Street 600, Harbin 150030, PR China
| | - Yang Hu
- College of Resources and Environment, Northeast Agricultural University, Changjiang Street 600, Harbin 150030, PR China
| | - Ying Zhang
- College of Resources and Environment, Northeast Agricultural University, Changjiang Street 600, Harbin 150030, PR China.
| |
Collapse
|
23
|
Hu C, Yang S, Zhang T, Ge Y, Chen Z, Zhang J, Pu Y, Liang G. Organoids and organoids-on-a-chip as the new testing strategies for environmental toxicology-applications & advantages. ENVIRONMENT INTERNATIONAL 2024; 184:108415. [PMID: 38309193 DOI: 10.1016/j.envint.2024.108415] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/13/2023] [Accepted: 01/01/2024] [Indexed: 02/05/2024]
Abstract
An increasing number of harmful environmental factors are causing serious impacts on human health, and there is an urgent need to accurately identify the toxic effects and mechanisms of these harmful environmental factors. However, traditional toxicity test methods (e.g., animal models and cell lines) often fail to provide accurate results. Fortunately, organoids differentiated from stem cells can more accurately, sensitively and specifically reflect the effects of harmful environmental factors on the human body. They are also suitable for specific studies and are frequently used in environmental toxicology nowadays. As a combination of organoids and organ-on-a-chip technology, organoids-on-a-chip has great potential in environmental toxicology. It is more controllable to the physicochemical microenvironment and is not easy to be contaminated. It has higher homogeneity in the size and shape of organoids. In addition, it can achieve vascularization and exchange the nutrients and metabolic wastes in time. Multi-organoids-chip can also simulate the interactions of different organs. These advantages can facilitate better function and maturity of organoids, which can also make up for the shortcomings of common organoids to a certain extent. This review firstly discussed the limitations of traditional toxicology testing platforms, leading to the introduction of new platforms: organoids and organoids-on-a-chip. Next, the applications of different organoids and organoids-on-a-chip in environmental toxicology were summarized and prospected. Since the advantages of the new platforms have not been sufficiently considered in previous literature, we particularly emphasized them. Finally, this review also summarized the opportunities and challenges faced by organoids and organoids-on-a-chip, with the expectation that readers will gain a deeper understanding of their value in the field of environmental toxicology.
Collapse
Affiliation(s)
- Chengyu Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Tianyi Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Yiling Ge
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
24
|
Bovio F, Perciballi E, Melchioretto P, Ferrari D, Forcella M, Fusi P, Urani C. Morphological and metabolic changes in microglia exposed to cadmium: Cues on neurotoxic mechanisms. ENVIRONMENTAL RESEARCH 2024; 240:117470. [PMID: 37871786 DOI: 10.1016/j.envres.2023.117470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Microglial cells play a key role in protecting the central nervous system from pathogens and toxic compounds and are involved in the pathogenesis of different neurodegenerative diseases. Cadmium is a widespread toxic heavy metal, released into the environment at a rate of 30,000 tons/year by anthropogenic activities; it is easily uptaken by the human body through diet and cigarette smoke, as well as by occupational exposure. Once inside the body, cadmium enters the cells and substitutes to zinc and other divalent cations altering many biological functions. Its extremely long half-life makes it a serious health threat. Recent data suggest a role for heavy metals in many neurodegenerative diseases; however, the role of cadmium is still to be elucidated. In this work we report the investigation of cadmium toxicity towards murine BV2 microglial cells, a widely used model for the study of neurodegeneration. Results show that increasing cadmium concentrations increase oxidative stress, a proposed mechanism of neurodegeneration, but also that BV2 cells can keep oxidative stress under control by increasing glutathione reduction. Moreover, cadmium induces alterations of cell morphology and metabolism leading to mitochondrial impairment, without switching the cells to Warburg effect. Finally cadmium induces the release of proinflammatory cytokines, but does not markedly switch BV2 cells to M1 phenotype.
Collapse
Affiliation(s)
- Federica Bovio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Elisa Perciballi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Pasquale Melchioretto
- Department of Earth and Environmental Sciences, University of Milano- Bicocca, Piazza della Scienza 1, 20126, Milan, Italy
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy
| | - Matilde Forcella
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy.
| | - Paola Fusi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126, Milan, Italy; Integrated Models for Prevention and Protection in Environmental and Occupational Health, Interuniversity Research Center, (MISTRAL), Italy.
| | - Chiara Urani
- Department of Earth and Environmental Sciences, University of Milano- Bicocca, Piazza della Scienza 1, 20126, Milan, Italy; Integrated Models for Prevention and Protection in Environmental and Occupational Health, Interuniversity Research Center, (MISTRAL), Italy
| |
Collapse
|
25
|
Deng P, Li J, Lu Y, Hao R, He M, Li M, Tan M, Gao P, Wang L, Hong H, Tao J, Lu M, Chen C, Ma Q, Yue Y, Wang H, Tian L, Xie J, Chen M, Luo Y, Yu Z, Zhou Z, Pi H. Chronic cadmium exposure triggered ferroptosis by perturbing the STEAP3-mediated glutathione redox balance linked to altered metabolomic signatures in humans. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167039. [PMID: 37716689 DOI: 10.1016/j.scitotenv.2023.167039] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Cadmium (Cd), a predominant environmental pollutant, is a canonical toxicant that acts on the kidneys. However, the nephrotoxic effect and underlying mechanism activated by chronic exposure to Cd remain unclear. In the present study, male mice (C57BL/6J, 8 weeks) were treated with 0.6 mg/L cadmium chloride (CdCl2) administered orally for 6 months, and tubular epithelial cells (TCMK-1 cells) were treated with low-dose (1, 2, and 3 μM) CdCl2 for 72 h (h). Our study results revealed that environmental Cd exposure triggered ferroptosis and renal dysfunction. Spatially resolved metabolomics enabled delineation of metabolic profiles and visualization of the disruption to glutathione homeostasis related to ferroptosis in mouse kidneys. Multiomics analysis revealed that chronic Cd exposure induced glutathione redox imbalance that depended on STEAP3-driven lysosomal iron overload. In particular, glutathione metabolic reprogramming linked to ferroptosis emerged as a metabolic hallmark in the blood of Cd-exposed workers. In conclusion, this study provides the first evidence indicating that chronic Cd exposure triggers ferroptosis and renal dysfunction that depend on STEAP3-mediated glutathione redox imbalance, greatly increasing our understanding of the metabolic reprogramming induced by Cd exposure in the kidneys and providing novel clues linking chronic Cd exposure to nephrotoxicity.
Collapse
Affiliation(s)
- Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jingdian Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rongrong Hao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Min Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Miduo Tan
- Department of Breast Surgery, Central Hospital of Zhuzhou City, Central South University, Zhuzhou 412000, Hunan, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Huihui Hong
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiawen Tao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Muxue Lu
- School of Medicine, Guangxi University, Nanning 530004, Guangxi, China
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yang Yue
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Hui Wang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China; State key Laboratory Of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
26
|
Xu Y, Hong H, Lin X, Tong T, Zhang J, He H, Yang L, Mao G, Hao R, Deng P, Yu Z, Pi H, Cheng Y, Zhou Z. Chronic cadmium exposure induces Parkinson-like syndrome by eliciting sphingolipid disturbance and neuroinflammation in the midbrain of C57BL/6J mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122606. [PMID: 37742865 DOI: 10.1016/j.envpol.2023.122606] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Cadmium (Cd) is known as a widespread environmental neurotoxic pollutant. Cd exposure is recently recognized as an etiological factor of Parkinson's disease (PD) in humans. However, the mechanism underlying Cd neurotoxicity in relation to Parkinsonism pathogenesis is unclear. In our present study, C57BL/6 J mice were exposed to 100 mg/L CdCl2 in drinking water for 8 weeks. It was found Cd exposure caused motor deficits, decreased DA neurons and induced neuropathological changes in the midbrain. Non-targeted lipidomic analysis uncovered that Cd exposure altered lipid profile, increased the content of proinflammatory sphingolipid ceramides (Cer), sphingomyelin (SM) and ganglioside (GM3) in the midbrain. In consistency with increased proinflammatory lipids, the mRNA levels of genes encoding sphingolipids biosynthesis in the midbrain were dysregulated by Cd exposure. Neuroinflammation in the midbrain was evinced by the up-regulation of proinflammatory cytokines at mRNA and protein levels. Blood Cd contents and lipid metabolites in Parkinsonism patients by ICP-MS and LC-MS/MS analyses demonstrated that elevated blood Cd concentration and proinflammatory lipid metabolites were positively associated with the score of Unified Parkinson's Disease Rating Scale (UPDRS). 3 ceramide metabolites in the blood showed good specificity as the candidate biomarkers to predict and monitor Parkinsonism and Cd neurotoxicity (AUC>0.7, p < 0.01). In summary, our present study uncovered that perturbed sphingomyelin lipid metabolism is related to the Parkinsonism pathogenesis and Cd neurotoxicity, partially compensated for the deficiency in particular metabolic biomarkers for Parkinsonism in relation to Cd exposure, and emphasized the necessity of reducing Cd exposure at population level.
Collapse
Affiliation(s)
- Yudong Xu
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Huihui Hong
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Xiqin Lin
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Tong
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Zhang
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Haotian He
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Gaofeng Mao
- Neurology Department, General Hospital of Center Theater Command, Wuhan, China
| | - Rongrong Hao
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yong Cheng
- Neurology Department, General Hospital of Center Theater Command, Wuhan, China
| | - Zhou Zhou
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China; Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
27
|
Yue Y, Zhang H, Deng P, Tan M, Chen C, Tang B, Li J, Chen F, Zhao Q, Li L, Hao R, Wang H, Luo Y, Tian L, Xie J, Chen M, Yu Z, Zhou Z, Pi H. Environmental cadmium exposure facilitates mammary tumorigenesis via reprogramming gut microbiota-mediated glutamine metabolism in MMTV-Erbb2 mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 897:165348. [PMID: 37429473 DOI: 10.1016/j.scitotenv.2023.165348] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/12/2023]
Abstract
Cadmium (Cd) is a heavy metal that has been widely reported to be linked to the onset and progression of breast cancer (BC). However, the mechanism of Cd-induced mammary tumorigenesis remains elusive. In our study, a transgenic mouse model that spontaneously develops tumors through overexpression of wild-type Erbb2 (MMTV-Erbb2) was constructed to investigate the effects of Cd exposure on BC tumorigenesis. The results showed that oral exposure to 3.6 mg/L Cd for 23 weeks dramatically accelerated tumor appearance and growth, increased Ki67 density and enhanced focal necrosis and neovascularization in the tumor tissue of MMTV-Erbb2 mice. Notably, Cd exposure enhanced glutamine (Gln) metabolism in tumor tissue, and 6-diazo-5-oxo-l-norleucine (DON), a Gln metabolism antagonist, inhibited Cd-induced breast carcinogenesis. Then our metagenomic sequencing and mass spectrometry-based metabolomics confirmed that Cd exposure disturbed gut microbiota homeostasis, especially Helicobacter and Campylobacter abundance remodeling, which altered the gut metabolic homeostasis of Gln. Moreover, intratumoral Gln metabolism profoundly increased under Cd-elevated gut permeability. Importantly, depletion of microbiota with an antibiotic cocktail (AbX) treatment led to a significant delay in the appearance of palpable tumors, inhibition of tumor growth, decrease in tumor weight, reduction in Ki67 expression and low-grade pathology in Cd-exposed MMTV-Erbb2 mice. Also, transplantation of Cd-modulated microbiota decreased tumor latency, accelerated tumor growth, increased tumor weight, upregulated Ki67 expression and exacerbated neovascularization as well as focal necrosis in MMTV-Erbb2 mice. In summary, Cd exposure induced gut microbiota dysbiosis, elevated gut permeability and increased intratumoral Gln metabolism, leading to the promotion of mammary tumorigenesis. This study provides novel insights into environmental Cd exposure-mediated carcinogenesis.
Collapse
Affiliation(s)
- Yang Yue
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Huadong Zhang
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing 400042, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Miduo Tan
- Department of Breast Surgery, The Affiliated Zhuzhou Hospital of Xiang Ya School of Medicine, Central South University, Zhuzhou 412000, Hunan, China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jingdian Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Fengqiong Chen
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing 400042, China
| | - Qi Zhao
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing 400042, China
| | - Ling Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Rongrong Hao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Hui Wang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
28
|
Xie L, Zhang X, Xie J, Xu Y, Li XJ, Lin L. Emerging Roles for DNA 6mA and RNA m6A Methylation in Mammalian Genome. Int J Mol Sci 2023; 24:13897. [PMID: 37762200 PMCID: PMC10531503 DOI: 10.3390/ijms241813897] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/25/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Epigenetic methylation has been shown to play an important role in transcriptional regulation and disease pathogenesis. Recent advancements in detection techniques have identified DNA N6-methyldeoxyadenosine (6mA) and RNA N6-methyladenosine (m6A) as methylation modifications at the sixth position of adenine in DNA and RNA, respectively. While the distributions and functions of 6mA and m6A have been extensively studied in prokaryotes, their roles in the mammalian brain, where they are enriched, are still not fully understood. In this review, we provide a comprehensive summary of the current research progress on 6mA and m6A, as well as their associated writers, erasers, and readers at both DNA and RNA levels. Specifically, we focus on the potential roles of 6mA and m6A in the fundamental biological pathways of the mammalian genome and highlight the significant regulatory functions of 6mA in neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Lin
- Guangdong Key Laboratory of Non-Human Primate Research, Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (L.X.); (X.Z.); (J.X.); (Y.X.); (X.-J.L.)
| |
Collapse
|