1
|
Xia N, Xue H, Li Y, Liu J, Lou Y, Li S, Wang Y, Lu J, Chen X. Potential Mechanisms and Effects of Dai Bai Jie Ethanol Extract in Preventing Acute Alcoholic Liver Injury. Curr Issues Mol Biol 2024; 47:3. [PMID: 39852118 PMCID: PMC11763393 DOI: 10.3390/cimb47010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025] Open
Abstract
This study investigated the protective effect of Dai Bai Jie (DBJ) extract against acute alcoholic liver injury (AALI) and elucidated its potential mechanism. The total saponin level in the DBJ extracts was measured using vanillin-chloroform acid colorimetry. To observe the preventive and protective effects of DBJ on AML-12 cells in an ethanol environment, the effective components of DBJ were identified. An alcohol-induced AALI mouse model was used to evaluate the efficacy of DBJ against AALI. For this purpose, alcohol dehydrogenase (ADH) and acetaldehyde dehydrogenase (ALDH) levels were assessed, liver function indices and oxidative and inflammatory markers were determined, and histopathological examinations were performed. Mechanistic investigations were conducted using RT-qPCR assays and immunohistochemical analysis to determine the protective effects of DBJ. The samples (DBJ-1, DBJ-2, and DBJ-3) were obtained by extracting DBJ with water, 50% ethanol, and 95% ethanol, yielding total saponin contents of 5.35%, 6.64%, and 11.83%, respectively. DBJ-3 was isolated and purified, and its components were identified by Ultra Performance Liquid Chromatography-Mass Spectrometry (UPLC-MS). DBJ-3 had the greatest effect on cell viability in an ethanol environment. Moreover, DBJ-3 reduced inflammatory infiltration, liver cell degeneration, and hemorrhage, while increasing ADH and ALDH levels in liver tissues. Additionally, DBJ-3 considerably decreased the serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol (TC), and triglyceride (TG) levels. DBJ-3 reduced malondialdehyde (MDA), reactive oxygen species (ROS), and inflammatory factors, such as tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin 6 (IL-6), while increasing superoxide dismutase (SOD) and glutathione S-transferase (GST) activities. Furthermore, DBJ-3 significantly increased alcohol dehydrogenase 1b (ADH1B) and aldehyde dehydrogenase 2 (ALDH2) expression at the gene and protein levels within alcohol metabolism pathways and reduced the nuclear factor kappa-B (NF-κB) gene and protein levels. These findings suggest that DBJ-3 can prevent AALI by enhancing alcohol metabolism via the regulation of ADH1B and ALDH2 and the modulation of the NF-κB pathway to improve antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Niantong Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Hongwei Xue
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Yihang Li
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong 666100, China;
| | - Jia Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Yang Lou
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Shuyang Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Yutian Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Juan Lu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Xi Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong 666100, China;
| |
Collapse
|
2
|
Chen J, Chen X, Tang A, Wang Z, Cheong KL, Liu X, Zhong S. Chondroitin sulfate-functionalized selenium nanoparticle-induced S-phase cell cycle arrest and apoptosis in HeLa Cells. J Food Sci 2024; 89:4469-4479. [PMID: 38837700 DOI: 10.1111/1750-3841.17137] [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: 01/31/2024] [Revised: 04/09/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024]
Abstract
This study aimed to evaluate the anti-cervical cancer activity of chondroitin sulfate-functionalized selenium nanoparticles (SeCS) and to elucidate their action mechanism. Cytotoxic effect of SeCS on HeLa cells was assessed by MTT assay. Further molecular mechanism of SeCS was analyzed by flow cytometric assay and western blotting. The results showed that treatment with SeCS resulted in a dose- and time-dependent inhibition in the proliferation of HeLa cells. The data obtained from flow cytometry demonstrated that SeCS inhibited HeLa cell growth via the induction of S-phase arrest and cell apoptosis. Further mechanism analysis found that SeCS down-regulated expression levels of cyclin A and CDK2 and up-regulated p21 expression, which contributed to S arrest. Moreover, SeCS increased the level of Bax and decreased the expression of Bcl-2, resulting in the release of cytochrome C from mitochondria and activating caspase-3/8/9 for caspase-dependent apoptosis. Meanwhile, intracellular reactive oxygen species (ROS) levels were elevated after SeCS treatment, suggesting that ROS might be upstream of SeCS-induced S-phase arrest and cell apoptosis. These data show that SeCS has anti-tumor effects and possesses the potential to become a new therapeutic agent or adjuvant therapy for cancer patients. PRACTICAL APPLICATION: In our previous study, we used chondroitin sulfate to stabilize nano-selenium to obtain SeCS to improve the bioactivity and stability of nano-selenium. We found that it possessed an inhibitory effect on HeLa cells. However, the molecular mechanism remains unclear. This study elucidated the mechanism of SeCS damage to HeLa cells. SeCS has the potential to become a new therapeutic agent or adjuvant therapy for cancer patients.
Collapse
Affiliation(s)
- Jianping Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Xuehua Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Anqi Tang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Zhuo Wang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Kit-Leong Cheong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Xiaofei Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
3
|
Ishikawa C, Mori N. Pivotal role of dihydroorotate dehydrogenase as a therapeutic target in adult T-cell leukemia. Eur J Haematol 2024; 113:99-109. [PMID: 38558052 DOI: 10.1111/ejh.14209] [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: 11/17/2023] [Revised: 03/10/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES We aimed to determine the role of dihydroorotate dehydrogenase (DHODH) in pathogenesis of adult T-cell leukemia (ATL) caused by human T-cell leukemia virus type 1 (HTLV-1) and the effects of its inhibition on the de novo pyrimidine biosynthesis pathway. METHODS Cell proliferation, viability, cycle, and apoptosis were analyzed using WST-8 assays, flow cytometry, and Hoechst 33342 staining. To elucidate the molecular mechanisms involved in the anti-ATL effects of DHODH knockdown and inhibition, RT-PCR and immunoblotting were conducted. RESULTS HTLV-1-infected T-cell lines aberrantly expressed DHODH. Viral infection and the oncoprotein, Tax, enhanced DHODH expression, while knockdown of DHODH decreased HTLV-1-infected T-cell growth. In addition, BAY2402234, a DHODH inhibitor, exerted an anti-proliferative effect, which was reversed by uridine supplementation. BAY2402234 induced DNA damage and S phase arrest by downregulating c-Myc, CDK2, and cyclin A and upregulating p53 and cyclin E. It also induced caspase-mediated apoptosis by the upregulation of pro-apoptotic and downregulation of anti-apoptotic proteins. Furthermore, BAY2402234 induced caspase-independent ferroptosis and necroptosis. It decreased phosphorylation of IKK, IκBα, PTEN, Akt, and its downstream targets, suggesting that inhibition of NF-κB and Akt signaling is involved in its anti-ATL action. CONCLUSION These findings highlight DHODH as a potential therapeutic target for treating ATL.
Collapse
Affiliation(s)
- Chie Ishikawa
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Health Sciences, Transdisciplinary Research Organization for Subtropics and Island Studies, University of the Ryukyus, Nishihara, Japan
| | - Naoki Mori
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| |
Collapse
|
4
|
Ren YL, Lei JT, Zhang TR, Lu P, Cui DD, Yang B, Zhao GY, Peng F, Cao ZX, Peng C, Li YZ. Isobavachalcone, a natural sirtuin 2 inhibitor, exhibits anti-triple-negative breast cancer efficacy in vitro and in vivo. Phytother Res 2024; 38:1815-1829. [PMID: 38349045 DOI: 10.1002/ptr.8143] [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: 04/28/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 04/10/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and lethal clinical subtype and lacks effective targeted therapies at present. Isobavachalcone (IBC), the main active component of Psoralea corylifolia L., has potential anticancer effects. Herein, we identified IBC as a natural sirtuin 2 (SIRT2) inhibitor and characterized the potential mechanisms underlying the inhibition of TNBC. Molecular dynamics analysis, enzyme activity assay, and cellular thermal shift assay were performed to evaluate the combination of IBC and SIRT2. The therapeutic effects, mechanism, and safety of IBC were analyzed in vitro and in vivo using cellular and xenograft models. IBC effectively inhibited SIRT2 enzyme activity with an IC50 value of 0.84 ± 0.22 μM by forming hydrogen bonds with VAL233 and ALA135 within its catalytic domain. In the cellular environment, IBC bound to and stabilized SIRT2, consequently inhibiting cellular proliferation and migration, and inducing apoptosis and cell cycle arrest by disrupting the SIRT2/α-tubulin interaction and inhibiting the downstream Snail/MMP and STAT3/c-Myc pathways. In the in vivo model, 30 mg/kg IBC markedly inhibited tumor growth by targeting the SIRT2/α-tubulin interaction. Furthermore, IBC exerted its effects by inducing apoptosis in tumor tissues and was well-tolerated. IBC alleviated TNBC by targeting SIRT2 and triggering the reactive oxygen species ROS/β-catenin/CDK2 axis. It is a promising natural lead compound for future development of SIRT2-targeting drugs.
Collapse
Affiliation(s)
- Ya-Li Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie-Ting Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting-Rui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan-Dan Cui
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Yang
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pharmacy, The Seventh People's Hospital of Chengdu, Chengdu, China
- Department of Pharmacy, Panzhihua Central Hospital, Dali University, Panzhihua, China
| | - Gui-Ying Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhi-Xing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Zhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
5
|
Sun W, Lv J, Guo S, Lv M. Cellular microenvironment: a key for tuning mesenchymal stem cell senescence. Front Cell Dev Biol 2023; 11:1323678. [PMID: 38111850 PMCID: PMC10725964 DOI: 10.3389/fcell.2023.1323678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Mesenchymal stem cells (MSCs) possess the ability to self-renew and differentiate into multiple cell types, making them highly suitable for use as seed cells in tissue engineering. These can be derived from various sources and have been found to play crucial roles in several physiological processes, such as tissue repair, immune regulation, and intercellular communication. However, the limited capacity for cell proliferation and the secretion of senescence-associated secreted phenotypes (SASPs) pose challenges for the clinical application of MSCs. In this review, we provide a comprehensive summary of the senescence characteristics of MSCs and examine the different features of cellular microenvironments studied thus far. Additionally, we discuss the mechanisms by which cellular microenvironments regulate the senescence process of MSCs, offering insights into preserving their functionality and enhancing their effectiveness.
Collapse
Affiliation(s)
| | | | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengzhu Lv
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Li J, Guo D, Wang F, Tian M, Wang C, Guo X, Xu B, Wang Y. Functional analysis of AccCDK2-like and AccCINP-like genes in Apis cerana cerana under pesticide and heavy metal stress. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 195:105540. [PMID: 37666611 DOI: 10.1016/j.pestbp.2023.105540] [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: 05/04/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 09/06/2023]
Abstract
Heavy metals and pesticides represent prominent sources of pollution in the natural habitat of Apis cerana cerana, potentially endangering their health through the induction of oxidative stress reactions. This study aimed to address this issue by isolating AccCDK2-like and AccCINP-like proteins from Apis cerana cerana and investigating their functional roles in honey bee resistance against pesticide and heavy metal stresses. Bioinformatics analysis revealed significant homology of these proteins with those found in other species. Functional studies confirmed their participation in interaction with each other, alongside demonstrating distinct patterns of expression and localization. Specifically, AccCDK2-like exhibited higher expression levels in prepupae and muscle tissues, while AccCINP-like showed maximal expression in brown pupae and abdomen. Furthermore, the expression levels of these proteins were found to be modulated in response to pesticide and heavy metal stresses. Notably, overexpression of AccCDK2-like and AccCINP-like led to a noticeable alteration in E. coli's ability to withstand external stresses. Additionally, silencing of the AccCDK2-like and AccCINP-like genes resulted in a significant reduction in antioxidant enzyme activity and the expression levels of genes related to antioxidant function. Consequently, the mortality rate of Apis cerana cerana under pesticide and heavy metal stresses conspicuously increased. Hence, our findings suggest that AccCDK2-like and AccCINP-like proteins potentially play a crucial role in the response of Apis cerana cerana to pesticide and heavy metal stress, likely by modulating the antioxidant pathway.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Dezheng Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Ming Tian
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Chen Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Xingqi Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China
| | - Ying Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, People's Republic of China.
| |
Collapse
|
7
|
He YH, Pan JX, Xu LM, Gu T, Chen YW. Ductular reaction in non-alcoholic fatty liver disease: When Macbeth is perverted. World J Hepatol 2023; 15:725-740. [PMID: 37397935 PMCID: PMC10308290 DOI: 10.4254/wjh.v15.i6.725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/03/2023] [Accepted: 04/24/2023] [Indexed: 06/25/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) or metabolic (dysfunction)-associated fatty liver disease is the leading cause of chronic liver diseases defined as a disease spectrum comprising hepatic steatosis, non-alcoholic steatohepatitis (NASH), liver fibrosis, cirrhosis, and hepatic carcinoma. NASH, characterized by hepatocyte injury, steatosis, inflammation, and fibrosis, is associated with NAFLD prognosis. Ductular reaction (DR) is a common compensatory reaction associated with liver injury, which involves the hepatic progenitor cells (HPCs), hepatic stellate cells, myofibroblasts, inflammatory cells (such as macrophages), and their secreted substances. Recently, several studies have shown that the extent of DR parallels the stage of NASH and fibrosis. This review summarizes previous research on the correlation between DR and NASH, the potential interplay mechanism driving HPC differentiation, and NASH progression.
Collapse
Affiliation(s)
- Yang-Huan He
- Department of Gastroenterology and Department of Geriatrics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Jia-Xing Pan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Lei-Ming Xu
- Department of Gastroenterology, School of Medicine, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China
| | - Ting Gu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Yuan-Wen Chen
- Department of Gastroenterology and Department of Geriatrics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| |
Collapse
|
8
|
Tian CM, Yang MF, Xu HM, Zhu MZ, Zhang Y, Yao J, Wang LS, Liang YJ, Li DF. Mesenchymal Stem Cell-derived Exosomes: Novel Therapeutic Approach for Inflammatory Bowel Diseases. Stem Cells Int 2023; 2023:4245704. [PMID: 37056457 PMCID: PMC10089786 DOI: 10.1155/2023/4245704] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/19/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
As double membrane-encapsulated nanovesicles (30-150 nm), exosomes (Exos) shuttle between different cells to mediate intercellular communication and transport active cargoes of paracrine factors. The anti-inflammatory and immunomodulatory activities of mesenchymal stem cell (MSC)-derived Exos (MSC-Exos) provide a rationale for novel cell-free therapies for inflammatory bowel disease (IBD). Growing evidence has shown that MSC-Exos can be a potential candidate for treating IBD. In the present review, we summarized the most critical advances in the properties of MSC-Exos, provided the research progress of MSC-Exos in treating IBD, and discussed the molecular mechanisms underlying these effects. Collectively, MSC-Exos had great potential for cell-free therapy in IBD. However, further studies are required to understand the full dimensions of the complex Exo system and how to optimize its effects.
Collapse
Affiliation(s)
- Cheng-mei Tian
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Emergency, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 Guangdong, China
| | - Mei-feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen, Guangdong, China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Bellanti F, Mangieri D, di Bello G, Lo Buglio A, Pannone G, Pedicillo MC, Fersini A, Dobrakowski M, Kasperczyk A, Kasperczyk S, Vendemiale G. Redox-Dependent Modulation of Human Liver Progenitor Cell Line Fate. Int J Mol Sci 2023; 24:1934. [PMID: 36768260 PMCID: PMC9916526 DOI: 10.3390/ijms24031934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Redox homeostasis is determinant in the modulation of quiescence/self-renewal/differentiation of stem cell lines. The aim of this study consisted of defining the impact of redox modifications on cell fate in a human hepatic progenitor line. To achieve this, the HepaRG cell line, which shows oval ductular bipotent characteristics, was used. The impact of redox status on the balance between self-renewal and differentiation of HepaRG cells was investigated using different methodological approaches. A bioinformatic analysis initially proved that the trans-differentiation of HepaRG toward bipotent progenitors is associated with changes in redox metabolism. We then exposed confluent HepaRG (intermediate differentiation phase) to oxidized (H2O2) or reduced (N-acetylcysteine) extracellular environments, observing that oxidation promotes the acquisition of a mature HepaRG phenotype, while a reduced culture medium stimulates de-differentiation. These results were finally confirmed through pharmacological modulation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2), a principal modulator of the antioxidant response, in confluent HepaRG. NRF2 inhibition led to intracellular pro-oxidative status and HepaRG differentiation, while its activation was associated with low levels of reactive species and de-differentiation. In conclusion, this study shows that both intra- and extracellular redox balance are crucial in the determination of HepaRG fate. The impact of redox status in the differentiation potential of HepaRG cells is significant on the utilization of this cell line in pre-clinical studies.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Giorgia di Bello
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Aurelio Lo Buglio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | | | - Alberto Fersini
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Michał Dobrakowski
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Aleksandra Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Sławomir Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
10
|
Peng J, He Q, Li S, Liu T, Zhang J. Hydrogen-Rich Water Mitigates LPS-Induced Chronic Intestinal Inflammatory Response in Rats via Nrf-2 and NF-κB Signaling Pathways. Vet Sci 2022; 9:621. [PMID: 36356098 PMCID: PMC9692594 DOI: 10.3390/vetsci9110621] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/18/2022] [Accepted: 11/02/2022] [Indexed: 04/04/2024] Open
Abstract
Long-term exposure to low-dose lipopolysaccharide can impair intestinal barriers, causing intestinal inflammation and leading to systemic inflammation. Hydrogen-rich water possesses antioxidant and anti-inflammatory functions and exerts inhibitory effects on various inflammatory diseases. In this study, we investigated whether oral hydrogen-rich water could prevent lipopolysaccharide-induced chronic intestinal inflammation. An experimental model was established by feeding hydrogen-rich water, followed by the injection of lipopolysaccharide (200 μg/kg) in the tail vein of rats after seven months. ELISA, Western blot, immunohistochemistry, and other methods were used to detect related cytokines, proteins related to the NF-κB and Nrf-2 signaling pathways, and tight-junction proteins to study the anti-inflammatory and antioxidant effects of hydrogen-rich water. The obtained results show that hydrogen-rich water significantly increased the levels of superoxide dismutase and structural proteins; activated the Nrf-2 signaling pathway; downregulated the expression of inflammatory factors cyclooxygenase-2, myeloperoxidase, and ROS; and decreased the activation of the NF-κB signaling pathway. These results suggest that hydrogen-rich water could protect against chronic intestinal inflammation in rats caused by lipopolysaccharide-induced activation of the NF-κB signaling pathway by regulating the Nrf-2 signaling pathway.
Collapse
Affiliation(s)
- Jin Peng
- Heilongjiang Key Laboratory for Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Qi He
- Heilongjiang Key Laboratory for Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Shuaichen Li
- Institute for Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Tao Liu
- Heilongjiang Key Laboratory for Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| | - Jiantao Zhang
- Heilongjiang Key Laboratory for Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
| |
Collapse
|
11
|
Qi L, Wang Y, Su S, Wang M, Jablonska E, Jia Y, Wang R, Hao S, Feng C, Li G, Jiang M, Du L, Sun H, Li Q, Wang T. Sodium selenite inhibits cervical cancer growth via ROS mediated AMPK/FOXO3a /GADD45a axis. Chem Biol Interact 2022; 367:110171. [PMID: 36108716 DOI: 10.1016/j.cbi.2022.110171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/26/2022] [Accepted: 09/04/2022] [Indexed: 11/03/2022]
Abstract
Selenium is a trace element that has been shown to inhibit the growth of various cancer cell types. However, its role in cervical cancer and its underlying mechanisms remains largely unknown. Herein, we explored the anti-cervical cancer effect of selenium and its potential mechanisms through xenograft and in vitro experiments. HeLa cell xenografts in female nude mice showed tumor growth retardation, with no obvious liver and kidney toxicity, after being intraperitoneally injected with 3 mg/kg sodium selenite (SS) for 14 days. Compared to the control group, selenium levels in the tumor tissue increased significantly after SS treatment. In vitro experiments, SS inhibited the viability of HeLa and SiHa cells, blocked the cell cycle at the S phase, and enhanced apoptosis. RNA-sequencing, Kyoto encyclopedia of genes and genomes pathway analysis showed that forkhead box protein O (FOXO) was a key regulatory signaling pathway for SS to exhibit anticancer effects. Gene Ontology analysis filtered multiple terms associated with apoptosis, anti-proliferation, and cell cycle arrest. Further research revealed that SS increased intracellular reactive oxygen species (ROS) and impaired mitochondrial function, which activated adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) via phosphorylation at Thr172, resulting in activation of FOXO3a and its downstream growth arrest and DNA damage-inducible alpha (GADD45a). In summary, SS exhibited anti-cervical cancer effects, and their mechanisms may be that SS is involved in inducing cell cycle arrest and potentiating cell apoptosis caused by ROS-dependent activation of the AMPK/FOXO3a/GADD45a axis.
Collapse
Affiliation(s)
- Lei Qi
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Yuanyuan Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Shengqi Su
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Mingxing Wang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Ewa Jablonska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Sw. Teresy 8 Street, Lodz, 91-348, Poland
| | - Yuehui Jia
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Ruixiang Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Shuxiu Hao
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Chen Feng
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Guijin Li
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Meijing Jiang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Linlin Du
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Huixin Sun
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Qi Li
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Tong Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
12
|
Xun T, Lin Z, Zhang M, Mo L, Chen Y, Wang X, Zhao J, Ye C, Feng H, Yang X. Advanced oxidation protein products upregulate ABCB1 expression and activity via HDAC2-Foxo3α-mediated signaling in vitro and in vivo. Toxicol Appl Pharmacol 2022; 449:116140. [PMID: 35753429 DOI: 10.1016/j.taap.2022.116140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 11/28/2022]
Abstract
The unpredictable pharmacokinetics of non-renal cleared drugs in chronic kidney disease (CKD) patients is associated with the activity of drug transporters. However, the mechanisms underlying regulation of drug transporters are yet to be established. In this study, we demonstrated the involvement of a HDAC2-Foxo3α pathway in advanced oxidation protein products (AOPPs)-induced ATP-binding cassette subfamily B member 1 (ABCB1) expression and activity. The correlation of AOPPs accumulation with concentration of cyclosporine in plasma was evaluated in 194 patients with transplantation. Molecular changes in acetylation of various histones and related regulatory molecules were examined in HepG2 cell cultures treated with AOPPs. Accumulation of AOPPs in serum in relation to molecular changes in HDAC2-Foxo3α in vivo were evaluated in 5/6 nephrectomy (5/6 nx) and oral adenine (Adenine) CKD rat models. Interestingly, the cyclosporine level was negatively correlated with AOPPs in plasma. In addition, AOPPs markedly suppressed the expression of histone deacetylase 2 (HDAC2), inducing ABCB1 expression and activity in vitro and in vivo. Importantly, AOPPs modulated phosphorylation of Foxo3α and the upstream Akt protein. Our findings indicate that AOPPs regulate the expression and activity of ABCB1 via reducing HDAC2 expression and activating Foxo3α-dependent signaling. The collective results support the utility of AOPPs as a potential target for drug and/or dosage adjustment in CKD patients. Targeting of AOPPs presents a novel approach to regulate non-renal clearance.
Collapse
Affiliation(s)
- Tianrong Xun
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhufen Lin
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Mimi Zhang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Liqian Mo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Chen
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaokang Wang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jingqian Zhao
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chunxiao Ye
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Haixing Feng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixiao Yang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
13
|
Li C, Zhan Y, Zhang R, Tao Q, Lang Z, Zheng J. 20(S)- Protopanaxadiol suppresses hepatic stellate cell activation via WIF1 demethylation-mediated inactivation of the Wnt/β-catenin pathway. J Ginseng Res 2022. [DOI: 10.1016/j.jgr.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
14
|
Age-related accumulation of advanced oxidation protein products promotes osteoclastogenesis through disruption of redox homeostasis. Cell Death Dis 2021; 12:1160. [PMID: 34907153 PMCID: PMC8671415 DOI: 10.1038/s41419-021-04441-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/16/2021] [Accepted: 11/25/2021] [Indexed: 11/27/2022]
Abstract
Enhanced osteoclastogenesis is one of the major causes of age-related bone loss. Aging is accompanied by accumulation of advanced oxidation protein products (AOPPs). However, whether AOPPs accumulation contributing to the osteoclastogenesis with aging remains unclear. Here, we showed that AOPPs accumulation was associated with the enhanced osteoclastogenesis and deterioration of bone microstructure in aged mice. In vitro, AOPPs directly induced osteoclastogenesis by interaction with receptor activator of nuclear factor κ B (RANK) and the receptor for advanced glycation end products (RAGE) in the primary bone marrow monocytes. Bindings of AOPPs to RANK and RAGE were able to activate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, trigger generation of reactive oxygen species, then induce phosphorylation of mitogen-activated protein kinases and c-fos, upregulation of the nuclear factor of activated T cell c1, eventually induce bone marrow monocytes to differentiate into mature osteoclasts. Chronic exposure to AOPPs enhanced osteoclastogenesis and bone loss in mice, which could be alleviated by NADPH oxidase inhibitor apocynin. Local injection of AOPPs into subperiosteal area induced bone resorption at the site of administration, which was similar to the effect of RANK ligand. Together, these results suggested that AOPPs could serve as a novel regulator of osteoclastogenesis and AOPPs accumulation might play an important role in the development of age-related bone loss.
Collapse
|
15
|
A network pharmacology approach to investigate the anticancer mechanism of cinobufagin against hepatocellular carcinoma via downregulation of EGFR-CDK2 signaling. Toxicol Appl Pharmacol 2021; 431:115739. [PMID: 34619160 DOI: 10.1016/j.taap.2021.115739] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest cancers with high mortality and poor prognosis, and the investigation on new approaches and effective drugs for HCC therapy is of great significance. In our study, we demonstrate that treatment with cinobufagin, a natural compound isolated from traditional chinese medicine Chansu, reduces proliferation and the colony formation capacity of the human hepatoma cells in vitro, in addition, cinobufagin induces mitotic arrest in human hepatoma cells. The results of a network pharmacology-based analysis show that EGFR, MAPK1, PTK2, CDK2, MAPK3, ESR1, CDK1, PRKCA, AR, and CSNK2A1 are the key targets involved in the anti-tumor activities of cinobufagin, additionally, several signaling pathways such as proteoglycans in cancer, pathways in cancer, HIF-1 signaling pathway, VEGF signaling pathway, ErbB signaling pathway, and PI3K-AKT signaling pathway are identified as the potential pathways involved in the inhibitory effects of cinobufagin against HCC. Furthermore, at the molecular level, we find that cinobufagin decreases EGFR expression and CDK2 activity in human hepatoma cells. Inhibition of EGFR or CDK2 expression could not only suppress the growth of tumor cells but also enhance the inhibitory effects of cinobufagin on the proliferative potential of human hepatoma cells. We also demonstrate that EGFR positively regulates CDK2 expression. Furthermore, EGFR inhibitor gefitinib or CDK2 inhibitor CVT-313 synergistically enhances anticancer effects of cinobufagin in human hepatoma cells. Taken together, these findings indicate that cinobufagin may exert antitumor effects by suppressing EGFR-CDK2 signaling, and our study suggests that cinobufagin may be a novel, promising anticancer agent for the treatment of HCC.
Collapse
|
16
|
Advanced Oxidation Protein Products Induce G1/G0-Phase Arrest in Ovarian Granulosa Cells via the ROS-JNK/p38 MAPK-p21 Pathway in Premature Ovarian Insufficiency. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6634718. [PMID: 34367464 PMCID: PMC8337115 DOI: 10.1155/2021/6634718] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/09/2021] [Accepted: 02/15/2021] [Indexed: 01/10/2023]
Abstract
The mechanism underlying the role of oxidative stress and advanced oxidation protein products (AOPPs) in the aetiology of premature ovarian insufficiency (POI) is poorly understood. Here, we investigated the plasma AOPP level in POI patients and the effects of AOPPs on granulosa cells both in vitro and in vivo. KGN cells were treated with different AOPP doses, and cell cycle distribution, intracellular reactive oxygen species (ROS), and protein expression levels were measured. Sprague–Dawley (SD) rats were treated daily with PBS, rat serum albumin, AOPP, or AOPP+ N-acetylcysteine (NAC) for 12 weeks to explore the effect of AOPPs on ovarian function. Plasma AOPP concentrations were significantly higher in both POI and biochemical POI patients than in controls and negatively correlated with anti-Müllerian hormone and the antral follicle count. KGN cells treated with AOPP exhibited G1/G0-phase arrest. AOPP induced G1/G0-phase arrest in KGN cells by activating the ROS-c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK)-p21 pathway. Pretreatment with NAC, SP600125, SB203580, and si-p21 blocked AOPP-induced G1/G0-phase arrest. In SD rats, AOPP treatment increased the proportion of atretic follicles, and NAC attenuated the adverse effects of AOPPs in the ovary. In conclusion, we provide mechanistic evidence that AOPPs may induce cell cycle arrest in granulosa cells via the ROS-JNK/p38 MAPK-p21 pathway and thus may be a novel biomarker of POI.
Collapse
|
17
|
Ethanolic and Aqueous Extracts of Avocado (Persea americana) Seeds Attenuates Doxorubicin-Induced Cardiotoxicity in Male Albino Rats. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2021. [DOI: 10.1007/s13369-020-04994-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
18
|
Bellanti F, di Bello G, Iannelli G, Pannone G, Pedicillo MC, Boulter L, Lu WY, Tamborra R, Villani R, Vendemiale G, Forbes SJ, Serviddio G. Inhibition of nuclear factor (erythroid-derived 2)-like 2 promotes hepatic progenitor cell activation and differentiation. NPJ Regen Med 2021; 6:28. [PMID: 34039998 PMCID: PMC8155039 DOI: 10.1038/s41536-021-00137-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 04/28/2021] [Indexed: 02/08/2023] Open
Abstract
The stem cell ability to self-renew and lead regeneration relies on the balance of complex signals in their microenvironment. The identification of modulators of hepatic progenitor cell (HPC) activation is determinant for liver regeneration and may improve cell transplantation for end-stage liver disease. This investigation used different models to point out the Nuclear factor (erythroid-derived 2)-like 2 (NRF2) as a key regulator of the HPC fate. We initially proved that in vivo models of biliary epithelial cells (BECs)/HPC activation show hepatic oxidative stress, which activates primary BECs/HPCs in vitro. NRF2 downregulation and silencing were associated with morphological, phenotypic, and functional modifications distinctive of differentiated cells. Furthermore, NRF2 activation in the biliary tract repressed the ductular reaction in injured liver. To definitely assess the importance of NRF2 in HPC biology, we applied a xenograft model by inhibiting NRF2 in the human derived HepaRG cell line and transplanting into SCID/beige mice administered with anti-Fas antibody to induce hepatocellular apoptosis; this resulted in effective human hepatocyte repopulation with reduced liver injury. To conclude, NRF2 inhibition leads to the activation and differentiation of liver progenitors. This redox-dependent transcription factor represents a potential target to regulate the commitment of undifferentiated hepatic progenitors into specific lineages.
Collapse
Affiliation(s)
- Francesco Bellanti
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Giorgia di Bello
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppina Iannelli
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Pannone
- Anatomical Pathology Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Carmela Pedicillo
- Anatomical Pathology Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Wei-Yu Lu
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston Birmingham, UK
| | - Rosanna Tamborra
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosanna Villani
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gianluigi Vendemiale
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Gaetano Serviddio
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
19
|
Cerium Oxide Nanoparticles: A New Therapeutic Tool in Liver Diseases. Antioxidants (Basel) 2021; 10:antiox10050660. [PMID: 33923136 PMCID: PMC8146351 DOI: 10.3390/antiox10050660] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress induced by the overproduction of free radicals or reactive oxygen species (ROS) has been considered as a key pathogenic mechanism contributing to the initiation and progression of injury in liver diseases. Consequently, during the last few years antioxidant substances, such as superoxide dismutase (SOD), resveratrol, colchicine, eugenol, and vitamins E and C have received increasing interest as potential therapeutic agents in chronic liver diseases. These substances have demonstrated their efficacy in equilibrating hepatic ROS metabolism and thereby improving liver functionality. However, many of these agents have not successfully passed the scrutiny of clinical trials for the prevention and treatment of various diseases, mainly due to their unspecificity and consequent uncontrolled side effects, since a minimal level of ROS is needed for normal functioning. Recently, cerium oxide nanoparticles (CeO2NPs) have emerged as a new powerful antioxidant agent with therapeutic properties in experimental liver disease. CeO2NPs have been reported to act as a ROS and reactive nitrogen species (RNS) scavenger and to have multi-enzyme mimetic activity, including SOD activity (deprotionation of superoxide anion into oxygen and hydrogen peroxide), catalase activity (conversion of hydrogen peroxide into oxygen and water), and peroxidase activity (reducing hydrogen peroxide into hydroxyl radicals). Consequently, the beneficial effects of CeO2NPs treatment have been reported in many different medical fields other than hepatology, including neurology, ophthalmology, cardiology, and oncology. Unlike other antioxidants, CeO2NPs are only active at pathogenic levels of ROS, being inert and innocuous in healthy cells. In the current article, we review the potential of CeO2NPs in several experimental models of liver disease and their safety as a therapeutic agent in humans as well.
Collapse
|
20
|
Xu M, Hang H, Huang M, Li J, Xu D, Jiao J, Wang F, Wu H, Sun X, Gu J, Kong X, Gao Y. DJ-1 Deficiency in Hepatocytes Improves Liver Ischemia-Reperfusion Injury by Enhancing Mitophagy. Cell Mol Gastroenterol Hepatol 2021; 12:567-584. [PMID: 33766785 PMCID: PMC8258983 DOI: 10.1016/j.jcmgh.2021.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS DJ-1 is universally expressed in various tissues and organs and is involved in the physiological processes in various liver diseases. However, the role of DJ-1 in liver ischemia-reperfusion (I/R) injury is largely unknown. METHODS In this study, we first examined the DJ-1 expression changes in the liver tissues of mice and clinical donor after hepatic I/R by both quantitative polymerase chain reaction and Western blotting assays. Then we investigated the role of DJ-1 in I/R injury by using a murine liver I/R model. RESULTS We demonstrated that DJ-1 down-regulation in both human and mouse liver tissues in response to I/R injury and Dj-1 deficiency in hepatocytes but not in myeloid cells could significantly ameliorate I/R induced liver injury and inflammatory responses. This hepatoprotective effect was dependent on enhanced autophagy in Dj-1 knockout mice, because inhibition of autophagy by 3-methyladenine and chloroquine could reverse the protective effect on hepatic I/R injury in Dj-1 knockout mice. CONCLUSIONS Dj-1 deficiency in hepatocytes significantly enhanced mitochondrial accumulation and protein stability of PARKIN, which in turn promotes the onset of mitophagy resulting in elevated clearance of damaged mitochondria during I/R injury.
Collapse
Affiliation(s)
- Min Xu
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shangha, China
| | - Hualian Hang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Miao Huang
- Department of Transplantation, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jichang Li
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shangha, China
| | - Dongwei Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junzhe Jiao
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shangha, China
| | - Fang Wang
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shangha, China
| | - Hailong Wu
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Xuehua Sun
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shangha, China
| | - Jinyang Gu
- Department of Transplantation, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xiaoni Kong
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shangha, China.
| | - Yueqiu Gao
- Central Laboratory, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shangha, China.
| |
Collapse
|
21
|
Jiang H, Liu J, Fan C, Wang J, Li W. lncRNAS56464.1 as a ceRNA promotes the proliferation of fibroblast‑like synoviocytes in experimental arthritis via the Wnt signaling pathway and sponges miR‑152‑3p. Int J Mol Med 2021; 47:17. [PMID: 33448322 PMCID: PMC7834957 DOI: 10.3892/ijmm.2021.4850] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/04/2020] [Indexed: 12/23/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that occurs in approximately 1.0% of the general population. In RA patients, physical disability and joint damage are the major prognostic factors, which are associated with a reduction in the quality of life and early mortality. At present, the exact molecular mechanism of RA remains elusive. Long noncoding RNAs (lncRNAs) have been revealed to play a regulatory role in the pathogenesis of RA. To reveal the function of lncRNAs in rheumatoid arthritis, lncRNAS56464.1 was screened to verify its targeting of the microRNA (miR)-152-3p/Wnt pathway and its effect on the proliferation of fibroblast-like synoviocytes (FLS). In the present study, based on the competing endogenous RNA (ceRNA) theory, siRNA was designed for transfection into FLS to calculate the lncRNAS56464.1 interference efficiency and then the effect of lncRNAS56464.1 interference on FLS proliferation was detected by MTT assay. Then, lncRNAS56464.1 targeting of the miR-152-3p/Wnt pathway was detected by a dual-luciferase reporter assay. In addition, RT-qPCR, immunofluorescence and western blotting techniques were employed to detect the expression of lncRNAS56464.1, miR-152-3p and some key genes of the Wnt signaling pathway in FLS after lncRNAS56464.1 interference. The results revealed that lncRNAS56464.1 could combine with miR-152-3p and promoted the proliferation of FLS. In addition, lncRNAS56464.1 interference could not only decrease the proliferation of FLS and the expression of Wnt1, β-catenin, c-Myc, cyclin D1, and p-GSK-3β/GSK-3β, but it also increased the expression of SFRP4. The present data indicated that lncRNAS56464.1 could target the miR-152-3p/Wnt pathway to induce synovial cell proliferation and then participate in the pathogenesis of RA.
Collapse
Affiliation(s)
- Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Jian Liu
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Chang Fan
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Jing Wang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Weiping Li
- School of Basic Medical Science, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
22
|
Allicin Attenuated Advanced Oxidation Protein Product-Induced Oxidative Stress and Mitochondrial Apoptosis in Human Nucleus Pulposus Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6685043. [PMID: 33381267 PMCID: PMC7758128 DOI: 10.1155/2020/6685043] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022]
Abstract
Intervertebral disc degeneration (IDD) is one of the most common chronic degenerative musculoskeletal disorders. Oxidative stress-induced apoptosis of the nucleus pulposus (NP) cells plays a key role during IDD progression. Advanced oxidation protein products (AOPP), novel biomarkers of oxidative stress, have been reported to function in various diseases due to their potential for disrupting the redox balance. The current study is aimed at investigating the function of AOPP in the oxidative stress-induced apoptosis of human NP cells and the alleviative effects of allicin during this process which was known for its antioxidant properties. AOPP were demonstrated to hamper the viability and proliferation of NP cells in a time- and concentration-dependent manner and cause cell apoptosis markedly. High levels of reactive oxygen species (ROS) and lipid peroxidation product malondialdehyde (MDA) were detected in NP cells after AOPP stimulation, which resulted in depolarized mitochondrial transmembrane potential (MTP). Correspondingly, higher levels of AOPP were discovered in the human degenerative intervertebral discs (IVD). It was also found that allicin could protect NP cells against AOPP-mediated oxidative stress and mitochondrial dysfunction via suppressing the p38-MAPK pathway. These results disclosed a significant role of AOPP in the oxidative stress-induced apoptosis of NP cells, which could be involved in the primary pathogenesis of IDD. It was also revealed that allicin could be a promising therapeutic approach against AOPP-mediated oxidative stress during IDD progression.
Collapse
|
23
|
Mert H, Açikkol S, Çalli İ, Çibuk S, Keskin S, Mert N. Advanced Oxidation Protein Product (AOPP) Levels in Second- and Third-Degree Thermal Burns. J Burn Care Res 2020; 42:207-211. [PMID: 33009548 DOI: 10.1093/jbcr/iraa127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Burn is a systemic injury affecting the entire organism according to its etiology and severity. The aim of this study was to investigate plasma AOPP levels before and after treatment of second- and third-degree thermal burn patients and determine the changes in this parameter, and also, to find out the relationship between AOPP level and hospitalization period and total body surface area (TBSA). The study material consisted of pediatric patients with the complaint of second- and third-degree thermal burns aged between 1 and 18 years, with a burn area exceeding 10%. Blood samples were taken twice before and after treatment. AOPP level in blood plasma was measured in ELISA. It was observed that in the second-degree thermal burn group, AOPP level was 25.85 ± 2.82 ng/ml before the treatment decreased to 22.16 ± 3.62 ng/ml after treatment, whereas in the third-degree thermal burn group before the treatment AOPP was 25.96 ± 3.49 ng/ml, and after the treatment dropped to 21.70 ± 3.79 ng/ml, decreases were significantly important (P < .05). There was no statistically significant difference between the two groups in terms of AOPP levels (P > .05). Correlation analyses in the second- and third-degree thermal burn group did not show any correlation between AOPP levels and burn area and length of hospitalization period. As a result, AOPP level has been studied, for the first time, in burn cases. In both groups, the level of AOPP increased due to oxidative stress before treatment and decreased after treatment.
Collapse
Affiliation(s)
- Handan Mert
- Department of Biochemistry, Faculty of Veterinary Medicine, Van Yuzuncu Yil University
| | - Suat Açikkol
- University of Health Sciences, Van Education and Research Hospital, Burn Unit
| | - İskan Çalli
- Department of General Surgery, Faculty of Medicine
| | | | - Sıddık Keskin
- Department of Biostatistics, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Nihat Mert
- Department of Biochemistry, Faculty of Veterinary Medicine, Van Yuzuncu Yil University
| |
Collapse
|
24
|
Oxidative stress-mediated mitochondrial dysfunction facilitates mesenchymal stem cell senescence in ankylosing spondylitis. Cell Death Dis 2020; 11:775. [PMID: 32943613 PMCID: PMC7498590 DOI: 10.1038/s41419-020-02993-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disease possessing a morbid serum microenvironment with enhanced oxidative stress. Long-term exposure to an oxidative environment usually results in cellular senescence alone with cellular dysfunction. Mesenchymal stem cells (MSCs) are a kind of stem cell possessing strong capabilities for immunoregulation, and senescent MSCs may increase inflammation and participate in AS pathogenesis. The objective of this study was to explore whether and how the oxidative serum environment of AS induces MSC senescence. Here, we found that AS serum facilitated senescence of MSCs in vitro, and articular tissues from AS patients exhibited higher expression levels of the cell cycle arrest-related proteins p53, p21 and p16. Importantly, the levels of advanced oxidative protein products (AOPPs), markers of oxidative stress, were increased in AS serum and positively correlated with the extent of MSC senescence induced by AS serum. Furthermore, MSCs cultured with AS serum showed decreased mitochondrial membrane potential and ATP production together with a reduced oxygen consumption rate. Finally, we discovered that AS serum-induced mitochondrial dysfunction resulted in elevated reactive oxygen species (ROS) in MSCs, and ROS inhibition successfully rescued MSCs from senescence. In conclusion, our data demonstrated that the oxidative serum environment of AS facilitated MSC senescence through inducing mitochondrial dysfunction and excessive ROS production. These results may help elucidate the pathogenesis of AS and provide potential targets for AS treatment.
Collapse
|
25
|
Sun Y, Lu Y, Saredy J, Wang X, Drummer Iv C, Shao Y, Saaoud F, Xu K, Liu M, Yang WY, Jiang X, Wang H, Yang X. ROS systems are a new integrated network for sensing homeostasis and alarming stresses in organelle metabolic processes. Redox Biol 2020; 37:101696. [PMID: 32950427 PMCID: PMC7767745 DOI: 10.1016/j.redox.2020.101696] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are critical for the progression of cardiovascular diseases, inflammations and tumors. However, the mechanisms of how ROS sense metabolic stress, regulate metabolic pathways and initiate proliferation, inflammation and cell death responses remain poorly characterized. In this analytic review, we concluded that: 1) Based on different features and functions, eleven types of ROS can be classified into seven functional groups: metabolic stress-sensing, chemical connecting, organelle communication, stress branch-out, inflammasome-activating, dual functions and triple functions ROS. 2) Among the ROS generation systems, mitochondria consume the most amount of oxygen; and nine types of ROS are generated; thus, mitochondrial ROS systems serve as the central hub for connecting ROS with inflammasome activation, trained immunity and immunometabolic pathways. 3) Increased nuclear ROS production significantly promotes cell death in comparison to that in other organelles. Nuclear ROS systems serve as a convergent hub and decision-makers to connect unbearable and alarming metabolic stresses to inflammation and cell death. 4) Balanced ROS levels indicate physiological homeostasis of various metabolic processes in subcellular organelles and cytosol, while imbalanced ROS levels present alarms for pathological organelle stresses in metabolic processes. Based on these analyses, we propose a working model that ROS systems are a new integrated network for sensing homeostasis and alarming stress in metabolic processes in various subcellular organelles. Our model provides novel insights on the roles of the ROS systems in bridging metabolic stress to inflammation, cell death and tumorigenesis; and provide novel therapeutic targets for treating those diseases. (Word count: 246).
Collapse
Affiliation(s)
- Yu Sun
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Yifan Lu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Jason Saredy
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xianwei Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Charles Drummer Iv
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ying Shao
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Fatma Saaoud
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Keman Xu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ming Liu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - William Y Yang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaohua Jiang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
26
|
Othmène YB, Hamdi H, Salem IB, Annabi E, Amara I, Neffati F, Najjar MF, Abid-Essefi S. Oxidative stress, DNA damage and apoptosis induced by tebuconazole in the kidney of male Wistar rat. Chem Biol Interact 2020; 330:109114. [PMID: 32735800 DOI: 10.1016/j.cbi.2020.109114] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 04/03/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
Tebuconazole (TEB) is a broad-spectrum conazole fungicide that has been used in agriculture in the control of foliar and soil-borne diseases of many crops. The present study has investigated the adverse effects of subchronic exposure to TEB on the kidney of male rats. Animals were divided into four equal groups and treated with TEB at increasing doses 0.9, 9 and 27 mg/kg body weight for 28 consecutive days. The results showed that TEB induced oxidative stress in the kidney demonstrated by an increase in malondialdehyde (MDA), protein carbonyl (PC), advanced oxidation protein product (AOPP) levels and DNA damage, as compared to the controls. Furthermore, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) activities were increased in the renal tissue of treated rats. Moreover, significant decrease in reduced glutathione (GSH) content in TEB-treated rats was observed, while oxidized glutathione (GSSG) levels were increased, thus a marked fall in GSH/GSSG ratio was registered in the kidney. Glutathione reductase (GR) activity showed a significant increase after TEB exposure. Moreover, TEB down-regulated the expression of Bcl2 and up-regulated the expression of Bax and caspase 3, which triggered apoptosis via the Bax/Bcl2 and caspase pathway. Also, TEB administration resulted in altered biochemical indicators of renal function and varying lesions in the overall histo-architecture of renal tissues. Taken together, our findings brought into light the renal toxicity induced by TEB, which was found to be significant at low doses.
Collapse
Affiliation(s)
- Yosra Ben Othmène
- University of Monastir, Faculty of Dental Medicine of Monastir, Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Rue Avicenne, 5000, Monastir, Tunisia
| | - Hiba Hamdi
- University of Monastir, Faculty of Dental Medicine of Monastir, Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Rue Avicenne, 5000, Monastir, Tunisia
| | - Intidhar Ben Salem
- University of Monastir, Faculty of Dental Medicine of Monastir, Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Rue Avicenne, 5000, Monastir, Tunisia; University of Sousse, Faculty of Medicine of Sousse, Rue Mohamed Karoui, 4000, Tunisia
| | - Emna Annabi
- University of Monastir, Faculty of Dental Medicine of Monastir, Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Rue Avicenne, 5000, Monastir, Tunisia
| | - Ines Amara
- University of Monastir, Faculty of Dental Medicine of Monastir, Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Rue Avicenne, 5000, Monastir, Tunisia
| | - Fadwa Neffati
- Fattouma Bourguiba University Hospital, Laboratory of Biochemistry-Toxicology, Avenue 1 Juin 1955, 5000, Monastir, Tunisia
| | - Mohamed Fadhel Najjar
- Fattouma Bourguiba University Hospital, Laboratory of Biochemistry-Toxicology, Avenue 1 Juin 1955, 5000, Monastir, Tunisia
| | - Salwa Abid-Essefi
- University of Monastir, Faculty of Dental Medicine of Monastir, Laboratory for Research on Biologically Compatible Compounds, LR01SE17, Rue Avicenne, 5000, Monastir, Tunisia.
| |
Collapse
|
27
|
Ma G, Liu Y, Wang Y, Wen Z, Li X, Zhai H, Miao L, Luo J. Liraglutide reduces hyperglycemia-induced cardiomyocyte death through activating glucagon-like peptide 1 receptor and targeting AMPK pathway. J Recept Signal Transduct Res 2020; 40:133-140. [PMID: 32013667 DOI: 10.1080/10799893.2020.1719517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective: Hyperglycemia-mediated cardiomyocyte damage is associated with inflammation and AMPK inactivation.Aim: The aim of our study is to explore the protective effects exerted by liraglutide on AMPK pathway and glucagon-like peptide 1 receptor in diabetic cardiomyopathy.Methods: Cardiomyocytes were treated with high-glucose stress and cardiomyocyte viability was determined via (3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide assay. Besides, LDH release, immunofluorescence, and qPCR were used to verify the influence of liraglutide on hyperglycemia-treated cardiomyocytes.Results: Hyperglycemia treatment caused inflammation response and oxidative stress were significantly elevated in cardiomyocytes. This alteration could be reversed by liraglutide. Besides, cell viability was reduced whereas apoptosis was increased after exposure to high glucose treatment. However, liraglutide treatment could attenuate apoptosis and reverse cell viability in cardiomyocyte. Further, we found that AMPK pathway was also activated and glucagon-like peptide 1 receptor expression was increased in response to liraglutide treatment.Conclusions: Liraglutide could attenuate hyperglycemia-mediated cardiomyocyte damage through reversing AMPK pathway and upregulating glucagon-like peptide 1 receptor.
Collapse
Affiliation(s)
- Guanqun Ma
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yingwu Liu
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yu Wang
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Zhinan Wen
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Xin Li
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Hu Zhai
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Li Miao
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Jieying Luo
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| |
Collapse
|
28
|
Zhang M, Zhou D, Ouyang Z, Yu M, Jiang Y. Sphingosine kinase 1 promotes cerebral ischemia-reperfusion injury through inducing ER stress and activating the NF-κB signaling pathway. J Cell Physiol 2020; 235:6605-6614. [PMID: 31985036 DOI: 10.1002/jcp.29546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/15/2020] [Indexed: 01/02/2023]
Abstract
Endoplasm reticulum stress and inflammation response have been found to be linked to cerebral ischemia-reperfusion (IR) injury. Sphingosine kinase 1 (SPHK1) has been reported to be a novel endoplasm reticulum regulator. The aim of our study is to figure out the role of SPHK1 in cerebral IR injury and verify whether it has an ability to regulate inflammation and endoplasm reticulum stress. Hydrogen peroxide was used to induce cerebral IR injury. Enzyme-linked immunosorbent assay, quantitative polymerase chain reaction, western blots, and immunofluorescence were used to measure the alterations of cell viability, inflammation response, and endoplasm reticulum stress. The results demonstrated that after exposure to hydrogen peroxide, cell viability was reduced whereas SPHK1 expression was significantly elevated. Knockdown of SPHK1 attenuated hydrogen peroxide-mediated cell death and reversed cell viability. Our data also demonstrated that SPHK1 deletion reduced endoplasm reticulum stress and alleviated inflammation response in hydrogen peroxide-treated cells. In addition, we also found that SHPK1 modulated endoplasm reticulum stress and inflammation response to through the NF-κB signaling pathway. Inhibition of NF-κB signaling pathway has similar results when compared with the cells with SPHK1 deletion. Altogether, our results demonstrated that SPHK1 upregulation, induced by hydrogen peroxide, is responsible for cerebral IR injury through inducing endoplasm reticulum stress and inflammation response in a manner working through the NF-κB signaling pathway. This finding provides new insight into the molecular mechanism to explain the neuron death induced by cerebral IR injury.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dingzhou Zhou
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhu Ouyang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengqiang Yu
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
29
|
Zhang Y, Zhang H, Shi W, Wang W. Mief1 augments thyroid cell dysfunction and apoptosis through inhibiting AMPK-PTEN signaling pathway. J Recept Signal Transduct Res 2020; 40:15-23. [PMID: 31960779 DOI: 10.1080/10799893.2020.1716799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objective: Inflammation-mediated thyroid cell dysfunction and apoptosis increases the like-hood of hypothyroidism.Aim: Our aim in the present study is to explore the role of mitochondrial elongation factor 1 (Mief1) in thyroid cell dysfunction induced by TNFα.Materials and methods: Different doses of TNFα were used to incubate with thyroid cells in vitro. The survival rate, apoptotic index and proliferation capacity of thyroid cells were measured. Cellular energy metabolism and endoplasmic reticulum function related to protein synthesis were detected.Results: In response to TNFα treatment, the levels of Mief1 were increased, coinciding with a drop in the viability of thyroid cells in vitro. Loss of Mief1 attenuates TNFα-induced cell death through reducing the ratio of cell apoptosis. Further, we found that Mief1 deletion reversed cell energy metabolism and this effect was attributable to mitochondrial protection. Mief1 knockdown sustained mitochondrial membrane potential and reduced mitochondrial ROS overproduction. In addition, Mief1 knockdown also reduced endoplasmic reticulum stress, as evidenced by decreased levels of Chop and Caspase-12. Finally, our data verified that TNFα treatment inhibited the activity of AMPK-PTEN pathway whereas Mief1 deletion reversed the activity of AMPK and thus promoted the upregulation of PTEN. However, inhibition of AMPK-PTEN pathways could abolish the beneficial effects exerted by Mief1 deletion on thyroid cells damage and dysfunction.Conclusions: Altogether, our data indicate that immune abnormality-mediated thyroid cell dysfunction and death are alleviated by Mief1 deletion possible driven through reversing the activity of AMPK-PTEN pathways.
Collapse
Affiliation(s)
- Yonglan Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Otorhinolaryngology Research Institute of Tianjin, Tianjin Key Laboratory of Auditory Speech Balance Medicine, Tianjin, People's Republic of China
| | - Haichao Zhang
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, Tianjin, People's Republic of China
| | - Wenjie Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Otorhinolaryngology Research Institute of Tianjin, Tianjin Key Laboratory of Auditory Speech Balance Medicine, Tianjin, People's Republic of China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Otorhinolaryngology Research Institute of Tianjin, Tianjin Key Laboratory of Auditory Speech Balance Medicine, Tianjin, People's Republic of China
| |
Collapse
|
30
|
Ahmed AE, Alshehri A, Al-Kahtani MA, Elbehairi SEI, Alshehri MA, Shati AA, Alfaifi MY, Al-Doais AA, Taha R, Morsy K, El-Mansi AA. Vitamin E and selenium administration synergistically mitigates ivermectin and doramectin-induced testicular dysfunction in male Wistar albino rats. Biomed Pharmacother 2020; 124:109841. [PMID: 31972360 DOI: 10.1016/j.biopha.2020.109841] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 01/09/2023] Open
Abstract
Avermectins are broad-spectrum antiparasitic drugs in veterinary and human medication. The current study aimed to examine the toxic effects of ivermectin (IVM) and doramectin (DRM), with or without co-treatment of vitamin E (Vit.E) and selenium (Se) on apoptosis, oxidative stress and male fertility in Wistar rats. Twenty five adult male animals were divided into five groups; G1; was control (CTL) received saline, G2; IVM (0.2 mg/kg b.w), G3; IVM plus Vit.E/Se (80/1.6 mg/kg b.w, respectively), G4; DRM (0.2 mg/kg b.w), and G5; DRM plus Vit.E/Se. Both IVM and DRM were given by subcutaneous (s.c) injections while Vit.E/Se was orally given. All treatments were administered once weekly for four consecutive weeks. By 24 h after the last treatment, the animals were sacrificed. Blood and tissue samples were collected for hematology, serobiochemistry, histopathology, and molecular assays for hepatic/ renal toxicities, oxidative stress, cell viability and fertility parameters. Apoptosis of the hepatic cells obtained from the treated rats was assayed by detection of annexin-V using the flow cytometric assay (FCA). The proliferating cellular nuclear antigen (PCNA) and DNA fragmentation in the treated rats' testicular tissues were also assayed. Moreover, the direct effects of IVM or DRM with or without concomitant administration of Vit.E/Se on testicular cells isolated from adult rat were also performed in vitro. Apoptosis of those cultured testicular cells in response to the different treatments was assayed by detection of the inhibition-concentration fifty (IC50) using the SRB method, and evaluating the viable versus apoptotic cells microscopically after staining with acridine orange-ethidium bromide (AO/EB). In conclusion, both avermectins induced apoptosis in the living and cultured cells, while those antioxidants; Vit.E and Se, reduced the oxidative stress and cytotoxicity both in vivo and in vitro, either. Furthermore, the reprotoxicity and reduced male fertility were seriously evoked by IVM, but not DRM with dramatic ameliorative effect of Vit.E/Se if concomitantly administered. Avermectins, especially ivermectin, should be given according to the dose recommended by the manufacturer company and repeated dosages should be given with Vit.E/Se.
Collapse
Affiliation(s)
- Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia; Department of Theriogenology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt.
| | - Ali Alshehri
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - M A Al-Kahtani
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | | | - Mohammed A Alshehri
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Mohammad Y Alfaifi
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Amin A Al-Doais
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Ramadan Taha
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia; Department of Zoology, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Ahmed A El-Mansi
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia; Zoology Dept., Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
31
|
Zhao Y, Chen E, Huang K, Xie Z, Zhang S, Wu D, Ji F, Zhu D, Xu X, Li L. Dynamic Alterations of Plasma Metabolites in the Progression of Liver Regeneration after Partial Hepatectomy. J Proteome Res 2020; 19:174-185. [PMID: 31802674 DOI: 10.1021/acs.jproteome.9b00493] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To elucidate the dynamic alterations of metabolites in rat plasma during liver regeneration and search for potential biomarkers of liver regeneration, 65 male Sprague-Dawley rats were divided into three groups: 70% partial hepatectomy group (PHx, n = 30), sham-operated group (Sham, n = 30), and pre-PHx group (pre-PHx, n = 5). Rats in the Sham and PHx groups were sacrificed after 30 min (min), 6 h (h), 24, 48, 72, and 168 h of surgery (n = 5 per time point). The gas chromatography-mass spectrometry-based metabolomic approach was used to identify the dynamic metabolites. Liver regeneration in the rats was evidenced by an increase in the liver weight/body weight ratio, expression of proliferating cell nuclear antigen, and yes-associated protein-1. Thirty-four differentially abundant metabolites between the Sham and PHx groups were identified, which were involved in arginine and proline metabolism, aminoacyl-tRNA biosynthesis, and cysteine and methionine metabolism pathways. Of these metabolites, low 1,5-anhydroglucitol may indicate proliferation of liver parenchymal cells during liver regeneration. Thus, a series of metabolic changes occurred with the progression of liver regeneration, and 1,5-anhydroglucitol could function as a novel hallmark of proliferation of liver parenchymal cells.
Collapse
Affiliation(s)
| | - Ermei Chen
- Department of Gastroenterology , Zhongshan Hospital Affiliated to Xiamen University , Xiamen 361004 , Fujian Province , China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ferreiro ME, Amarilla MS, Glienke L, Méndez CS, González C, Jacobo PV, Sobarzo CM, De Laurentiis A, Ferraris MJ, Theas MS. The inflammatory mediators TNFα and nitric oxide arrest spermatogonia GC-1 cell cycle. Reprod Biol 2019; 19:329-339. [PMID: 31757605 DOI: 10.1016/j.repbio.2019.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 10/08/2019] [Accepted: 11/02/2019] [Indexed: 01/01/2023]
Abstract
During an inflammatory process of the testis, the network of somatic, immune, and germ cell interactions is altered leading to organ dysfunction. In testicular biopsies of infertile men, spermatogenesis impairment is associated with reduced spermatogonia proliferation, increased number of immune cells, and content of pro-inflammatory cytokines. TNFα-TNFR and nitric oxide (NO)-NO synthase systems are up-regulated in models of testicular damage and in human testis with maturation arrest. The purpose of this study was to test the hypothesis that TNFα-TNFR system and NO alter the function of spermatogonia in the inflamed testis. We studied the effect of TNFα and NO on GC-1 spermatogonia cell cycle progression and death by flow cytometry. GC-1 cells expressed TNFR1 and TNFR2 (immunofluorescence). TNFα (10 and 50 ng/ml) and DETA-Nonoate (0.5 and 2 mM), a NO releaser, increased the percentage of cells in S-phase of the cell cycle and reduced the percentage in G1, inducing also cell apoptosis. TNFα effect was not mediated by oxidative stress unlike NO, since the presence of N-acetyl-l-cysteine (2.5 and 5.0 mM) prevented NO induced cell cycle arrest and death. GC-1 spermatogonia overpass NO induced cell cycle arrest but no TNFα, since after removal of NO, spermatogonia progressed through the cell cycle. We propose TNFα and NO might contribute to impairment of spermatogenesis by preventing adequate functioning of the spermatogonia population. Our results showed that TNFα and NO impaired spermatogonia cell cycle, inducing GC-1 arrest in the S phase.
Collapse
Affiliation(s)
- María Eugenia Ferreiro
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - María Sofía Amarilla
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Leilane Glienke
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Cinthia Soledad Méndez
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Candela González
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnósticos (CEBBAD), Universidad Maimónides, Buenos Aires, Argentina
| | - Patricia Verónica Jacobo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Cristian Marcelo Sobarzo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Andrea De Laurentiis
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO) CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Jimena Ferraris
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - María Susana Theas
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular, Cátedra II de Histología, Buenos Aires, Argentina, CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina.
| |
Collapse
|
33
|
Qiang Y, Ma F, Wang Z, Nie Z, Xu L, Ding P, Ma X. LukS-PV induces cell cycle arrest and apoptosis through p38/ERK MAPK signaling pathway in NSCLC cells. Biochem Biophys Res Commun 2019; 521:846-852. [PMID: 31708104 DOI: 10.1016/j.bbrc.2019.10.181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 10/26/2019] [Indexed: 12/16/2022]
Abstract
Non-small-cell lung cancer (NSCLC) accounts for nearly 85% of lung cancer cases. LukS-PV, one of the two components of Panton-Valentine leucocidin (PVL), is produced by Staphylococcus aureus. The present study showed that LukS-PV can induce apoptosis in human acute myeloid leukemia (AML) lines (THP-1 and HL-60). However, the role of LukS-PV in NSCLC is unclear. In this study, we treated NSCLC cell lines A549 and H460 and a normal lung cell line, 16HBE, with LukS-PV and investigated the biological roles of LukS-PV in NSCLC. Cells were treated with varying concentrations of LukS-PV and cell viability was evaluated by CCK8 and EdU assay. Flow cytometry was used to detect cell apoptosis and analyze the cell cycle, and the expression of apoptosis and cell cycle-associated proteins and genes were identified by western blotting analysis and qRT-polymerase chain reaction, respectively. We found that LukS-PV inhibited the proliferation of NSCLC cells but had little cytotoxicity in normal lung cells. LukS-PV induced NSCLC cell apoptosis and increased the BAX/BCL-2 ratio, triggering S-phase arrest in A549 and H460 cells while increasing P21 expression and decreasing CDK2, cyclin D1, and cyclin A2 expression. We also observed increased P-p38 and P-ERK in NSCLC cells treated with LukS-PV. Treatment of NSCLC with LukS-PV combined with p38 and ERK inhibitors reversed the pro-apoptotic and pro-cell cycle arrest effects of LukS-PV. Overall, these findings indicate that LukS-PV has anti-tumor effects in NSCLC and may contribute to the development of anti-cancer agents.
Collapse
Affiliation(s)
- Yawen Qiang
- Department of Clinical Laboratory, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fan Ma
- Department of Clinical Laboratory, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ziran Wang
- Department of Clinical Laboratory, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhengchao Nie
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Liangfei Xu
- Department of Clinical Laboratory, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Pengsheng Ding
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoling Ma
- Department of Clinical Laboratory, Affiliated Provincial Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
34
|
Córdoba-Jover B, Arce-Cerezo A, Ribera J, Pauta M, Oró D, Casals G, Fernández-Varo G, Casals E, Puntes V, Jiménez W, Morales-Ruiz M. Cerium oxide nanoparticles improve liver regeneration after acetaminophen-induced liver injury and partial hepatectomy in rats. J Nanobiotechnology 2019; 17:112. [PMID: 31672158 PMCID: PMC6822381 DOI: 10.1186/s12951-019-0544-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023] Open
Abstract
Background and aims Cerium oxide nanoparticles are effective scavengers of reactive oxygen species and have been proposed as a treatment for oxidative stress-related diseases. Consequently, we aimed to investigate the effect of these nanoparticles on hepatic regeneration after liver injury by partial hepatectomy and acetaminophen overdose. Methods All the in vitro experiments were performed in HepG2 cells. For the acetaminophen and partial hepatectomy experimental models, male Wistar rats were divided into three groups: (1) nanoparticles group, which received 0.1 mg/kg cerium nanoparticles i.v. twice a week for 2 weeks before 1 g/kg acetaminophen treatment, (2) N-acetyl-cysteine group, which received 300 mg/kg of N-acetyl-cysteine i.p. 1 h after APAP treatment and (3) partial hepatectomy group, which received the same nanoparticles treatment before partial hepatectomy. Each group was matched with vehicle-controlled rats. Results In the partial hepatectomy model, rats treated with cerium oxide nanoparticles showed a significant increase in liver regeneration, compared with control rats. In the acetaminophen experimental model, nanoparticles and N-acetyl-cysteine treatments decreased early liver damage in hepatic tissue. However, only the effect of cerium oxide nanoparticles was associated with a significant increment in hepatocellular proliferation. This treatment also reduced stress markers and increased cell cycle progression in hepatocytes and the activation of the transcription factor NF-κB in vitro and in vivo. Conclusions Our results demonstrate that the nanomaterial cerium oxide, besides their known antioxidant capacities, can enhance hepatocellular proliferation in experimental models of liver regeneration and drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Bernat Córdoba-Jover
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain
| | - Altamira Arce-Cerezo
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain
| | - Jordi Ribera
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain
| | - Montse Pauta
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain
| | - Denise Oró
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain
| | - Gregori Casals
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain.,Working Group for the Biochemical Assessment of Hepatic Disease-SEQC-ML, Barcelona, Spain
| | - Guillermo Fernández-Varo
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain
| | - Eudald Casals
- Vall d'Hebron Institut of Research (VHIR), Barcelona, Spain.,Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, China
| | - Victor Puntes
- Vall d'Hebron Institut of Research (VHIR), Barcelona, Spain.,Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Wladimiro Jiménez
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain.,Department of Biomedicine-Biochemistry Unit, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Manuel Morales-Ruiz
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, CIBERehd, 170 Villarroel St., 08036, Barcelona, Spain. .,Working Group for the Biochemical Assessment of Hepatic Disease-SEQC-ML, Barcelona, Spain. .,Department of Biomedicine-Biochemistry Unit, School of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
35
|
Wang S, Hu H, Zhong B, Shi D, Qing X, Cheng C, Deng X, Zhang Z, Shao Z. Bruceine D inhibits tumor growth and stem cell-like traits of osteosarcoma through inhibition of STAT3 signaling pathway. Cancer Med 2019; 8:7345-7358. [PMID: 31631559 PMCID: PMC6885873 DOI: 10.1002/cam4.2612] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 09/19/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Patients with osteosarcoma exhibiting resistance to chemotherapy or presenting with metastasis usually have a poor prognosis. Osteosarcoma stem cells (OSCs) are a potential cause of tumor metastasis, relapse, and chemotherapy resistance. Therefore, it is necessary to develop novel therapeutic drugs, which not only kill osteosarcoma cells but also target OSCs. This study aims to explore the anti‐osteosarcoma effects of Bruceine D (BD), a natural compound derived from Brucea javanica, and investigate its underlying mechanisms. Results demonstrated that BD could significantly inhibit cell proliferation and migration, induce cell cycle arrest, and promote apoptosis in osteosarcoma cells. Besides, BD could also suppress the sphere‐forming and self‐renewal ability of OSCs. Mechanistically, the inhibitory role of BD on osteosarcoma cell growth and migration including OSC stemness was partially executed through the inhibition of STAT3 signaling pathway. More importantly, BD showed significant anti‐osteosarcoma activity without obvious side effects in vivo. Collectively, the results of this study demonstrated that BD exerts a strong inhibitory effect on tumor growth and stem cell like traits of osteosarcoma which may be partially due to STAT3 inhibition, suggesting that BD maybe a promising therapeutic candidate against osteosarcoma.
Collapse
Affiliation(s)
- Shangyu Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongzhi Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binlong Zhong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Deyao Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangcheng Qing
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyu Deng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Luceri C, Bigagli E, Agostiniani S, Giudici F, Zambonin D, Scaringi S, Ficari F, Lodovici M, Malentacchi C. Analysis of Oxidative Stress-Related Markers in Crohn's Disease Patients at Surgery and Correlations with Clinical Findings. Antioxidants (Basel) 2019; 8:antiox8090378. [PMID: 31489956 PMCID: PMC6771139 DOI: 10.3390/antiox8090378] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022] Open
Abstract
Crohn' disease (CD) patients are at high risk of postoperative recurrence and new tools for the assessment of disease activity are needed to prevent long-term complications. In these patients, the over-production of ROS generated by inflamed bowel tissue and inflammatory cells activates a pathogenic cascade that further exacerbates inflammation and leads to increased oxidative damage to DNA, proteins, and lipids. We measured the products of protein/lipid oxidation and the total antioxidant capacity (ferric reducing ability of plasma, FRAP) in the serum of CD patients with severe disease activity requiring surgery with the aim to characterize their redox status and identify associations between oxidative stress-related markers and their clinical characteristics. At the systemic level, CD was associated with increased levels of protein and lipid oxidation products when compared to healthy volunteers, even though the FRAP values were similar. Advanced oxidation protein product (AOPP) levels showed the highest difference between patients and the controls (11.25, 5.02-15.15, vs. 1.36, 0.75-2.70, median, interquartile range; p < 0.0001) and the analysis of receiver operating characteristic (ROC) curves, indicated for AOPP, the best area under the curve (AUC) value for CD prediction. Advanced glycated end-products (AGEs) were also significantly higher in CD patients (p < 0.01), which is of interest since AOPP and AGEs are both able to activate the membrane receptor for advanced glycation end products (RAGE) involved in inflammatory diseases. Thiobarbituric acid reactive substance (TBARS) levels were significantly higher in CD patients with ileal localization and aggressive disease behavior, in smokers, and in patients suffering from allergies. In conclusion, our data indicate that circulating oxidative stress biomarkers may be attractive candidates as disease predictors as well as for clinical or therapeutic monitoring of CD. Our results also suggest that AOPP/AGEs and RAGE signaling may represent a pathogenic factor and a potential therapeutic target in CD.
Collapse
Affiliation(s)
- Cristina Luceri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy.
| | - Elisabetta Bigagli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy.
| | - Sara Agostiniani
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy
| | - Francesco Giudici
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Daniela Zambonin
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Stefano Scaringi
- Department of Experimental and Clinical Medicine, Surgery Unit IBD, Careggi-University Hospital (AOUC), 50139 Florence, Italy
| | - Ferdinando Ficari
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Maura Lodovici
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy
| | - Cecilia Malentacchi
- Department of Biomedical Experimental and Clinical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| |
Collapse
|
37
|
Park S, Lee JY, Lim W, You S, Song G. Butylated Hydroxyanisole Exerts Neurotoxic Effects by Promoting Cytosolic Calcium Accumulation and Endoplasmic Reticulum Stress in Astrocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9618-9629. [PMID: 31381342 DOI: 10.1021/acs.jafc.9b02899] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Astrocytes provide nutritional support, regulate inflammation, and perform synaptic functions in the human brain. Although butylated hydroxyanisole (BHA) is a well-known antioxidant, several studies in animals have indicated BHA-mediated liver toxicity, retardation in reproductive organ development and learning, and sleep deficit. However, the specific effects of BHA on human astrocytes and the underlying mechanisms are yet unclear. Here, we investigated the antigrowth effects of BHA through cell cycle arrest and downregulation of regulatory protein expression. The typical cell proliferative signaling pathways, phosphoinositide 3-kinase/protein kinase B and extracellular signal-regulated kinase 1/2, were downregulated in astrocytes after BHA treatment. BHA increased the levels of pro-apoptotic proteins, such as BAX, cytochrome c, cleaved caspase 3, and cleaved caspase 9, and decreased the level of anti-apoptotic protein BCL-XL. It also increased the cytosolic calcium level and the expression of endoplasmic reticulum stress proteins. Treatment with BAPTA-AM, a calcium chelator, attenuated the increased levels of ER stress proteins and cleaved members of the caspase family. We further performed an in vivo evaluation of the neurotoxic effect of BHA on zebrafish embryos and glial fibrillary acidic protein, a representative astrocyte biomarker, in a gfap:eGFP zebrafish transgenic model. Our results provide clear evidence of the potent cytotoxic effects of BHA on human astrocytes, which lead to disruption of the brain and nerve development.
Collapse
Affiliation(s)
- Sunwoo Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology , Korea University , Seoul 02841 , Republic of Korea
| | - Jin-Young Lee
- Department of Biochemistry , Medical College of Wisconsin , Milwaukee , Wisconsin 53226 , United States
| | - Whasun Lim
- Department of Food and Nutrition , Kookmin University , Seoul 02707 , Republic of Korea
| | - Seungkwon You
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology , Korea University , Seoul 02841 , Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology , Korea University , Seoul 02841 , Republic of Korea
| |
Collapse
|
38
|
Yu W, Mei X, Zhang Q, Zhang H, Zhang T, Zou C. Yap overexpression attenuates septic cardiomyopathy by inhibiting DRP1-related mitochondrial fission and activating the ERK signaling pathway. J Recept Signal Transduct Res 2019; 39:175-186. [PMID: 31354091 DOI: 10.1080/10799893.2019.1641822] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Context: Yes-associated protein (Yap) has been linked to several cardiovascular disorders, but the role of this protein in septic cardiomyocytes is not fully understood. Objective: The aim of our study was to explore the influence of Yap in septic cardiomyopathy in vivo and in vitro. Materials and methods: In the current study, Yap transgenic mice and Yap adenovirus-mediated gain-of-function assays were used in an LPS-established septic cardiomyopathy model. Mitochondrial function and mitochondrial fission were determined through western blotting, immunofluorescence analysis and ELISA. Results: Our results demonstrated that Yap expression was downregulated by LPS, whereas Yap overexpression sustained cardiac function and attenuated cardiomyocyte death. The functional exploration revealed that LPS treatment induced cardiomyocyte mitochondrial stress, as manifested by mitochondrial superoxide overproduction, cardiomyocyte ATP deprivation, and caspase-9 apoptosis activation. Furthermore, we demonstrated that LPS-mediated mitochondrial damage was controlled by mitochondrial fission. However, Yap overexpression reduced mitochondrial fission and therefore improved mitochondrial function. A molecular investigation revealed that Yap overexpression inhibited mitochondrial fission by reversing ERK activity, and the inhibition of the ERK pathway promoted DRP1 upregulation and thereby mediated mitochondrial fission activation in the presence of Yap overexpression. Conclusions: Overall, our results suggest that the cause of septic cardiomyopathy appears to be connected with Yap downregulation. The overexpression of Yap can attenuate myocardial inflammation injury through the reduction of DRP1-related mitochondrial fission in an ERK pathway activation-dependent manner.
Collapse
Affiliation(s)
- Wancheng Yu
- a Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , China
| | - Xu Mei
- b Department of Geriatrics, Shandong University Qilu Hospital , Jinan , China
| | - Qian Zhang
- a Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , China
| | - Haizhou Zhang
- a Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , China
| | - Tao Zhang
- a Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , China
| | - Chengwei Zou
- a Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , China
| |
Collapse
|
39
|
Fan J, Zhu Q, Wu Z, Ding J, Qin S, Liu H, Miao P. Protective effects of irisin on hypoxia-reoxygenation injury in hyperglycemia-treated cardiomyocytes: Role of AMPK pathway and mitochondrial protection. J Cell Physiol 2019; 235:1165-1174. [PMID: 31268170 DOI: 10.1002/jcp.29030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/12/2019] [Indexed: 12/22/2022]
Abstract
Recent evidence has verified the cardioprotective actions of irisin in different diseases models. However, the beneficial action of irisin on hypoxia-reoxygenation (HR) injury under high glucose stress has not been described. Herein our research investigated the influence of irisin on HR-triggered cardiomyocyte death under high glucose stress. HR model was established in vitro under high glucose treatment. The results illuminated that HR injury augmented apoptotic ratio of cardiomyocyte under high glucose stress; this effect could be abolished by irisin via modulating mitochondrial function. Irisin treatment attenuated cellular redox stress, improved cellular ATP biogenetics, sustained mitochondria potential, and impaired mitochondrion-related cell death. At the molecular levels, irisin treatment activated the 5'-adenosine monophosphate-activated protein kinase (AMPK) pathway and the latter protected cardiomyocyte and mitochondria against HR injury under high glucose stress. Altogether, our results indicated a novel role of irisin in HR-treated cardiomyocyte under high glucose stress. Irisin-activated AMPK pathway and the latter sustained cardiomyocyte viability and mitochondrial function.
Collapse
Affiliation(s)
- Jiamao Fan
- Department of Cardiology, Linfen Central Hospital, Linfen, China
| | - Qing Zhu
- Department of Cardiology, Linfen Central Hospital, Linfen, China.,Institutes of Biomedical Sciences, Shanghai Medical School, Fudan University, Shanghai, China
| | - Zhenhua Wu
- Department of Cardiology, Linfen Central Hospital, Linfen, China
| | - Jiao Ding
- Department of Cardiology, Linfen Central Hospital, Linfen, China
| | - Shuai Qin
- Department of Cardiovascular Surgery, Linfen Central Hospital, Linfen, China
| | - Hui Liu
- Department of Cardiovascular Surgery, Linfen Central Hospital, Linfen, China
| | - Pengfei Miao
- Department of Cardiology, Linfen Central Hospital, Linfen, China
| |
Collapse
|
40
|
Shi J, Sun S, Liao Y, Tang J, Xu X, Qin B, Qin C, Peng L, Luo M, Bai L, Xie F. Advanced oxidation protein products induce G1 phase arrest in intestinal epithelial cells via a RAGE/CD36-JNK-p27kip1 mediated pathway. Redox Biol 2019; 25:101196. [PMID: 31014575 PMCID: PMC6859530 DOI: 10.1016/j.redox.2019.101196] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/28/2019] [Accepted: 04/08/2019] [Indexed: 01/06/2023] Open
Abstract
Intestinal epithelial cell (IEC) cycle arrest has recently been found to be involved in the pathogenesis of Crohn's disease (CD). However, the mechanism underlying the regulation of this form of cell cycle arrest, remains unclear. Here, we investigated the roles that advanced oxidation protein products (AOPPs) may play in regulating IEC cycle arrest. Plasma AOPPs levels and IEC cycle distributions were evaluated in 12 patients with CD. Molecular changes in various cyclins, cyclin-dependent kinases (CDKs), and other regulatory molecules were examined in cultured immortalized rat intestinal epithelial (IEC-6) cells after treatment with AOPPs. The in vivo effects exerted by AOPPs were evaluated using a normal C57BL/6 mouse model with an acute AOPPs challenge. Interestingly, plasma AOPPs levels were elevated in active CD patients and correlated with IEC G1 phase arrest. In addition, IEC treatment with AOPPs markedly reduced the expression of cyclin E and CDK2, thus sensitizing epithelial cells to cell cycle arrest both in vitro and in vivo. Importantly, we found that AOPPs induced IEC G1 phase arrest by modulating two membrane receptors, RAGE and CD36. Furthermore, phosphorylation of c-jun N-terminal kinase (JNK) and the expression of p27kip1 in AOPPs-treated cells were subsequently increased and thus affected cell cycle progression. Our findings reveal that AOPPs influence IEC cycle progression by reducing cyclin E and CDK2 expression through RAGE/CD36-depedent JNK/p27kip1 signaling. Consequently, AOPPs may represent a potential therapeutic molecule. Targeting AOPPs may offer a novel approach to managing CD.
Collapse
Affiliation(s)
- Jie Shi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Shibo Sun
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yan Liao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jing Tang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaoping Xu
- Department of Gastroenterology, Hunan Provincial People's Hospital, Changsha, Hunan, 410005, China
| | - Biyan Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Caolitao Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lishan Peng
- Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Mengshi Luo
- Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lan Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Fang Xie
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
41
|
Inhibitory effect of melatonin on Mst1 ameliorates myocarditis through attenuating ER stress and mitochondrial dysfunction. J Mol Histol 2019; 50:405-415. [PMID: 31256303 DOI: 10.1007/s10735-019-09836-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/20/2019] [Indexed: 12/27/2022]
Abstract
Viral myocarditis has been found to be one of the leading causes of sudden death in young adults. However, no effective drugs have been developed to intervene the progression of myocarditis. Accordingly, the present study is carried out to explore the protective role played by melatonin in the setting of viral myocarditis with a focus on Mst1-Hippo pathway, mitochondrial dysfunction and ER stress. Cardiac function was determined via echocardiographic examination. Mitochondrial function and ER stress were detected via ELISA, western blots, and immunofluorescence. Our data demonstrated that virus injection induced cardiac dysfunction as evidenced by reduced contractile function in myocardium. Besides, LDH release assay and western blotting analysis demonstrated that cardiomyocyte death was activated by virus injection. Interestingly, melatonin treatment improved cardiac function and repressed virus-mediated cardiomyocyte apoptosis. At the molecular levels, mitochondrial dysfunction was induced by virus infection, as indicated by mitochondrial membrane potential reduction, mPTP opening rate elevation and caspase-9-related apoptosis activation. Besides, ER stress parameters were also elevated in virus-treated cardiomyocytes. Interestingly, melatonin treatment maintained mitochondrial dysfunction and repressed ER stress. To the end, we found that Mst1 was upregulated by virus infection; this effect was attenuated through supplementation with melatonin. However, Mst1 overexpression reduced the beneficial impact exerted by melatonin on cardiomyocyte viability, mitochondrial function and ER homeostasis. Our study illustrated that melatonin treatment attenuated viral myocarditis via sustaining cardiomyocyte viability, repressing mitochondrial dysfunction and inhibiting ER stress in a manner dependent on Mst1 inhibition.
Collapse
|
42
|
Feng Y, Niu R, Cheng X, Wang K, Du Y, Peng X, Chen F. ATPR-induced differentiation and G0/G1 phase arrest in acute promyelocytic leukemia by repressing EBP50/NCF1 complex to promote the production of ROS. Toxicol Appl Pharmacol 2019; 379:114638. [PMID: 31254567 DOI: 10.1016/j.taap.2019.114638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022]
Abstract
Our previous study has demonstrated that 4-amino-2-trifluoromethyl-phenyl Retinate (ATPR) can induce human leukemia NB4 cells differentiation and G0/G1 phase arrest, but the underlying mechanism is still unclear. In this study, we used proteomics to screen differentially expressed protein profiles in NB4 cells before and after ATPR treatment in vitro. We analyzed the peptides digested from total cellular proteins by reverse phase LC-MS/MS and then performed label-free quantitative analysis. We found 27 significantly up-regulated proteins in the ATPR group compared to the control group. NCF1 was the most significantly changed protein. Immunoprecipitation and double immunofluorescent staining showed that EBP50 bind to NCF1. We further explored the potential molecular mechanism of EBP50/NCF1 complex in ATPR-induced differentiation and G0/G1 phase arrest. The results showed that ATPR remarkably reduced the expression of EBP50 in vivo and in vitro. Interestingly, the reduction of EBP50 contributed to ROS release by modulating the subcellular localization of NCF1. The reduction of EBP50 also contributed to G0/G1 phase arrest by inhibiting CyclinD1, CyclinA2 and CDK4, as well as promoting the differentiation of NB4 cells by increasing the expression of CD11b. Furthermore, we found that the overexpression of EBP50 restrained the effects of ATPR on differentiation and G0/G1 phase arrest in NB4 cells. These results suggest that ATPR-induced differentiation and G0/G1 phase arrest in acute promyelocytic leukemia (APL) by repressing EBP50/NCF1 complex to promote the production of ROS, and the results from in vivo experiments were consistent with those from in vitro studies. Therefore, our finding results suggest that EBP50 may be a new target for ATPR in the treatment of APL.
Collapse
Affiliation(s)
- Yubin Feng
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Ruowen Niu
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Xin Cheng
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Ke Wang
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Yan Du
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Xiaoqing Peng
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China
| | - Feihu Chen
- The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, Anhui, China.
| |
Collapse
|
43
|
Zhao Y, Zhang L, Ouyang X, Jiang Z, Xie Z, Fan L, Zhu D, Li L. Advanced oxidation protein products play critical roles in liver diseases. Eur J Clin Invest 2019; 49:e13098. [PMID: 30838641 DOI: 10.1111/eci.13098] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/26/2019] [Accepted: 03/03/2019] [Indexed: 01/24/2023]
Abstract
There is a complex oxidant and antioxidant system that maintains the redox homoeostasis in the liver. While suffering from exogenous or endogenous risk factors, the balance between oxidants and antioxidants is disturbed and excessive reactive oxygen species are generated, resulting in oxidative stress. Oxidative stress is prevalent in various liver diseases and is thought to be involved in their pathophysiology. Advanced oxidation protein products are generated under conditions of oxidative damage and are newly described protein markers of oxidative stress. Previous studies have underscored the universal pathogenic roles of oxidation protein products in various diseases. However, investigations into how these products participate in the development of liver diseases have been superficial and insufficient. In this review, we highlight the current understanding of the roles of advanced oxidation protein products in liver disease pathogenesis and the underlying mechanisms. Moreover, we summarize the current studies on advanced oxidation protein products in infectious and noninfectious, acute and chronic liver diseases. Different strategies for targeting these advanced oxidation protein products and future perspectives, which may pave the way for developing new therapeutic strategies, will also be discussed here.
Collapse
Affiliation(s)
- Yalei Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Lingjian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xiaoxi Ouyang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Zhengyi Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Zhongyang Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Linxiao Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
44
|
Lazzarino G, Listorti I, Bilotta G, Capozzolo T, Amorini AM, Longo S, Caruso G, Lazzarino G, Tavazzi B, Bilotta P. Water- and Fat-Soluble Antioxidants in Human Seminal Plasma and Serum of Fertile Males. Antioxidants (Basel) 2019; 8:E96. [PMID: 30978904 PMCID: PMC6523754 DOI: 10.3390/antiox8040096] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) are physiologically involved in functions like sperm maturation, capacitation and acrosome reaction, but their excess is involved in male infertility. Antioxidants in seminal plasma (SP) are an important factor balancing physiologic and harmful ROS activities. In this study, we determined and compared the full profiles of the water- and fat-soluble antioxidants in SP and serum of 15 healthy fertile subjects (ranging between the ages of 35 and 42 years). Ejaculates were obtained after 2⁻5 days of sexual abstinence. After liquefaction and withdrawal of an aliquot for the sperm count, samples were centrifuged to obtain SP. Thirty min after semen donation, a venous blood sample was collected from each subject. Donors with lower SP concentrations of ascorbic acid (n = 5) or α-tocopherol (n = 5) received a 4 week oral administration of either vitamin C (100 mg/day) or vitamin E (30 mg/day). They were then re-assayed to determine the SP and serum levels of ascorbic acid and α-tocopherol. SP and serum samples were properly processed and analyzed by HPLC methods suitable to determine water (ascorbic acid, glutathione (GSH) and uric acid) and fat-soluble (all-trans-retinoic acid, all-trans-retinol, α-tocopherol, carotenoids and coenzyme Q10) antioxidants. Data demonstrate that only ascorbic acid is higher in SP than in serum (SP/serum ratio = 4.97 ± 0.88). The other water-soluble antioxidants are equally distributed in the two fluids (GSH SP/serum ratio = 1.14 ± 0.34; uric acid SP/serum ratio = 0.82 ± 0.12). All fat-soluble antioxidants are about 10 times less concentrated in SP than in serum. In donors treated with vitamin C or vitamin E, ascorbic acid and α-tocopherol significantly increased in both fluids. However, the SP/serum ratio of ascorbic acid was 4.15 ± 0.45 before and 3.27 ± 0.39 after treatment, whilst those of α-tocopherol were 0.11 ± 0.03 before and 0.10 ± 0.02 after treatment. The results of this study, by showing the peculiar composition in water- and fat-soluble antioxidants SP, indicate that it is likely that still-unknown mechanisms allow ascorbic acid accumulation in SP against a concentration gradient. SP mainly relies its defenses on water- rather than fat-soluble antioxidants and on the mechanisms ensuring their transfer from serum.
Collapse
Affiliation(s)
- Giacomo Lazzarino
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Largo F. Vito 1, 00168 Rome, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy.
| | - Ilaria Listorti
- Alma Res Fertility Center, Centro di Fecondazione Assistita Alma Res, Via Parenzo 12, 00199 Rome, Italy.
| | - Gabriele Bilotta
- Alma Res Fertility Center, Centro di Fecondazione Assistita Alma Res, Via Parenzo 12, 00199 Rome, Italy.
| | - Talia Capozzolo
- Alma Res Fertility Center, Centro di Fecondazione Assistita Alma Res, Via Parenzo 12, 00199 Rome, Italy.
| | - Angela Maria Amorini
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Salvatore Longo
- LTA-Biotech srl, Viale Don Orione, 3D, 95047 Paternò (CT), Italy.
| | - Giuseppe Caruso
- Oasi Research Institute-IRCCS, Via Conte Ruggero, 73, 94018 Troina (EN), Italy.
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
- LTA-Biotech srl, Viale Don Orione, 3D, 95047 Paternò (CT), Italy.
| | - Barbara Tavazzi
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Largo F. Vito 1, 00168 Rome, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy.
| | - Pasquale Bilotta
- Alma Res Fertility Center, Centro di Fecondazione Assistita Alma Res, Via Parenzo 12, 00199 Rome, Italy.
| |
Collapse
|
45
|
Song J, Zhao W, Lu C, Shao X. LATS2 overexpression attenuates the therapeutic resistance of liver cancer HepG2 cells to sorafenib-mediated death via inhibiting the AMPK-Mfn2 signaling pathway. Cancer Cell Int 2019; 19:60. [PMID: 30923462 PMCID: PMC6423758 DOI: 10.1186/s12935-019-0778-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022] Open
Abstract
Background Effective therapy for hepatocellular carcinoma (HCC) is currently an imperative issue, and sorafenib is a first-line drug for the treatment of HCC. However, the clinical benefit of sorafenib is often impaired by drug resistance. Accordingly, the present study was conducted to investigate the molecular mechanisms involving sorafenib resistance, with a focus on large tumor suppressor 2 (LATS2) and mitophagy. Methods HepG2 liver cancer cells were treated with sorafenib and infected with adenovirus-loaded LATS2 (Ad-LATS2). Cell death, proliferation and migration were measured via western blotting analysis, immunofluorescence and qPCR. Mitochondrial function and mitophagy were determined via western blotting and immunofluorescence. Results Our data indicated that LATS2 expression was repressed by sorafenib treatment, and overexpression of LATS2 could further enhance sorafenib-mediated apoptosis in HepG2 liver cancer cells. At the molecular level, mitochondrial stress was triggered by sorafenib treatment, as evidenced by decreased mitochondrial membrane potential, increased mitochondrial ROS production, more cyc-c release into the nucleus, and elevated mitochondrial pro-apoptotic proteins. However, in response to mitochondrial damage, mitophagy was activated by sorafenib treatment, whereas LATS2 overexpression effectively inhibited mitophagy activity and thus augmented sorafenib-mediated mitochondrial stress. Subsequently, we also demonstrated that the AMPK–MFN2 signaling pathway was involved in mitophagy regulation after exposure to sorafenib treatment and/or LATS2 overexpression. Inhibition of the AMPK pathway interrupted mitophagy and thus enhanced the antitumor property of sorafenib, similar to the results obtained via overexpression of LATS2. Conclusions Altogether, our findings revealed the importance of the LATS2/AMPK/MFN2/mitophagy axis in understanding sorafenib resistance mechanisms, with a potential application to increase the sensitivity response of sorafenib in the treatment of liver cancer.
Collapse
Affiliation(s)
- Jie Song
- 1Department of Hepatopancreatobiliary Medicine, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Wei Zhao
- 2Department of Pharmacy, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Chang Lu
- 3Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Xue Shao
- 1Department of Hepatopancreatobiliary Medicine, The Second Hospital of Jilin University, Changchun, 130000 China
| |
Collapse
|
46
|
Sun J, Li P, Yang J. Repressing of NHERF1 inhibits liver cancer progression by promoting the production of ROS. Biochem Biophys Res Commun 2019; 509:8-15. [PMID: 30581004 DOI: 10.1016/j.bbrc.2018.11.121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 11/20/2018] [Indexed: 01/13/2023]
Abstract
NHERF1/EBP50 is a PDZ-scaffold protein initially identified as an organizer and modulator of transporters and channels at the apical side of epithelia via actin-binding ezrin-moesin-radixin proteins. Presently, hepatocellular carcinoma (HCC) is one of the most deadly cancers in the world and has no effective therapeutic strategies. In the present study, we attempted to explore the role of NHERF1 in regulating liver cancer progression. The results indicated that NHERF1 was significantly expressed in liver tumor samples compared to the corresponding adjacent normal tissues. HCC patients with low NHERF1 exhibited better survival rate. Additionally, repressing NHERF1 expression markedly down-regulated the cell proliferation. G0/G1 transition was highly induced by NHERF1 knockdown, accompanied with reduced expressions of Cyclin D1 and cyclin-dependent kinase 4 (CDK4), as well as the enhanced expression of p27, phosphatase and tensin homolog (PTEN) and p53. Moreover, NHERF1 suppression significantly induced apoptosis in liver cancer cells by promoting the activation of Caspase-3 and poly (ADP-ribose) polymerase (PARP). We also observed a remarkable increase of reactive oxygen species (ROS) production in NHERF1-knockdown cells, along with c-Jun-N-terminal kinase (JNK) phosphorylation. Importantly, suppressing ROS production abolished NHERF1 knockdown-induced JNK activation. Moreover, cell cycle-regulatory proteins meditated by NHERF1 knockdown in liver cancer cells were abrogated by the pre-treatment of ROS scavenger. Further, restraining ROS generation also diminished NHERF1 knockdown-induced apoptosis. In vivo, we also found that NHERF1 knockdown markedly reduced the tumor growth. In conclusion, the results suggested that NHERF1 played an essential role in regulating liver cancer progression, and repressing NHERF1 expression exhibited significant anticancer effects via the induction of G0/G1 phase arrest, apoptosis and ROS generation.
Collapse
Affiliation(s)
- Jing Sun
- Department of Diagnostic Radiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Department of Medical Technology, Xi'an Medical University, Xi'an, 710061, China
| | - Peng Li
- Department of Medical Technology, Xi'an Medical University, Xi'an, 710061, China
| | - Jian Yang
- Department of Diagnostic Radiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
47
|
Zhang J, Pei Y, Yang W, Yang W, Chen B, Zhao X, Long S. Cytoglobin ameliorates the stemness of hepatocellular carcinoma via coupling oxidative-nitrosative stress signals. Mol Carcinog 2018; 58:334-343. [PMID: 30365183 DOI: 10.1002/mc.22931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 09/22/2018] [Accepted: 10/23/2018] [Indexed: 12/18/2022]
Abstract
Cancer stem cells (CSCs) account for tumor self-renewal and heterogeneity. Oxidative-nitrosative stress (ONS) is an independent etiologic factor throughout tumorigenesis. Emerging evidences indicated that the interaction of ONS with CSCs contributes to tumor progression and resistance to chemoradiotherapy. Cytoglobin (Cygb) is a member of human hexacoordinate hemoglobin family and acts as a dynamic mediator of redox homeostasis. We observed that Cygb is significantly deregulated in human hepatocellular carcinoma (HCC) tissue and its decrease aggravates the growth of liver cancer stem cells (LCSCs) and increases the subpopulation of CD133(+) LCSCs. Cygb restoration inhibits HCC proliferation and LCSC growth, and decreases the subpopulation of CD133 (+) LCSCs in vitro. We found that Cygb absence promotes LCSC phenotypes and PI3 K/AKT activation, whereas Cygb restoration inhibits LCSC phenotypes and PI3 K/AKT activation. Furthermore, exogenous antioxidants can eliminate the inhibitory effect of Cygb to LCSC growth and phenotypes, as well as PI3 K/AKT activation. Collectively, this study demonstrated that cytoglobin functions as a tumor suppressor and targets CSCs at an ONS-dependent manner. Thus, Cygb restoration could be a novel and promising therapeutic strategy against HCC with aberrant ROS/RNS accumulation.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Pathology, the Affiliated Hospital of Guizhou Medical University, Guiyang, PR China.,Department of Pathology, Graduate School of Medicine, Guizhou Medical University, Guiyang, PR China.,Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences/Stem Cell and Tissue Engineering Research Center, Guizhou Medical University, Guiyang, PR China
| | - YuanYuan Pei
- Department of Pathology, the Affiliated Hospital of Guizhou Medical University, Guiyang, PR China
| | - Wen Yang
- Department of Pathology, the Affiliated Hospital of Guizhou Medical University, Guiyang, PR China
| | - WenXiu Yang
- Department of Pathology, the Affiliated Hospital of Guizhou Medical University, Guiyang, PR China.,Department of Pathology, Graduate School of Medicine, Guizhou Medical University, Guiyang, PR China
| | - BoXin Chen
- Department of Immunology, Basic School of Medicine, Guizhou Medical University, Guiyang, PR China
| | - Xing Zhao
- Department of Immunology, Basic School of Medicine, Guizhou Medical University, Guiyang, PR China
| | - Shiqi Long
- Department of Immunology, Basic School of Medicine, Guizhou Medical University, Guiyang, PR China
| |
Collapse
|
48
|
Li C, Liu Q, Xie L. Suppressing NLRP2 expression accelerates hepatic steatosis: A mechanism involving inflammation and oxidative stress. Biochem Biophys Res Commun 2018; 507:22-29. [PMID: 30454891 DOI: 10.1016/j.bbrc.2018.10.132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by lipid accumulation and inflammation in the liver, contributing to a broad spectrum of severe pathologies, such as metabolic syndrome and hepatocellular carcinoma. Presently, the pathogenesis that attributes to NAFLD has not been fully understood. NLRP2 has been shown to inhibit the NF-κB signaling, and thus may contribute to regulate the inflammatory response. However, its role in NAFLD is largely unclear. In the study, we found that NLRP2 was markedly decreased in liver tissues of individuals with severe steatosis, or in a genetic deficiency (ob/ob) mice. High fat diet (HFD) feeding also led to a significant reduction of NLRP2 in liver of mice. Then, the wild type (WT) and NLRP2 knockout (KO) mice were used to further explore the role of NLRP2 in the NAFLD progression. NLRP2 knockout mice exhibited severer metabolic syndrome and hepatic steatosis after HFD administration, as evidenced by the increased body weight, liver histological changes and lipid accumulation. Moreover, HFD feeding-induced inflammation was significantly accelerated by the loss of NLRP2, as evidenced by the increased expression of pro-inflammatory cytokines and activation of nuclear factor κB (NF-κB) pathway. In addition, oxidative stress triggered by HFD was further promoted by NLRP2 deletion through repressing NF-E2-related factor 2 (Nrf2) pathway. In vitro, we surprisingly found that promoting Nrf2 activation could attenuate NLRP2 knockout-accelerated inflammation and reactive oxygen species (ROS) generation. Therefore, our study indicated that NLRP2 might be a potential target for developing effective therapeutic strategy to prevent NAFLD progression.
Collapse
Affiliation(s)
- Chen Li
- Department of Gastroenterology, Xuzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, 221000, China
| | - Qing Liu
- Department of Oncology, Xuzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, 221000, China
| | - Liqun Xie
- Department of Traditional Chinese Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210009, China.
| |
Collapse
|
49
|
Zhang Q, Sun S, Zhu C, Xie F, Cai Q, Sun H, Chen G, Liang X, Xie H, Shi J, Liao Y, Zhou J. Expression of Allograft Inflammatory Factor-1 (AIF-1) in Hepatocellular Carcinoma. Med Sci Monit 2018; 24:6218-6228. [PMID: 30188879 PMCID: PMC6139115 DOI: 10.12659/msm.908510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Allograft inflammatory factor-1 (AIF-1) is a cytoplasmic protein cloned from activated macrophages in human and rat allografts. AIF-1 has been identified as a modulator of inflammatory response, and recently published studies have shown its increased expression in carcinogenesis. However, there are still limited data on the potential functional role of AIF-1 in hepatocellular carcinoma (HCC). Material/Methods We evaluated the expression of AIF-1 in 104 cases of paired HCC and adjacent non-cancerous liver tissues using immunohistochemistry, Western blotting, and qPCR analysis, and sought to determine whether its expression was correlated with clinicopathological features. In vitro assays, including cell proliferation and migration assays, were used to study the effects of AIF-1 knockdown in L02 human hepatocyte, and Huh7 and SMMC7721 liver cancer cell lines. Results Expression of AIF-1 was increased in HCC compared to adjacent normal liver tissues and was positively correlated with median tumor size (p=0.046), number of tumor deposits (p=0.009), the Barcelona Clinic Liver Cancer (BCLC) stage (p=0.004), and portal vein tumor thrombus (PVTT) (p<0.001). Huh7 and SMMC7721 human HCC cells demonstrated upregulated AIF-1 expression compared to normal hepatocytes. Small interfering RNA (siRNA)-mediated silencing of AIF-1 expression resulted in a reduction in cell proliferation and migration in human HCC cells. Conclusions These findings suggest AIF-1 may have roles as a diagnostic or prognostic biomarker and a promising therapeutic target in HCC.
Collapse
Affiliation(s)
- Qifan Zhang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Shibo Sun
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Chen Zhu
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Fang Xie
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Qing Cai
- Department of Hepatobiliary Surgery and Liver Transplantation Center, Guangzhou General Hospital of Guangzhou Military Area, Guangzhou, Guangdong, China (mainland)
| | - Hang Sun
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Gang Chen
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Xiaolu Liang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Haorong Xie
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Jie Shi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Yan Liao
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Jie Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
50
|
Huang AM, Lin KW, Lin WH, Wu LH, Chang HC, Ni C, Wang DL, Hsu HY, Su CL, Shih C. 1-Hydroxy-3-[( E )-4-(piperazine-diium)but-2-enyloxy]-9,10-anthraquinone ditrifluoroactate induced autophagic cell death in human PC3 cells. Chem Biol Interact 2018; 281:60-68. [DOI: 10.1016/j.cbi.2017.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/27/2017] [Accepted: 12/05/2017] [Indexed: 01/19/2023]
|