1
|
Wei X, Ge Y, Zheng Y, Zhao S, Zhou Y, Chang Y, Wang N, Wang X, Zhang J, Zhang X, Hu L, Tan Y, Jia Q. Hybrid EMT Phenotype and Cell Membrane Tension Promote Colorectal Cancer Resistance to Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413882. [PMID: 39985376 PMCID: PMC12005738 DOI: 10.1002/advs.202413882] [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: 10/29/2024] [Revised: 01/26/2025] [Indexed: 02/24/2025]
Abstract
Intratumoral heterogeneity, including epithelial-mesenchymal transition (EMT), is one major cause of therapeutic resistance. The induction of ferroptosis, an iron-dependent death, has the potential in overcoming this resistance to traditional treatment modalities. However, the roles of distinct EMT phenotypes in ferroptosis remain an enigma. This study reports that 3D soft fibrin microenvironment confers colorectal cancer (CRC) cells hybrid EMT phenotype and high level of resistance to ferroptosis. The activation of histone acetylation and WNT/β-catenin signaling drives this EMT phenotypic transition, which promotes the defense of 3D CRCs against ferroptosis via glutathione peroxidases/ferritin signaling axis. Unexpectedly, E-cadherin knockout in 3D but not 2D CRCs mediates an integrin β3 marked-late hybrid EMT state and further enhances the resistance to ferroptosis via integrin-mediated tension and mitochondrial reprogramming. The inhibition of integrin αvβ3-mediated tension and WNT/β-catenin-mediated hybrid EMT sensitizes 3D CRCs with and without E-cadherin deficiency to ferroptosis in vivo, respectively. Further, the EMT phenotype of patient-derived tumoroids is associated with CRC therapeutic resistance. In summary, this study uncovers previously unappreciated roles of hybrid EMT and cell membrane tension in ferroptosis, which not only predict the treatment efficacy but also potentiate the development of new ferroptosis-based targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xiaowei Wei
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yutong Ge
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Yaolin Zheng
- Department of RespiratoryCritical Care and Sleep MedicineXiang'an Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamen361102China
| | - Sunyan Zhao
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yuhan Zhou
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yuhan Chang
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Nuofan Wang
- School of MedicineSoutheast UniversityNanjing210009China
| | - Xiumei Wang
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Juan Zhang
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Xuanchang Zhang
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Liqiao Hu
- Guangzhou National LaboratoryGuangzhou510005China
| | - Youhua Tan
- The Hong Kong Polytechnic UniversityShenzhen Research InstituteShenzhen518000China
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Qiong Jia
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjing210006China
| |
Collapse
|
2
|
Yan X, Chen X, Zhang X, Qureshi A, Wang Y, Tang X, Hu T, Zhuang H, Ran X, Ma G, Luo P, Shen L. Proteomic analysis of the effects of Dictyophora polysaccharide on arsenic-induced hepatotoxicity in rats. Exp Mol Pathol 2024; 138:104910. [PMID: 38876078 DOI: 10.1016/j.yexmp.2024.104910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
Arsenic (As) is a highly toxic environmental toxicant and a known human carcinogen. Long-term exposure to As can cause liver injury. Dictyophora polysaccharide (DIP) is a biologically active natural compound found in the Dictyophora with excellent antioxidation, anti-inflammation, and immune protection properties. In this study, the Sprague-Dawley (SD) rat model of As toxicity was established using a feeding method, followed by DIP treatment in rats with As-induced liver injury. The molecular mechanisms of As toxicity to the rat liver and the protective effect of DIP were investigated by proteomic studies. The results showed that 172, 328 and 191 differentially expressed proteins (DEPs) were identified between the As-exposed rats versus control rats (As/Ctrl), DIP treated rats versus As-exposed rats (DIP+As/As), and DIP treated rats versus control rats (DIP+As /Ctrl), respectively. Among them, the expression of 90 DEPs in the As/Ctrl groups was reversed by DIP treatment. As exposure caused dysregulation of metabolic pathways, mitochondria, oxidative stress, and apoptosis-related proteins in the rat liver. However, DIP treatment changed or restored the levels of these proteins, which attenuated the damage to the livers of rats caused by As exposure. The results provide new insights into the mechanisms of liver injury induced by As exposure and the treatment of DIP in As poisoning.
Collapse
Affiliation(s)
- Xi Yan
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Xiaolu Chen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Xinglai Zhang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Ayesha Qureshi
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Yi Wang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Ting Hu
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Xiaoqian Ran
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Guanwei Ma
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China
| | - Peng Luo
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China.
| | - Liming Shen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, PR China; College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
3
|
Lv Y, Wang H, Zheng D, Shi M, Bi D, Hu Q, Zhi H, Lou D, Li J, Wei S, Hu Y. Environmental arsenic pollution induced liver oxidative stress injury by regulating miR-155 through inhibition of AUF1. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 922:171237. [PMID: 38423337 DOI: 10.1016/j.scitotenv.2024.171237] [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: 11/24/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
Arsenic (As), a common environmental pollutant, has become a hot topic in recent years due to its potentially harmful effects. Liver damage being a central clinical feature of chronic arsenic poisoning. However, the underlying mechanisms remain unclear. We demonstrated that arsenic can lead to oxidative stress in the liver and result in structural and functional liver damage, significantly correlated with the expression of AUF1, Dicer1, and miR-155 in the liver. Interestingly, knockdown AUF1 promoted the up-regulatory effects of arsenic on Dicer1 and miR-155 and the inhibitory effects on SOD1, which exacerbated oxidative damage in rat liver. However, overexpression of AUF1 reversed the up-regulatory effects of arsenic on Dicer1 and miR-155, restored arsenic-induced SOD1 depletion, and attenuated liver oxidative stress injury. Further, we verified the mechanism and targets of miR-155 in regulating SOD1 by knockdown/overexpression of miR-155 and nonsense mutant SOD1 3'UTR experiments. In conclusion, these results powerfully demonstrate that arsenic inhibits AUF1 protein expression, which in turn reduces the inhibitory effect on Dicer1 expression, which promotes miR-155 to act on the SOD1 3'UTR region after high expression, thus inhibiting SOD1 protein expression and enzyme activity, and inducing liver injury. This finding provides a new perspective for the mechanism research and targeted prevention of arsenic poisoning, as well as scientific evidence for formulating strategies to prevent and control environmental arsenic pollution.
Collapse
Affiliation(s)
- Ying Lv
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Hongling Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Dan Zheng
- Guiyang Maternity and Child Health Hospital, Guiyang 550003, Guizhou, China
| | - Mingyang Shi
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Dingnian Bi
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Qian Hu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Haiyan Zhi
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Didong Lou
- Department of Forensic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China; Key Laboratory of Traditional Chinese Medicine Toxicology in Forensic Medicine, Guizhou Education Department, Guiyang 550025, Guizhou, China
| | - Jun Li
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Shaofeng Wei
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Yong Hu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China.
| |
Collapse
|
4
|
Zhang XY, Li Y. PHD-BAH Domain in ASH1L Could Recognize H3K4 Methylation and Regulate the Malignant Behavior of Cholangiocarcinoma. Anticancer Agents Med Chem 2024; 24:1264-1274. [PMID: 39034728 DOI: 10.2174/0118715206312004240712072532] [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: 04/03/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Histone methyltransferase absent, small, or homeotic discs1-like (ASH1L) is composed of su(var)3-9, enhancer of zeste, trithorax (SET) domain, pleckstrin homology domain (PHD) domain, middle (MID) domain, and bromo adjacent homology (BAH) domain. The SET domain of ASH1L is known to mediate mediate H3K36 dimethylation (H3K36me2) modification. However, the specific functions of the PHD-BAH domain remain largely unexplored. This study aimed to explore the biological function of the PHD-BAH domain in ASH1L. METHODS We employed a range of techniques, including a prokaryotic fusion protein expression purification system, pull-down assay, Isothermal Titration Calorimetry (ITC), polymerase chain reaction (PCR), and sitedirected mutagenesis, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR-Cas9) gene editing, cell culture experiment, western blot, cell proliferation assay, and cell apoptosis test. RESULTS The PHD-BAH domain in ASH1L preferentially binds to the H3K4me2 peptide over H3K4 monomethylation (H3K4me1) and H3K4 trimethylation (H3K4me3) peptide. Notably, the W2603A mutation within the PHD-BAH domain could disrupt the interaction with H3K4me2 in vitro. Compared with wild-type Cholangiocarcinoma (CHOL) cells, deletion of the PHD-BAH domain in ASH1L led to increased CHOL cell apoptosis and reduced cell proliferation (P < 0.001). Additionally, the W2603A mutation affected the regulation of the proteasome 20S subunit beta (PSMB) family gene set. CONCLUSION W2603A mutation was crucial for the interaction between the PHD-BAH domain and the H3K4me2 peptide. ASH1L regulated CHOL cell survival and proliferation through its PHD-BAH domain by modulating the expression of the PSMB family gene set.
Collapse
Affiliation(s)
- Xiang-Yu Zhang
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yue Li
- External Cooperation Liaison Office, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, China
| |
Collapse
|
5
|
Lv Y, Wang Y, Yao J, He J, Lin C, Bai G, Tu C. The role of FGF9-mediated TGF-β1/Smad signaling in enamel hypoplasia induced by exposure to fluoride and SO 2 in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115243. [PMID: 37454483 DOI: 10.1016/j.ecoenv.2023.115243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Many geographical areas of the world are polluted by both fluoride and sulfur dioxide (SO2). However, the effects of simultaneous exposure to fluoride and SO2 on teeth are unknown. Fibroblast growth factor-9 (FGF9) and transforming growth factor-β1 (TGF-β1) are key signaling molecules in enamel development. The purpose of the study was to explore the effects of co-exposure to fluoride and sulfur dioxide on enamel and to investigate the role and mechanism of FGF9 and TGF-β1. First, sodium fluoride (NaF) and SO2 derivatives were used to construct rat models and evaluate the enamel development of rats. Then, TGF-β1 (cytokine) treatment, SIS3 (inhibitor) treatment and FGF9 gene knockdown were used to explore the mechanism of enamel damage in vitro. The results showed that enamel column crystals in the exposed group were characterized by enamel hypoplasia, as indicated by alterations such as disarrangement of enamel column crystals, space widening and breakage. Ameloblasts also showed pathological changes such as ribosome loss, mitochondrial swelling, nuclear fragmentation and chromatin aggregation. The protein expression of FGF9 was higher and the protein expression of AMBN, TGF-β1 and p-Smad2/3 protein was lower in the groups treated with fluoride and SO2 individually or in combination compared with the control group. Further studies showed that TGF-β1 significantly upregulated p-Smad2/3 and AMBN protein expression and reduced the inhibitory effects of fluoride and SO2; furthermore, SISI blocked the effect of TGF-β1. In addition, knockdown of FGF9 upregulated TGF-β1 protein expression, further activated Smad2/3 phosphorylation, eliminated the inhibitory effects of fluoride and SO2, and increased the protein expression of AMBN. In brief, the study confirms that co-exposure to fluoride and SO2 can result in enamel hypoplasia in rats and indicates that the underlying mechanism may be closely related to the effect of FGF9 on enamel matrix protein secretion through inhibition of the TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Ying Lv
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Yang Wang
- Infrastructure Construction Department, Guizhou Medical University, Guiyang 550025, China
| | - Jin Yao
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Jiaojiao He
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Changhu Lin
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Guohui Bai
- Key Laboratory of Oral Disease Research, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Chenglong Tu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
6
|
Müller FA, Stamou M, Englert FH, Frenzel O, Diedrich S, Suter-Dick L, Wambaugh JF, Sturla SJ. In vitro to in vivo extrapolation and high-content imaging for simultaneous characterization of chemically induced liver steatosis and markers of hepatotoxicity. Arch Toxicol 2023; 97:1701-1721. [PMID: 37046073 PMCID: PMC10182956 DOI: 10.1007/s00204-023-03490-8] [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: 01/21/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023]
Abstract
Chemically induced steatosis is characterized by lipid accumulation associated with mitochondrial dysfunction, oxidative stress and nucleus distortion. New approach methods integrating in vitro and in silico models are needed to identify chemicals that may induce these cellular events as potential risk factors for steatosis and associated hepatotoxicity. In this study we used high-content imaging for the simultaneous quantification of four cellular markers as sentinels for hepatotoxicity and steatosis in chemically exposed human liver cells in vitro. Furthermore, we evaluated the results with a computational model for the extrapolation of human oral equivalent doses (OED). First, we tested 16 reference chemicals with known capacities to induce cellular alterations in nuclear morphology, lipid accumulation, mitochondrial membrane potential and oxidative stress. Then, using physiologically based pharmacokinetic modeling and reverse dosimetry, OEDs were extrapolated from data of any stimulated individual sentinel response. The extrapolated OEDs were confirmed to be within biologically relevant exposure ranges for the reference chemicals. Next, we tested 14 chemicals found in food, selected from thousands of putative chemicals on the basis of structure-based prediction for nuclear receptor activation. Amongst these, orotic acid had an extrapolated OED overlapping with realistic exposure ranges. Thus, we were able to characterize known steatosis-inducing chemicals as well as data-scarce food-related chemicals, amongst which we confirmed orotic acid to induce hepatotoxicity. This strategy addresses needs of next generation risk assessment and can be used as a first chemical prioritization hazard screening step in a tiered approach to identify chemical risk factors for steatosis and hepatotoxicity-associated events.
Collapse
Affiliation(s)
- Fabrice A Müller
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Marianna Stamou
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Felix H Englert
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Ole Frenzel
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Sabine Diedrich
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Laura Suter-Dick
- School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, 4132, Muttenz, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), 4001, Basel, Switzerland
| | - John F Wambaugh
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, Durham, NC, 27711, USA
| | - Shana J Sturla
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland.
| |
Collapse
|
7
|
Bi D, Zheng D, Shi M, Hu Q, Wang H, Zhi H, Lou D, Zhang A, Hu Y. Role of SESTRIN2/AMPK/ULK1 pathway activation and lysosomes dysfunction in NaAsO 2-induced liver injury under oxidative stress. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 254:114751. [PMID: 36907090 DOI: 10.1016/j.ecoenv.2023.114751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/10/2023] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
Arsenic, a serious environmental poison to human health, is widely distributed in nature. As the main organ of arsenic metabolism, liver is easily damaged. In the present study, we found that arsenic exposure can cause liver injury in vivo and in vitro, to date the underlying mechanism of which is yet unclear. Autophagy is a process that depends on lysosomes to degrade damaged proteins and organelles. Here, we reported that oxidative stress can be induced and then activated the SESTRIN2/AMPK/ULK1 pathway, damaged lysosomes, and finally induced necrosis upon arsenic exposure in rats and primary hepatocytes, which was characterized by lipidation of LC3II, the accumulation of P62 and the activation of RIPK1 and RIPK3. Similarly, lysosomes function and autophagy can be damaged under arsenic exposure, which can be alleviated after NAC treatment and aggravated by Leupeptin treatment in primary hepatocytes. Moreover, we also found that the transcription and protein expressions of necrotic-related indicators RIPK1 and RIPK3 in primary hepatocytes were decreased after P62 siRNA. Taken together, the results revealed that arsenic can induce oxidative stress, activate SESTRIN2/AMPK/ULK1 pathway to damage lysosomes and autophagy, and eventually induce necrosis to damage liver.
Collapse
Affiliation(s)
- Dingnian Bi
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Dan Zheng
- Guiyang Maternity and Child Health Hospital, Guizhou, PR China
| | - Mingyang Shi
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Qian Hu
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Hongling Wang
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Haiyan Zhi
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Didong Lou
- Department of Forensic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, PR China; Key Laboratory of Traditional Chinese Medicine Toxicology in Forensic Medicine, Guizhou Education Department, Guiyang 550025, Guizhou, PR China
| | - Aihua Zhang
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Yong Hu
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| |
Collapse
|
8
|
Bi D, Shi M, Hu Q, Wang H, Lou D, Zhang A, Hu Y. LC/MS/MS-Based Liver Metabolomics to Identify Chronic Liver Injury Biomarkers Following Exposure to Arsenic in Rats. Biol Trace Elem Res 2022; 200:4355-4369. [PMID: 34981423 DOI: 10.1007/s12011-021-03026-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022]
Abstract
Arsenic is a widespread natural metalloid element. Long-term chronic exposure to arsenic can lead to different degrees of liver injury. Although the etiology of this disease is well known, to date, the underlying mechanism of arsenic-induced liver injury remains unclear, and no specific treatment exists because of the complexity of arsenic. In the present study, potential biomarkers and metabolic pathways in the livers of Wistar rats treated with arsenic for 24 weeks were investigated using an integrated metabolic approach with an LC-Orbitrap Q Exactive™ HF-X mass spectrometer. Markedly increased liver levels of arsenic, alanine aminotransferase (ALT), alkaline phosphatase (ALP), and total bile acid (TBA) were detected in the arsenic treatment groups (P < 0.05). Furthermore, histopathological examination of liver tissues showed obviously swollen, loose cytoplasm and increased necrosis in the arsenic treatment groups compared with those in the control group (P < 0.05). Metabonomics results showed that 109 metabolites (variable importance in the projection (VIP) > 1; fold change > 2 or < 0.5; P adjusted < 0.05) changed significantly after exposure to arsenic and included 71 upregulated metabolites and 38 downregulated metabolites. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that 6 metabolic pathways with statistical significance-phenylalanine metabolism, pyruvate metabolism, glycolysis/gluconeogenesis, citrate cycle (TCA cycle), thiamine metabolism, and vitamin B6 metabolism-were selected, and 13 differential metabolites were detected to be involved in regulating these metabolic pathways. The present study could help identify potential biomarkers and their functions, as well as metabolic pathways, likely providing evidence for the early diagnosis, prevention, and mechanistic study of arsenism.
Collapse
Affiliation(s)
- Dingnian Bi
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Mingyang Shi
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Qian Hu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Hongling Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Didong Lou
- Department of Forensic Medicine, Guizhou University of Traditional Chinese Medicine, Guizhou, 550025, China
- Key Laboratory of Traditional Chinese Medicine Toxicology in Forensic Medicine, Guizhou Education Department, Guizhou, 550025, China
| | - Aihua Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Yong Hu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
9
|
Liu J, Mi J, Liu S, Chen H, Jiang L. PSMB5 overexpression is correlated with tumor proliferation and poor prognosis in hepatocellular carcinoma. FEBS Open Bio 2022; 12:2025-2041. [PMID: 36062301 PMCID: PMC9623531 DOI: 10.1002/2211-5463.13479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/14/2022] [Accepted: 08/11/2022] [Indexed: 01/25/2023] Open
Abstract
Aberrant expression of members of the proteasome subunit beta (PSMB) family (including PSMB2, PSMB4, PSMB7 and PSMB8) has been reported in hepatocellular carcinoma (HCC). However the role of PSMB5 in HCC is unclear. To address this issue, we examined the expression of PSMB5 in HCC tissues using the The Cancer Genome Atlas, International Cancer Genome Consortium and Gene Expression Omnibus databases. A quantitative real-time PCR and immunohistochemistry were performed to validate the expression of PSMB5 in HCC. The survival mutation status and immune cell infiltration of PSMB5 were also evaluated in HCC. We then examined the effect of knocking down PSMB5 expression through RNA interference in the HCC cell line Huh7. High expression of PSMB5 was observed in HCC tissues and was associated with poor prognosis. PSMB5 expression and clinical characteristics were then incorporated to build a prognostic nomogram. We observed that PSMB5 expression was closely related to the abundance of B cells, CD4+ T cells, CD8+ T cells, dendritic cell macrophages and neutrophils. Moreover silencing of PSMB5 in Huh7 significantly suppressed cell proliferation and migration at the same time as increasing apoptosis. Inhibition of the phosphatidylinositol-3-kinase/Akt/mechanistic target of rapamycin pathway was observed after PSMB5 downregulation in Huh7 cells. Our findings suggest that PSMB5 may promote the proliferation of HCC cells by inactivating the phosphatidylinositol-3-kinase/Akt/mechanistic target of rapamycin signaling pathway and thus PSMB5 may have potential as a biomarker for diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Jun Liu
- Guangxi Medical UniversityNanningChina
| | - Jinglin Mi
- Department of Radiation OncologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | | | | | - Li Jiang
- Department of Radiation OncologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
10
|
Bi D, Shi M, Zheng D, Hu Q, Wang H, Peng L, Lou D, Zhang A, Hu Y. Mechanism underlying the targeted regulation of the SOD1 3'UTR by the AUF1/Dicer1/miR-155/SOD1 pathway in sodium arsenite-induced liver injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:113990. [PMID: 35998476 DOI: 10.1016/j.ecoenv.2022.113990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 06/13/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Arsenic (As) is a natural hepatotoxicity inducer that is ubiquitous in water, soil, coal, and food. Studies have found that arsenite exposure elicits increased mRNA transcription and decreased protein expression of SOD1 in vivo and in vitro; however, the specific mechanisms remain unclear. Here, we established a model of arsenic-induced chronic liver injury by providing rats with drinking water containing different concentrations of sodium arsenite (NaAsO2) and found that NaAsO2 exposure decreased the mRNA and protein levels of AUF1 and the protein level of SOD1 and elevated the mRNA and protein levels of Dicer1 and miR-155 and the mRNA level of SOD1. Overexpression of AUF1 under NaAsO2 stress in vitro induced Dicer1 mRNA and protein expression and decreased miR-155 levels, which could be reversed by AUF1 siRNA. In addition, miR-155 overexpression downregulated SOD1 mRNA and protein levels, although this change was inhibited after transfection with an miR-155 inhibitor. Taken together, our findings showed that NaAsO2 could upregulate Dicer1 mRNA and protein, thereby increasing miR-155 expression by downregulating AUF1 mRNA and protein expression. A dual-luciferase reporter assay indicated that miR-155 decreased the mRNA and protein levels of SOD1 by targeting the SOD1 3'UTR, resulting in liver injury. This study provides an important research basis for further understanding the factors underlying arsenic-induced liver injury to improve the prevention and control strategies for arsenism.
Collapse
Affiliation(s)
- Dingnian Bi
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Mingyang Shi
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Dan Zheng
- Guiyang Maternity and Child Health Hospital, Guizhou, PR China
| | - Qian Hu
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Hongling Wang
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Liuyu Peng
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Didong Lou
- Department of Forensic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, PR China; Key Laboratory of Traditional Chinese Medicine Toxicology in Forensic Medicine, Guizhou Education Department, Guiyang 550025, Guizhou, PR China
| | - Aihua Zhang
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Yong Hu
- Key Laboratory of Enviromental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| |
Collapse
|
11
|
PSMB5 Alleviates Ulcerative Colitis by Inhibiting ROS-Dependent NLRP3 Inflammasome-Mediated Pyroptosis. DISEASE MARKERS 2022; 2022:2329904. [PMID: 36061354 PMCID: PMC9439905 DOI: 10.1155/2022/2329904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 12/04/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease. Intestinal mucosal injury is a significant factor in UC. Pyroptosis is a kind of programmed cell death induced by inflammatory caspases. Proteasome 20S subunit beta 5 (PSMB5) promotes cell viability. The purpose of this study was to determine the impact of PSMB5 on intestinal mucosal injury and to elucidate the underlying processes in dextran sulfate sodium- (DSS-) induced UC mice. Kunming (KM) mice received 3% DSS for 5 days to induce UC. We collected clinical symptoms, body weight, colon length, and histological changes. MDA (malondialdehyde) and SOD (superoxide dismutase) levels were determined using an ELISA assay. RT-PCR was used to assess the expression of IL-1β and IL-18. PSMB5 demonstrated a significant effect against UC by increasing body weight and colon length and decreasing DAI (disease activity index), colon macroscopic damage index (CMDI), histological injury scores, and reactive oxygen species (ROS), MDA, and SOD levels, thereby alleviating histopathological changes and inhibiting oxidative stress. HIEC-6 cells were exposed to lipopolysaccharide (LPS) condition with or without PSMB5, along with caspase-1 inhibitor (Z-VAD-FMK), NLRP3 inhibitor (MCC950), and ROS scavenger N-acetylcysteine (NAC). The viability of the cells, the release of lactate dehydrogenase (LDH), and intracellular ROS generation were determined using assay kits. Western blot analysis was used to determine the levels of NLRP3, ASC, cleaved caspase-1 (p20), pro-IL-1β, IL-1β, pro-IL-18, and IL-18. PSMB5 overexpression enhanced the inflammatory damage in LPS-treated HIEC-6 cells by activating the NLRP3 inflammasome and mediating pyroptosis, as demonstrated by increased LDH release and lower cell viability, as well as increased expression of NLRP3, ASC, cleaved caspase-1 (p20), IL-1, and IL-18. Meanwhile, NAC protected HIEC-6 cells from LPS-induced damage by reversing the activation of the NLRP3 inflammasome-mediated pyroptosis. In conclusion, PSMB5 may lower HIEC-6 cell susceptibility to LPS and ameliorate UC-induced HIEC-6 cell damage by decreasing ROS generation and hence inhibiting NLRP3-mediated pyroptosis.
Collapse
|
12
|
Hu T, Shen L, Huang Q, Wu C, Zhang H, Zeng Q, Wang G, Wei S, Zhang S, Zhang J, Khan NU, Shen X, Luo P. Protective Effect of Dictyophora Polysaccharides on Sodium Arsenite-Induced Hepatotoxicity: A Proteomics Study. Front Pharmacol 2021; 12:749035. [PMID: 34899304 PMCID: PMC8660860 DOI: 10.3389/fphar.2021.749035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022] Open
Abstract
The purpose of this study is to understand the mechanism of sodium arsenite (NaAsO2)-induced apoptosis of L-02 human hepatic cells, and how Dictyophora polysaccharide (DIP) protects L-02 cells from arsenic-induced apoptosis. The results revealed that DIP pretreatment inhibited NaAsO2 induced L-02 cells apoptosis by increasing anti-apoptotic Bcl-2 expression and decreasing pro-apoptotic Bax expression. Proteomic analysis showed that arsenic treatment disrupted the expression of metabolism and apoptosis associated proteins, including ribosomal proteins (RPs). After pretreatment with DIP, the expression levels of these proteins were reversed or restored. For the first time, it was observed that the significant decrease of cytoplasmic RPs and the increase of mitochondrial RPs were related to human normal cell apoptosis induced by arsenic. This is also the first report that the protective effect of DIP on cells was related to RPs. The results highlight the relationship between RPs and apoptosis, as well as the relationship between RPs and DIP attenuating arsenic-induced apoptosis.
Collapse
Affiliation(s)
- Ting Hu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guiyang, China
| | - Liming Shen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Qun Huang
- School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guiyang, China
| | - Changyan Wu
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Huajie Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Qibing Zeng
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guiyang, China
| | - Guoze Wang
- School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guiyang, China
| | - Shaofeng Wei
- School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guiyang, China
| | - Shuling Zhang
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Jun Zhang
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Naseer Ullah Khan
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiangchun Shen
- Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Peng Luo
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| |
Collapse
|