1
|
Chen X, Xie X, Liu C, Chen L, Zhang M, Zhang Y. Maduramicin ammonium impairs autophagic flux through activating AMPK-mediated eIF2α-ATF4 endoplasmic reticulum stress pathway in skeletal muscle. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:2952-2959. [PMID: 39660505 DOI: 10.1002/jsfa.14071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Maduramicin ammonium (MA), a widely used coccidiostat, has been reported to cause skeletal muscle degeneration in animals and even humans. In this study, we explore the underlying mechanism of its toxicity in skeletal muscle. RESULTS First, we observed that MA impaired autophagic flux which was evidenced by increased protein level of LC3-II and p62 in skeletal myoblast C2C12 and L6 cell lines and rectus femoris muscle tissues of rats and broilers. Then, we found that MA induced eIF2α phosphorylation and ATF4 expression in the cells and tissues. Co-treatment with ISRIB attenuated MA-induced LC3-II and p62 in C2C12 and L6 cells, suggesting that MA-induced eIF2α-ATF4 pathway contributed to impairment of autophagic flux in the cells. Lastly, we showed that MA activated AMPK signaling in skeletal muscle, since the phosphorylation of AMPK was increased by MA treatment in skeletal myoblast cell lines and muscle tissues. Furthermore, in AMPK downregulated C2C12 cells, MA-induced LC3-II, ATF4 and phosphorylation of eIF2α was reversed, supporting that AMPK was involved in the regulation of the eIF2α-ATF4 pathway and autophagic flux during MA exposure. CONCLUSION Our findings showed that MA impairs autophagic flux through activating the AMPK-induced eIF2α-ATF4 pathway in skeletal muscle. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xin Chen
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| | - Xin Xie
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
| | - Chang Liu
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
| | - Lin Chen
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
| | - Meng Zhang
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
| | - Yumei Zhang
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
2
|
Xie ZF, Liu HM, Zhao JF, Gao Y, Zhao YL, Zheng JY, Pei XW, Zhang N, Tian G. AMD1, a cardiotoxicity target for Maduramicin. BMC Pharmacol Toxicol 2025; 26:55. [PMID: 40069794 PMCID: PMC11895246 DOI: 10.1186/s40360-025-00897-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/07/2025] [Indexed: 03/15/2025] Open
Abstract
OBJECTIVE The aim of this study was to investigate AMD1 cardiotoxicity function for Maduramicin (Mad). METHODS SD rats were divided into control (Control) group and Mad treatment (3.5 mg/kg) group (Mad). After treatment with Mad for seven days, the levels of LDH and CK-MB in serum were detected, H&E staining and TUNEL staining were performed. In vitro, 1.0 μm Mad was used for the subsequently experiment, observing cell apoptosis from Flow cytometry. Caspase-3 and AMD1 were detected in Western blotting. Flow cytometry and Western blotting were also performed after use of siRNA-AMD1-1. Then, analysis AMD1 potential function in cardiotoxicity from bioinformatics techniques including GO, KEGG, PPI, immune infiltration and molecular docking. RESULT Maduramicin has myocardial toxic effects in vivo and vitro, which with AMD1 raised. When AMD1 was knocked down, toxic effects of Mad were alleviated. Apoptosis, proliferation and inflammation were the major pathophysiological changes in myocardial apoptosis process with AMD1-knockdown. This process involved in IL1A, IL1B, PTGS2, VEGFA, VEGFC and HBEFG, as hub genes related AMD1 cardiotoxicity function for Maduramicin. AMD1 was knocked down, their microenvironment changes: Effector memory CD4 T cell and Natural killer cell were more infiltrated, and Mast cell were less infiltrated. CONCLUSION Mad exerted cardiotoxic effects by upregulating the AMD1 gene, which may be associated with cell apoptosis, proliferation and inflammatory response. AMD1 also had cardiotoxicity function, by the impact of both myocardial cells and the microenvironment they live.
Collapse
Affiliation(s)
- Zi-Feng Xie
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Han-Meng Liu
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Jia-Fan Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Yuan Gao
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Yuan-Long Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Jia-Yue Zheng
- Stomatology Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Xiao-Wei Pei
- Department of Physical Medicine and Rehabilitation, Linghai Daling River Hospital, Linghai, Liaoning, 121200, China
| | - Ning Zhang
- Department of Hematology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Ge Tian
- Department of Cardiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, China.
| |
Collapse
|
3
|
Wang K, Wen J, Liang T, Hu H, Li S, Shen L, Ren T, Yao Y, Xie J, Ding J, Chen J, Tang YD, Zhu Y, Gao C. Enhancing miR-19a/b induced cardiomyocyte proliferation in infarcted hearts by alleviating oxidant stress and controlling miR-19 release. Biomaterials 2025; 312:122732. [PMID: 39088913 DOI: 10.1016/j.biomaterials.2024.122732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/03/2024]
Abstract
Fully restoring the lost population of cardiomyocytes and heart function remains the greatest challenge in cardiac repair post myocardial infarction. In this study, a pioneered highly ROS-eliminating hydrogel was designed to enhance miR-19a/b induced cardiomyocyte proliferation by lowering the oxidative stress and continuously releasing miR-19a/b in infarcted myocardium in situ. In vivo lineage tracing revealed that ∼20.47 % of adult cardiomyocytes at the injected sites underwent cell division in MI mice. In MI pig the infarcted size was significantly reduced from 40 % to 18 %, and thereby marked improvement of cardiac function and increased muscle mass. Most importantly, our treatment solved the challenge of animal death--all the treated pigs managed to live until their hearts were harvested at day 50. Therefore, our strategy provides clinical conversion advantages and safety for healing damaged hearts and restoring heart function post MI, which will be a powerful tool to battle cardiovascular diseases in patients.
Collapse
Affiliation(s)
- Kai Wang
- The State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou 310009, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jun Wen
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Tian Liang
- Department of Cardiology, the Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shifen Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Liyin Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Tanchen Ren
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, the Second Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jieqi Xie
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jinghai Chen
- Department of Cardiology, the Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Yi-Da Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Yang Zhu
- The State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou 310009, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China.
| | - Changyou Gao
- The State Key Laboratory of Transvascular Implantation Devices, Zhejiang University, Hangzhou 310009, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312099, China.
| |
Collapse
|
4
|
Cheng B, Zhang H, Zhao W, Jiang S, Wu Z, Li H, Liu S, Zhang H. The highly hazardous veterinary drug "maduramicin" and its toxicokinetics in rats. Heliyon 2024; 10:e39620. [PMID: 39640695 PMCID: PMC11620213 DOI: 10.1016/j.heliyon.2024.e39620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 12/07/2024] Open
Abstract
Background Maduramicin (MAD) is an anticoccidial veterinary drug, but it frequently causes fatal poisonings in poultry, livestock, or humans. However, there is no specific antidote or guidance on first aid for MAD poisoning. Aim The aim of the present study is to evaluate the acute toxicity and toxicokinetics of MAD after oral exposure, so as to make a foundation for developing diagnostic and therapeutic protocols for human intoxication. Methods Five groups of rats (eight-to-nine-week-old male Wistar rats) were orally administered MAD via gavage at doses of 0, 4.64, 10.0, 21.5, or 46.4 mg/kg bw for only one time. The survival rates of the rats were observed over the following 14 days to assess acute toxicity. To evaluate the toxic effects of MAD, two doses (4.8 mg/kg bw and 10 mg/kg bw) were orally administered via gavage. Biochemical parameters including creatine kinase, lactate dehydrogenase, aspartate aminotransferase, alanine aminotransferase, urea, creatinine, serum myoglobin, and urinary myoglobin were measured. Liver, kidney, heart, and hind limb skeletal muscle samples from severely poisoned rats were obtained for pathological examination. For toxicokinetic analysis, samples of serum, urine, and feces from the 4.8 mg/kg bw dose group were analyzed using high-performance liquid chromatography-tandem mass spectrometry. Results The LD50 of MAD in male Wistar rats was determined to be 6.81 mg/kg bw. In the 10 mg/kg bw group, elevated serum urea levels and increased myoglobin levels in both serum and urine indicated renal injury and potential muscle damage. Toxicokinetics in serum revealed that following oral administration of 4.8 mg/kg bw MAD, peak serum concentration of 59.8 ± 8.9 μg/L was achieved at 30.0 ± 13.9 h. MAD exhibited a slow elimination from the blood with an elimination half-life of 72.9 ± 36.8 h and a mean residence time of 79.6 ± 25.5 h. Additionally, fecal excretion of MAD was found to be greater than urinary excretion. Conclusion MAD is a highly toxic veterinary drug which requires careful handling. The primary effects of poisoning include kidney injury and suspected rhabdomyolysis. It is excreted very slowly after oral administration. Promoting toxin excretion in individuals poisoned by MAD could potentially serve as an effective treatment method until a specific antidote is identified.
Collapse
Affiliation(s)
- Bowen Cheng
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Huarui Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Wenjin Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Shaofeng Jiang
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Zhijun Wu
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Huiling Li
- Occupational Disease and Poisoning Department of Beijing Chaoyang Hospital, Beijing 100020, China
| | - Shuai Liu
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Hongshun Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| |
Collapse
|
5
|
He S, Chen Y, Wang H, Li S, Wei Y, Zhang H, Gao Q, Wang F, Zhang R. Neuroprotective effects of chlorogenic acid: Modulation of Akt/Erk1/2 signaling to prevent neuronal apoptosis in Parkinson's disease. Free Radic Biol Med 2024; 222:275-287. [PMID: 38925315 DOI: 10.1016/j.freeradbiomed.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024]
Abstract
As a prevalent neurodegenerative disorder, Parkinson's disease is associated with oxidative stress. Our recent investigations revealed that reactive oxygen species (ROS) and PD-toxins like 6-hydroxydopamine (6-OHDA) can induce neuronal apoptosis through over-activation of Akt signaling. Chlorogenic acid (CGA), a natural acid phenol abundant in the human diet, is well-documented for its ability to mitigate intracellular ROS. In this study, we utilized CGA to treat experimental models of PD both in vitro and in vivo. Our study results demonstrated that SH-SY5Y and primary neurons exhibited cell apoptosis in response to 6-OHDA. Pretreatment with CGA significantly attenuated PD toxins-induced large amount of ROS, inhibiting Erk1/2 activation, preventing Akt inhibition, and hindering neuronal cell death. Combining the Erk1/2 inhibitor U0126 with CGA could reverse 6-OHDA-induced Akt inhibition, ROS, and apoptosis in the cells. Crucially, the Akt activator SC79 and ROS scavenger NAC both could eliminate excessive ROS via Akt and Erk1/2 signaling pathways, and CGA further potentiated these effects in PD models. Behavioral experiments revealed that CGA could alleviate gait abnormalities in PD model mice. The neuroprotective effects have been demonstrated in several endocrine regions and in the substantia nigra tissue, which shows the positive tyrosine hydroxylase (TH). Overall, our results suggest that CGA prevents the activation of Erk1/2 and inactivation of Akt by removing excess ROS in PD models. These findings propose a potential strategy for mitigating neuronal degeneration in Parkinson's disease by modulating the Akt/Erk1/2 signaling pathway through the administration of CGA and/or the use of antioxidants to alleviate oxidative stress.
Collapse
Affiliation(s)
- Shuai He
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Yuxiang Chen
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Hui Wang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Shupei Li
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Yu Wei
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Hui Zhang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Qian Gao
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Fengsong Wang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China.
| | - Ruijie Zhang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China.
| |
Collapse
|
6
|
Wang W, Zheng P, Yan L, Chen X, Wang Z, Liu Q. Mechanism of non-thermal atmospheric plasma in anti-tumor: influencing intracellular RONS and regulating signaling pathways. Free Radic Res 2024; 58:333-353. [PMID: 38767976 DOI: 10.1080/10715762.2024.2358026] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
Non-thermal atmospheric plasma (NTAP) has been proven to be an effective anti-tumor tool, with various biological effects such as inhibiting tumor proliferation, metastasis, and promoting tumor cell apoptosis. At present, the main conclusion is that ROS and RNS are the main effector components of NTAP, but the mechanisms of which still lack systematic summary. Therefore, in this review, we first summarized the mechanism by which NTAP directly or indirectly causes an increase in intracellular RONS concentration, and the multiple pathways dysregulation (i.e. NRF2, PI3K, MAPK, NF-κB) induced by intracellular RONS. Then, we generalized the relationship between NTAP induced pathways dysregulation and the various biological effects it brought. The summary of the anti-tumor mechanism of NTAP is helpful for its further research and clinical transformation.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Peijia Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Liang Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaoman Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhicheng Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Qi Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Niu J, Guo W, Lu A, Han G, Wang G, Peng B, Zhao J. Comparison with gastric cancer-associated genes reveals the role of ferroptosis-related genes in eosinophils of asthma patients: A bioinformatic study. Medicine (Baltimore) 2023; 102:e35002. [PMID: 37832131 PMCID: PMC10578675 DOI: 10.1097/md.0000000000035002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/08/2023] [Indexed: 10/15/2023] Open
Abstract
Ferroptosis-inducing agents (FIAs) induced lipid-peroxidation-independent ferroptosis in eosinophils, thus ameliorating airway inflammation in asthmatic mice. Differences in ferroptosis-related genes (FerrGs) between eosinophils and cells in which FIAs induce canonical ferroptosis are supposed to contribute to this noncanonical ferroptosis but remain unclear. This study aims to explore these differences. This study used gastric cancer cells (GCCs) in stomach adenocarcinoma as the representative of cells in which FIAs induce canonical ferroptosis. FerrGs in Ferroptosis Database V2 respectively intersected with differentially expressed genes (DEGs) of eosinophils (E-MTAB-4660 dataset) and GCCs (GEPIA2 Stomach adenocarcinoma dataset) to obtain original ferroptosis DEGs (FerrDEGs). Then, they were subjected to Venn analysis to identify FerrDEGs shared by them and FerrDEGs exclusively expressed in eosinophils or GCCs. Identified genes were subjected to functional enrichment analysis, protein-protein interactions analysis, Hub genes analysis, and construction of the LncRNA-mediated ceRNA network. Sixty-six original FerrDEGs in eosinophils and 110 original FerrDEGs in GCCs were obtained. Venn analysis identified that eosinophils and GCCs shared 19 FerrDEGs that presented opposite expression directions and were involved in the ferroptosis pathway. Four upregulated and 20 downregulated FerrDEGs were exclusively expressed in eosinophils and GCCs, respectively. The former were enriched only in glycerolipid metabolism, while the latter were not enriched in pathways. Forty downregulated and 68 upregulated FerrDEGs were solely expressed in eosinophils and GCCs, respectively. The former was associated with the FoxO signaling pathway; the latter was related to glutathione metabolism and they were all implicated in autophagy. PPI analysis shows that the top 10 Hub genes of 66 original FerrDEGs and 44 exclusive FerrDEGs in eosinophils shared 9 genes (STAT3, NFE2L2, MAPK8, PTEN, MAPK3, TLR4, SIRT1, BECN1, and PTGS2) and they were also involved in the FoxO signaling pathway and autophagy pathway. Among them, PTEN is involved in forming a ceRNA network containing 3 LncRNAs, 3 miRNAs and 3 mRNAs. In contrast to FerrGs in cells in which FIAs induce canonical ferroptosis, the FerrGs in eosinophils differ in expression and in the regulation of ferroptosis, FoxO signaling pathway, and autophagy. It lays the groundwork for targeted induction of eosinophils lipid-peroxidation-independent ferroptosis in asthma.
Collapse
Affiliation(s)
- Jianfei Niu
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Wei Guo
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Aiyangzi Lu
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Guanxiong Han
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Guanqun Wang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Bihui Peng
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Jiping Zhao
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Jiang W, Zhong S, Chen Z, Qian J, Huang X, Zhang H, Wen L, Zhang Y, Yao G. 2D-CuPd nanozyme overcome tamoxifen resistance in breast cancer by regulating the PI3K/AKT/mTOR pathway. Biomaterials 2023; 294:121986. [PMID: 36623325 DOI: 10.1016/j.biomaterials.2022.121986] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/28/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023]
Abstract
Tamoxifen is the most commonly used treatment for estrogen-receptor (ER) positive breast cancer patients, but its efficacy is severely hampered by resistance. PI3K/AKT/mTOR pathway inhibition was proven to augment the benefit of endocrine therapy and exhibited potential for reversing tamoxifen-induced resistance. However, the vast majority of PI3K inhibitors currently approved for clinical use are unsatisfactory in terms of safety and efficacy. We developed two-dimensional CuPd (2D-CuPd) nanosheets with oxidase and peroxidase nanozyme activities to offer a novel solution to inhibit the activity of the PI3K/AKT/mTOR pathway. 2D-CuPd exhibit superior dual nanozyme activities converting hydrogen peroxide accumulated in drug-resistant cells into more lethal hydroxyl radicals while compensating for the insufficient superoxide anion produced by tamoxifen. The potential clinical utility was further demonstrated in an orthotopically implanted tamoxifen-resistant PDX breast cancer model. Our results reveal a novel nanozyme ROS-mediated protein mechanism for the regulation of the PI3K subunit, illustrate the cellular pathways through which increased p85β protein expression contributes to tamoxifen resistance, and reveal p85β protein as a potential therapeutic target for overcoming tamoxifen resistance. 2D-CuPd is the first reported nanomaterial capable of degrading PI3K subunits, and its high performance combined with further materials engineering may lead to the development of nanozyme-based tumor catalytic therapy.
Collapse
Affiliation(s)
- Wenwei Jiang
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, P. R. China
| | - Suqin Zhong
- School of Medicine, School of Biomedical Sciences and Engineering, South China University of Technology, 510006, Guangzhou, P. R. China
| | - Ziying Chen
- School of Medicine, School of Biomedical Sciences and Engineering, South China University of Technology, 510006, Guangzhou, P. R. China
| | - Jieying Qian
- School of Medicine, School of Biomedical Sciences and Engineering, South China University of Technology, 510006, Guangzhou, P. R. China
| | - Xiaowan Huang
- School of Medicine, School of Biomedical Sciences and Engineering, South China University of Technology, 510006, Guangzhou, P. R. China
| | - Hao Zhang
- School of Medicine, School of Biomedical Sciences and Engineering, South China University of Technology, 510006, Guangzhou, P. R. China
| | - Longping Wen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, P. R. China.
| | - Yunjiao Zhang
- School of Medicine, School of Biomedical Sciences and Engineering, South China University of Technology, 510006, Guangzhou, P. R. China; National Engineering Research Center for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, 510006, Guangzhou, P. R. China.
| | - Guangyu Yao
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, P. R. China.
| |
Collapse
|
9
|
Pan L, Sun X, Che H, Li M. CTRP9 mitigates vascular endothelial cell injury in patients with hypertensive heart disease by inhibiting PI3K/Akt/mTOR axis. Am J Transl Res 2022; 14:6596-6603. [PMID: 36247301 PMCID: PMC9556454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 03/26/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To investigate the mechanism of factor-alpha-related protein 9 (CTRP9) in mitigating the vascular endothelial cell (VEC) injury in patients with hypertensive heart disease (HHD) by the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) axis. METHODS 43 patients with HHD admitted to our hospital from February 2018 to February 2019 were included in the study group, and another 39 healthy controls from the same period were the reference group. The total protein of transfected VECs was detected by western blotting, and the proliferation rate of the VECs was determined by Cell Counting Kit-8 (CCK-8). The levels of CTRP9, high sensitivity C-reactive protein (hs-CRP), thrombomodulin (TM), and von Willebrand factor (vWF) were detected by ELISA. The mechanism of CTRP9 in alleviating VEC injury in HHD patients by inhibiting the PI3K/Akt/mTOR axis was analyzed. RESULTS The two groups did not differ in terms of their general data (P>0.05). The CTRP9 level in the study group was higher than in the reference group (P<0.001). Study group had higher levels of endothelin-1 (ET-1), hs-CRP, TM, vWF (P<0.001), and markedly lower phospho-PI3K (p-PI3K) and phospho-protein kinase B (p-AKT) protein levels (P<0.05). Compared to the reference group, the proliferation capacity of trophoblast cells in the study group was sharply decreased (P<0.05). The study group had lower phosphorylation levels of PI3K, Akt, and mTOR proteins than the reference group (P<0.05). Phosphorylation of Akt occurred at 15 min and reached its peak at 30 min. A drastically reduced invasion capacity of VECs was observed in the study group compared to the reference group (P<0.05). CONCLUSIONS CTRP9 mitigates VEC injury in patients with HHD by inhibiting the PI3K/Akt/mTOR axis.
Collapse
Affiliation(s)
- Lingyun Pan
- Department of Emergency Medicine, The Second People’s Hospital of Liaocheng, The Second Hospital of Liaocheng Affiliated to Shandong First Medical UniversityLiaocheng, Shandong, China
| | - Xiaocui Sun
- Department of Emergency Medicine, The Second People’s Hospital of Liaocheng, The Second Hospital of Liaocheng Affiliated to Shandong First Medical UniversityLiaocheng, Shandong, China
| | - Haixia Che
- Department of Emergency Medicine, The Second People’s Hospital of Liaocheng, The Second Hospital of Liaocheng Affiliated to Shandong First Medical UniversityLiaocheng, Shandong, China
| | - Mingzhe Li
- Shandong ENT Hospital Affiliated to Shandong UniversityJinan, Shandong, China
| |
Collapse
|
10
|
Chen K, Li G, Cui H, Du Y, Zheng J, Zhang Q, Li F, Shang H, Lei H. Systems pharmacology and GC-MS metabolomics reveal the efficacy and mechanisms of zedoary oil on acute liver injury induced by oxidative stress. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154295. [PMID: 35802998 DOI: 10.1016/j.phymed.2022.154295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Zedoray oil (ZO) is the main component of Curcuma zedoaria, one traditional herb used for dispersing stasis clinically in China. Previously, the potential of ZO was discovered against lethal and acute liver injury (ALI) mice with little impact on the immune, which deserved further study. METHODS An approach combined systems pharmacology with GC-MS metabolomics was applied for predicting pathways affected by ZO. Subsequently, H2O2 and tertbutyl hydroperoxide (t-BHP) were respectively applied to induce the ALI model in vitro for validation. RESULTS First, systems pharmacology and intracellular metabolites suggested that ZO might regulate oxidative stress via PI3K/Akt/FoxO1 pathway, TCA cycle, pantothenate, and CoA biosynthesis, beta-alanine metabolism, and propanoate metabolism. Further, levels of ALT, AST, ROS, T-AOC, MDA, GR, ΔΨm, and related proteins affected by ZO had been detected to validate the above mechanisms using dual cell models. CONCLUSION ZO could protect the L02 cells against ALI by regulating the PI3K/Akt/FoxO1 pathway, as well as restore the function of mitochondria and redox imbalance damaged by toxicants. This work has uncovered the nonimmune mechanisms of ZO against ALI to provide the basis for relevant research and disease treatment.
Collapse
Affiliation(s)
- Kedian Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Guoping Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Chinese Academy of Inspection and Quarantine, Beijing 100176, China
| | - Herong Cui
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Yawen Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaxin Zheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qi Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Feifei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Haimin Lei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
11
|
Shen Y, Wang X, Shen X, Wang Y, Wang S, Zhang Y, Yao X, Xu Y, Sang M, Pan J, Qin Y, Zhou Q, Shen J. Geniposide Possesses the Protective Effect on Myocardial Injury by Inhibiting Oxidative Stress and Ferroptosis via Activation of the Grsf1/GPx4 Axis. Front Pharmacol 2022; 13:879870. [PMID: 35600863 PMCID: PMC9117627 DOI: 10.3389/fphar.2022.879870] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/30/2022] [Indexed: 12/23/2022] Open
Abstract
Reactive oxygen species (ROS) produced in the ischemic myocardium can induce cardiomyocyte injury and death, resulting in cardiac remodeling. Ferroptosis, known as a newly type of cell death caused by iron-dependent oxidative stress, which is an essential death mechanism in cardiomyocytes. However, it is unclear whether oxidative stress products can further induce ferroptosis and aggravate cardiomyocyte injury. Geniposide (GEN), a major active component of Gardenia jasminoides J. Ellis, possesses the natural antioxidant activity and cardioprotective effect. Herein, we evaluated the role of ferroptosis in myocardial oxidative injury and the protective effect of GEN on myocardial ferroptosis. We first detected iron overload, massive ROS, and lipid peroxidation in ferric ammonium citrate (FAC)-treated cardiomyocytes, which were typical characteristics of ferroptosis. The iron overload-induced oxidative stress and ferroptosis aggravated cardiomyocyte injury, which were significantly alleviated by GEN treatment. Similar phenotypic changes of ferroptosis were consistently discovered in hydrogen peroxide (H2O2)-induced cells, which were reversed by GEN treatment as well. Interestingly, the RNA-binding protein Grsf1, which directly upregulated Gpx4 at the translational level, was activated by GEN following myocardial oxidative injury. The specific knockdown of Grsf1 increased their sensitivity to ferroptosis and weakened the cardioprotective effect of GEN in H2O2-treated cardiomyocytes. Moreover, GEN treatment reduced iron overload and lipid peroxidation in myocardial infarction (MI) rats, thereby fighting against the cardiac ischemic injury. Collectively, our study revealed the pathogenesis of oxidative stress and ferroptosis associated with myocardial ischemia, and indicated the antioxidant and anti-ferroptosis effects of GEN on preventing myocardial injury by activating the Grsf1/GPx4 axis, serving as a potential therapeutic target.
Collapse
Affiliation(s)
- Yuehong Shen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Xindong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Xinyu Shen
- Department of Biostatistics, School of Global Public Health, New York University, New York, NY, United States
| | - Yue Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Shulin Wang
- Zhenjiang Hospital Affiliated to Nanjing University of Chinese Medicine (Zhenjiang Hospital of Traditional Chinese Medicine), Zhenjiang, China
| | - Yunyun Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Xiaoming Yao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Yijiao Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Ming Sang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Jiamin Pan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Yu Qin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
| | - Qian Zhou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
- *Correspondence: Qian Zhou, ; Jianping Shen,
| | - Jianping Shen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nangjing, China
- *Correspondence: Qian Zhou, ; Jianping Shen,
| |
Collapse
|
12
|
Peng Y, Li Y, Li H, Yu J. Shikonin attenuates kidney tubular epithelial cells apoptosis, oxidative stress, and inflammatory response through nicotinamide adenine dinucleotide phosphate oxidase 4/PTEN pathway in acute kidney injury of sepsis model. Drug Dev Res 2022; 83:1111-1124. [PMID: 35417044 DOI: 10.1002/ddr.21936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 11/10/2022]
Abstract
Natural compounds were used in the treatment of acute kidney injury (AKI) caused by sepsis. This study investigated the function of shikonin from the roots of Arnebia purpurea in sepsis-induced AKI model. The target genes of shikonin were predicted by traditional Chinese medicine integrative database (TCMID). The markers of kidney injury, oxidative stress, and inflammatory factors were measured by enzyme-linked immunosorbent assay (ELISA). The pathological changes of kidney tubules were assessed by Hematoxylin and Eosin staining. Apoptosis of kidney tubular epithelial cells (KTECs) was detected by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling. Protein expression was measured by western blot. Shikonin significantly improved kidney injury induced by cecal ligation and perforation (CLP). Besides, shikonin reduced KTECs apoptosis, malondialdehyde (MDA), reactive oxygen species (ROS), interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α) levels, while augmented SOD and IL-10 levels. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase4 (NOX4) was predicted a target gene of shikonin. The expression of NOX4 was significantly inhibited in shikonin-treated group and the levels of phosphatidylinositol 3,4,5-trisphosphate 3-phosphate and dual specificity protein phosphate (PTEN) and p-p65 were decreased, while level of p-Akt was elevated. In vitro experiments, shikonin inhibited cell apoptosis, inflammatory, and ROS in human HK-2 cells and rat TECs. Shikonin downregulated expression of NOX4, PTEN and p-p65, and upregulated p-AKT and Bcl-2 expression in HK2 cells treated with lipopolysaccharide (LPS). Moreover, overexpression of NOX4 enhanced the effect of LPS on the expression level of PTEN, p-p65, p-AKT, and Bcl-2, which was reversed by the addition of shikonin. Taken together, shikonin could improve sepsis-induced AKI in rats, and attenuate the LPS induced KTECs apoptosis, oxidative stress, and inflammatory reaction via modulating NOX4/PTEN/AKT pathway.
Collapse
Affiliation(s)
- Yanqin Peng
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuan Li
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hao Li
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Junhua Yu
- Department of Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
13
|
Liu L, Liu X, Zhao L, Liu Y. 1,8-cineole alleviates bisphenol A-induced apoptosis and necroptosis in bursa of Fabricius in chicken through regulating oxidative stress and PI3K/AKT pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112877. [PMID: 34634738 DOI: 10.1016/j.ecoenv.2021.112877] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 06/13/2023]
Abstract
Bisphenol A (BPA), an important chemical raw material, is now a ubiquitous environmental contaminant. As an endocrine disruptor similar to estrogen, BPA increases the risk of various metabolic and chronic diseases. BPA has immunotoxicity to humans and animals. 1,8-cineole (CIN) is a plant-derived monoterpene with antioxidant and antiapoptosis actions. However, there are no reports about whether CIN could antagonize the BPA-induced apoptosis and necroptosis in bursa of Fabricius (BF) of chicken. This study was to elucidate the ameliorative mechanism of CIN on the apoptosis and necroptosis in BF induced by BPA. 120 broilers (1-day-old) were randomly divided into four groups: control group, CIN group, CIN and BPA co-treatment group, and BPA group. TUNEL analysis results, histopathological variations, and the overexpression of proapoptosis biomakers (Caspase 3, Bax, Cyt-c, and p53) and necroptosis pathway-related factors (RIPK1, RIPK3, MLKL, and FADD) indicated that BPA exposure induced the apoptosis and necroptosis in chicken BF. Moreover, BPA treatment elevated the levels of oxidative stress indexes (MDA, iNOS, and NO) and weaken antioxidases activity (SOD, GPx, and CAT) and total antioxidant capacity in chicken BF. BPA administration also lessened the expression of PI3K and AKT and promoted HSPs (HSP27, HSP40, HSP60, and HSP70) activation. whereas CIN supplementation prominently mitigated BPA-caused these changes and the apoptosis and necroptosis damages. In brief, this study illuminated that CIN could protect the chicken BF against BPA-induced apoptosis and necroptosis through restraining oxidative stress and activating PI3K/AKT pathway.
Collapse
Affiliation(s)
- Lili Liu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China.
| | - Xiangling Liu
- The Second Clinical Medical School, Harbin Medical University, Harbin 150086, PR China
| | - Liangyou Zhao
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China
| | - Yuan Liu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China
| |
Collapse
|
14
|
Huang J, Zhao K, Li M, Chen Y, Liang X, Li J. Development of an immunomagnetic bead clean-up ELISA method for detection of Maduramicin using single-chain antibody in chicken muscle. FOOD AGR IMMUNOL 2021. [DOI: 10.1080/09540105.2021.1998388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Jingjie Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal–Derived Food Safety, Beijing Laboratory of Food Quality and Safety, Beijing, People’s Republic of China
| | - Kunxia Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal–Derived Food Safety, Beijing Laboratory of Food Quality and Safety, Beijing, People’s Republic of China
| | - Miao Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal–Derived Food Safety, Beijing Laboratory of Food Quality and Safety, Beijing, People’s Republic of China
| | - Yingxian Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal–Derived Food Safety, Beijing Laboratory of Food Quality and Safety, Beijing, People’s Republic of China
| | - Xueyan Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal–Derived Food Safety, Beijing Laboratory of Food Quality and Safety, Beijing, People’s Republic of China
| | - Jiancheng Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal–Derived Food Safety, Beijing Laboratory of Food Quality and Safety, Beijing, People’s Republic of China
| |
Collapse
|
15
|
Li F, Yang H, Cao Y, Li D, Ma J, Liu P. DOX-loaded silver nanotriangles and photothermal therapy exert a synergistic antibreast cancer effect via ROS/ERK1/2 signaling pathway. NANOTECHNOLOGY 2021; 33:075101. [PMID: 34749347 DOI: 10.1088/1361-6528/ac378c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
The combination of multiple therapies has been proved to be more effective than a single therapy for many cancers. This study aimed to investigate the synergistic antibreast cancer effect of doxorubicin-loaded silver nanotriangles (DOX-AgNTs) combined with near-infrared (NIR) irradiation and explore the underlying mechanism. AgNTs were prepared by a chemical method and DOX was loaded via electrostatic adsorption. Characterization was performed by transmission electron microscopy, ultraviolet-visible spectroscopy and dynamic light scattering. The viability of MDA-MB-231 cells was detected by using MTT assay to evaluate the synergistic anticancer effect of DOX-AgNTs combined with NIR irradiation. The intracellular reactive oxygen species (ROS) level and cell apoptosis were analyzed by flow cytometry. Mitochondrial membrane potential (MMP) was measured with fluorescence microscopy. The mechanism was further investigated with ROS scavenger N-acetylcysteine and specific inhibitors of extracellular signal-regulated kinase 1/2 (ERK1/2), C-jun N-terminal kinase and p38 pathways. Characterization results revealed that the prepared AgNTs were mostly triangular and the mean edge length was about 126 nm. The combination of DOX-AgNTs and NIR exhibited a superior synergistic anticancer effect over single DOX-AgNTs or photothermal therapy (PTT). N-acetylcysteine and ERK1/2 inhibitor U0126 were found to significantly rescue the decreased cell viability, declined MMP and increased apoptosis induced by the combined treatment. Our results suggested that DOX-AgNTs combined with PTT performed a synergistic antibreast cancer effect. The synergy might be closely associated with the excessive production of ROS, changed MMP and the activation of ERK1/2 signaling pathway. These findings might provide a new perspective for the development of breast cancer treatments with excellent efficacy.
Collapse
Affiliation(s)
- Fan Li
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Huiquan Yang
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Yuyu Cao
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Dongdong Li
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Jing Ma
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, People's Republic of China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
16
|
Yiming Z, Zhaoyi L, Jing L, Jinliang W, Zhiqiang S, Guangliang S, Shu L. Cadmium induces the thymus apoptosis of pigs through ROS-dependent PTEN/PI3K/AKT signaling pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:39982-39992. [PMID: 33765263 DOI: 10.1007/s11356-021-13517-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/15/2021] [Indexed: 06/12/2023]
Abstract
Cadmium (Cd) is a transition metal that is toxic to living organisms in the environment and endangers living organisms. To explore whether Cd induces apoptosis in pig thymus and its possible mechanism, the role Cd induction of the PTEN/PI3K/Akt pathway in apoptosis of thymus cells was studied in pigs. We found that Cd exposure (the feed is treated with Cd) significantly increased Cd accumulation in the thymus of pigs. The TUNEL assay confirmed the typical apoptotic characteristics of thymus in Cd group. Moreover, in the Cd group, the activities of antioxidant indices decreased significantly, while the levels of oxidative stress indexes increased significantly, and the mRNA levels of GSH, CAT, Gpx1, GST, SOD1, and SOD2 decreased obviously. Moreover, the mRNA and protein levels of PTEN/PI3K/AKT and apoptosis-related genes were detected by qPCR and western blotting. The results show that the expressions of PI3K and AKT decreased, while the expression of PTEN increased, indicating that pathway activated. With the PTEN/PI3K/AKT pathway regulating, Bcl-2 expression decreased. Conversely, the mRNA and protein expression of apoptosis-related genes were up-regulated. In conclusion, accumulation of Cd in the pigs caused oxidative damage to immune tissues. In addition, Cd-induced oxidative stress activates the PTEN/PI3K/AKT pathway, inducing apoptosis in the thymus of pigs.
Collapse
Affiliation(s)
- Zhang Yiming
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Liu Zhaoyi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Lan Jing
- Quality and Safety Institute of Agricultural Products, Heilongjiang Academy of Agricultural Sciences, Harbin, 150000, China
| | - Wang Jinliang
- Shandong Binzhou Anim Sci & Vet Med Acad, Binzhou, 256600, People's Republic of China
| | - Shen Zhiqiang
- Shandong Binzhou Anim Sci & Vet Med Acad, Binzhou, 256600, People's Republic of China
| | - Shi Guangliang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Li Shu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
17
|
Gao X, Ji C, Wang J, Song X, Zuo R, Zhang J, Chen X, Ji H, Peng L, Guo D, Jiang S. Maduramicin induces cardiotoxicity via Rac1 signaling-independent methuosis in H9c2 cells. J Appl Toxicol 2021; 41:1937-1951. [PMID: 33890316 DOI: 10.1002/jat.4175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022]
Abstract
Maduramicin frequently induces severe cardiotoxicity in target and nontarget animals in clinic. Apoptotic and non-apoptotic cell death mediate its cardiotoxicity; however, the underlying non-apoptotic cell death induced by maduramicin remains unclear. In current study, a recently described non-apoptotic cell death "methuosis" caused by maduramicin was defined in mammalian cells. Rat myocardial cell H9c2 was used as an in vitro model, showing excessively cytoplasmic vacuolization upon maduramicin (0.0625-5 μg/mL) exposure for 24 h. Maduramicin-induced reversible cytoplasmic vacuolization of H9c2 cells in a time- and concentration-dependent manner. The vacuoles induced by maduramicin were phase lucent with single membrane and were not derived from the swelling of organelles such as mitochondria, endoplasmic reticulum, lysosome, and Golgi apparatus. Furthermore, maduramicin-induced cytoplasmic vacuoles are generated from micropinocytosis, which was demonstrated by internalization of extracellular fluid-phase marker Dextran-Alexa Fluor 488 into H9c2 cells. Intriguingly, these cytoplasmic vacuoles acquired some characteristics of late endosomes and lysosomes rather than early endosomes and autophagosomes. Vacuolar H+ -ATPase inhibitor bafilomycin A1 efficiently prevented the generation of cytoplasmic vacuoles and decreased the cytotoxicity of H9c2 cells triggered by maduramicin. Mechanism studying indicated that maduramicin activated H-Ras-Rac1 signaling pathway at both mRNA and protein levels. However, the pharmacological inhibition and siRNA knockdown of Rac1 rescued maduramicin-induced cytotoxicity of H9c2 cells but did not alleviate cytoplasmic vacuolization. Based on these findings, maduramicin induces methuosis in H9c2 cells via Rac-1 signaling-independent seriously cytoplasmic vacuolization.
Collapse
Affiliation(s)
- Xiuge Gao
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Chunlei Ji
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Junqi Wang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xinhao Song
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Runan Zuo
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jingjing Zhang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaorong Chen
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hui Ji
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Lin Peng
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Dawei Guo
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shanxiang Jiang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
18
|
Li ZL, Mi J, Lu L, Luo Q, Liu X, Yan YM, Jin B, Cao YL, Zeng XX, Ran LW. The main anthocyanin monomer of Lycium ruthenicum Murray induces apoptosis through the ROS/PTEN/PI3K/Akt/caspase 3 signaling pathway in prostate cancer DU-145 cells. Food Funct 2021; 12:1818-1828. [PMID: 33527955 DOI: 10.1039/d0fo02382e] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anthocyanins have been reported to have effective chemopreventive activity. Lycium ruthenicum Murray is rich in anthocyanins and exhibits many biological activities. The purpose of this study was to investigate the effects and possible biological mechanism of the main anthocyanin monomer (Pt3G) of Lycium ruthenicum Murray on prostate cancer DU-145 cells. The cell proliferation was detected by methyl thiazolyl tetrazolium assay. The cell apoptosis rates were assessed by flow cytometric analysis and TUNEL assay. The expressions of apoptosis related proteins were evaluated by western blotting. Our data demonstrated that Pt3G inhibited cell proliferation, induced apoptosis and promoted cell cycle arrest at the S phase in a concentration-dependent manner (0, 100, 200 and 400 μg mL-1). Furthermore, it was shown that Pt3G decreased the mitochondrial membrane permeability through regulating the expressions of Bax and Bcl-2. Western blot analysis indicated that Pt3G significantly increased the expression of PTEN and then activated the PI3K/Akt-mediated caspase 3 pathway. In addition, our results also suggested that Pt3G activated the PTEN gene to induce the apoptosis of DU-145 cells by stimulating the overproduction of ROS. To sum up, these results indicate that Pt3G inhibits cell proliferation and induces apoptosis through the ROS/PTEN/PI3K/Akt/caspase 3 signaling pathway in prostate cancer DU-145 cells. Therefore, Pt3G of Lycium ruthenicum Murray may be a potential anti-proliferative agent for the prevention or treatment of prostate cancer.
Collapse
Affiliation(s)
- Zhan-Long Li
- College of Public Health and Management, Ningxia Medical University, Yinchuan 750004, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Huang MZ, Zhang ZD, Yang YJ, Liu XW, Qin Z, Li JY. Aspirin Eugenol Ester Protects Vascular Endothelium From Oxidative Injury by the Apoptosis Signal Regulating Kinase-1 Pathway. Front Pharmacol 2020; 11:588755. [PMID: 33658932 PMCID: PMC7919194 DOI: 10.3389/fphar.2020.588755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/09/2020] [Indexed: 11/29/2022] Open
Abstract
Aspirin eugenol ester (AEE) is a new potential pharmaceutical compound possessing anti-inflammatory, anti-cardiovascular disease, and antioxidative stress activity. The pharmacological activities of AEE are partly dependent on its regulation of cell apoptosis. However, it is still unclear how AEE inhibits cell apoptosis on the basis of its antioxidative stress effect. This study aimed to reveal the vascular antioxidative mechanism of AEE in response to H2O2-induced oxidative stress in HUVECs and paraquat-induced oxidative stress in rats. In the different intervention groups of HUVECs and rats, the expression of ASK1, ERK1/2, SAPK/JNK, and p38 and the phosphorylation levels of ERK1/2, SAPK/JNK, and p38 were measured. The effects of ASK1 and ERK1/2 on the anti-apoptotic activity of AEE in the oxidative stress model were probed using the corresponding inhibitors ASK1 and ERK1/2. The results showed that in the HUVECs, 200 μM H2O2 treatment significantly increased the phosphorylation of SAPK/JNK and the level of ASK1 but decreased the phosphorylation of ERK1/2, while in the HUVECs pretreated with AEE, the H2O2-induced changes were significantly ameliorated. The findings were observed in vitro and in vivo. Moreover, inhibition of ASK1 and ERK1/2 showed that ASK1 plays a vital role in the protective effect of AEE on H2O2-induced apoptosis. All findings suggested that AEE protects the vascular endothelium from oxidative injury by mediating the ASK1 pathway.
Collapse
Affiliation(s)
- Mei-Zhou Huang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhen-Dong Zhang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Ya-Jun Yang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Xi-Wang Liu
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Zhe Qin
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Jian-Yong Li
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| |
Collapse
|
20
|
Wang L, Wang L, Shi X, Xu S. Chlorpyrifos induces the apoptosis and necroptosis of L8824 cells through the ROS/PTEN/PI3K/AKT axis. JOURNAL OF HAZARDOUS MATERIALS 2020; 398:122905. [PMID: 32768820 DOI: 10.1016/j.jhazmat.2020.122905] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/26/2020] [Accepted: 05/06/2020] [Indexed: 06/11/2023]
Abstract
Excessive chlorpyrifos (CPF) in the environment causes toxicity to nontarget organisms by triggering oxidative stress. Phosphatase and tensin homolog deleted on chromosome ten (PTEN) plays an important role in controlling apoptosis and necrosis by negatively regulating the phosphatidylinositol 3-kinase/threonine kinase (PI3K/AKT) pathway. However, the effects of different concentrations of CPF on grass fish liver cell injury and the role of the ROS/PTEN/PI3K/AKT axis remain poorly understood. In this study, L8824 cells treated with different concentrations of CPF (0, 40, 60, or 80 μM) were used as the research object. The results showed that the median inhibitory concentration (IC50) was 112.226 μM. As the CPF concentrations increased, the ROS and MDA levels increased, and the T-AOC levels and SOD/GPx/GST activities decreased. As PTEN expression increased, PI3K/AKT, BCL-2, and Caspase-8 expression dramatically decreased. Conversely, RIPK1/RIPK3/MLKL and Bax/Cyt-c/Caspase-3 expression increased. Additionally, necroptosis increased in a dose-dependent manner, while apoptosis first increased and then decreased. In conclusion, our study showed that CPF could trigger oxidative stress and induce apoptosis and necroptosis in fish liver cells by regulating the ROS/PTEN/PI3K/AKT axis, and the type of damage induced was dose-dependent. These results are meaningful for toxicological studies of CPF and efforts to protect the ecosystem.
Collapse
Affiliation(s)
- Lanqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Lanxi Wang
- College of Basic Medicine, Harbin Medical University, Harbin 150081, PR China
| | - Xu Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
21
|
Gao X, Ruan X, Ji H, Peng L, Qiu Y, Yang D, Song X, Ji C, Guo D, Jiang S. Maduramicin triggers methuosis-like cell death in primary chicken myocardial cells. Toxicol Lett 2020; 333:105-114. [PMID: 32736005 DOI: 10.1016/j.toxlet.2020.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 06/28/2020] [Accepted: 07/26/2020] [Indexed: 02/08/2023]
Abstract
Maduramicin frequently induces severe cardiotoxicity in broiler chickens as well as in humans who consume maduramicin accidentally. Apoptosis and non-apoptotic cell death occur concurrently in the process of maduramicin-induced cardiotoxicity; however, the underlying mechanism of non-apoptotic cell death is largely unknown. Here, we report the relationship between maduramicin-caused cytoplasmic vacuolization and methuosis-like cell death as well as the underlying mechanism in primary chicken myocardial cells. Maduramicin induced a significant increase of cytoplasmic vacuoles with a degree of cell specificity in primary chicken embryo fibroblasts and chicken hepatoma cells (LMH), along with a decrease of ATP and an increase of LDH. The accumulated vacuoles were partly derived from cellular endocytosis rather than the swelling of endoplasm reticulum, lysosomes, and mitochondria. Moreover, the broad-spectrum caspase inhibitor carbobenzoxy-Val-Ala-Asp-fluoromethylketone (z-VAD-fmk) did not prevent maduramicin-induced cytoplasmic vacuolization. DNA ladder and cleavage of PARP were not observed in chicken myocardial cells during maduramicin exposure. Pretreatment with 3-methyladenine (3-MA) and cholorquine (CQ) of chicken myocardial cells did not attenuate cytoplasmic vacuolization and cytotoxicity, although LC3 and p62 were activated. Bafilomycin A1 almost completely prevented the generation of cytoplasmic vacuoles and significantly attenuated cytotoxicity induced by maduramicin, along with downregulation of K-Ras and upregulation of Rac1. Taken together, "methuosis" due to excessive cytoplasmic vacuolization mediates the cardiotoxicity of maduramicin. This provides new insights for understanding a nonclassical form of cell death in the field of drug-induced cytotoxicity.
Collapse
Affiliation(s)
- Xiuge Gao
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Xiangchun Ruan
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China; Laboratory of Veterinary Pharmacology and Toxicology, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Hui Ji
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Lin Peng
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Yawei Qiu
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Dan Yang
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Xinhao Song
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Chunlei Ji
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Dawei Guo
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China.
| | - Shanxiang Jiang
- Joint International Research Laboratory of Animal Health and Food Safety, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China.
| |
Collapse
|
22
|
Genomics-driven discovery of the biosynthetic gene cluster of maduramicin and its overproduction in Actinomadura sp. J1-007. ACTA ACUST UNITED AC 2020; 47:275-285. [DOI: 10.1007/s10295-019-02256-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023]
Abstract
Abstract
Maduramicin is the most efficient and possesses the largest market share of all anti-coccidiosis polyether antibiotics (ionophore); however, its biosynthetic gene cluster (BGC) has yet to been identified, and the associated strains have not been genetically engineered. Herein, we performed whole-genome sequencing of a maduramicin-producing industrial strain of Actinomadura sp. J1-007 and identified its BGC. Additionally, we analyzed the identified BGCs in silico to predict the biosynthetic pathway of maduramicin. We then developed a conjugation method for the non-spore-forming Actinomadura sp. J1-007, consisting of a site-specific integration method for gene overexpression. The maduramicin titer increased by 30% to 7.16 g/L in shake-flask fermentation following overexpression of type II thioesterase MadTE that is the highest titer at present. Our findings provide insights into the biosynthetic mechanism of polyethers and provide a platform for the metabolic engineering of maduramicin-producing microorganisms for overproduction and development of maduramicin analogs in the future.
Collapse
|