1
|
Li S, Wang Y, Lv Z, Wang Q, Jia T, Zhai Z, Fang W. Angiotensin 1-7 restrains vascular injury of extracorporeal membrane oxygenation by inhibiting ferroptosis. Int Immunopharmacol 2024; 142:113177. [PMID: 39298820 DOI: 10.1016/j.intimp.2024.113177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Angiotensin 1-7 (Ang1-7) is the classical end product of angiotensin II, which has the effects of dilating blood vessels, protecting endothelial cells, anti-hypertension, improving cardiac function, and inhibiting atherosclerosis. We hypothesize that Ang1-7 inhibits human umbilical vein endothelial cells (HUVEC) ferroptosis through NF-κB/P53 signal pathway, and reduces extracorporeal membrane oxygenation (ECMO) vascular injury. METHODS Cultured HUVEC were seeded into 15 wells and randomly divided into five groups: the control group and four experimental groups (erastin, erastin + Ang1-7, erastin + Ang1-7 + Betulinic acid, erastin + Betulinic acid). After stimulation, cell viability, lactate dehydrogenase (LDH), malondialdehyde (MDA), and superoxide dismutase (SOD) activity were measured. The effects of Ang1-7 on HUVEC microstructure, antioxidant enzymes (ferritin heavy chain 1 (FTH1), cystine/glutamic acid reverse transport solute carrier family 7 members 11 (SLC7A11 or XCT), superoxide dismutase-2 (SOD-2) and glutathione peroxidase 4 (GPX4)), NF-κB, P-NF-κB, P53, and P-P53). RESULTS Erastin stimulation promoted HUVEC lipid peroxidation, decreased antioxidant enzyme expression, increased P-NF-κB, P53, and P-P53 expressions, and damaged HUVEC mitochondrial structure. Ang1-7 alleviated the effect of erastin on HUVEC, which was destroyed by Betulinic acid. CONCLUSION Angiotensin1-7 pretreatment inhibited vascular endothelial cells' ferroptosis and alleviated ECMO vessel injury through NF-κB /P53 signal pathway.
Collapse
Affiliation(s)
- Shengqiang Li
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China; Department of Physiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuping Wang
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhen Lv
- Department of Cardiology, Zibo First Hospital, Zibo 255200, China
| | - Qizhi Wang
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tong Jia
- Department of Geratology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Zhenzhen Zhai
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China.
| | - Wei Fang
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
2
|
Yang F, Hu D, Du S, Wu L, Gong M, Zhang Y, Yang X, Yang Y, Chen R, Xu Y, Zeng Q. Assessing the double-edged of extracellular signal-regulated kinase/CCAAT-enhancer-binding protein beta signaling pathway in arsenic-induced skin damage and its potential foodborne interventions. ENVIRONMENTAL TOXICOLOGY 2023; 38:2867-2880. [PMID: 37565747 DOI: 10.1002/tox.23922] [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: 05/31/2023] [Revised: 07/06/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
Arsenic exposure is a major environmental public health challenge worldwide. As typical manifestations for arsenic exposure, the pathogenesis of arsenic-induced skin lesions has not been fully elucidated, as well as the lack of effective control measures. In this study, we first determined the short-term and high-dose arsenic exposure can increase the apoptosis rates, while long-term low-dose arsenic exposure decrease the apoptosis rates. Then, the HaCaT cells with knockdown and overexpression of CCAAT-enhancer-binding protein β (CEBPB) and extracellular signal-regulated kinase (ERK) were constructed. The results demonstrate that knockdown of CEBPB and ERK can reduce NaAsO2 -induced cell apoptosis by inhibiting ERK/CEBPB signaling pathway and vice versa. Further cells were treated with Kaji-Ichigoside F1 (KF1). The results clearly show that KF1 can decrease the arsenic-induced cell apoptosis rates and the expression of ERK/CEBPB signaling pathway-related genes. These results provide evidence that ERK/CEBPB signaling pathway acts as a double-edged sword in arsenic-induced skin damage. Another interesting finding was that KF1 can alleviate arsenic-induced skin cell apoptosis by inhibiting the ERK/CEBPB signaling pathway. This study will contribute to a deeper understanding of the mechanisms of arsenic-induced skin cell apoptosis, and our findings will help to identify a potential food-borne intervention in arsenic detoxification.
Collapse
Affiliation(s)
- Fan Yang
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Dexiu Hu
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Sufei Du
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Liping Wu
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Maoyuan Gong
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yuhong Zhang
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Xingcan Yang
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yang Yang
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Ruobi Chen
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yuyan Xu
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
| | - Qibing Zeng
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education & School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Ecological Food Creation Engineering Research Center, Guizhou Medical University, Guiyang, China
| |
Collapse
|
3
|
Wen W, Zha S, Cheng H, Qi J, Chen Q, Gu Y. As3MT is related to relative RNAs and base modifications of p53 in workers exposed to arsenic. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:62094-62103. [PMID: 36940027 DOI: 10.1007/s11356-023-26457-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/10/2023] [Indexed: 05/10/2023]
Abstract
As3MT is the key enzyme involved in the methylation metabolism of arsenic. It is associated with DNA methylation closely also. This study is to explore the relationships between As3MT and epigenetic changes, and how p53 and relative ncRNAs and mRNAs play roles in the process. In this study, workers from four arsenic plants and individuals who resided in villages far away from the four plants were recruited. Arsenic compounds, relative indices, 28 relative RNAs, and base modifications of exons 5-8 of p53 were detected separately. Several methods were used to analyze the associations between them. Results shown that As3MT RNA was closely associated with all selected lncRNAs, miRNAs, and mRNAs related to miRNA production and maturation, tumorigenesis, and base modifications of p53. There probably exists causal relationship. Base modifications of exons 7 and 8 of p53 had significant synergistic effects on the expression of As3MT RNA and a series of genetic indices. But miR-190, miR-548, and base modifications of exon 5 of p53 had substantial inhibitory effects. Arsenic compounds and relative indices of metabolic transformation may have limited roles. The main novel finding in the present study is that As3MT play special and significant roles in the genotoxicity and carcinogenesis which could be coordinated operation with p53, and influenced by epigenetic factors largely, such as lncRNAs and miRNAs. P53 and relative ncRNAs and mRNAs may regulate the process by interacting with As3MT. The changes may initiate by arsenic, but probability through indirect relationship.
Collapse
Affiliation(s)
- Weihua Wen
- Yunnan Center for Disease Control and Prevention, No.158, Dongsi Street, Kunming, 650022, Yunnan, China.
| | - Shun Zha
- Yunnan Center for Disease Control and Prevention, No.158, Dongsi Street, Kunming, 650022, Yunnan, China
| | - Huirong Cheng
- Yunnan Center for Disease Control and Prevention, No.158, Dongsi Street, Kunming, 650022, Yunnan, China
| | - Jun Qi
- Yunnan Center for Disease Control and Prevention, No.158, Dongsi Street, Kunming, 650022, Yunnan, China
| | - Qian Chen
- Public Health College, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yun Gu
- Public Health College, Kunming Medical University, Kunming, 650500, Yunnan, China
| |
Collapse
|
4
|
So KY, Oh SH. Arsenite-induced cytotoxicity is regulated by poly-ADP ribose polymerase 1 activation and parthanatos in p53-deficient H1299 cells: The roles of autophagy and p53. Biochem Biophys Res Commun 2023; 656:78-85. [PMID: 36958258 DOI: 10.1016/j.bbrc.2023.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Arsenic is a double-edged sword metalloid since it is both an environmental carcinogen and a chemopreventive agent. Arsenic cytotoxicity can be dependent or independent of the tumor suppressor p53. However, the effects and the underlying molecular mechanisms of arsenic cytotoxicity in p53-deficient cells are still unclear. Here, we report a distinctive cell death mode via PARP-1 activation by arsenic in p53-deficient H1299 cells. H1299 (p53-/-) cells showed higher sensitivity to sodium arsenite (NaAR) than H460 (p53+/+) cells. H460 cells induced canonical apoptosis through caspase-dependent poly-ADP ribose polymerase 1 (PARP-1) cleavage and induced the expression of phospho-p53 and p21. However, H1299 cells induced poly-ADP-ribose (PAR) polymer accumulation and caspase-independent parthanatos, which was inhibited by 3-aminobenzamide (AB) and nicotinamide (NAM). Fractionation studies revealed the mitochondrial translocation of PAR polymers and nuclear translocation of the apoptosis-inducing factor (AIF). Although the exposure of NaAR to p53-overexpressing H1299 cells increased the PAR polymer levels, it inhibited parthanatos by inducing p21 and phospho-p53 expression. LC3-II and p62 accumulated in a NaAR dose- and exposure time-dependent manner, and this accumulation was further enhanced by autophagy inhibition, indicating that arsenic inhibits autophagic flux. p53 overexpression led to a decrease in the p62 levels, an increase in the LC3-II levels, and reduced parthanatos, indicating that arsenic induces p53-dependent functional autophagy. These results show that the NaAR-induced cytotoxicity in p53-deficient H1299 cells is regulated by PARP-1 activation-mediated parthanatos, which is promoted by autophagy inhibition. This suggests that PARP-1 activation could be used as an effective therapeutic approach for arsenic toxicity in p53-deficient cells.
Collapse
Affiliation(s)
- Keum-Young So
- Department of Anesthesiology and Pain Medicine, 309 Pilmundaero, Dong-gu, Gwangju, 61452, Republic of Korea
| | - Seon-Hee Oh
- School of Medicine, Chosun University, 309 Pilmundaero, Dong-gu, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
5
|
Mukherjee AG, Valsala Gopalakrishnan A. The interplay of arsenic, silymarin, and NF-ĸB pathway in male reproductive toxicity: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114614. [PMID: 36753973 DOI: 10.1016/j.ecoenv.2023.114614] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Arsenic toxicity is one of the most trending reasons for several malfunctions, particularly reproductive toxicity. The exact mechanism of arsenic poisoning is a big question mark. Exposure to arsenic reduces sperm count, impairs fertilization, and causes inflammation and genotoxicity through interfering with autophagy, epigenetics, ROS generation, downregulation of essential protein expression, metabolite changes, and hampering several signaling cascades, particularly by the alteration of NF-ĸB pathway. This work tries to give a clear idea about the different aspects of arsenic resulting in male reproductive complications, often leading to infertility. The first part of this article explains the implications of arsenic poisoning and the crosstalk of the NF-ĸB pathway in male reproductive toxicity. Silymarin is a bioactive compound that exerts anti-cancer and anti-inflammatory properties and has demonstrated hopeful outcomes in several cancers, including colon cancer, breast cancer, and skin cancer, by downregulating the hyperactive NF-ĸB pathway. The next half of this article thus sheds light on silymarin's therapeutic potential in inhibiting the NF-ĸB signaling cascade, thus offering protection against arsenic-induced male reproductive toxicity.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| |
Collapse
|
6
|
Hsa_circ_0005050 interacts with ILF3 and affects cell apoptosis and proliferation by disrupting the balance between p53 and p65. Chem Biol Interact 2022; 368:110208. [DOI: 10.1016/j.cbi.2022.110208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/15/2022] [Accepted: 10/03/2022] [Indexed: 11/19/2022]
|
7
|
Ma N, Guo J, Wu X, Liu Z, Yao T, Zhao Q, Li B, Tian F, Yan X, Zhang W, Qiu Y, Gao Y. Meta-analysis of TLR4 pathway-related protein alterations induced by arsenic exposure. Biol Trace Elem Res 2022; 201:3290-3299. [PMID: 36166114 DOI: 10.1007/s12011-022-03426-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/15/2022] [Indexed: 11/02/2022]
Abstract
Arsenic is a toxic metal, which ultimately leads to cell apoptosis. TLR4 signaling pathway played a key role in immunomodulatory. Therefore, alterations in related proteins on the TLR4 signaling pathway induced by arsenic exposure was systematically reviewed and analyzed by meta-analysis. Some databases were searched including PubMed, Web of Science, China National Knowledge Infrastructure (CNKI), and WANFANG MED ONLINE. The results of NF-κB, IKK, NF-κBp65, phospho-NF-κBp65, and TLR4 expressions were analyzed by Review Manage 5.3. In the arsenic intervention group, NF-κB, phospho-NF-κBp65, and TLR4 expression levels were higher than the control group, respectively. SMD and 95%CI were 11.29 (6.34, 16.24), 4.71(1.73, 7.68), and 5.79 (-4.22, 15.80). Compared to controls, in the exposed group, IKK levels were found to be 38.11-fold higher (Z = 0.97; P = 0.33); NF-κBp65 levels were found to be 0.92-fold higher (Z = 3.33; P = 0.0009) for normal cells and tissue, while IKK levels were found to be 5.18-fold lower (Z = 5.34; P < 0.0001); NF-κBp65 levels were found to be 2.01-fold lower (Z = 3.87; P = 0.0001) for abnormal cells. With comparing of low dose, high dose of arsenic exposure was found to reduce the expression of NF-κB, but increase the expression of NF-κBp65. This review supports the alterations in related proteins on the TLR4 signaling pathway induced by arsenic exposure, which is helpful to provide theoretical basis for the mechanism of toxicity of arsenic-induced immune system damage.
Collapse
Affiliation(s)
- Nanxin Ma
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Jian Guo
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Xiaolong Wu
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Zhenzhong Liu
- School of Public Health, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Tian Yao
- The First Hospital of Shanxi Medical University, Shanxi, 030001, China
| | - Qian Zhao
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Ben Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Fengjie Tian
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Xiaoyan Yan
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Wenping Zhang
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Yulan Qiu
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China
| | - Yi Gao
- Department of Toxicology, School of Public Health, Shanxi Medical University, 56 Xin-Jian South Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
8
|
Dutta S, Gorain B, Choudhury H, Roychoudhury S, Sengupta P. Environmental and occupational exposure of metals and female reproductive health. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62067-62092. [PMID: 34558053 DOI: 10.1007/s11356-021-16581-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/13/2021] [Indexed: 06/13/2023]
Abstract
Untainted environment promotes health, but the last few decades experienced steep upsurge in environmental contaminants posing detrimental physiological impact. The responsible factors mainly include the exponential growth of human population, havoc rise in industrialization, poorly planned urbanization, and slapdash environment management. Environmental degradation can increase the likelihood of human exposure to heavy metals, resulting in health consequences such as reproductive problems. As a result, research into metal-induced causes of reproductive impairment at the genetic, epigenetic, and biochemical levels must be strengthened further. These metals impact upon the female reproduction at all strata of its regulation and functions, be it development, maturation, or endocrine functions, and are linked to an increase in the causes of infertility in women. Chronic exposures to the heavy metals may lead to breast cancer, endometriosis, endometrial cancer, menstrual disorders, and spontaneous abortions, as well as pre-term deliveries, stillbirths. For example, endometriosis, endometrial cancer, and spontaneous abortions are all caused by the metalloestrogen cadmium (Cd); lead (Pb) levels over a certain threshold can cause spontaneous abortion and have a teratogenic impact; toxic amounts of mercury (Hg) have an influence on the menstrual cycle, which can lead to infertility. Impact of environmental exposure to heavy metals on female fertility is therefore a well-known fact. Thus, the underlying mechanisms must be explained and periodically updated, given the growing evidence on the influence of increasing environmental heavy metal load on female fertility. The purpose of this review is to give a concise overview of how heavy metal affects female reproductive health.
Collapse
Affiliation(s)
- Sulagna Dutta
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom, Malaysia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Hira Choudhury
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | | | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Malaysia.
| |
Collapse
|
9
|
Jang JY, Im E, Choi YH, Kim ND. Mechanism of Bile Acid-Induced Programmed Cell Death and Drug Discovery against Cancer: A Review. Int J Mol Sci 2022; 23:7184. [PMID: 35806184 PMCID: PMC9266679 DOI: 10.3390/ijms23137184] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
Bile acids are major signaling molecules that play a significant role as emulsifiers in the digestion and absorption of dietary lipids. Bile acids are amphiphilic molecules produced by the reaction of enzymes with cholesterol as a substrate, and they are the primary metabolites of cholesterol in the body. Bile acids were initially considered as tumor promoters, but many studies have deemed them to be tumor suppressors. The tumor-suppressive effect of bile acids is associated with programmed cell death. Moreover, based on this fact, several synthetic bile acid derivatives have also been used to induce programmed cell death in several types of human cancers. This review comprehensively summarizes the literature related to bile acid-induced programmed cell death, such as apoptosis, autophagy, and necroptosis, and the status of drug development using synthetic bile acid derivatives against human cancers. We hope that this review will provide a reference for the future research and development of drugs against cancer.
Collapse
Affiliation(s)
- Jung Yoon Jang
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Korea; (J.Y.J.); (E.I.)
| | - Eunok Im
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Korea; (J.Y.J.); (E.I.)
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea;
| | - Nam Deuk Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Korea; (J.Y.J.); (E.I.)
| |
Collapse
|
10
|
Wu L, Yang F, Du S, Hu T, Wei S, Wang G, Zeng Q, Luo P. Inorganic arsenic promotes apoptosis of human immortal keratinocytes through the TGF-β1/ERK signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:1321-1331. [PMID: 35142421 DOI: 10.1002/tox.23486] [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: 01/22/2022] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Chronic exposure to high-dose inorganic arsenic through groundwater, air, or food remains a major environmental public health issue worldwide. Apoptosis, a method of cell death, has recently become a hot topic of research in biology and medicine. Previous studies have demonstrated that extracellular signal-regulated kinase (ERK) is related to arsenic-induced apoptosis. However, the reports are contradictory, and the knowledge of the above-mentioned mechanisms and their mutual regulation remains limited. In this study, the associations between the TGF-β1/ERK signaling pathway and arsenic-induced cell apoptosis were confirmed using the HaCaT cell model. The relative expressions of the indicators of the TGF-β1/ERK signaling pathway, apoptosis-related genes (cytochrome C, caspase-3, caspase-9, cleaved caspase-3, cleaved caspase-9, and Bax), the mitochondrial membrane potential, and the total apoptosis rate were significantly increased (P < .05), while the expression of the antiapoptosis gene Bcl-2 was significantly decreased (P < .05) in cells of the group exposed to arsenic. Moreover, the results demonstrated that the ERK inhibitor (PD98059) and TGF-β1 inhibitor (LY364947) could inhibit the activation of the ERK signaling pathway, thereby reducing the mitochondrial membrane potential, the total apoptosis rate, and the expression of pro-apoptosis-related genes in the cells, while the expression of the antiapoptosis gene Bcl-2 was significantly increased (P < .05). By contrast, the recombinant human TGF-β1 could promote apoptosis of the HaCaT cells by increasing the activation of the ERK signaling pathway (P < .05). These results indicate that inorganic arsenic promotes the apoptosis of human immortal keratinocytes through the TGF-β1/ERK signaling pathway.
Collapse
Affiliation(s)
- Liping Wu
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Fan Yang
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Sufei Du
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Ting Hu
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guizhou Medical University, Guiyang, China
| | - Shaofeng Wei
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guizhou Medical University, Guiyang, China
| | - Guoze Wang
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guizhou Medical University, Guiyang, China
| | - Qibing Zeng
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guizhou Medical University, Guiyang, China
| | - Peng Luo
- The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Research Center of Food Nutrition and Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
11
|
Lei R, Zhou M, Zhang S, Luo J, Qu C, Wang Y, Guo P, Huang R. Potential role of PRKCSH in lung cancer: bioinformatics analysis and a case study of Nano ZnO. NANOSCALE 2022; 14:4495-4510. [PMID: 35254362 DOI: 10.1039/d1nr08133k] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
PRKCSH, also known as glucosidase II beta, functions as a contributor to lung tumorigenesis by regulating the cell cycle in a p53-dependent manner under severe environmental stress. However, the prognostic value and molecular mechanisms by which the level of PRKCSH is significantly increased in cancer cells are not clearly understood. Here, we first generated a biological profile of PRKCSH expression changes in cancers by analysing bioinformatic data from cancer databases. We found that higher PRKCSH expression was correlated with a poorer prognosis and greater infiltration of most immune cell types in patients with lung cancer. In particular, PRKCSH expression showed significant negative correlations with the level of STAT6 (r = -0.31, p < 0.001) in lung cancer tissues. We further found that PRKCSH deficiency promoted G2/M arrest in response to zinc oxide nanoparticle (Nano ZnO) treatment in A549 cells. With regard to the mechanism, PRKCSH deficiency may induce STAT6 translocation to the nucleus to activate p53 expression through binding to the p53 promoter region from -365 bp to +126 bp. Eventually, activated p53 contributed to Nano-ZnO-induced G2/M arrest in lung cancer cells. Taken together, our data provide new insights into immunotherapy target choices and the prognostic value of PRKCSH. Since the G2/M cell cycle checkpoint is crucial for lung cancer prognosis, targeting PRKCSH expression to suppress the activation of the STAT6/p53 pathway is a potential therapeutic strategy for managing lung cancer.
Collapse
Affiliation(s)
- Ridan Lei
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Meiling Zhou
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Shusheng Zhang
- Changsha Stomatological Hospital, Changsha, Hunan Province, China.
| | - Jinhua Luo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Can Qu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Yin Wang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Peiyu Guo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| |
Collapse
|
12
|
Miao Z, Miao Z, Wang S, Wu H, Xu S. Exposure to imidacloprid induce oxidative stress, mitochondrial dysfunction, inflammation, apoptosis and mitophagy via NF-kappaB/JNK pathway in grass carp hepatocytes. FISH & SHELLFISH IMMUNOLOGY 2022; 120:674-685. [PMID: 34954370 DOI: 10.1016/j.fsi.2021.12.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Imidacloprid (IMI) is a neonicotinoid compound widely used in agriculture production, causing surface water pollution and threatening non-target organisms. The aim of this study was to analyze the effects of IMI on grass carp (Ctenopharyngodon idellus) liver cell (L8824) injury. The L8824 cells were exposed to different doses of IMI (65 mg/L, 130 mg/L and 260 mg/L) for 24 h. Our results demonstrated that exposure IMI significantly suppressed the activity of anti-oxidant enzymes (SOD, CAT and T-AOC) and accumulated oxidase (MDA) levels, and promoting reactive oxygen species (ROS) generation in L8824 cells. Additionally, mitochondrial membrane potential (ΔΨ m), mitochondria-derived ROS and ATP content and the MitoTracker Green indicated that IMI aggravated mitochondrial dysfunction, thereby inducing inflammation and enhancing pro-inflammatory genes (NF-kappaB, TNFα, IL-1β and IL-6) expressions. However, the addition of 2 mM N-acetyl-l-cysteine (NAC) can reverse these adverse effects of high-dose IMI- induced. Hence, ROS is the main factor of IMI-induced mitochondrial dysfunction and inflammation. We further found that exposure to IMI induced apoptosis, which is characterized by promoting release of cytochrome c (Cyt-C), and increasing the expression of Bcl-2-Associated X (BAX), cysteinyl aspartate specific proteinases (Caspase 9 and 3), decreasing Bcl-2 level. Immunofluorescent staining, qRT-PCR and Western Blot results indicated that IMI exposure also activated mitophagy, which was demonstrated by the expression of mitophagy-related genes (BNIP3, LC3B and P62). Conversely, scavenging JNK by SP600125(10 μM) alleviated the expression of mitochondrial apoptosis and mitophagy-related gene induced by high-dose IMI. Therefore, these results of study demonstrated that IMI-induced oxidative stress to regulate mitochondrial dysfunction, thus causing inflammation, mitochondrial apoptosis and mitophagy in grass carp hepatocytes through NF-kappaB/JNK pathway.
Collapse
Affiliation(s)
- Zhiruo Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Zhiying Miao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shengchen Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Hao Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
13
|
Wang J, Shi K, Li S, Chen L, Liu W, Wu X, Shen Y, Sun Y, Cheng J, Wu X, Xu Q. Meisoindigo attenuates dextran sulfate sodium-induced experimental colitis via its inhibition of TAK1 in macrophages. Int Immunopharmacol 2021; 101:108239. [PMID: 34653728 DOI: 10.1016/j.intimp.2021.108239] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022]
Abstract
At present, inflammatory bowel disease (IBD) seriously threatens human health, and its treatment is a huge challenge for people. In our studies, we found that meisoindigo, a derivative of indirubin, significantly ameliorated dextran sulfate sodium (DSS)-induced experimental colitis in mice. Meisoindigo treatment markedly elevated the level of glutathione, while suppressed the activities of alkaline phosphatase and myeloperoxidase in colonic tissues. Moreover, the mRNA expression of vascular cell adhesion molecule 1, intercellular adhesion molecule 1, cyclooxygenase-2 which are important colitis-related molecules and the levels of the inflammatory cytokines interleukin (IL)-18, IL-1β, IL-6, tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS) were suppressed dose-dependently following treatment with meisoindigo. Immunofluorescence results indicated that meisoindigo inhibited macrophage infiltration and nuclear factor (NF)-κB activation in colons from DSS-treated mice. Therefore, mouse RAW264.7 and human THP-1 cells were treated with lipopolysaccharide (LPS) alone or combined adenosine triphosphate to activate NF-κB pathway in vitro. It was shown that meisoindigo reduced the elevated levels of NO, IL-18, IL-1β and TNF-α after LPS treatment in both cells. In addition, meisoindigo showed inhibitory effects on NF-κB by using a luciferase reporter gene that depends on NF-κB. Through molecular docking, microscale thermophoresis and cellular thermal shift assay. It was further found that meisoindigo targeted transforming growth factor β activated kinase-1 (TAK1), which is an important regulator in the upstream of NF-κB pathway. In conclusion, our findings show that meisoindigo can alleviate IBD effectively at low doses, and negatively regulate proinflammatory responses by inhibiting the activation of TAK1, which provides new ideas for clinical anti-inflammatory therapy.
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Ke Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Shuaifei Li
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Lu Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China; Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Wentao Liu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Xudong Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yan Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | | | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
14
|
Mwaeni VK, Nyariki JN, Jillani N, Omwenga G, Ngugi M, Isaac AO. Coenzyme Q 10 protected against arsenite and enhanced the capacity of 2,3-dimercaptosuccinic acid to ameliorate arsenite-induced toxicity in mice. BMC Pharmacol Toxicol 2021; 22:19. [PMID: 33827703 PMCID: PMC8028750 DOI: 10.1186/s40360-021-00484-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/16/2021] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Arsenic poisoning affects millions of people. The inorganic forms of arsenic are more toxic. Treatment for arsenic poisoning relies on chelation of extracellularly circulating arsenic molecules by 2,3-dimecaptosuccinic acid (DMSA). As a pharmacological intervention, DMSA is unable to chelate arsenic molecules from intracellular spaces. The consequence is continued toxicity and cell damage in the presence of DMSA. A two-pronged approach that removes extracellular arsenic, while protecting from the intracellular arsenic would provide a better pharmacotherapeutic outcome. In this study, Coenzyme Q10 (CoQ10), which has been shown to protect from intracellular organic arsenic, was administered separately or with DMSA; following oral exposure to sodium meta-arsenite (NaAsO2) - a very toxic trivalent form of inorganic arsenic. The aim was to determine if CoQ10 alone or when co-administered with DMSA would nullify arsenite-induced toxicity in mice. METHODS Group one represented the control; the second group was treated with NaAsO2 (15 mg/kg) daily for 30 days, the third, fourth and fifth groups of mice were given NaAsO2 and treated with 200 mg/kg CoQ10 (30 days) and 50 mg/kg DMSA (5 days) either alone or in combination. RESULTS Administration of CoQ10 and DMSA resulted in protection from arsenic-induced suppression of RBCs, haematocrit and hemoglobin levels. CoQ10 and DMSA protected from arsenic-induced alteration of WBCs, basophils, neutrophils, monocytes, eosinophils and platelets. Arsenite-induced dyslipidemia was nullified by administration of CoQ10 alone or in combination with DMSA. Arsenite induced a drastic depletion of the liver and brain GSH; that was significantly blocked by CoQ10 and DMSA alone or in combination. Exposure to arsenite resulted in significant elevation of liver and kidney damage markers. The histological analysis of respective organs confirmed arsenic-induced organ damage, which was ameliorated by CoQ10 alone or when co-administered with DMSA. When administered alone, DMSA did not prevent arsenic-driven tissue damage. CONCLUSIONS Findings from this study demonstrate that CoQ10 and DMSA separately or in a combination, significantly protect against arsenic-driven toxicity in mice. It is evident that with further pre-clinical and clinical studies, an adjunct therapy that incorporates CoQ10 alongside DMSA may find applications in nullifying arsenic-driven toxicity.
Collapse
Affiliation(s)
- Victoria K Mwaeni
- Department of Biochemistry and Biotechnology, Technical University of Kenya, P. O. Box 52428, Nairobi, 00200, Kenya
| | - James N Nyariki
- Department of Biochemistry and Biotechnology, Technical University of Kenya, P. O. Box 52428, Nairobi, 00200, Kenya
| | - Ngalla Jillani
- Institute of Primate Research, P.O. Box 24481, Karen, Nairobi, 00502, Kenya
| | - George Omwenga
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, P.O. Box 43844-00100, Nairobi, Kenya
| | - Mathew Ngugi
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, P.O. Box 43844-00100, Nairobi, Kenya
| | - Alfred Orina Isaac
- Department of Pharmaceutical Sciences and Technology, Technical University of Kenya, P. O. Box 52428, Nairobi, 00200, Kenya.
| |
Collapse
|
15
|
Yang Y, Liu C, Xie T, Wang D, Chen X, Ma L, Zhang A. Role of inhibiting Chk1-p53 pathway in hepatotoxicity caused by chronic arsenic exposure from coal-burning. Hum Exp Toxicol 2021; 40:1141-1152. [PMID: 33501840 DOI: 10.1177/0960327120988880] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Arsenic is a naturally occurring environmental toxicant, chronic exposure to arsenic can cause multiorgan damage, except for typical skin lesions, liver damage is the main problem for health concern in population with arsenic poisoning. Abnormal apoptosis is closely related to liver-related diseases, and p53 is one of the important hallmark proteins in apoptosis progression. This study was to investigate whether arsenic poisoning-induced hepatocyte apoptosis and the underlying role of p53 signaling pathway. A rat model of arsenic poisoning was established by feeding corn powder for 90 days, which was baked with high arsenic coal, then were treated with Ginkgo biloba extract (GBE) for 45 days by gavage. The results showed that arsenic induced liver damage, increased hepatocyte apoptosis and elevated the expression level of Chk1 and the ratios of p-p53/p53 and Bax/Bcl-2 in liver tissues, which were significantly attenuated by GBE. Additionally, to further demonstrate the potential apoptosis-associated mechanism, L-02 cells were pre-incubated with p53 inhibitor pifithrin-α (PFTα), ataxia telangiectasia-mutated (ATM)/ataxia telangiectasia-mutated and Rad3-related (ATR) inhibitor (CGK733) or GBE, then treated with sodium arsenite (NaAsO2) for 24 h. The results showed that GBE, PFTα or CGK733 significantly reduced arsenic-induced Chk1 expression and the ratios of p-p53/p53 and Bax/Bcl-2. In conclusion, Chk1-p53 pathway was involved in arsenic poisoning-induced hepatotoxicity, and inhibiting of Chk1-p53 pathway ameliorated hepatocyte apoptosis caused by coal-burning arsenic poisoning. The study provides a pivotal clue for understanding of the mechanism of arsenic poisoning-induced liver damage, and possible intervention strategies.
Collapse
Affiliation(s)
- Yuan Yang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, 74628Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Chunyan Liu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, 74628Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Tingting Xie
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, 74628Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Dapeng Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, 74628Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Xiong Chen
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, 74628Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Lu Ma
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, 74628Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Aihua Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, 74628Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
16
|
Wang M, Tan J, Jiang C, Li S, Wu X, Ni G, He Y. Inorganic arsenic influences cell apoptosis by regulating the expression of MEG3 gene. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2021; 43:475-484. [PMID: 33033900 DOI: 10.1007/s10653-020-00740-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/27/2020] [Indexed: 06/11/2023]
Abstract
Arsenic is a wildly distributed carcinogen in the environment. Arsenic-induced apoptosis has been extensively studied in therapeutics and toxicology. LncRNA MEG3 has been extensively studied as apoptosis regulatory gene in recent years. However, it stays unclear regarding how the mechanism of MEG3 regulates arsenic-induced apoptosis. Our focus was to explore the effects of MEG3 on arsenic-induced apoptosis. MTS assay was used to test cell viability, and qRT-PCR was for the examination of gene expressions. The effect of the apoptosis and necrosis after knockdown MEG3 was detected with double staining. Our results demonstrated that MEG3 expression was positively correlated with the concentration of three arsenic species (inorganic arsenic (iAs), monomethylarsonic acid (MMA) and dimethylarsinic acid (DMA)) (p < 0.05). The ability of iAs to induce MEG3 expression was much higher compared with that induced by MMA and DMA. In addition, our experiments confirmed that MEG3 knockdown increased cell viability and arsenic-induced apoptosis, but cell viability decreased after iAs treatment. Moreover, LncRNA MEG3 regulated apoptosis via down-regulate API5 while up-regulate CASP7, CCND3 and APAF1. It is further proved that arsenic-induced apoptosis increased after the knockdown of MEG3, which regulates these genes. These findings provide experimental evidence and possible mechanisms for subsequent research on the effects of arsenic on health.
Collapse
Affiliation(s)
- Mengjie Wang
- School of Public Health, Kunming Medical University, No.1168 Chunrongxi Road Chenggong District, Kunming, Yunnan Province, China
| | - Jingwen Tan
- School of Public Health, Kunming Medical University, No.1168 Chunrongxi Road Chenggong District, Kunming, Yunnan Province, China
| | - Chenglan Jiang
- School of Public Health, Kunming Medical University, No.1168 Chunrongxi Road Chenggong District, Kunming, Yunnan Province, China
| | - Shuting Li
- School of Public Health, Kunming Medical University, No.1168 Chunrongxi Road Chenggong District, Kunming, Yunnan Province, China
| | - Xinan Wu
- School of Public Health, Kunming Medical University, No.1168 Chunrongxi Road Chenggong District, Kunming, Yunnan Province, China
| | - Guanghui Ni
- College of Pharmaceutic Science, Yunnan University of Chinese Medicine, No.1076 Yuhua Road Chenggong District, Kunming, Yunnan Province, China.
| | - Yuefeng He
- School of Public Health, Kunming Medical University, No.1168 Chunrongxi Road Chenggong District, Kunming, Yunnan Province, China.
| |
Collapse
|
17
|
Carrà G, Lingua MF, Maffeo B, Taulli R, Morotti A. P53 vs NF-κB: the role of nuclear factor-kappa B in the regulation of p53 activity and vice versa. Cell Mol Life Sci 2020; 77:4449-4458. [PMID: 32322927 PMCID: PMC11104960 DOI: 10.1007/s00018-020-03524-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/06/2020] [Accepted: 04/06/2020] [Indexed: 12/18/2022]
Abstract
The onco-suppressor p53 is a transcription factor that regulates a wide spectrum of genes involved in various cellular functions including apoptosis, cell cycle arrest, senescence, autophagy, DNA repair and angiogenesis. p53 and NF-κB generally have opposing effects in cancer cells. While p53 activity is associated with apoptosis induction, the stimulation of NF-κB has been demonstrated to promote resistance to programmed cell death. Although the transcription factor NF-κB family is considered as the master regulator of cancer development and maintenance, it has been mainly studied in relation to its ability to regulate p53. This has revealed the importance of the crosstalk between NF-κB, p53 and other crucial cell signaling pathways. This review analyzes the various mechanisms by which NF-κB regulates the activity of p53 and the role of p53 on NF-κB activity.
Collapse
Affiliation(s)
- Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy.
| | | | - Beatrice Maffeo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Riccardo Taulli
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy.
| |
Collapse
|
18
|
Zhang K, Hussain T, Wang J, Li M, Wang W, Ma X, Liao Y, Yao J, Song Y, Liang Z, Zhou X, Xu L. Sodium Butyrate Abrogates the Growth and Pathogenesis of Mycobacterium bovis via Regulation of Cathelicidin (LL37) Expression and NF-κB Signaling. Front Microbiol 2020; 11:433. [PMID: 32265874 PMCID: PMC7096352 DOI: 10.3389/fmicb.2020.00433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium bovis is the causative agent of bovine tuberculosis, has been identified a serious threat to human population. It has been found that sodium butyrate (NaB), the inhibitor of histone deacetylase, can promote the expression of cathelicidin (LL37) and help the body to resist a variety of injuries. In the current study, we investigate the therapeutic effect of NaB on the regulation of host defense mechanism against M. bovis infection. We found an increased expression of LL37 in M. bovis infected THP-1 cells after NaB treatment. In contrast, NaB treatment significantly down-regulated the expression of Class I HDAC in THP-1 cells infected with M. bovis. Additionally, NaB reduced the expression of phosphorylated P65 (p-P65) and p-IκBα, indicating the inhibition of nuclear factor-κB (NF-κB) signaling. Furthermore, we found that NaB treatment reduced the production of inflammatory cytokines (IL-1β, TNF-α, and IL-10) and a key anti-apoptotic marker protein Bcl-2 in THP-1 cell infected with M. bovis. Notably, mice showed high resistance to M. bovis infection after NaB treatment. The reduction of viable M. bovis bacilli indicates that NaB-induced inhibition of M. bovis infection mediated by upregulation of LL37 and inhibition of NF-κB signaling pathway. These observations illustrate that NaB mediate protective immune responses against M. bovis infection. Overall, these results suggest that NaB can be exploited as a therapeutic strategy for the control of M. bovis in animals and human beings.
Collapse
Affiliation(s)
- Kai Zhang
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Tariq Hussain
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.,College of Veterinary Sciences, The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Jie Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Mengying Li
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Wenjia Wang
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Xiaojing Ma
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Yi Liao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiao Yao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yinjuan Song
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhengmin Liang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lihua Xu
- School of Agriculture, Ningxia University, Yinchuan, China
| |
Collapse
|
19
|
Chen S, Tang Y, Yang C, Li K, Huang X, Cao J. Silencing CDC25A inhibits the proliferation of liver cancer cells by downregulating IL‑6 in vitro and in vivo. Int J Mol Med 2020; 45:743-752. [PMID: 31922225 PMCID: PMC7015122 DOI: 10.3892/ijmm.2020.4461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023] Open
Abstract
Cell division cycle 25A (CDC25A) is a core regulator of the cell cycle that has a dual‑specific phosphatase activity, which is closely associated with the occurrence and development of a tumor, and is overexpressed in liver cancer. However, the molecular mechanism of CDC25A in the development of liver cancer remains unclear. The purpose of the present study was to further investigate the effect of CDC25A on cell proliferation in vitro and in vivo and to investigate whether an interaction exists between CDC25A and interleukin (IL)‑6 in liver cancer. An Affymetrix human gene expression profiling chip screened differentially expressed genes in HepG2 cells with silenced CDC25A and the IL‑6 signaling pathway was revealed to be significantly inhibited (P<0.05). In the present study, the effects of CDC25A on cell proliferation and migration were analyzed using cell cycle, MTT and Transwell assays. Reverse transcription‑quantitative PCR, western blot and immunohistochemistry analyses confirmed that silencing the CDC25A gene downregulated the expression of IL‑6 in HepG2 cells and the mRNA and protein expression of IL‑1β, mitogen‑activated protein kinase kinase kinase 14 (NIK) and nuclear factor‑κB (NF‑κB), which are regulatory molecules upstream of IL‑6. In addition, silencing CDC25A by short hairpin RNA inhibited the development of liver cancer xenograft tumor types in nude mice, and decreased the expression of IL‑1β, NIK, NF‑κB and IL‑6 in xenograft tumor types. In conclusion, silencing CDC25A significantly inhibited the proliferation of liver cancer cells in vitro and in vivo, potentially via an interaction with IL‑6 through the downregulation of the IL‑1β/NIK/NF‑κB signaling axis.
Collapse
Affiliation(s)
- Si Chen
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yanping Tang
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chun Yang
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Kezhi Li
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoqing Huang
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ji Cao
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
20
|
Ouyang Z, Tan T, Liu C, Duan J, Wang W, Guo X, Zhang Q, Li Z, Huang Q, Dou P, Liu T. Targeted delivery of hesperetin to cartilage attenuates osteoarthritis by bimodal imaging with Gd 2(CO 3) 3@PDA nanoparticles via TLR-2/NF-κB/Akt signaling. Biomaterials 2019; 205:50-63. [PMID: 30903825 DOI: 10.1016/j.biomaterials.2019.03.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/16/2019] [Accepted: 03/14/2019] [Indexed: 02/05/2023]
Abstract
The progressive degeneration of cartilage marks the advancement of osteoarthritis (OA), which requires specific targeted treatment for effective cartilage repair. However, there is still no efficient cartilage delivery system or novel magnetic resonance (MR) contrast agent (CA). Herein, we report the synthesis of a novel class of MR CA, Gd2(CO3)3-based nanoparticles (NPs), from a simpler and "greener" approach than previous ones. After the coating of polydopamine (PDA) onto the Gd2(CO3)3 core, we further anchored a cartilage-targeting peptide and loaded hesperetin (Hes) into NPs (Hes-Gd2(CO3)3@PDA-PEG-DWpeptide, HGdPDW), showing excellent cartilage affinity and MR suitability. Additionally, the synthesized HGdPDW exerted significant protective effects against IL-1β stimulation, as shown by the decreased apoptosis and inflammation and increased maturation of chondrocytes in vitro. More importantly, RNA-seq analyses showed the significant reduction of TLR-2 in IL-1β-treated chondrocytes, and this reduction was followed by the inactivation of NF-κB/Akt signaling, leading to the protective effect of HGdPDW. By the establishment of anterior cruciate ligament transection (ACLT) OA mice, the bimodal MRI/IVIS imaging demonstrated the effective cartilage-binding ability of HGdPDW in OA knees with low cytotoxicity, which alleviated the gradual degeneration of articular cartilage in vivo by inhibiting TLR-2 in chondrocytes. Taken together, these results suggest that HGdPDW could target cartilage effectively, thereby protecting chondrocytes from apoptosis and inflammation via TLR-2/NF-κB/Akt signaling. We hope this new class of MRI CA could be applied in not only other fields using MRI technology but also the treatment of general cartilage-related diseases; this application will undoubtedly extend the treatment of OA clinically.
Collapse
Affiliation(s)
- Zhengxiao Ouyang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China
| | - Tingting Tan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Department of Metabolism & Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Chunfeng Liu
- Department of Orthopedics, Suzhou Kowloon Hospital Affiliated to School of Medicine, Shanghai Jiao Tong University, Suzhou, 215021, PR China
| | - Juan Duan
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Wanchun Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Xiaoning Guo
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Qing Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Qianli Huang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China
| | - Pengcheng Dou
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China.
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China.
| |
Collapse
|