1
|
Gwak YH, Shiwakoti S, Ko JY, Sim HH, Lee JH, Kim HJ, Oak MH. Paeonol from Paeonia lactiflora attenuates calcification of aortic valvular interstitial cell by inhibiting activation of redox-sensitive NFκB/AKT/ERK1/2 pathway. Food Sci Biotechnol 2025; 34:2269-2279. [PMID: 40351722 PMCID: PMC12064534 DOI: 10.1007/s10068-025-01838-7] [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: 10/26/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 05/14/2025] Open
Abstract
Calcific aortic valve disease (CAVD) is a life-threatening cardiovascular condition without effective pharmacological treatments. Paeonol, a bioactive compound from the roots of Paeonia lactiflora, is known for its anti-inflammatory, antioxidant, and anti-atherosclerotic properties. While it has shown benefits in treating several cardiovascular conditions, its role in CAVD remains unexplored, requiring further investigation. This study explores paeonol's potential to inhibit calcification in porcine valve interstitial cells (VICs) and to elucidate the underlying mechanism. Calcification was induced using a pro-calcifying medium (PCM), and the anti-calcification effects of paeonol were evaluated. Paeonol significantly reduced level of calcification and its markers such as RUNX2, osteopontin, and BMP2. It also lowered reactive oxygen species levels, thereby reducing oxidative stress, which is known to activate several calcification pathways. Paeonol inhibited these pathways by reducing phosphorylation of NFκB, AKT, and ERK1/2, thereby preventing their activation and suggesting its potential as a therapeutic agent to mitigate CAVD progression.
Collapse
Affiliation(s)
- Yeon-Hyang Gwak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554 Republic of Korea
| | - Saugat Shiwakoti
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554 Republic of Korea
- Convergence Center for Green Anti-Aging Research, Muan-Gun, 58554 Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554 Republic of Korea
- Convergence Center for Green Anti-Aging Research, Muan-Gun, 58554 Republic of Korea
| | - Hwan-Hee Sim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554 Republic of Korea
- Convergence Center for Green Anti-Aging Research, Muan-Gun, 58554 Republic of Korea
| | - Ji-Hyeok Lee
- Division of Commercialization Support, Honam National Institute of Biological Resources, Mokpo, 58762 Republic of Korea
| | - Hyun Jung Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554 Republic of Korea
- Convergence Center for Green Anti-Aging Research, Muan-Gun, 58554 Republic of Korea
| | - Min-Ho Oak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554 Republic of Korea
- Convergence Center for Green Anti-Aging Research, Muan-Gun, 58554 Republic of Korea
| |
Collapse
|
2
|
Shen B, Wen Y, Li S, Zhou Y, Chen J, Yang J, Zhao C, Wang J. Paeonol ameliorates hyperlipidemia and autophagy in mice by regulating Nrf2 and AMPK/mTOR pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155839. [PMID: 38943694 DOI: 10.1016/j.phymed.2024.155839] [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: 11/16/2023] [Revised: 05/31/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Hyperlipidemia, inadequate diet, and excessive medication increase the risk of cardiovascular disease. Paeonl (Pae), a phenolic compound found in Peony and Angelica dahurica, can alleviate lipid metabolism disorders and lipotoxicity. However, the molecular mechanism of Pae alleviating hyperlipidemia remains unclear and needs to be further explored. PURPOSE In this study, we explored whether Pae can prevent hyperlipidemia and investigated the molecular mechanisms. METHODS The effects of Pae (30, 45, 60mg·kg-1) on hyperlipidemia in Tyloapol-induced WT mice and Nrf2 knockout mice (Pae: 60mg·kg-1) were detected by oil red O staining, HE staining, TG, TC and other indexes. The expression levels of proinflammatory mediators, key lipid proteins and autophagy signaling pathway proteins were analyzed by enzyme-linked immunosorbent assay, western blot and immunofluorescence. The molecular mechanism of Pae alleviating hyperlipidemia was explored through molecular docking technique and in vivo and in vitro experiments. RESULTS Several studies indicated that Pae effectively improved tyloxapol (Ty)-induced lipid metabolism disorder, as evidenced by decreased triglyceride content, increased carnitine palmitoyltransferase 1 (CPT1), and Sirtuin 1 (Sirt1) protein expression. In addition, Pae ameliorated hyperlipidemia by activating the AMPK/ACC and PI3K/mTOR pathways. Interestingly, the therapeutic effect of Pae on hyperlipidemia was markedly reduced in Nrf2-/- mice. Molecular docking results indicated that Pae and Nrf2 exhibited good binding ability, suggesting that Nrf2 is a core target mediating the effects of Pae in the treatment of hyperlipidemia. Taken together, Pae alleviated hyperlipidemia in vivo and ameliorated lipid accumulation in vitro by activating AMPK/ACC and PI3K/mTOR signaling pathways via Nrf2 binding. CONCLUSION Our data suggest that paeonol can ameliorate hyperlipidemia and autophagy in mice by regulating Nrf2 and AMPK/mTOR pathways, and it has potential therapeutic value in the occurrence and development of hyperlipidemia.
Collapse
Affiliation(s)
- Bingyu Shen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yongqiang Wen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shengxin Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yi Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Junlin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiaqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chenxu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
3
|
Wang Z, Liu Y, Wang X, Wang X, Wu Y, Song Y, Xu J, Xue C. Sea cucumber plasmalogen enhance lipophagy to alleviate abnormal lipid accumulation induced by high-fat diet. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159495. [PMID: 38609006 DOI: 10.1016/j.bbalip.2024.159495] [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: 11/16/2023] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Sea cucumber phospholipids, including the plasmalogen (PlsEtn) and plasmanylcholine (PakCho), have been shown to play a regulatory role in lipid metabolism disorders, but their mechanism of action remains unclear. Therefore, high-fat diet (HFD) and palmitic acid were used to establish lipid accumulation models in mice and HepG2 cells, respectively. Results showed that PlsEtn can reduce lipid deposition both in vivo and in vitro. HFD stimulation abnormally activated lipophagy through the phosphorylation of the AMPK/ULK1 pathway. The lipophagy flux monitor revealed abnormalities in the fusion stage of lipophagy. Of note, only PlsEtn stimulated the dynamic remodeling of the autophagosome membrane, which was indicated by the significantly decreased LC3 II/I ratio and p62 level. In all experiments, the effect of PlsEtn was significantly higher than that of PakCho. These findings elucidated the mechanism of PlsEtn in alleviating lipid accumulation, showed that it might be a lipophagy enhancer, and provided new insights into the high-value utilization of sea cucumber as an agricultural resource.
Collapse
Affiliation(s)
- Zhigao Wang
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China.
| | - Yanjun Liu
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China.
| | - Xiaoxu Wang
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China.
| | - Xincen Wang
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China; Institute of Nutrition and Health, Qingdao University, No. 308 Ningxia Road, Qingdao, Shandong 266071, China.
| | - Yuan Wu
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China.
| | - Yu Song
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China.
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China.
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, No. 1299, Sanshan Road, Qingdao, Shandong Province 266003, China; Qingdao Marine Science and Technology Center, Qingdao 266235, China.
| |
Collapse
|
4
|
Gao M, Dong L, Yang Y, Yan J, Liang Y, Ma X, Zhou M, Wu H, Liu Y, Dai M. The anti-atherosclerotic effect of Paeonol against the lipid accumulation in macrophage-derived foam cells by inhibiting ferroptosis via the SIRT1/NRF2/GPX4 signaling pathway. Biochem Biophys Res Commun 2024; 708:149788. [PMID: 38518720 DOI: 10.1016/j.bbrc.2024.149788] [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: 12/07/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/24/2024]
Abstract
Atherosclerosis (AS) is the underlying cause of many severe vascular diseases and is primarily characterized by abnormal lipid metabolism. Paeonol (Pae), a bioactive compound derived from Paeonia Suffruticosa Andr., is recognized for its significant role in reducing lipid accumulation. Our research objective is to explore the link between lipid buildup in foam cells originating from macrophages and the process of ferroptosis, and explore the effect and mechanism of Pae on inhibiting AS by regulating ferroptosis. In our animal model, ApoE-deficient mice, which were provided with a high-fat regimen to provoke atherosclerosis, were administered Pae. The treatment was benchmarked against simvastatin and ferrostatin-1. The results showed that Pae significantly reduced aortic ferroptosis and lipid accumulation in the mice. In vitro experiments further demonstrated that Pae could decrease lipid accumulation in foam cells induced by oxidized low-density lipoprotein (LDL) and challenged with the ferroptosis inducer erastin. Crucially, the protective effect of Pae against lipid accumulation was dependent on the SIRT1/NRF2/GPX4 pathway, as SIRT1 knockdown abolished this effect. Our findings suggest that Pae may offer a novel therapeutic approach for AS by inhibiting lipid accumulation through the suppression of ferroptosis, mediated by the SIRT1/NRF2/GPX4 pathway. Such knowledge has the potential to inform the creation of novel therapeutic strategies aimed at regulating ferroptosis within the context of atherosclerosis.
Collapse
Affiliation(s)
- Menglong Gao
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Lishun Dong
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Yulong Yang
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Jinjin Yan
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Yuning Liang
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Xiaolin Ma
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Min Zhou
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Hongfei Wu
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China
| | - Yarong Liu
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China.
| | - Min Dai
- School of Pharmacy, Anhui University of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, No. 350 Longzihu Road, Hefei, 230012, China.
| |
Collapse
|
5
|
Li R, Yi Q, Wang J, Miao Y, Chen Q, Xu Y, Tao M. Paeonol promotes longevity and fitness in Caenorhabditis elegans through activating the DAF-16/FOXO and SKN-1/Nrf2 transcription factors. Biomed Pharmacother 2024; 173:116368. [PMID: 38471269 DOI: 10.1016/j.biopha.2024.116368] [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: 11/23/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Paeonol, as one of the most abundant plant-derived polyphenols, has multiple bioactivities including anti-inflammatory, anti-tumor, and anti-cardiovascular diseases. Nevertheless, the anti-aging effects and related mechanisms of paeonol are rarely reported. In this study, we found that paeonol significantly prolonged the mean lifespan of Caenorhabditis elegans (C. elegans) by 28.49% at a dose of 200 μM. Moreover, paeonol promoted the health of C. elegans by increasing the body bending and pharyngeal pumping rates and reducing the lipofuscin accumulation level. Meanwhile, paeonol induced the expression of stress-related genes or proteins by activating the transcription factors DAF-16/FOXO, SKN-1/Nrf2, and HSF-1, which in turn enhanced oxidative and thermal stress tolerance. The mechanism behind the anti-aging effect of paeonol occurred by down-regulating the insulin/IGF-1 signaling (IIS) pathway. Our findings shed new light on the application of paeonol for longevity promotion and human health.
Collapse
Affiliation(s)
- Rong Li
- College of Bioengineering/Hubei Engineering Research Center for Specialty Flowers Biological Breeding, Jingchu University of Technology, Jingmen, People's Republic of China
| | - Qingping Yi
- College of Bioengineering/Hubei Engineering Research Center for Specialty Flowers Biological Breeding, Jingchu University of Technology, Jingmen, People's Republic of China
| | - Jinsong Wang
- College of Bioengineering/Hubei Engineering Research Center for Specialty Flowers Biological Breeding, Jingchu University of Technology, Jingmen, People's Republic of China
| | - Yuanxin Miao
- College of Bioengineering/Hubei Engineering Research Center for Specialty Flowers Biological Breeding, Jingchu University of Technology, Jingmen, People's Republic of China
| | - Qingchan Chen
- College of Bioengineering/Hubei Engineering Research Center for Specialty Flowers Biological Breeding, Jingchu University of Technology, Jingmen, People's Republic of China
| | - Yan Xu
- College of Bioengineering/Hubei Engineering Research Center for Specialty Flowers Biological Breeding, Jingchu University of Technology, Jingmen, People's Republic of China.
| | - Mingfang Tao
- Hubei Key Laboratory of Nutritional Quality and Safety of Agro-products, Institute of Agricultural Quality Standards and Detection Technology, Hubei Academy of Agricultural Sciences, Wuhan, People's Republic of China.
| |
Collapse
|
6
|
Li R, Zhou J, Zhang X, Wang Y, Wang J, Zhang M, He C, Zhuang P, Chen H. Construction of the Gal-NH 2/mulberry leaf polysaccharides-lysozyme/luteolin nanoparticles and the amelioration effects on lipid accumulation. Int J Biol Macromol 2023; 253:126780. [PMID: 37699459 DOI: 10.1016/j.ijbiomac.2023.126780] [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: 03/06/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023]
Abstract
Luteolin is a kind of natural flavonoid with great potential for lipid accumulation intervention. However, the poor water solubility and non-targeted release greatly diminish its efficiency. In this study, 4-aminophenyl β-D-galactopyranoside (Gal-NH2)/mulberry leaf polysaccharides- lysozyme/luteolin nanoparticles (Gal-MPL/Lut) were fabricated via amide reaction, self-assembly process and electrostatic interaction. The nanoparticles could hepatic-target of Lut and enhance action on liver tissue by specific recognition of asialoglycoprotein receptor (ASGPR). Physicochemical characterization of the nanoparticles showed a spherical shape with a uniform particle size distribution (77.8 ± 2.6 nm) with a polydispersity index (PDI) of 0.22 ± 0.06. Subsequently, in HepG2 cells model, administration with hepatic-targeted Gal-MPL/Lut nanoparticles promoted the cellular uptake of Lut, and regulated lipid metabolism manifested by remarkably inhibiting total cholesterol (TC) and triglyceride (TG) expression levels through the modulation of PI3K/SIRT-1/FAS/CEBP-α signaling pathway. This study provides a promising strategy for a highly hepatic-targeted therapy to ameliorate lipid accumulation using natural medicines facilitated by nano-technology.
Collapse
Affiliation(s)
- Ruilin Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Jingna Zhou
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Xiaoyu Zhang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Yajie Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Jia Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Min Zhang
- Tianjin Agricultural University, Tianjin 300384, PR China; State Key Laboratory of Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, PR China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao
| | - Pengwei Zhuang
- Haihe Laboratory of Modern Chinese Medicine, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haixia Chen
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| |
Collapse
|
7
|
Kose T, Moreno-Fernandez J, Vera-Aviles M, Sharp PA, Latunde-Dada GO. Ferulic acid protects HepG2 cells and mouse liver from iron-induced damage. Biochem Biophys Rep 2023; 35:101521. [PMID: 37560439 PMCID: PMC10407627 DOI: 10.1016/j.bbrep.2023.101521] [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: 03/30/2023] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023] Open
Abstract
Liver as iron storage organ is particularly susceptible to oxidative stress-induced injury from excess iron. Thus, antioxidant therapies are often used to reverse oxidative damage and protect cells and tissues. This study investigated the protective effects of phenolic acids; ferulic acid (FA) and its metabolite, ferulic acid 4-O-sulfate disodium salt (FAS) against oxidative stress under iron overload conditions in mouse and HepG2 cells. Cells were exposed to FA or FAS and then treated with iron-induced oxidative stress complex of 50 μmol/L FAC and 20 μmol/L of 8-hydroxyquinoline 8HQ (8HQ-FAC). Iron dextran was injected intraperitoneally on alternate days for 10 days to induce the iron overload condition in BALB/c mice. The study revealed that the phenolic acids were protective against ROS production, lipid peroxidation and antioxidant depletion in HepG2 cells and liver tissues of BALB/c mice during iron-induced oxidative stress. The protective function of phenolic acids was achieved by the transcriptional activation of nuclear factor erythroid-2-related factor 2 (Nrf2) to regulate antioxidant genes. In conclusion, the study provides evidence that FA has the potential as a therapeutic agent against iron-related diseases such as T2D.
Collapse
Affiliation(s)
- Tugba Kose
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, Franklin-Wilkins-Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Jorge Moreno-Fernandez
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, Franklin-Wilkins-Building, 150 Stamford Street, London, SE1 9NH, UK
- Department of Physiology, University of Granada, 18071, Granada, Spain
| | - Mayra Vera-Aviles
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, Franklin-Wilkins-Building, 150 Stamford Street, London, SE1 9NH, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Paul A. Sharp
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, Franklin-Wilkins-Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Gladys O. Latunde-Dada
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, Franklin-Wilkins-Building, 150 Stamford Street, London, SE1 9NH, UK
| |
Collapse
|
8
|
Zhu M, Wei C, Wang H, Han S, Cai L, Li X, Liao X, Che X, Li X, Fan L, Qiu G. SIRT1 mediated gastric cancer progression under glucose deprivation through the FoxO1-Rab7-autophagy axis. Front Oncol 2023; 13:1175151. [PMID: 37293593 PMCID: PMC10244632 DOI: 10.3389/fonc.2023.1175151] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/28/2023] [Indexed: 06/10/2023] Open
Abstract
Purpose Silent mating type information regulator 2 homolog 1 (SIRT1) and autophagy have a two-way action (promoting cell death or survival) on the progression and treatment of gastric cancer (GC) under different conditions or environments. This study aimed to investigate the effects and underlying mechanism of SIRT1 on autophagy and the malignant biological behavior of GC cells under conditions of glucose deprivation (GD). Materials and methods Human immortalized gastric mucosal cell GES-1 and GC cell lines SGC-7901, BGC-823, MKN-45 and MKN-28 were utilized. A sugar-free or low-sugar (glucose concentration, 2.5 mmol/L) DMEM medium was used to simulate GD. Additionally, CCK8, colony formation, scratches, transwell, siRNA interference, mRFP-GFP-LC3 adenovirus infection, flow cytometry and western blot assays were performed to investigate the role of SIRT1 in autophagy and malignant biological behaviors (proliferation, migration, invasion, apoptosis and cell cycle) of GC under GD and the underlying mechanism. Results SGC-7901 cells had the longest tolerance time to GD culture conditions, which had the highest expression of SIRT1 protein and the level of basal autophagy. With the extension of GD time, the autophagy activity in SGC-7901 cells also increased. Under GD conditions, we found a close relationship between SIRT1, FoxO1 and Rab7 in SGC-7901 cells. SIRT1 regulated the activity of FoxO1 and upregulated the expression of Rab7 through deacetylation, which ultimately affected autophagy in GC cells. In addition, changing the expression of FoxO1 provided feedback on the expression of SIRT1 in the cell. Reducing SIRT1, FoxO1 or Rab7 expression significantly inhibited the autophagy levels of GC cells under GD conditions, decreased the tolerance of GC cells to GD, enhanced the inhibition of GD in GC cell proliferation, migration and invasion and increased apoptosis induced by GD. Conclusion The SIRT1-FoxO1-Rab7 pathway is crucial for the autophagy and malignant biological behaviors of GC cells under GD conditions, which could be a new target for the treatment of GC.
Collapse
Affiliation(s)
- Mengke Zhu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Chao Wei
- Clinical Medicine Teaching and Research Section, Xi’an Health School, Xi’an, Shaanxi, China
| | - Haijiang Wang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shangning Han
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lindi Cai
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaowen Li
- Department of General Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xinhua Liao
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiangming Che
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lin Fan
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Guanglin Qiu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
9
|
Resveratrol Improves the Progression of Osteoarthritis by Regulating the SIRT1-FoxO1 Pathway-Mediated Cholesterol Metabolism. Mediators Inflamm 2023; 2023:2936236. [PMID: 36643587 PMCID: PMC9833897 DOI: 10.1155/2023/2936236] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 01/06/2023] Open
Abstract
Osteoarthritis (OA) is considered a metabolic disorder. This study investigated the effect of resveratrol (RES) on cholesterol accumulation in osteoarthritic articular cartilage via the silent information regulator 1 (SIRT1)/forkhead transcription factor (FoxO1) pathway. Interleukin (IL)-1β-treated chondrocytes that mimic OA chondrocytes were used in in vitro experiments. The optimal RES concentration was selected based on the results of chondrocyte proliferation in the Cell Counting Kit-8 assay. Western blotting, immunofluorescence, and reverse transcription-quantitative polymerase chain reaction were performed. For the animal experiments, mice were randomly divided into the RES group (n = 15), medial meniscus destabilization group (n = 15), and sham group (n = 15), and each group received the same dose of RES or saline. Articular cartilage tissue was obtained eight weeks after surgery for relevant histological analysis. Clinical tissue test results suggest that downregulation of the SIRT1/FoxO1 pathway is associated with cholesterol buildup in OA chondrocytes. For the in vitro studies, RES increased the expression of SIRT1 and phosphorylation of FoxO1 in IL-1β-treated chondrocytes, promoted the expression of cholesterol efflux factor liver X receptor alpha (LXRα), and inhibited the expression of cholesterol synthesis-associated factor sterol-regulatory element binding proteins 2 (SREBP2). This reduced IL-1β-induced chondrocytes cholesterol accumulation. SIRT1 inhibition prevented the RES-mediated reduction in cholesterol buildup. Inhibiting FoxO1 but not SIRT1 reduced FoxO1 phosphorylation and increased cholesterol buildup in cultured chondrocytes. Additionally, in vivo experiments have shown that RES can alleviate cholesterol buildup and pathological changes in OA cartilage. Our findings suggest that RES regulates cholesterol buildup in osteoarthritic articular cartilage via the SIRT1/FoxO1 pathway, thereby improving the progression of OA.
Collapse
|
10
|
Xing H, Liang C, Wang C, Xu X, Hu Y, Qiu B. Metformin mitigates cholesterol accumulation via the AMPK/SIRT1 pathway to protect osteoarthritis chondrocytes. Biochem Biophys Res Commun 2022; 632:113-121. [PMID: 36206595 DOI: 10.1016/j.bbrc.2022.09.074] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
Abstract
In this study, we aim to investigate the effect of metformin on cholesterol synthesis and efflux-related genes in chondrocytes during osteoarthritis (OA) and explore the underlying mechanisms. Primary chondrocytes were harvested from Wistar rat cartilage and divided into control and treatment groups. Chondrocytes in the treatment group were treated with interleukin-1β (IL-1β) mimicking the inflammatory environment of osteoarthritis. Subsequently, RT-qPCR, Western blotting, immunofluorescence staining, and Cell Counting Kit-8 (CCK-8) were conducted. Significant reductions in phosphorylated AMP-activated protein kinase (p-AMPK) and silent information regulator 1 (SIRT1) protein expression were observed in both human OA chondrocytes and cultured primary murine chondrocytes treated with IL-1β, while AMP-activated protein kinase (AMPK) was not inhibited. Moreover, in the presence of IL-1β, metformin significantly increased the expression of p-AMPK and SIRT1 at the protein and mRNA level. Meanwhile, metformin could reverse IL-1β-induced cartilage extracellular matrix degradation in chondrocytes from the rat model of OA (treated by IL-β) by activating the AMPK/SIRT1 pathway. Moreover, metformin activated AMPK and SIRT1, mediated by the activation of SREBP-2 and HMGCR in OA chondrocytes. Inhibiting AMPK/SIRT1 activity by its specific inhibitor could suppress IL-1β-induced expression of LXRα, ABCA1 and ApoA1 and cholesterol efflux. Thus, metformin inhibits cholesterol synthesis and promotes cholesterol efflux by activating the AMPK/SIRT1 pathway in OA chondrocytes. This study improves our understanding of the effect of metformin on cholesterol accumulation in OA chondrocytes.
Collapse
Affiliation(s)
- Hengte Xing
- Department of Orthopedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, 430000, China
| | - Chuancai Liang
- Department of Orthopedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, 430000, China
| | - Chenyu Wang
- Department of Orthopedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, 430000, China
| | - Xiongfeng Xu
- Department of Orthopedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, 430000, China
| | - Yong Hu
- Department of Orthopedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, 430000, China.
| | - Bo Qiu
- Department of Orthopedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, 430000, China.
| |
Collapse
|
11
|
Yu W, Ilyas I, Aktar N, Xu S. A review on therapeutical potential of paeonol in atherosclerosis. Front Pharmacol 2022; 13:950337. [PMID: 35991897 PMCID: PMC9385965 DOI: 10.3389/fphar.2022.950337] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
The morbidity and mortality of atherosclerotic cardiovascular disease (ASCVD) is increasing year by year. Cortex Moutan is a traditional Chinese medicinal herb that has been widely used for thousands of years to treat a wide variety of diseases in Eastern countries due to its heat-clearing and detoxifying effects. Paeonol is a bioactive monomer extracted from Cortex Moutan, which has anti-atherosclerotic effects. In this article, we reviewed the pharmacological effects of paeonol against experimental atherosclerosis, as well as its protective effects on vascular endothelial cells, smooth muscle cells, macrophages, platelets, and other important cell types. The pleiotropic effects of paeonol in atherosclerosis suggest that it can be a promising therapeutic agent for atherosclerosis and its complications. Large-scale randomized clinical trials are warranted to elucidate whether paeonol are effective in patients with ASCVD.
Collapse
Affiliation(s)
- Wei Yu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, Anhui, China
- Anhui Renovo Pharmaceutical Co., Ltd., Hefei, Anhui, China
- *Correspondence: Wei Yu, ; Suowen Xu,
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Nasrin Aktar
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- *Correspondence: Wei Yu, ; Suowen Xu,
| |
Collapse
|
12
|
Sabir U, Irfan HM, Alamgeer, Umer I, Niazi ZR, Asjad HMM. Phytochemicals targeting NAFLD through modulating the dual function of forkhead box O1 (FOXO1) transcription factor signaling pathways. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:741-755. [PMID: 35357518 DOI: 10.1007/s00210-022-02234-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/18/2022] [Indexed: 02/06/2023]
Abstract
Literature evidence reveals that natural compounds are potential candidates for ameliorating obesity-associated non-alcoholic fatty liver disease (NAFLD) by targeting forkhead box O1 (FOXO1) transcription factor. FOXO1 has a dual and complex role in regulating both increase and decrease in lipid accumulation in hepatocytes and adipose tissues (AT) at different stages of NAFLD. In insulin resistance (IR), it is constitutively expressed, resulting in increased hepatic glucose output and lipid metabolism irregularity. The studies on different phytochemicals indicate that dysregulation of FOXO1 causes disturbance in cellular nutrients homeostasis, and the natural entities have an enduring impact on the mitigation of these abnormalities. The current review communicates and evaluates certain phytochemicals through different search engines, targeting FOXO1 and its downstream cellular pathways to find lead compounds as potential therapeutic agents for treating NAFLD and related metabolic disorders. The findings of this review confirm that polyphenols, flavonoids, alkaloids, terpenoids, and anthocyanins are capable of modulating FOXO1 and associated signaling pathways, and they are potential therapeutic agents for NAFLD and related complications. HIGHLIGHTS: • FOXO1 has the potential to be targeted by novel drugs from natural sources for the treatment of NAFLD and obesity. • FOXO1 regulates cellular autophagy, inflammation, oxidative stress, and lipogenesis through alternative mechanisms. • Phytochemicals treat NAFLD by acting on FOXO1 or SREBP1c and PPARγ transcription factor signaling pathways.
Collapse
Affiliation(s)
- Usman Sabir
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Hafiz Muhammad Irfan
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan.
| | - Alamgeer
- Punjab University College of Pharmacy, University of the Punjab Lahore, Lahore, Pakistan
| | - Ihtisham Umer
- Pharmacy Department, Comsat International University Lahore Campus, Lahore, Pakistan
| | | | | |
Collapse
|
13
|
Ni HY, Yu L, Zhao XL, Wang LT, Zhao CJ, Huang H, Zhu HL, Efferth T, Gu CB, Fu YJ. Seed oil of Rosa roxburghii Tratt against non-alcoholic fatty liver disease in vivo and in vitro through PPARα/PGC-1α-mediated mitochondrial oxidative metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153919. [PMID: 35104757 DOI: 10.1016/j.phymed.2021.153919] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/07/2021] [Accepted: 12/30/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), characterized by hepatic steatosis and hepatocyte injury, is an obesity-induced metabolic dysregulation with few available therapeutic options. Enhancement of the mitochondrial function was considered as an effective treatment for NALFD. Unsaturated fatty acids (UFAs) have been shown to have beneficial effects on metabolic syndrome disease such as hyperlipidemia, coronary artery disease and cardiovascular diseases. The seed oil of Rosa roxburghii Tratt (ORRT) was of high quality in terms of its high amount of unsaturated fatty acids. However, the effects of ORRT on NALFD have not been reported so far. PURPOSE The study aimed to evaluate the protective effects and molecular mechanism of ORRT for the treatment of NAFLD in vivo and in vitro. METHODS The beneficial effects, especially improving the mitochondrial function, and the potential mechanism of ORRT on NAFLD were studied both in vivo and in vitro. Lipid levels were determined by triglyceride (TG), total cholesterol (TC), and Oil Red O staining. Oxidative stress and inflammation were assessed by detecting antioxidant enzyme activity, MDA content, and ELISA assay. Blood TG, TC, HDL-c and LDL-c levels were measured in HFD mice. Western blot analyses were used to determine the levels of the protein involved in fatty acid oxidation, oxidative metabolism, and mitochondria biogenesis and function. The mitochondrial membrane potential level was measured by JC-1 staining to teste the effect of ORRT on mitochondrial function in vitro. GW6471 (inhibitor of PPARα) was used to confirm the relationship between PPARα and PGC-1α. RESULTS ORRT significantly restrained NAFLD progression by attenuating lipid accumulation, oxidative stress and inflammatory response. Furthermore, ORRT upregulated thermogenesis-related gene expressions, such as uncoupling protein 1 (UCP1) and p38 mitogen-activated protein kinase (p38 MAPK). The results showed that the expression of key genes involved in fatty acid oxidation (e.g., CPT-1α, ACADL, PPARα) and in mitochondrial biogenesis and function (e.g., TFAM, NRF1, PGC-1α, and COX IV) was significantly increased. Together with the observed MMP improvement, these findings suggested that ORRT activated the mitochondrial oxidative pathway. Additionally, GW6471 inhibited the ORRT on promoting the expression of PGC-1α, CPT-1α, and ACADL. In conclusion, ORRT possessed the potential to prevent lipid accumulation via the PPARα/PGC-1α signaling pathway, which could be developed as a natural health-promoting oil against NAFLD.
Collapse
Affiliation(s)
- Hai-Yan Ni
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Liang Yu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Xue-Lian Zhao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Li-Tao Wang
- College of Forestry, Beijing Forestry University, Beijing 100083, China
| | - Chun-Jian Zhao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Han Huang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Han-Lin Zhu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Thomas Efferth
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Cheng-Bo Gu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China.
| | - Yu-Jie Fu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China; College of Forestry, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
14
|
Shang P, Liu Y, Jia J. Paeonol inhibits inflammatory response and protects chondrocytes by upregulating sirtuin 1. Can J Physiol Pharmacol 2022; 100:283-290. [PMID: 35235465 DOI: 10.1139/cjpp-2021-0319] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Paeonol is the bioactive component in Paeonia lactiflora Pall., Cynanchum paniculatum and Paeonia × suffruticosa Andr. Paeonol has been previously demonstrated to inhibit the release of tumor necrosis factor α (TNF-α) and interluekin 6 (IL-6) in chondrocytes. Sirtuin 1 (SIRT1) is downregulated in degraded cartilage and paeonol could induce nuclear accumulation of SIRT1. Therefore, the present study aims to investigate the possible role of paeonol in chondrocyte inflammation and cartilage protection in osteoarthritis (OA) as well as its regulation of SIRT1. Primary chondrocytes from rat knee joints were transfected with short hairpin (sh) - SIRT1 and (or) paeonol prior to IL-1β exposure, and then inflammatory response, apoptosis, and extracellular matrix (ECM) degradation in the cells were evaluated concurrent with the activation of the nuclear factor κβ (NF-κβ) signaling pathway. Increased levels of TNF-α, IL-17, IL-6, matrix metalloproteinase 1 (MMP-1), MMP-3, and MMP-13 along with decreased tissue inhibitor of metalloproteinases 1 and type II collagen levels were found in IL-1β-stimulated chondrocytes. Chondrocyte apoptosis was elevated and the NF-κβ signaling pathway was activated in response to IL-1β treatment. Paeonol enhanced SIRT1 expression to inactivate the NF-κβ signaling pathway, thereby ameliorating inflammatory cytokine secretion, ECM degradation, and chondrocyte apoptosis. In conclusion, the results of the present study confirm the potential of paeonol as a candidate OA drug.
Collapse
Affiliation(s)
- Peng Shang
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Ying Liu
- Department of Oncology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Junqing Jia
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
15
|
Chidamide Suppresses the Growth of Cholangiocarcinoma by Inhibiting HDAC3 and Promoting FOXO1 Acetylation. Stem Cells Int 2022; 2022:3632549. [PMID: 35126526 PMCID: PMC8816583 DOI: 10.1155/2022/3632549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022] Open
Abstract
Inhibitors for histone deacetylases (HDACs) have been identified as epigenetic drug targets to treat a variety of malignancies through several molecular mechanisms. The present study is aimed at investigating the mechanism underlying the possible antitumor effect of the HDAC inhibitor chidamide (CDM) on cholangiocarcinoma (CCA). Microarray-based gene expression profiling was conducted to predict the expression of HDACs in CCA, which was validated in clinical tissue samples from CCA patients. Next, the proliferation, migration, invasion, autophagy, and apoptosis of human CCA QBC939 and SNU308 cells were measured following treatment with CDM at different concentrations. The acetylation level of FOXO1 in the nucleus and cytoplasm of QBC939 and SNU308 cells was determined after overexpression and suppression of HDAC3. A QBC939-implanted xenograft nude mouse model was established for further exploration of CDM roles in vitro. HDAC3 was prominently expressed in CCA tissues and indicated a poor prognosis for patients with CCA. CDM significantly inhibited cell proliferation, migration, and invasion of QBC939 and SNU308 cells, while inducing their autophagy and apoptosis by reducing the expression of HDAC3. CDM promoted FOXO1 acetylation by inhibiting HDAC3, thereby inducing cell autophagy. Additionally, CDM inhibited tumor growth in vivo via HDAC3 downregulation and FOXO1 acetylation induction. Overall, this study reveals that CDM can exhibit antitumor effects against CCA by promoting HDAC3-mediated FOXO1 acetylation, thus identifying a new therapeutic avenue for the treatment of CCA.
Collapse
|
16
|
Jin R, Ren H, Liao M, Shang J, Wang D, Li M, Liu N. A dipeptidyl peptidase IV inhibitory peptide relieves palmitic acid-induced endoplasmic reticulum stress in HepG2 cells independent of inhibiting dipeptidyl peptidase IV activity. Food Funct 2021; 12:10773-10782. [PMID: 34609396 DOI: 10.1039/d1fo02283k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The peptide VLATSGPG (VLA) is known to inhibit dipeptidyl peptidase IV (DPP-IV), although its mechanism in relieving endoplasmic reticulum (ER) stress is unclear. In this study, we found that treating HepG2 cells with 1.0 mM VLA reduced DPP-IV activity by 73.7 ± 4.8% without changing the DPP-IV mRNA expression level. In addition, 1.0 and 0.5 mM VLA alleviated palmitic acid (PA)-induced cell death and intracellular calcium imbalances. The levels of apoptosis-related proteins (caspase-3, caspase-9, and CHOP) were reduced by VLA treatment, which was presumed to be related to ER stress. Further studies confirmed that VLA alleviated PA-induced morphological damage to the ER and reduced the levels of the ER stress marker proteins (BIP, p-PERK, and p-IRE1α). VLA alleviated PA-induced ER stress in HepG2 cells independent of DPP-IV enzymatic activity inhibition. These findings have implications for developing novel treatment approaches for liver diseases caused by ER stress.
Collapse
Affiliation(s)
- Ritian Jin
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China. .,Key Lab of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.,Harbin Tengning Technology Co., Ltd, Harbin, 150010, China
| | - Haowei Ren
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China. .,Key Lab of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.,Harbin Tengning Technology Co., Ltd, Harbin, 150010, China
| | - Minhe Liao
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China. .,Key Lab of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.,Harbin Tengning Technology Co., Ltd, Harbin, 150010, China
| | - Jiaqi Shang
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China. .,Key Lab of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.,Harbin Tengning Technology Co., Ltd, Harbin, 150010, China
| | - Dangfeng Wang
- College of Food Science and Technology, Bohai University, Food Safety Key Lab of Liaoning Province, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, Liaoning, 121013, China.,College of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Meng Li
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China. .,Key Lab of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
| | - Ning Liu
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China. .,Key Lab of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.,Harbin Tengning Technology Co., Ltd, Harbin, 150010, China
| |
Collapse
|
17
|
Wu M, Yu Z, Li X, Zhang X, Wang S, Yang S, Hu L, Liu L. Paeonol for the Treatment of Atherosclerotic Cardiovascular Disease: A Pharmacological and Mechanistic Overview. Front Cardiovasc Med 2021; 8:690116. [PMID: 34368250 PMCID: PMC8333700 DOI: 10.3389/fcvm.2021.690116] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022] Open
Abstract
With improvement in living standards and average life expectancy, atherosclerotic cardiovascular disease incidences and mortality have been increasing annually. Paeonia suffruticosa, a natural herb, has been used for the treatment of atherosclerotic cardiovascular disease for thousands of years in Eastern countries. Paeonol is an active ingredient extracted from Paeonia suffruticosa. Previous studies have extensively explored the clinical benefits of paeonol. However, comprehensive reviews on the cardiovascular protective effects of paeonol have not been conducted. The current review summarizes studies reporting on the protective effects of paeonol on the cardiovascular system. This study includes studies published in the last 10 years. The biological characteristics of Paeonia suffruticosa, pharmacological mechanisms of paeonol, and its toxicological and pharmacokinetic characteristics were explored. The findings of this study show that paeonol confers protection against atherosclerotic cardiovascular disease through various mechanisms, including inflammation, platelet aggregation, lipid metabolism, mitochondria damage, endoplasmic reticulum stress, autophagy, and non-coding RNA. Further studies should be conducted to elucidate the cardiovascular benefits of paeonol.
Collapse
Affiliation(s)
- Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zongliang Yu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoya Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaonan Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Songzi Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lanqing Hu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Longtao Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Tian F, Ying HM, Wang YY, Cheng BN, Chen J. MiR-542-5p Inhibits Hyperglycemia and Hyperlipoidemia by Targeting FOXO1 in the Liver. Yonsei Med J 2020; 61:780-788. [PMID: 32882762 PMCID: PMC7471073 DOI: 10.3349/ymj.2020.61.9.780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/18/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE This research was designed to investigate how miR-542-5p regulates the progression of hyperglycemia and hyperlipoidemia. MATERIALS AND METHODS An in vivo model with diabetic db/db mice and an in vitro model with forskolin/dexamethasone (FSK/DEX)-induced primary hepatocytes and HepG2 cells were employed in the study. Bioinformatics analysis was conducted to identify the expression of candidate miRNAs in the liver tissues of diabetic and control mice. H&E staining revealed liver morphology in diabetic and control mice. Pyruvate tolerance tests, insulin tolerance tests, and intraperitoneal glucose tolerance test were utilized to assess insulin resistance. ELISA was conducted to evaluate blood glucose and insulin levels. Red oil O staining showed lipid deposition in liver tissues. Luciferase reporter assay was used to depict binding between miR-542-5p and forkhead box O1 (FOXO1). RESULTS MiR-542-5p expression was under-expressed in the livers of db/db mice. Further in vitro experiments revealed that FSK/DEX, which mimics the effects of glucagon and glucocorticoids, induced cellular glucose production in HepG2 cells and in primary hepatocytes cells. Notably, these changes were reversed by miR-542-5p. We found that transcription factor FOXO1 is a target of miR-542-5p. Further in vivo study indicated that miR-542-5p overexpression decreases FOXO1 expression, thereby reversing increases in blood glucose, blood lipids, and glucose-related enzymes in diabetic db/db mice. In contrast, anti-miR-542-5p exerted an adverse influence on blood glucose and blood lipid metabolism, and its stimulatory effects were significantly inhibited by sh-FOXO1 in normal control mice. CONCLUSION Collectively, our results indicated that miR-542-5p inhibits hyperglycemia and hyperlipoidemia by targeting FOXO1.
Collapse
Affiliation(s)
- Fang Tian
- Department of Endocrinology, Xixi Hospital of Hangzhou Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Min Ying
- Department of Endocrinology, Xixi Hospital of Hangzhou Affiliated to Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yuan Yuan Wang
- Department of Endocrinology, Xixi Hospital of Hangzhou Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Ning Cheng
- Department of Endocrinology, Xixi Hospital of Hangzhou Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Juan Chen
- Department of Endocrinology, Xixi Hospital of Hangzhou Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|