1
|
An G, Song J, Ying W, Lim W. Overview of the hazardous impacts of metabolism-disrupting chemicals on the progression of fatty liver diseases. Mol Cell Toxicol 2025; 21:387-397. [PMID: 40160987 PMCID: PMC11947047 DOI: 10.1007/s13273-025-00521-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 04/02/2025]
Abstract
Background Given the global increase in obesity, metabolic dysfunction-associated steatotic liver disease (MASLD) is a major health concern. Because the liver is the primary organ for xenobiotic metabolism, the impact of environmental stressors on liver homeostasis and MASLD has garnered significant interest over the past few decades. The concept of metabolism-disrupting chemicals (MDCs) has been introduced to underscore the importance of environmental factors in metabolic homeostasis. Recent epidemiological and biological studies suggest a causal link between exposure to MDCs and prevalence and progression of MASLD. Objective This review aims to introduce the emerging concept of MDCs and their representative toxic mechanisms. In particular, this review focuses on broadening the understanding of their impacts on MASLD or metabolic dysfunction-associated steatohepatitis (MASH) progression. Result Recent research has highlighted the environmental contaminants, such as heavy metals, microplastics, and pesticides, have the potential to influence hepatic metabolism and aggravate MASLD/MASH progression. These MDCs not only directly affect lipid metabolism in hepatocytes but also affect other cell types, such as immune cells and stellate cells, as well as the gut-liver axis. Conclusion Collectively, these findings contribute to establishing a well-defined adverse outcome pathway and identify novel therapeutic options for liver diseases associated with pollutants.
Collapse
Affiliation(s)
- Garam An
- Department of Biological Sciences, Institute of Basic Science, College of Science, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Jisoo Song
- Department of Biological Sciences, Institute of Basic Science, College of Science, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093 USA
| | - Whasun Lim
- Department of Biological Sciences, Institute of Basic Science, College of Science, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| |
Collapse
|
2
|
Elhemiely AA, Darwish A. Pharmacological and biochemical insights into lead-induced hepatotoxicity: Pathway interplay and the protective effects of arbutin via the oral and intraperitoneal routes in silico and in vivo. Int Immunopharmacol 2024; 142:112968. [PMID: 39226827 DOI: 10.1016/j.intimp.2024.112968] [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: 06/24/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Lead acetate (PbAc), a hazardous heavy metal, poses significant threats to human health and the environment because of widespread industrial exposure. PbAc exposure leads to liver injury primarily through oxidative stress and the disruption of key regulatory pathways. Understanding these mechanisms and exploring protective agents are vital for mitigating PbAc-induced hepatotoxicity. Therefore, we aimed to investigate the molecular pathways implicated in PbAc-induced liver damage, focusing on Sirt-1, Nrf2 (HO-1, NQO1, and SOD), Akt-1/GSK3β, m-TOR, and P53. Additionally, we aimed to assess the hepatoprotective effects of arbutin, which is administered orally and intraperitoneally, to determine the most effective delivery method. METHODOLOGY In silico analyses were conducted to identify relevant protein networks associated with Sirt-1 and AKT-1/GSK-3B pathways. The pharmacodynamic properties of arbutin were examined, followed by molecular docking studies to elucidate its interactions with the selected protein network. In vivo preclinical studies were carried out on adult male rats randomly assigned to 6 different treatment groups, including PbAc exposure and PbAc exposure treated with arbutin either orally or intraperitoneally. RESULTS PbAc exposure led to hepatic oxidative stress, as evidenced by elevated MDA levels and SIRT-1 inhibition, disrupting antioxidant pathways and activating antiautophagic and proapoptotic pathways, ultimately resulting in hepatocyte necrosis. Both oral and intraperitoneal arbutin administration effectively modifed these effects, with intraperitoneal delivery showing superior efficacy in mitigating PbAc-induced histological, immunological, and biochemical alterations. CONCLUSION This study provides insights into the molecular mechanisms underlying PbAc-induced liver injury and highlights the hepatoprotective potential of arbutin. These findings suggest that arbutin, particularly when administered intraperitoneally, holds promise as a therapeutic agent for combating PbAc-induced hepatotoxicity.
Collapse
Affiliation(s)
| | - Alshaymaa Darwish
- Department of Biochemistry, Faculty of Pharmacy, Sohag University, Sohag, Egypt.
| |
Collapse
|
3
|
Shen W, Yang M, Chen H, He C, Li H, Yang X, Zhuo J, Lin Z, Hu Z, Lu D, Xu X. FGF21-mediated autophagy: Remodeling the homeostasis in response to stress in liver diseases. Genes Dis 2024; 11:101027. [PMID: 38292187 PMCID: PMC10825283 DOI: 10.1016/j.gendis.2023.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/23/2023] [Accepted: 05/09/2023] [Indexed: 02/01/2024] Open
Abstract
Liver diseases are worldwide problems closely associated with various stresses, such as endoplasmic reticulum stress. The exact interplay between stress and liver diseases remains unclear. Autophagy plays an essential role in maintaining homeostasis, and recent studies indicate tight crosstalk between stress and autophagy in liver diseases. Once the balance between damage and autophagy is broken, autophagy can no longer resist injury or maintain homeostasis. In recent years, FGF21 (fibroblast growth factor 21)-induced autophagy has attracted much attention. FGF21 is regarded as a stress hormone and can be up-regulated by an abundance of signaling pathways in response to stress. Also, increased FGF21 activates autophagy by a complicated signaling network in which mTOR plays a pivotal role. This review summarizes the mechanism of FGF21-mediated autophagy and its derived application in the defense of stress in liver diseases and offers a glimpse into its promising prospect in future clinical practice.
Collapse
Affiliation(s)
- Wei Shen
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Modan Yang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Hao Chen
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Chiyu He
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Huigang Li
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xinyu Yang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Jianyong Zhuo
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Zuyuan Lin
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Zhihang Hu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Di Lu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
- National Center for Healthcare Quality Management in Liver Transplant, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
4
|
Wang N, Gao X, Huo Y, Li Y, Cheng F, Zhang Z. Lead exposure aggravates glucose metabolism disorders through gut microbiota dysbiosis and intestinal barrier damage in high-fat diet-fed mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:3057-3068. [PMID: 38057285 DOI: 10.1002/jsfa.13197] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Lead (Pb) is an ancient toxic metal and is still a major public health issue. Our previous study found that Pb exposure promotes metabolic disorders in obese mice, but the molecular mechanisms remain unclear. The present study explored the effects of Pb exposure on glucose homeostasis in mice fed a normal diet (ND) and high-fat diet (HFD) from the perspective of gut microbiota. RESULTS Pb exposure had little effect on glucose metabolism in ND mice, but exacerbated hyperglycemia and insulin resistance, and impaired glucose tolerance in HFD mice. Pb exposure impaired intestinal tight junctions and mucin expression in HFD mice, increasing intestinal permeability and inflammation. Moreover, Pb exposure altered the composition and structure of the gut microbiota and decreased short-chain fatty acids (SCFAs) levels in HFD mice. Correlation analysis revealed that the gut microbiota and SCFAs were significantly correlated with the gut barrier and glucose homeostasis. Furthermore, the fecal microbiota transplantation from Pb-exposed HFD mice resulted in glucose homeostasis imbalance, intestinal mucosal structural damage and inflammation in recipient mice. However, Pb did not exacerbate the metabolic toxicity in HFD mice under depleted gut microbiota. CONCLUSION The findings of the present study suggest that Pb induces impairment of glucose metabolism in HFD mice by perturbing the gut microbiota. Our study offers new perspectives on the mechanisms of metabolic toxicity of heavy metals and demonstrates that the gut microbiota may be a target of action for heavy metal exposure. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Nana Wang
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, Suzhou, China
| | - Xue Gao
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, Suzhou, China
| | - Yuan Huo
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, Suzhou, China
| | - Yuting Li
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, Suzhou, China
| | - Fangru Cheng
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, Suzhou, China
| | - Zengli Zhang
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Lu YY, Lu L, Ren HY, Hua W, Zheng N, Huang FY, Wang J, Tian M, Huang Q. The size-dependence and reversibility of polystyrene nanoplastics-induced lipid accumulation in mice: Possible roles of lysosomes. ENVIRONMENT INTERNATIONAL 2024; 185:108532. [PMID: 38422876 DOI: 10.1016/j.envint.2024.108532] [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: 01/03/2024] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
Nanoplastics (NPs) continue to accumulate in global aquatic and terrestrial systems, posing a potential threat to human health through the food chain and/or other pathways. Both in vivo and in vitro studies have confirmed that the liver is one of the main organs targeted for the accumulation of NPs in living organisms. However, whether exposure to NPs induces size-dependent disorders of liver lipid metabolism remains controversial, and the reversibility of NPs-induced hepatotoxicity is largely unknown. In this study, the effects of long-term exposure to environmentally relevant doses of polystyrene nanoplastics (PS-NPs) on lipid accumulation were investigated in terms of autophagy and lysosomal mechanisms. The findings indicated that hepatic lipid accumulation was more pronounced in mice exposed to 100 nm PS-NPs compared to 500 nm PS-NPs. This effect was effectively alleviated after 50 days of self-recovery for 100 nm and 500 nm PS-NPs exposure. Mechanistically, although PS-NPs exposure activated autophagosome formation through ERK (mitogen-activated protein kinase 1)/mTOR (mechanistic target of rapamycin kinase) signaling pathway, the inhibition of Rab7 (RAB7, member RAS oncogene family), CTSB (cathepsin B), and CTSD (cathepsin D) expression impaired lysosomal function, thereby blocking autophagic flux and contributing to hepatic lipid accumulation. After termination of PS-NPs exposure, lysosomal exocytosis was responsible for the clearance of PS-NPs accumulated in lysosomes. Furthermore, impaired lysosomal function and autophagic flux inhibition were effectively alleviated. This might be the main reason for the alleviation of PS-NPs-induced lipid accumulation after recovery. Collectively, we demonstrate for the first time that lysosomes play a dual role in the persistence and reversibility of hepatotoxicity induced by environmental relevant doses of NPs, which provide novel evidence for the prevention and intervention of liver injury associated with nanoplastics exposure.
Collapse
Affiliation(s)
- Yan-Yang Lu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Lu Lu
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hong-Yun Ren
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Weizhen Hua
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Nengxing Zheng
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Fu-Yi Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Jiani Wang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Meiping Tian
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Qingyu Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China.
| |
Collapse
|
6
|
Tinkov AA, Aschner M, Santamaria A, Bogdanov AR, Tizabi Y, Virgolini MB, Zhou JC, Skalny AV. Dissecting the role of cadmium, lead, arsenic, and mercury in non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. ENVIRONMENTAL RESEARCH 2023; 238:117134. [PMID: 37714366 DOI: 10.1016/j.envres.2023.117134] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
The objective of the present study was to review the existing epidemiological and laboratory findings supporting the role of toxic metal exposure in non-alcoholic fatty liver disease (NAFLD). The existing epidemiological studies demonstrate that cadmium (Cd), lead (Pb), arsenic (As), and mercury (Hg) exposure was associated both with an increased risk of NAFLD and altered biochemical markers of liver injury. Laboratory studies demonstrated that metal exposure induces hepatic lipid accumulation resulting from activation of lipogenesis and inhibition of fatty acid β-oxidation due to up-regulation of sterol regulatory element-binding protein 1 (SREBP-1), carbohydrate response element binding protein (ChREBP), peroxisome proliferator-activated receptor γ (PPARγ), and down-regulation of PPARα. Other metabolic pathways involved in this effect may include activation of reactive oxygen species (ROS)/extracellular signal-regulated kinase (ERK) and inhibition of AMP-activated protein kinase (AMPK) signaling. The mechanisms of hepatocyte damage during development of metal-induced hepatic steatosis were shown to involve oxidative stress, endoplasmic reticulum stress, pyroptosis, ferroptosis, and dysregulation of autophagy. Induction of inflammatory response contributing to progression of NAFLD to non-alcoholic steatohepatitis (NASH) upon toxic metal exposure was shown to be mediated by up-regulation of nuclear factor κB (NF-κB) and activation of NRLP3 inflammasome. Moreover, epigenetic effects of the metals, as well as their effect on gut microbiota and gut wall integrity were also shown to mediate their role in NAFLD development. Despite being demonstrated for Cd, Pb, and As, the contribution of these mechanisms into Hg-induced NAFLD is yet to be estimated. Therefore, further studies are required to clarify the intimate mechanisms underlying the relationship between heavy metal and metalloid exposure and NAFLD/NASH to reveal the potential targets for treatment and prevention of metal-induced NAFLD.
Collapse
Affiliation(s)
- Alexey A Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003, Yaroslavl, Russia; Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, 10461, NY, USA
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Alfred R Bogdanov
- Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Russian State Social University, 129226, Moscow, Russia; Municipal State Hospital No. 13 of the Moscow City Health Department, 115280, Moscow, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, 20059, USA
| | - Miriam B Virgolini
- Departamento de Farmacología Otto Orsingher, Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Ji-Chang Zhou
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China
| | - Anatoly V Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003, Yaroslavl, Russia; Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia
| |
Collapse
|
7
|
Chen J, Zheng D, Cai Z, Zhong B, Zhang H, Pan Z, Ling X, Han Y, Meng J, Li H, Chen X, Zhang H, Liu L. Increased DNMT1 Involvement in the Activation of LO2 Cell Death Induced by Silver Nanoparticles via Promoting TFEB-Dependent Autophagy. TOXICS 2023; 11:751. [PMID: 37755761 PMCID: PMC10537645 DOI: 10.3390/toxics11090751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023]
Abstract
The accumulation of exogenous silver nanoparticles (AgNPs) will terminally bring about liver injury, including cell death, where DNA methylation tends to be a crucial epigenetic modulator. The change in the cell autophagy level verified to be closely associated with hepatocyte death has been followed with wide interest. But the molecular toxicological mechanisms of AgNPs in relation to DNA methylation, autophagy, and cell death remain inconclusive. To address the issue above, in LO2 cells treated with increasing concentrations of AgNPs (0, 5, 10, and 20 μg/mL), a cell cytotoxicity assay was performed to analyze the level of cell death, which also helped to choose an optimal concentration for next experiments. An immunofluorescence assay was used to determine the autophagic flux as well as TFEB translocation, with qRT-PCR and western blot being used to analyze the expression level of autophagy-related genes and proteins. According to our findings, in the determination of cell viability, 20 μg/mL (AgNPs) was adopted as the best working concentration. LO2 cell death, autophagy, and TFEB nuclear translocation were induced by AgNPs, which could be inhibited by lysosome inhibitor chloroquine (CQ) or siRNA specific for TFEB. Moreover, AgNP exposure led to DNA hypermethylation, with DNMT1 taking part mainly, which could be obviously prevented by 5-Aza-2'-deoxycytidine (5-AzaC) or trichostatin A (TSA) treatment or DNMT1 knockout in LO2 cells. Our studies suggest that through TFEB-dependent cell autophagy, increased DNMT1 may facilitate cell death induced by AgNPs.
Collapse
Affiliation(s)
- Jialong Chen
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China;
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Dongyan Zheng
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Ziwei Cai
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Bohuan Zhong
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Haiqiao Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Zhijie Pan
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Xiaoxuan Ling
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Yali Han
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Jinxue Meng
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Huifang Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Xiaobing Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - He Zhang
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China;
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| | - Linhua Liu
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China;
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (D.Z.); (Z.C.); (B.Z.); (H.Z.); (Z.P.); (X.L.); (Y.H.); (J.M.); (H.L.); (X.C.)
| |
Collapse
|
8
|
Li Q, Feng Y, Wang R, Liu R, Ba Y, Huang H. Recent insights into autophagy and metals/nanoparticles exposure. Toxicol Res 2023; 39:355-372. [PMID: 37398566 PMCID: PMC10313637 DOI: 10.1007/s43188-023-00184-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 03/08/2023] [Accepted: 04/04/2023] [Indexed: 07/04/2023] Open
Abstract
Some anthropogenic pollutants, such as heavy metals and nanoparticles (NPs), are widely distributed and a major threat to environmental safety and public health. In particular, lead (Pb), cadmium (Cd), chromium (Cr), arsenic (As), and mercury (Hg) have systemic toxicity even at extremely low concentrations, so they are listed as priority metals in relation to their significant public health burden. Aluminum (Al) is also toxic to multiple organs and is linked to Alzheimer's disease. As the utilization of many metal nanoparticles (MNPs) gradually gain traction in industrial and medical applications, they are increasingly being investigated to address potential toxicity by impairing certain biological barriers. The dominant toxic mechanism of these metals and MNPs is the induction of oxidative stress, which subsequently triggers lipid peroxidation, protein modification, and DNA damage. Notably, a growing body of research has revealed the linkage between dysregulated autophagy and some diseases, including neurodegenerative diseases and cancers. Among them, some metals or metal mixtures can act as environmental stimuli and disturb basal autophagic activity, which has an underlying adverse health effect. Some studies also revealed that specific autophagy inhibitors or activators could modify the abnormal autophagic flux attributed to continuous exposure to metals. In this review, we have gathered recent data about the contribution of the autophagy/mitophagy mediated toxic effects and focused on the involvement of some key regulatory factors of autophagic signaling during exposure to selected metals, metal mixtures, as well as MNPs in the real world. Besides this, we summarized the potential significance of interactions between autophagy and excessive reactive oxygen species (ROS)-mediated oxidative damage in the regulation of cell survival response to metals/NPs. A critical view is given on the application of autophagy activators/inhibitors to modulate the systematic toxicity of various metals/MNPs.
Collapse
Affiliation(s)
- Qiong Li
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Yajing Feng
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Ruike Wang
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Rundong Liu
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Yue Ba
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Hui Huang
- Department of Environmental Health and Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| |
Collapse
|
9
|
Zhao YS, Li JY, Li ZC, Wang LL, Gan CL, Chen J, Jiang SY, Aschner M, Ou SY, Jiang YM. Sodium Para-aminosalicylic Acid Inhibits Lead-Induced Neuroinflammation in Brain Cortex of Rats by Modulating SIRT1/HMGB1/NF-κB Pathway. Neurochem Res 2023; 48:238-249. [PMID: 36063295 PMCID: PMC9825627 DOI: 10.1007/s11064-022-03739-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 01/11/2023]
Abstract
Lead (Pb) is considered to be a major environmental pollutant and occupational health hazard worldwide which may lead to neuroinflammation. However, an effective treatment for Pb-induced neuroinflammation remains elusive. The aim of this study was to investigate the mechanisms of Pb-induced neuroinflammation, and the therapeutic effect of sodium para-aminosalicylic acid (PAS-Na, a non-steroidal anti-inflammatory drug) in rat cerebral cortex. The results indicated that Pb exposure induced pathological damage in cerebral cortex, accompanied by increased levels of inflammatory factors tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β). Moreover, Pb decreased the expression of silencing information regulator 2 related enzyme 1 (SIRT1) and brain-derived neurotrophic factor (BDNF), and increased the levels of high mobile group box 1 (HMGB1) expression and p65 nuclear factor-κB (NF-κB) phosphorylation. PAS-Na treatment ameliorated Pb-induced histopathological changes in rat cerebral cortex. Moreover, PAS-Na reduced the Pb-induced increase of TNF-α and IL-1β levels concomitant with a significant increase in SIRT1 and BDNF levels, and a decrease in HMGB1 and the phosphorylation of p65 NF-κB expression. Thus, PAS-Na may exert anti-inflammatory effects by mediating the SIRT1/HMGB1/NF-κB pathway and BDNF expression. In conclusion, in this novel study PAS-Na was shown to possess an anti-inflammatory effect on cortical neuroinflammation, establishing its efficacy as a potential treatment for Pb exposures.
Collapse
Affiliation(s)
- Yue-Song Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Jun-Yan Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Hengyang Center for Disease Control and Prevention, Hengyang, China
| | - Zhao-Cong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lei-Lei Wang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Cui-Liu Gan
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Jing Chen
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Si-Yang Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Shi-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
10
|
Miao Z, Miao Z, Teng X, Xu S. Melatonin alleviates lead-induced intestinal epithelial cell pyroptosis in the common carps (Cyprinus carpio) via miR-17-5p/TXNIP axis. FISH & SHELLFISH IMMUNOLOGY 2022; 131:127-136. [PMID: 36202203 DOI: 10.1016/j.fsi.2022.09.071] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
Lead (Pb) has been concerned as one of the most severe hazardous contaminants, because it can cause pyroptosis in multiple tissues of mammals and birds. Melatonin (Mel) has attracted much interest for its role in governing intestinal injury via microRNAs (miRNAs). To explore the effect of Mel on Pb exposure-induced intestinal epithelial cell pyroptosis in common carps by regulating miR-17-5p/TXNIP axis, the Pb exposure and Pb-Mel treated models were constructed in vivo. The results elucidated that the suppressed expression of miR-17-5p and intensified level of TXNIP were primarily detected in Pb-exposed gut tissues, and both abolished with Mel addition, along with downregulated Pb-mediated elevated expression of NLRP3, CASP1, IL1β and GSDMD. Additionally, the targeting relationship between miR-17-5p and TXNIP were demonstrated by dual-luciferase reporter assay, and on this basis, miR-17-5p NC, mimic and inhibitor cell models were established. Thereby, Thereby, the expression of TXNIP in the miR-17-5p mimic groups was significant lower in the Pb-exposure but still elevated than the Control group, and the expression of NLRP3 and NLRP3-dependent pyrotposis-related genes performed consistent alterations. Noticeably, the expression of TXNIP suppressed with Mel addition even in the miR-17-5p inhibitor cell model, resulting in the inactivation of NLRP3 inflammasome-dependent pyroptosis. Overall, we draw the conclusion as Mel attenuates Pb-induced intestinal epithelial cell pyroptosis via miR-17-5p/TXNIP axis. The present study provides a novel perspective for toxicological mechanism of Pb, and new insights for the detoxification mechanism of Mel.
Collapse
Affiliation(s)
- Zhiying Miao
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Zhiruo Miao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Shiwen Xu
- 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, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
11
|
Guan S, Zhang R, Zhao Y, Meng Z, Lu J. 1,3-Dichloro-2-propanol induced ferroptosis through Nrf2/ARE signaling pathway in hepatocytes. ENVIRONMENTAL TOXICOLOGY 2022; 37:2515-2528. [PMID: 35870111 DOI: 10.1002/tox.23615] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/16/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
1,3-Dichloro-2-propanol (1,3-DCP) is a representative chloropropane environmental contaminant with multiple toxicities. Ferroptosis is a novel iron-dependent form of regulated cell death that is closely associated with the accumulation of lipid peroxides, Fe2+ and reactive oxygen species (ROS). In this study, we found that 1,3-DCP could induce mouse liver injury via ferroptosis. Administrating of C57BL/6J mice with 12.5, 25, and 50 mg/kg 1,3-DCP for 4 weeks via oral gavage, the data showed that 1,3-DCP exposure led to the pathological changes in mouse livers, remarkably induced accumulation of malondialdehyde (MDA) and Iron, reduction of glutathione (GSH), and changed in the expression of ferroptosis marker proteins glutathione peroxidase 4 (GPX4) and acyl-CoA synthetase-4 (ACSL4). Then, we also proved the results with HepG2 cells in vitro. The data showed that treatment 1,3-DCP significantly triggered the ferroptosis in vitro. Furthermore, we found that the ferroptosis-related signal pathways were significantly activated in mice livers and HepG2 cells in response to 1,3-DCP exposure. The data showed that 1,3-DCP induced ferroptosis by inhibiting nuclear factor erythroid 2-related factor 2 (Nrf2) translocation into nuclear and thereby suppressing the expression of its downstream target proteins including GPX4, ferritin heavy chain (FTH), ferroportin (FPN), cystine/glutamate transporter xCT (SLC7A11), and heme oxygenase 1 (HO-1). Taken together, our findings confirmed that 1,3-DCP induced ferroptosis via the Nrf2/ARE signaling pathway in hepatocytes. Our works provide new toxicity mechanisms of 1,3-DCP with ferroptosis on hepatocytes injury.
Collapse
Affiliation(s)
- Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| | - Ranran Zhang
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Yanan Zhao
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Zhuoqun Meng
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, People's Republic of China
| |
Collapse
|