1.Arsenic trioxide preconditioning attenuates hepatic ischemia- reperfusion injury in mice: Role of ERK/AKT and autophagy.
Chaoqun WANG ; Hongjun YU ; Shounan LU ; Shanjia KE ; Yanan XU ; Zhigang FENG ; Baolin QIAN ; Miaoyu BAI ; Bing YIN ; Xinglong LI ; Yongliang HUA ; Zhongyu LI ; Dong CHEN ; Bangliang CHEN ; Yongzhi ZHOU ; Shangha PAN ; Yao FU ; Hongchi JIANG ; Dawei WANG ; Yong MA
Chinese Medical Journal 2025;138(22):2993-3003
BACKGROUND:
Arsenic trioxide (ATO) is indicated as a broad-spectrum medicine for a variety of diseases, including cancer and cardiac disease. While the role of ATO in hepatic ischemia/reperfusion injury (HIRI) has not been reported. Thus, the purpose of this study was to identify the effects of ATO on HIRI.
METHODS:
In the present study, we established a 70% hepatic warm I/R injury and partial hepatectomy (30% resection) animal models in vivo and hepatocytes anoxia/reoxygenation (A/R) models in vitro with ATO pretreatment and further assessed liver function by histopathologic changes, enzyme-linked immunosorbent assay, cell counting kit-8, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay. Small interfering RNA (siRNA) for extracellular signal-regulated kinase (ERK) 1/2 was transfected to evaluate the role of ERK1/2 pathway during HIRI, followed by ATO pretreatment. The dynamic process of autophagic flux and numbers of autophagosomes were detected by green fluorescent protein-monomeric red fluorescent protein-LC3 (GFP-mRFP-LC3) staining and transmission electron microscopy.
RESULTS:
A low dose of ATO (0.75 μmol/L in vitro and 1 mg/kg in vivo ) significantly reduced tissue necrosis, inflammatory infiltration, and hepatocyte apoptosis during the process of hepatic I/R. Meanwhile, ATO obviously promoted the ability of cell proliferation and liver regeneration. Mechanistically, in vitro studies have shown that nontoxic concentrations of ATO can activate both ERK and phosphoinositide 3-kinase-serine/threonine kinase (PI3K-AKT) pathways and further induce autophagy. The hepatoprotective mechanism of ATO, at least in part, relies on the effects of ATO on the activation of autophagy, which is ERK-dependent.
CONCLUSION
Low, non-toxic doses of ATO can activate ERK/PI3K-AKT pathways and induce ERK-dependent autophagy in hepatocytes, protecting liver against I/R injury and accelerating hepatocyte regeneration after partial hepatectomy.
Animals
;
Arsenic Trioxide
;
Autophagy/physiology*
;
Reperfusion Injury/prevention & control*
;
Mice
;
Male
;
Proto-Oncogene Proteins c-akt/physiology*
;
Arsenicals/therapeutic use*
;
Oxides/therapeutic use*
;
Liver/metabolism*
;
Extracellular Signal-Regulated MAP Kinases/metabolism*
;
Mice, Inbred C57BL
2.Investigation of the Effects of Arsenic Trioxide Combined with Deslorelin on Proliferation and Apoptosis of Jurkat Cells Based on Wnt/β-Catenin Pathway.
Journal of Experimental Hematology 2025;33(3):640-647
OBJECTIVE:
To investigate the effect of Arsenic trioxide (ATO) combined with Norcantharidin (NCTD) on the proliferation and apoptosis of Jurkat cells, and to evaluate its effect on the proliferation and apoptosis of acute T-lymphoblastic leukemia (T-ALL) based on the Wnt/β-catenin signaling pathway.
METHODS:
Jurkat cell lines were used as the study subjects and treated with different concentrations of ATO (0, 2, 4, 8, 16 μmol/L) and NCTD (0, 10, 25, 50, 100 μmol/L) for 72 hours, and the cell proliferation was detected by CCK-8. Meanwhile, flow cytometry was used to detect the apoptosis rate, EdU staining to detect cell proliferation viability, cell clone formation assay to assess cell cloning ability, Transwell assay to assess cell invasion ability, and Western blot to detect apoptosis and the expression of Wnt/β-catenin signaling pathway-related proteins.
RESULTS:
Compared with the control group, both ATO and NCTD effectively inhibited Jurkat cell proliferation when used alone, and the inhibition effect was more significant when used in combination ( P < 0.05). The combination significantly increased the apoptosis rate of Jurkat cells ( P < 0.05). Meanwhile, the combination significantly decreased the proliferation vitality and clone formation ability of the cells ( P < 0.05), and inhibited the invasion ability of Jurkat cells ( P < 0.05). Western blot analysis showed that the combination of ATO and NCTD significantly up-regulated the expression of pro-apoptotic proteins Bax and E-cadherin, and down-regulated the expression of anti-apoptotic proteins Bcl-2, c-myc and Cyclin D1 ( P < 0.05).
CONCLUSION
The combination of ATO and NCTD had a synergistic effect in inhibiting proliferation and promoting apoptosis in Jurkat cells, which may be related to the inhibition of Wnt/β-catenin signaling pathway.
Humans
;
Apoptosis/drug effects*
;
Jurkat Cells
;
Cell Proliferation/drug effects*
;
Arsenic Trioxide
;
Wnt Signaling Pathway/drug effects*
;
Bridged Bicyclo Compounds, Heterocyclic/pharmacology*
;
beta Catenin/metabolism*
;
Arsenicals/pharmacology*
;
Oxides/pharmacology*
3.Microorganism-mediated arsenic reduction and its environmental effects.
Teng MAO ; Guoliang CHEN ; Zhihui QU
Chinese Journal of Biotechnology 2024;40(12):4480-4492
Arsenic (As) is a common toxic pollution element. The microorganism-mediated transformation of arsenic forms is an important part in the biogeochemical cycle of As. In the various microbial metabolic processes involving As, the coupling reduction of As has a great impact on the environment and is a process that is easily overlooked. From the biogeochemical cycle of As, this review introduces the microorganism-mediated methane oxidation, anaerobic ammonium oxidation, and iron (Fe)-sulfur (S) oxidation coupled with As reduction. Organic matter, pH, and redox potential are the main factors affecting the coupling reduction. After the coupling reduction, the toxicity and migration of As are greatly enhanced, which may increase the risk of As pollution. Therefore, it is of great significance to clarify the influences of carbon, nitrogen, Fe, S and other elements on the coupling process and explore more microbial processes coupled with As reduction for the prevention and control of As pollution.
Arsenic/metabolism*
;
Oxidation-Reduction
;
Bacteria/metabolism*
;
Environmental Pollutants/metabolism*
;
Biodegradation, Environmental
;
Methane/metabolism*
;
Iron/metabolism*
;
Ammonium Compounds/metabolism*
4.CircRNA-0028171 regulates arsenic trioxide-induced apoptosis in vascular endothelial cells.
Ji-Chen WU ; Sai-Di JIN ; Jia-Hang SONG ; Xin-Qi LIU ; Wen-Jun MA ; Lin CHANG ; Xiao-Xiang GUAN ; Ming-Yu ZHANG ; Jia-Qi LIU ; Hui FU ; Ying WANG ; Chao-Qian XU
Acta Physiologica Sinica 2022;74(5):763-772
The present study was aimed to investigate the effects of circRNA-0028171 on the apoptosis of vascular endothelial cells induced by arsenic trioxide (As2O3). Human umbilical vein endothelial cells (HUVECs) were treated with 0-15 μmol/L As2O3 for 24 h. Then, cellular viability was measured by MTT assay. The expression levels of circRNA-0028171, Bcl-2 and Bax mRNA were detected by real-time quantitative PCR. Bcl-2/Bax protein ratio was detected by Western blot. Whether circRNA-0028171 was involved in the regulation of HUVECs by As2O3 was investigated by transfection with overexpression plasmid of circRNA-0028171 and siRNA. The results showed that compared with the control group, As2O3 group showed decreased cellular viability, reduced Bcl-2/Bax mRNA and protein ratios, and significantly lower expression of circRNA-0028171. Overexpression of circRNA-0028171 inhibited apoptosis of HUVECs induced by As2O3. Knockdown of circRNA-0028171 by siRNA promoted As2O3-induced apoptosis in HUVECs. These results suggest that circRNA-0028171 is involved in the vascular endothelial cell apoptosis induced by As2O3.
Humans
;
Arsenic Trioxide/pharmacology*
;
RNA, Circular
;
bcl-2-Associated X Protein/metabolism*
;
RNA, Small Interfering/metabolism*
;
Apoptosis
;
Proto-Oncogene Proteins c-bcl-2/metabolism*
;
Human Umbilical Vein Endothelial Cells/metabolism*
;
RNA, Messenger/metabolism*
5.Effects of arsenic and its main metabolites on A549 cell apoptosis and the expression of pro-apoptotic genes Bad and Bik.
Qian ZHOU ; Jin Yao YIN ; Jing Wen TAN ; Shu Ting LI ; Cheng Lan JIANG ; Yue Feng HE
Chinese Journal of Industrial Hygiene and Occupational Diseases 2022;40(9):661-667
Objective: To investigate the effect of arsenic and its main metabolites on the apoptosis of human lung adenocarcinoma cell line A549 and the expression of pro-apoptotic genes Bad and Bik. Methods: In October 2020, A549 cells were recovered and cultured, and the cell viability was detected by the cell counting reagent CCK-8 to determine the concentration and time of sodium arsenite exposure to A549. The study was divided into NaAsO(2) exposure groups and metobol: le expoure groups: the metabolite comparison groups were subdivided into the control group, the monomethylarsinic acid exposure group (60 μmol/L) , and the dimethylarsinic acid exposure group (60 μmol/L) ; sodium arsenite dose groups were subdivided into 4 groups: control group (0) , 20, 40, 60 μmol/L sodium arsenite NaAsO(2). Hoechst 33342/propidium iodide double staining (Ho/PI) was used to observe cell apoptosis and real-time quantitative polymerase chain reaction (qRT-PCR) was used to detect the expression levels of Bad and Bik mRNA in cells after exposure. Western blotting was used to detect the protein expressions of Bad, P-Bad-S112, Bik, cleaved Bik and downstream proteins poly ADP-ribose polymerase PARP1 and cytochrome C (Cyt-C) , using spectrophotometry to detect the activity changes of caspase 3, 6, 8, 9. Results: Compared with the control group, the proportion of apoptotic cells in the 20, 40, and 60 μmol/L NaAsO(2) dose groups increased significantly (P<0.01) , and the expression levels of Bad, Bik mRNA, the protein expression levels of Bad, P-Bad-S112, Bik, cleaved Bik, PARP1, Cyt-C were increased (all P<0.05) , and the activities of Caspase 3, 6, 8, and 9 were significantly increased with significantly differences (P<0.05) . Compared with the control group, the expression level of Bad mRNA in the DMA exposure group (1.439±0.173) was increased with a significant difference (P=0.024) , but there was no significant difference in the expression level of Bik mRNA (P=0.788) . There was no significant differences in the expression levels of Bad and Bik mRNA in the poison groups (P=0.085, 0.063) . Compared with the control group, the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to MMA were 0.696±0.023, 0.707±0.014, 0.907±0.031, 1.032±0.016, and there was no significant difference between the two groups (P=0.469, 0.669, 0.859, 0.771) ; the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to DMA were 0.698±0.030, 0.705±0.022, 0.908±0.015, 1.029±0.010, and there was no difference between the two groups (P=0.479, 0.636, 0.803, 0.984) . Conclusion: Sodium arsenite induces the overexpression of Bad and Bik proteins, initiates the negative feedback regulation of phosphorylated Bad and the degradation of Bik, activates the downstream proteins PARP1, Cyt-C and Caspase pathways, and mediates the apoptosis of A549 cells.
A549 Cells
;
Adenosine Diphosphate Ribose/pharmacology*
;
Apoptosis
;
Apoptosis Regulatory Proteins
;
Arsenic
;
Arsenites
;
Cacodylic Acid/pharmacology*
;
Caspase 3
;
Caspases/pharmacology*
;
Cytochromes c/pharmacology*
;
Humans
;
Mitochondrial Proteins/pharmacology*
;
Poisons
;
Propidium/pharmacology*
;
RNA, Messenger
;
Sincalide/pharmacology*
;
Sodium Compounds
;
bcl-Associated Death Protein/metabolism*
6.The research progress of metabolism and occupational biological exposure index metabolism of inorganic arsenic and its compounds.
Chinese Journal of Industrial Hygiene and Occupational Diseases 2022;40(11):876-880
Arsenic is a common metal-like element. Drinking arsenic-containing water and occupational exposure to arsenic are the main ways exposure to arsenic for population. Long-term exposure to arsenic can cause various organs dysfunction and cancer. After entering the body, inorganic arsenic is mainly methylated into monomethyl arsenic and dimethyl arsenic in the liver. Only a small part of inorganic arsenic is metabolized in the kidneys and lungs, and finally the metabolites of arsenic are excreted in the urine. understanding the biological characteristics of arsenic absorption, metabolism, and distribution in the body and formulating biological indicators related to occupational exposure to arsenic and can provide a scientific basis for the prevention and treatment of arsenic-related diseases. This article will review the biological monitoring indicators of occupational exposure to arsenic and the metabolic process of arsenic in the body.
Arsenicals
;
Arsenic
;
Occupational Exposure
;
Drinking Water
;
Liver/metabolism*
7.Effects of maternal exposure to arsenic on social behavior and related gene expression in F2 male mice.
Soe-Minn HTWAY ; Takehiro SUZUKI ; Sanda KYAW ; Keiko NOHARA ; Tin-Tin WIN-SHWE
Environmental Health and Preventive Medicine 2021;26(1):34-34
BACKGROUND:
Arsenic is a developmental neurotoxicant. It means that its neurotoxic effect could occur in offspring by maternal arsenic exposure. Our previous study showed that developmental arsenic exposure impaired social behavior and serotonergic system in C3H adult male mice. These effects might affect the next generation with no direct exposure to arsenic. This study aimed to detect the social behavior and related gene expression changes in F2 male mice born to gestationally arsenite-exposed F1 mice.
METHODS:
Pregnant C3H/HeN mice (F0) were given free access to tap water (control mice) or tap water containing 85 ppm sodium arsenite from days 8 to 18 of gestation. Arsenite was not given to F1 or F2 mice. The F2 mice were generated by mating among control F1 males and females, and arsenite-F1 males and females at the age of 10 weeks. At 41 weeks and 74 weeks of age respectively, F2 males were used for the assessment of social behavior by a three-chamber social behavior apparatus. Histological features of the prefrontal cortex were studied by ordinary light microscope. Social behavior-related gene expressions were determined in the prefrontal cortex by real time RT-PCR method.
RESULTS:
The arsenite-F2 male mice showed significantly poor sociability and social novelty preference in both 41-week-old group and 74-week-old group. There was no significant histological difference between the control mice and the arsenite-F2 mice. Regarding gene expression, serotonin receptor 5B (5-HT 5B) mRNA expression was significantly decreased (p < 0.05) in the arsenite-F2 male mice compared to the control F2 male mice in both groups. Brain-derived neurotrophic factor (BDNF) and dopamine receptor D1a (Drd1a) gene expressions were significantly decreased (p < 0.05) only in the arsenite-F2 male mice of the 74-week-old group. Heme oxygenase-1 (HO-1) gene expression was significantly increased (p < 0.001) in the arsenite-F2 male mice of both groups, but plasma 8-hydroxy-2'-deoxyguanosine (8-OHdG) and cyclooxygenase-2 (COX-2) gene expression were not significantly different. Interleukin-1β (IL-1β) mRNA expression was significantly increased only in 41-week-old arsenite-F2 mice.
CONCLUSIONS
These findings suggest that maternal arsenic exposure affects social behavior in F2 male mice via serotonergic system in the prefrontal cortex. In this study, COX-2 were not increased although oxidative stress marker (HO-1) was increased significantly in arsnite-F2 male mice.
Animals
;
Arsenic/toxicity*
;
Arsenites/toxicity*
;
Behavior, Animal/drug effects*
;
Environmental Pollutants/toxicity*
;
Female
;
Gene Expression/drug effects*
;
Genetic Markers
;
Male
;
Maternal Exposure/adverse effects*
;
Mice
;
Mice, Inbred C3H
;
Oxidative Stress/genetics*
;
Prefrontal Cortex/drug effects*
;
Pregnancy
;
Prenatal Exposure Delayed Effects/psychology*
;
Reverse Transcriptase Polymerase Chain Reaction
;
Serotonin/metabolism*
;
Social Behavior
;
Sodium Compounds/toxicity*
8.Arsenic Trioxide Combining Leflunomide Activates Nrf2-ARE-HO-1 Signaling Pathway and Protects Heart Xenografts.
Teng-da WANG ; Song-Lin XU ; Zheng-Yi YU ; Shao-Bin NI ; Cheng ZHANG ; Zhi-Xing JIAO
Chinese journal of integrative medicine 2021;27(10):760-766
OBJECTIVE:
To investigate the molecular mechanisms underlying the effects of arsenic trioxide (As
METHODS:
Transplantation of LVG hamster hearts to Lewis rats was performed by anastomosis of vessels in the neck using end-to-end anastomosis with a non-suture cuff technique. Four groups of recipient rats (n=6 in each) were treated with normal saline (control), As
RESULTS:
Expression of Nrf2-ARE-HO-1 signaling pathway was upregulated in heart xenografts in rats treated with As
CONCLUSION
Combination treatment with As
Animals
;
Arsenic Trioxide
;
Cricetinae
;
Heart Transplantation
;
Heme Oxygenase-1/metabolism*
;
Heterografts
;
Leflunomide
;
NF-E2-Related Factor 2/metabolism*
;
Rats
;
Rats, Inbred Lew
;
Signal Transduction
9.Involvement of PML proteins in treatment of acute promyelocytic leukemia with arsenic trioxide.
Rui HAO ; Lide SU ; Yiming SHAO ; Na BU ; Liya MA ; Hua NARANMANDURA
Journal of Zhejiang University. Medical sciences 2018;47(5):541-551
Promyelocytic leukemia (PML) protein, a tumor suppressor, plays an important role in patients with acute promyelocytic leukemia (APL) receiving arsenic trioxide (AsO) therapy. APL is a M3 subtype of acute myeloid leukemia (AML), which is characterized by expression of PML-RARα (P/R) fusion protein, leading to the oncogenesis. AsO is currently used as the first-line drug for patients with APL, and the mechanism may be:AsO directly binds to PML part of P/R protein and induces multimerization of related proteins, which further recruits different functional proteins to reform PML nuclear bodies (PML-NBs), and finally it degraded by SUMOylation and ubiquitination proteasomal pathway. Gene mutations may lead to relapse and drug resistance after AsO treatment. In this review, we discuss the structure and function of PML proteins; the pathogenesis of APL induced by P/R fusion protein; the involvement of PML protein in treatment of APL patient with AsO; and explain how PML protein mutations could cause resistance to AsO therapy.
Antineoplastic Agents
;
therapeutic use
;
Arsenic Trioxide
;
therapeutic use
;
Drug Resistance, Neoplasm
;
genetics
;
Humans
;
Leukemia, Promyelocytic, Acute
;
drug therapy
;
Mutation
;
Oncogene Proteins, Fusion
;
metabolism
;
Promyelocytic Leukemia Protein
;
chemistry
;
genetics
;
metabolism
10.Increased PIT1 and PIT2 Expression in Streptozotocin (STZ)-induced Diabetic Mice Contributes to Uptake of iAs(V).
Sha Li YU ; Ling Fei XU ; Jun Xia WU ; Chen Juan YAO ; Qiao Yun HU ; Chun Xue ZHANG ; Xin Yuan ZHAO ; Hai Yan WEI ; Xiao Ke WANG ; Gang CHEN
Biomedical and Environmental Sciences 2017;30(11):792-801
OBJECTIVEThis study aimed to investigate the susceptibility of mice with streptozotocin(STZ)-induced diabetes mellitus (TIDM) to the uptake of pentavalent inorganic arsenic (iAsV) and the possible molecular mechanism.
METHODSTIDM was induced in mice by STZ. TIDM and normal mice were treated with 15.0 mg/kg Na2HAsO4·12H2O by intragastric administration. Then, the concentrations of arsenic in various tissues were measured by atomic fluorescence spectrometry. The gene expression levels of Pit1 and Pit2 were quantified by real-time RT-PCR, and their protein levels were detected by Western blotting in mouse heart, kidney, and liver tissues.
RESULTSThe concentrations of arsenic in STZ-induced TIDM mouse tissues were higher at 2 h after intragastric administration of Na2HAsO4·12H2O. Compared with the levels in normal mice, PIT1 and PIT2, which play a role in the uptake of iAsV, were upregulated in the livers and hearts of TIDM mice. PIT1 but not PIT2 was higher in TIDM mouse kidneys. The upregulation of Pit1 and Pit2 expression could be reversed by insulin treatment.
CONCLUSIONThe increased uptake of iAsV in TIDM mouse tissues may be associated with increased PIT1 and/or PIT2 expression.
Animals ; Arsenic ; pharmacokinetics ; Diabetes Mellitus, Experimental ; metabolism ; Environmental Pollutants ; pharmacokinetics ; Gene Expression Regulation ; physiology ; Male ; Mice ; Mice, Inbred ICR ; Sodium-Phosphate Cotransporter Proteins, Type III ; genetics ; metabolism ; Transcription Factor Pit-1 ; genetics ; metabolism

Result Analysis
Print
Save
E-mail