1
|
Hou K, Liu L, Fang ZH, Zong WX, Sun D, Guo Z, Cao L. The role of ferroptosis in cardio-oncology. Arch Toxicol 2024; 98:709-734. [PMID: 38182913 DOI: 10.1007/s00204-023-03665-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024]
Abstract
With the rapid development of new generations of antitumor therapies, the average survival time of cancer patients is expected to be continuously prolonged. However, these therapies often lead to cardiotoxicity, resulting in a growing number of tumor survivors with cardiovascular disease. Therefore, a new interdisciplinary subspecialty called "cardio-oncology" has emerged, aiming to detect and treat cardiovascular diseases associated with tumors and antitumor therapies. Recent studies have highlighted the role of ferroptosis in both cardiovascular and neoplastic diseases. The balance between intracellular oxidative stress and antioxidant defense is crucial in regulating ferroptosis. Tumor cells can evade ferroptosis by upregulating multiple antioxidant defense pathways, while many antitumor therapies rely on downregulating antioxidant defense and promoting ferroptosis in cancer cells. Unfortunately, these ferroptosis-inducing antitumor therapies often lack tissue specificity and can also cause injury to the heart, resulting in ferroptosis-induced cardiotoxicity. A range of cardioprotective agents exert cardioprotective effects by inhibiting ferroptosis. However, these cardioprotective agents might diminish the efficacy of antitumor treatment due to their antiferroptotic effects. Most current research on ferroptosis only focuses on either tumor treatment or heart protection but rarely considers both in concert. Therefore, further research is needed to study how to protect the heart during antitumor therapies by regulating ferroptosis. In this review, we summarized the role of ferroptosis in the treatment of neoplastic diseases and cardiovascular diseases and also attempted to propose further research directions for ferroptosis in the field of cardio-oncology.
Collapse
Affiliation(s)
- Kai Hou
- Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Chest Hospital, Tianjin, 300222, China.
- Chest Hospital, Tianjin University, Tianjin, 300222, China.
- Pu'er People's Hospital, Yunnan, 665000, China.
| | - Lin Liu
- Institute of Natural Sciences, MOE-LSC, School of Mathematical Sciences, CMA-Shanghai, SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | - Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, 08854, USA
| | - Daqiang Sun
- Tianjin Medical University, Tianjin, 300070, China
- Tianjin Chest Hospital, Tianjin, 300222, China
- Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Zhigang Guo
- Tianjin Medical University, Tianjin, 300070, China
- Tianjin Chest Hospital, Tianjin, 300222, China
- Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Lu Cao
- Tianjin Chest Hospital, Tianjin, 300222, China.
- Chest Hospital, Tianjin University, Tianjin, 300222, China.
| |
Collapse
|
2
|
Ding Q, Pi A, Hao L, Xu T, Zhu Q, Shu L, Yu X, Wang W, Si C, Li S. Genistein Protects against Acetaldehyde-Induced Oxidative Stress and Hepatocyte Injury in Chronic Alcohol-Fed Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1930-1943. [PMID: 36653166 DOI: 10.1021/acs.jafc.2c05747] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Alcohol-related liver disease (ALD) is one of the most prevalent forms of liver disease in the world. Acetaldehyde, an intermediate product of alcohol catabolism, is a cause of liver injury caused by alcohol. This study was designed to evaluate the protective role and mechanism(s) of genistein against acetaldehyde-induced liver injury in the pathological process of ALD. We found that genistein administration significantly ameliorated alcohol-induced hepatic steatosis, injury, and inflammation in mice. Genistein supplementation markedly reversed hepatic oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, and hepatocellular apoptosis in both alcohol-fed mice liver and acetaldehyde-treated hepatocytes. The mechanistic experiments revealed that the restoration of genistein administration rescued heme oxygenase-1 (HO-1) reduction at both transcriptional and protein levels in either alcohol-fed mice liver or acetaldehyde-treated hepatocytes, and the beneficial aspects derived from genistein were abolished in antioxidase heme oxygenase-1 (HO-1)-deficient hepatocytes. Moreover, we confirmed that genistein administration-restored hepatic nuclear factor erythroid 2-related factor 2 (NRF2), a key transcriptional regulator of HO-1, was involved in the protective role of genistein in ALD. This study demonstrated that genistein ameliorated acetaldehyde-induced oxidative stress and liver injury by restoring the hepatic NRF2-HO-1 signaling pathway in response to chronic alcohol consumption. Therefore, genistein may serve as a potential therapeutic choice for the treatment of ALD.
Collapse
Affiliation(s)
- Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
- College of Animal Science, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Aiwen Pi
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
| | - Liuyi Hao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
| | - Tiantian Xu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
| | - Qin Zhu
- Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, Zhejiang, P. R. China
| | - Long Shu
- Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, Zhejiang, P. R. China
| | - Xiaolong Yu
- Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, Zhejiang, P. R. China
| | - Weiguang Wang
- Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, Zhejiang, P. R. China
| | - Caijuan Si
- Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, Zhejiang, P. R. China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
- Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou 310013, Zhejiang, P. R. China
| |
Collapse
|
3
|
Song L, Wang J, Gong M, Zhang Y, Li Y, Wu X, Qin L, Duan Y. Detoxification technology and mechanism of processing with Angelicae sinensis radix in reducing the hepatotoxicity induced by rhizoma Dioscoreae bulbiferae in vivo. Front Pharmacol 2022; 13:984858. [PMID: 36249801 PMCID: PMC9554241 DOI: 10.3389/fphar.2022.984858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022] Open
Abstract
Rhizoma Dioscoreae Bulbiferae (RDB) was effective on relieving cough and expectorant but accompanied by severe toxicity, especially in hepatotoxicity. A previous study found that processing with Angelicae Sinensis Radix (ASR) reduced RDB-induced hepatotoxicity. However, up to now, the optimized processing process of ASR-processed RDB has not been explored or optimized, and the detoxification mechanism is still unknown. This study evaluated the detoxification technology and possible mechanism of processing with ASR on RDB-induced hepatotoxicity. The optimized processing process of ASR-processed RDB was optimized by the content of diosbulbin B (DB), the levels of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and histopathological analysis. The processing detoxification mechanism was evaluated by detecting the antioxidant levels of nuclear factor E2 related factor 2 (Nrf2) and its downstream heme oxygenase 1 (HO-1), quinone oxidoreductase 1 (NQO1), glutamylcysteine ligase catalytic subunit (GCLM), and the levels of downstream antioxidant factors of Nrf2. Besides, the antitussive and expectorant efficacy of RDB was also investigated. This work found that processing with ASR attenuated RDB-induced hepatotoxicity, which can be verified by reducing the levels of ALT, AST, and ALP, and reversing the pathological changes of liver histomorphology. And the optimized processing process of ASR-processed RDB is “processing at a mass ratio of 100:20 (RDB:ASR) and a temperature of 140°C for 10 min.” Further results corroborated that the intervention of processed products of ASR-processed RDB remarkably upregulated the Nrf2/HO-1/NQO1/GCLM protein expression levels in liver, and conserved antitussive and expectorant efficacy of RDB. The above findings comprehensively indicated that the optimized processing process of ASR-processed RDB was “processing at a mass ratio of 100:20 (RDB:ASR) and a temperature of 140°C for 10 min,” and the processing detoxification mechanism involved enhancing the level of Nrf2-mediated antioxidant defense in liver as a key target organ.
Collapse
Affiliation(s)
- Lingling Song
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Junming Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P. R. China, Henan University of Chinese Medicine, Zhengzhou, China
- *Correspondence: Junming Wang,
| | - Mingzhu Gong
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yueyue Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yamin Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaohui Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lingyu Qin
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yaqian Duan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
4
|
Kim HY, Ahn SB, Hong JM, Oh JH, Saeed WK, Kim GS, Kim H, Kang JK, Kang S, Jun DW. BTT-105 ameliorates hepatic fibrosis in non-alcoholic fatty liver animal model. FASEB J 2021; 35:e21979. [PMID: 34694029 DOI: 10.1096/fj.202002656rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 09/08/2021] [Accepted: 09/22/2021] [Indexed: 12/29/2022]
Abstract
BTT-105 (1-O-hexyl-2,3,5-trimethylhydroquinone), a hydroquinone derivative, is a potent anti-oxidant that was safe and tolerable in phase I clinical trial. This study examined the anti-fibrotic effect of BTT-105 in a mouse model of non-alcoholic fatty liver disease (NAFLD) along with the underlying mechanisms. In vivo, efficacy of BTT-105 evaluated from three kinds of NAFLD models (methionine/choline deficient diet (MCD), high fat diet (HF) and western diet (WD)). Metabolomics and transcriptomics profiling analysis in liver tissues were conducted. In vitro, anti-fibrotic effect of BTT-105 assessed in human hepatic stellated cells (HSCs) and primary mouse HSCs. BTT-105 improved NAFLD activity score in three kinds of NAFLD animal models (MCD, HF, and WD). BTT-105 also decreased levels of hepatic pro-collagen and collagen fibers deposition in liver tissue. Metabolome and transcriptome analysis revealed that BTT-105 decreased lipid metabolites and increased antioxidants in NAFLD mice. In HepG2 cells, BTT-105 enhanced Nrf2-ARE reporter activity in a dose-dependent manner and increased the levels of antioxidant gene expression. BTT-105 showed inhibition of HSCs activation and migration. Gene expression profiling and protein expression showed that BTT-105 increased Nrf2 activation as well as decreased PI3K-Akt pathway in activated HSCs. BTT-105 attenuated ameliorates steatohepatitis and hepatic fibrosis.
Collapse
Affiliation(s)
- Hye Young Kim
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Sang Bong Ahn
- Department of Internal Medicine, Eulji University School of Medicine, Seoul, Republic of Korea
| | | | - Ju Hee Oh
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Waqar Khalid Saeed
- Department of Internal Medicine, Hanyang University School of Medicine, Seoul, Republic of Korea
| | - Gyu Sik Kim
- Biotoxtech Co., Ltd., Cheongju, Republic of Korea
| | - Hyun Kim
- Biotoxtech Co., Ltd., Cheongju, Republic of Korea
| | | | - Sukmo Kang
- Biotoxtech Co., Ltd., Cheongju, Republic of Korea
| | - Dae Won Jun
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea.,Department of Internal Medicine, Hanyang University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
5
|
Tao J, Krutsenko Y, Moghe A, Singh S, Poddar M, Bell A, Oertel M, Singhi AD, Geller D, Chen X, Lujambio A, Liu S, Monga SP. Nuclear factor erythroid 2-related factor 2 and β-Catenin Coactivation in Hepatocellular Cancer: Biological and Therapeutic Implications. Hepatology 2021; 74:741-759. [PMID: 33529367 PMCID: PMC8326305 DOI: 10.1002/hep.31730] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/08/2020] [Accepted: 01/03/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS HCC remains a major unmet clinical need. Although activating catenin beta-1 (CTNNB1) mutations are observed in prominent subsets of HCC cases, these by themselves are insufficient for hepatocarcinogenesis. Coexpression of mutant CTNNB1 with clinically relevant co-occurrence has yielded HCCs. Here, we identify cooperation between β-catenin and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling in HCC. APPROACH AND RESULTS Public HCC data sets were assessed for concomitant presence of CTNNB1 mutations and either mutations in nuclear factor erythroid-2-related factor-2 (NFE2L2) or Kelch like-ECH-associated protein 1 (KEAP1), or Nrf2 activation by gene signature. HCC development in mice and similarity to human HCC subsets was assessed following coexpression of T41A-CTNNB1 with either wild-type (WT)-, G31A-, or T80K-NFE2L2. Based on mammalian target of rapamycin complex 1 activation in CTNNB1-mutated HCCs, response of preclinical HCC to mammalian target of rapamycin (mTOR) inhibitor was investigated. Overall, 9% of HCC cases showed concomitant CTNNB1 mutations and Nrf2 activation, subsets of which were attributable to mutations in NFE2L2/KEAP1. Coexpression of mutated CTNNB1 with mutant NFE2L2, but not WT-NFE2L2, led to HCC development and mortality by 12-14 weeks. These HCCs were positive for β-catenin targets, like glutamine synthetase and cyclin-D1, and Nrf2 targets, like NAD(P)H quinone dehydrogenase 1 and peroxiredoxin 1. RNA-sequencing and pathway analysis showed high concordance of preclinical HCC to human HCC subset showing activation of unique (iron homeostasis and glioblastoma multiforme signaling) and expected (glutamine metabolism) pathways. NFE2L2-CTNNB1 HCC mice were treated with mTOR inhibitor everolimus (5-mg/kg diet ad libitum), which led to >50% decrease in tumor burden. CONCLUSIONS Coactivation of β-catenin and Nrf2 is evident in 9% of all human HCCs. Coexpression of mutant NFE2L2 and mutant CTNNB1 led to clinically relevant HCC development in mice, which responded to mTOR inhibitors. Thus, this model has both biological and therapeutic implications.
Collapse
Affiliation(s)
- Junyan Tao
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Yekaterina Krutsenko
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Akshata Moghe
- Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Department of MedicineUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Sucha Singh
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Minakshi Poddar
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Aaron Bell
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Michael Oertel
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Aatur D Singhi
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - David Geller
- Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Department of SurgeryUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver CenterUniversity CaliforniaSan FranciscoCA
| | - Amaia Lujambio
- Department of Oncological SciencesTisch Cancer InstitutePrecision Immunology Institute, and Liver Cancer ProgramIcahn School of Medicine at Mount SinaiNew YorkNY
| | - Silvia Liu
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Satdarshan P Monga
- Department of PathologyUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Pittsburgh Liver Research CenterUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA.,Department of MedicineUniversity of PittsburghSchool of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| |
Collapse
|
6
|
1-O-Hexyl-2,3,5-Trimethylhydroquinone Ameliorates the Development of Preeclampsia through Suppression of Oxidative Stress and Endothelial Cell Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8839394. [PMID: 33542786 PMCID: PMC7840260 DOI: 10.1155/2021/8839394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/26/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
1-O-Hexyl-2,3,5-trimethylhydroquinone (HTHQ), a potent nuclear factor-E2-related factor 2 (Nrf2) activator, has potent antioxidant activity by scavenging reactive oxygen species (ROS). However, the role of HTHQ on the development of preeclampsia (PE) and the underlying mechanisms have barely been explored. In the present study, PE model was induced by adenovirus-mediated overexpression of soluble fms-like tyrosine kinase 1 (sFlt-1) in pregnant mice. The results showed that HTHQ treatment significantly relieved the high systolic blood pressure (SBP) and proteinuria and increased the fetal weight and fetal weight/placenta weight in preeclamptic mice. Furthermore, we found that HTHQ treatment significantly decreased soluble endoglin (sEng), endothelin-1 (ET-1), and activin A and restored vascular endothelial growth factor (VEGF) in preeclamptic mice. In addition, HTHQ treatment inhibited oxidative stress and endothelial cell apoptosis by increasing the levels of Nrf2 and its downstream haemoxygenase-1 (HO-1) protein. In line with the data in vivo, we discovered that HTHQ treatment attenuated oxidative stress and cell apoptosis in human umbilical vein endothelial cells (HUVECs) following hypoxia and reperfusion (H/R), and the HTHQ-mediated protection was lost after transfected with siNrf2. In conclusion, these results suggested that HTHQ ameliorates the development of preeclampsia through suppression of oxidative stress and endothelial cell apoptosis.
Collapse
|
7
|
Wang J, Lin Z, Yang Z, Liu X. lncRNA Eif4g2 improves palmitate-induced dysfunction of mouse β-cells via modulation of Nrf2 activation. Exp Cell Res 2020; 396:112291. [PMID: 32956705 DOI: 10.1016/j.yexcr.2020.112291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
Chronic oxidative stress resulting from hyperlipidemia is thought to be a key pathogenic driver of pancreatic β-cell dysfunction in leading to the onset of type 2 diabetes mellitus (T2DM). Long non-coding RNAs (lncRNAs) have been increasingly recognized to regulate dysfunction within pancreatic β-cells in the context of T2DM. In the present study, we sought to comprehensively analyze the roles of lncRNAs in dysfunctional β-cells and mouse islets. Analyses of INS-1E cells were performed by RNA-seq and qRT-PCR after treating with or without 0.5 mM palmitate for 4 days, leading us to identify the novel lncRNA Eif4g2 (lncEif4g2) as a functional regulator within these cells. When we overexpressed lncEif4g2 in INS-1E β-cells and mouse islets, this was sufficient for the reversal of palmitate-mediated reductions in cell viability, insulin production, ATP production by mitochondria, and creation of intracellular reactive oxygen species (ROS) and the dysfunction of mouse islets, with nuclear factor erythroid 2 related factor 2 (Nrf2) activation also being observed. In contrast, when lncEif4g2 was knocked down this led INS-1E cells and mouse islets to become more sensitive to palmitate-induced dysfunction, with reduced Nrf2 nuclear translocation also being detected. When antioxidants were used to treat INS-1E cells and mouse islets, however, these negative effects were reversed. Additional functional analyses revealed lncEif4g2 to be capable of directly binding to miR-3074-5p in β-cells, with the expression of lncEif4g2 and miR-3074-5p being negatively correlated with one another. We further found that cAMP-responsive element binding-protein (CREB) was a miR-3074-5p target gene in these cells, thus at least in part serving as a functional mediator of the lncEif4g2/miR-3074-5p axis within dysfunctional β-cells. In summary, our results thus reveal that lncEif4g2 is able to indirectly regulate the expression of CREB via targeting miR-3074-5p in INS-1E cells and mouse islets, thereby leading to enhanced Nrf2 activation.
Collapse
Affiliation(s)
- Jing Wang
- Department of Endocrinology, The 1st Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Zijing Lin
- Department of Endocrinology, The 1st Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zhuowen Yang
- Department of Gerontology, The 1st Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xiaomin Liu
- Department of Endocrinology, The 1st Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
8
|
Zhou Y, Jin Y, Yu H, Shan A, Shen J, Zhou C, Zhao Y, Fang H, Wang X, Wang J, Fu Y, Wang R, Li R, Zhang J. Resveratrol inhibits aflatoxin B1-induced oxidative stress and apoptosis in bovine mammary epithelial cells and is involved the Nrf2 signaling pathway. Toxicon 2019; 164:10-15. [PMID: 30946912 DOI: 10.1016/j.toxicon.2019.03.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/12/2019] [Accepted: 03/31/2019] [Indexed: 12/22/2022]
Abstract
Aflatoxins are widely occurring food contaminants that are particularly harmful to dairy products and cows. The plant polyphenol resveratrol has been reported to have a good effect on increasing the resistance of cells toward toxins. Therefore, we measured the effects of aflatoxin B1 and resveratrol on the viability of the MAC-T cow mammary epithelial cell line. The appropriate treatment concentrations were assayed (12.81 μM aflatoxin B1 and 43.81 μM resveratrol) to verify the protective effect of resveratrol toward mammary epithelial cells. The results showed that resveratrol alleviates aflatoxin B1-induced cytotoxicity, including the increase in ROS and the decrease in mitochondrial membrane potential (MMP) and apoptosis in MAC-T cells. The expression of mRNA transcripts (including Nrf2, Keap1, NQO1, HO-1, SOD2 and HSP70) for components of the Nrf2 signaling pathway was evaluated by real-time fluorescent quantitative PCR, with resveratrol also exhibiting a good regulatory effect. Thus, resveratrol was shown to have an ameliorating effect on aflatoxin toxicity in MAC-T cells.
Collapse
Affiliation(s)
- Yongfeng Zhou
- College of Animal Science, Jilin University, Changchun, China
| | - Yongcheng Jin
- College of Animal Science, Jilin University, Changchun, China
| | - Hao Yu
- College of Animal Science, Jilin University, Changchun, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Jinglin Shen
- College of Animal Science, Jilin University, Changchun, China
| | - Changhai Zhou
- College of Animal Science, Jilin University, Changchun, China
| | - Yun Zhao
- College of Animal Science, Jilin University, Changchun, China
| | - Hengtong Fang
- College of Animal Science, Jilin University, Changchun, China
| | - Xin Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Junmei Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Yurong Fu
- College of Animal Science, Jilin University, Changchun, China
| | - Rui Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Ruihua Li
- College of Animal Science, Jilin University, Changchun, China
| | - Jing Zhang
- College of Animal Science, Jilin University, Changchun, China.
| |
Collapse
|
9
|
Kim J, Shin SH, Kang JK, Kim JW. HX-1171 attenuates pancreatic β-cell apoptosis and hyperglycemia-mediated oxidative stress via Nrf2 activation in streptozotocin-induced diabetic model. Oncotarget 2018; 9:24260-24271. [PMID: 29849938 PMCID: PMC5966269 DOI: 10.18632/oncotarget.24916] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/04/2017] [Indexed: 12/20/2022] Open
Abstract
Streptozotocin (STZ) acts specifically on pancreatic beta cells, inducing cell destruction and cell dysfunction, resulting in diabetes. Many studies have reported that nuclear factor-erythroid 2-related factor 2 (Nrf2), a main regulator of antioxidant expression, prevents and improves diabetes-related diseases. In this study, we investigated the antidiabetic effect of the newly discovered Nrf2 activator, HX-1171, in the STZ-induced diabetic mouse model. HX-1171 enhanced insulin secretion by reducing STZ-induced cell apoptosis, and decreased intracellular reactive oxygen species (ROS) generation by upregulating the expression of antioxidant enzymes through Nrf2 activation in INS-1 pancreatic beta cells. In STZ-induced diabetic mice, HX-1171 administration significantly lowered blood glucose levels and restored blood insulin levels. In the STZ-only injected mice, the pancreatic islets showed morphological changes and loss of function, whereas the HX-1171-treated group was similar to that of the control group. These results suggest that HX-1171 may be developed as a promising therapeutic agent for diabetes-related diseases.
Collapse
Affiliation(s)
- Jimin Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, Republic of Korea
| | - Su-Hyun Shin
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, Republic of Korea
| | - Jong-Koo Kang
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Jae Wha Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
10
|
Kirby RJ, Divlianska DB, Whig K, Bryan N, Morfa CJ, Koo A, Nguyen KH, Maloney P, Peddibhotla S, Sessions EH, Hershberger PM, Smith LH, Malany S. Discovery of Novel Small-Molecule Inducers of Heme Oxygenase-1 That Protect Human iPSC-Derived Cardiomyocytes from Oxidative Stress. J Pharmacol Exp Ther 2017; 364:87-96. [PMID: 29101218 DOI: 10.1124/jpet.117.243717] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/31/2017] [Indexed: 01/09/2023] Open
Abstract
Oxidative injury to cardiomyocytes plays a critical role in cardiac pathogenesis following myocardial infarction. Transplantation of stem cell-derived cardiomyocytes has recently progressed as a novel treatment to repair damaged cardiac tissue but its efficacy has been limited by poor survival of transplanted cells owing to oxidative stress in the post-transplantation environment. Identification of small molecules that activate cardioprotective pathways to prevent oxidative damage and increase survival of stem cells post-transplantation is therefore of great interest for improving the efficacy of stem cell therapies. This report describes a chemical biology phenotypic screening approach to identify and validate small molecules that protect human-induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) from oxidative stress. A luminescence-based high-throughput assay for cell viability was used to screen a diverse collection of 48,640 small molecules for protection of hiPSC-CMs from peroxide-induced cell death. Cardioprotective activity of "hit" compounds was confirmed using impedance-based detection of cardiomyocyte monolayer integrity and contractile function. Structure-activity relationship studies led to the identification of a potent class of compounds with 4-(pyridine-2-yl)thiazole scaffold. Examination of gene expression in hiPSC-CMs revealed that the hit compound, designated cardioprotectant 312 (CP-312), induces robust upregulation of heme oxygenase-1, a marker of the antioxidant response network that has been strongly correlated with protection of cardiomyocytes from oxidative stress. CP-312 therefore represents a novel chemical scaffold identified by phenotypic high-throughput screening using hiPSC-CMs that activates the antioxidant defense response and may lead to improved pharmacological cardioprotective therapies.
Collapse
Affiliation(s)
- R Jason Kirby
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Daniela B Divlianska
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Kanupriya Whig
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Nadezda Bryan
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Camilo J Morfa
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Ada Koo
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Kevin H Nguyen
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Patrick Maloney
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Satayamaheshwar Peddibhotla
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - E Hampton Sessions
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Paul M Hershberger
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Layton H Smith
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Siobhan Malany
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| |
Collapse
|